1
|
Ayele K, Wakimoto H, Nauwynck HJ, Kaufman HL, Rabkin SD, Saha D. Understanding the interplay between oHSV and the host immune system: Implications for therapeutic oncolytic virus development. Mol Ther 2024:S1525-0016(24)00854-2. [PMID: 39741405 DOI: 10.1016/j.ymthe.2024.12.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/05/2024] [Accepted: 12/27/2024] [Indexed: 01/03/2025] Open
Abstract
Oncolytic herpes simplex viruses (oHSV) preferentially replicate in cancer cells while inducing antitumor immunity, and thus, they are often referred to as in situ cancer vaccines. OHSV infection of tumors elicits diverse host immune responses comprising both innate and adaptive components. Although the innate and adaptive immune responses primarily target the tumor, they also contribute to antiviral immunity, limiting viral replication/oncolysis. OHSV-encoded proteins use various mechanisms to evade host antiviral pathways and immune recognition, favoring oHSV replication, oncolysis, and spread. In general, oHSV infection and replication within tumors results in a series of sequential events, such as oncolysis and release of tumor and viral antigens, dendritic cell-mediated antigen presentation, T cell priming and activation, T cell trafficking and infiltration to tumors, and T cell recognition of cancer cells, leading to tumor (and viral) clearance. These sequential events align with all steps of the cancer-immunity cycle. However, a comprehensive understanding of the interplay between oHSV and host immune responses is crucial to optimize oHSV-induced antitumor immunity and efficacy. Therefore, this review aims to elucidate oHSV's communication with innate and adaptive immune systems and use such interactions to improve oHSV's potential as a potent immunovirotherapeutic agent against cancer.
Collapse
Affiliation(s)
- Kalkidan Ayele
- Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Elk Grove, CA 95757, USA
| | - Hiroaki Wakimoto
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Hans J Nauwynck
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Howard L Kaufman
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Samuel D Rabkin
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Dipongkor Saha
- Department of Biology, College of Science and Technology, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, USA.
| |
Collapse
|
2
|
Galli JD, Horton M, Durr E, Heidecker GJ, Freed D, Fridman A, Wang D, Zhang L. Evaluation of HSV-2 gE Binding to IgG-Fc and Application for Vaccine Development. Vaccines (Basel) 2022; 10:vaccines10020184. [PMID: 35214644 PMCID: PMC8879737 DOI: 10.3390/vaccines10020184] [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: 12/08/2021] [Revised: 01/08/2022] [Accepted: 01/11/2022] [Indexed: 02/04/2023] Open
Abstract
Glycoprotein E (gE) and glycoprotein I (gI) are expressed as a heterodimer on the surface of Herpes simplex virus (HSV). Glycoprotein E binds Fc domain of immunoglobulin G (IgG) and inhibits activities mediated by the IgG Fc domain, contributing to immune evasion by HSV. It has been reported that HSV type 1 gE (gE-1) is capable of binding IgG Fc as a monomer and in a heterodimeric complex with gI, with the heterodimer having 50- to100-fold greater affinity for Fc than gE alone. We report the production of both a soluble form of HSV type 2 gE (gE-2) and a soluble HSV-2 gE/gI heterodimer (gE-2/gI-2). Characterization of soluble gE-2 by surface plasmon resonance (SPR) demonstrates that it is incapable of binding human IgG or the IgG Fc domain. Co-expression with HSV-2 gI (gI-2) and purification of the gE-2/gI-2 heterodimer enable gE-2 to bind human IgG through its Fc domain. We hypothesize that functional epitopes of wildtype gE-2 may be masked by plasma IgG Fc and affect the immunogenicity of the gE-2/gI-2 heterodimer as a vaccine antigen. A series of gE-2 mutations within the surface-exposed Fc:gE-2 interface was designed, and gE-2 mutants were co-expressed with gI-2. Evaluation of twelve gE-2 mutant heterodimers by SPR assay identified nine gE-2 mutations which abrogated or reduced Fc binding while maintaining heterodimer formation with gI. Vaccinating rabbits with the four most Fc-binding deficient gE-2/gI-2 heterodimers elicited comparable anti-heterodimer binding antibody titers and statistically significantly higher serum neutralization antibody levels than wildtype heterodimers. Taken together, these data support the concept of rational antigen design for improved vaccine candidates.
Collapse
Affiliation(s)
- Jennifer D. Galli
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, PA 19486, USA; (M.H.); (E.D.); (G.J.H.); (D.F.); (D.W.); (L.Z.)
- Correspondence:
| | - Melanie Horton
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, PA 19486, USA; (M.H.); (E.D.); (G.J.H.); (D.F.); (D.W.); (L.Z.)
| | - Eberhard Durr
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, PA 19486, USA; (M.H.); (E.D.); (G.J.H.); (D.F.); (D.W.); (L.Z.)
| | - Gwendolyn J. Heidecker
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, PA 19486, USA; (M.H.); (E.D.); (G.J.H.); (D.F.); (D.W.); (L.Z.)
| | - Daniel Freed
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, PA 19486, USA; (M.H.); (E.D.); (G.J.H.); (D.F.); (D.W.); (L.Z.)
| | - Arthur Fridman
- Data Science and Scientific Informatics, Merck & Co., Inc., Rahway, NJ 07065, USA;
| | - Dai Wang
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, PA 19486, USA; (M.H.); (E.D.); (G.J.H.); (D.F.); (D.W.); (L.Z.)
| | - Lan Zhang
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, PA 19486, USA; (M.H.); (E.D.); (G.J.H.); (D.F.); (D.W.); (L.Z.)
| |
Collapse
|
3
|
Persistent Infection with Herpes Simplex Virus 1 and Alzheimer's Disease-A Call to Study How Variability in Both Virus and Host may Impact Disease. Viruses 2019; 11:v11100966. [PMID: 31635156 PMCID: PMC6833100 DOI: 10.3390/v11100966] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/14/2019] [Accepted: 10/14/2019] [Indexed: 02/06/2023] Open
Abstract
Increasing attention has focused on the contributions of persistent microbial infections with the manifestation of disease later in life, including neurodegenerative conditions such as Alzheimer’s disease (AD). Current data has shown the presence of herpes simplex virus 1 (HSV-1) in regions of the brain that are impacted by AD in elderly individuals. Additionally, neuronal infection with HSV-1 triggers the accumulation of amyloid beta deposits and hyperphosphorylated tau, and results in oxidative stress and synaptic dysfunction. All of these factors are implicated in the development of AD. These data highlight the fact that persistent viral infection is likely a contributing factor, rather than a sole cause of disease. Details of the correlations between HSV-1 infection and AD development are still just beginning to emerge. Future research should investigate the relative impacts of virus strain- and host-specific factors on the induction of neurodegenerative processes over time, using models such as infected neurons in vitro, and animal models in vivo, to begin to understand their relationship with cognitive dysfunction.
Collapse
|
4
|
Jenks JA, Goodwin ML, Permar SR. The Roles of Host and Viral Antibody Fc Receptors in Herpes Simplex Virus (HSV) and Human Cytomegalovirus (HCMV) Infections and Immunity. Front Immunol 2019; 10:2110. [PMID: 31555298 PMCID: PMC6742691 DOI: 10.3389/fimmu.2019.02110] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 08/21/2019] [Indexed: 12/01/2022] Open
Abstract
Herpesvirus infections are a leading cause of neurodevelopmental delay in newborns and end-organ disease in immunocompromised patients. One leading strategy to reduce the disease burden of herpesvirus infections such as herpes simplex virus (HSV) and human cytomegalovirus (HCMV) is to prevent primary acquisition by vaccination, yet vaccine development remains hampered by limited understanding of immune correlates of protection against infection. Traditionally, vaccine development has aimed to increase antibody titers with neutralizing function, which involves the direct binding of antibodies to viral particles. However, recent research has explored the numerous other responses that can be mediated by engagement of the antibody constant region (Fc) with Fc receptors (FcR) present on immune cells or with complement molecules. These functions include antiviral responses such as antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). Uniquely, herpesviruses encode FcR that can act as distractor receptors for host antiviral IgG, thus enabling viral evasion of host defenses. This review focuses on the relative roles of neutralizing and non-neutralizing functions antibodies that target herpesvirus antigens for HSV and HCMV, as well as the roles of Fc-FcR interactions for both host defenses and viral escape.
Collapse
Affiliation(s)
- Jennifer A Jenks
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, United States
| | - Matthew L Goodwin
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, United States
| | - Sallie R Permar
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, United States.,Department of Pediatrics, Children's Health and Discovery Institute, Durham, NC, United States
| |
Collapse
|
5
|
Ma W, He H, Wang H. Oncolytic herpes simplex virus and immunotherapy. BMC Immunol 2018; 19:40. [PMID: 30563466 PMCID: PMC6299639 DOI: 10.1186/s12865-018-0281-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 12/06/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Oncolytic viruses have been proposed to be employed as a potential treatment of cancer. Well targeted, they will serve the purpose of cracking tumor cells without causing damage to normal cells. In this category of oncolytic viral drugs human pathogens herpes simplex virus (HSV) is especially suitable for the cause. Although most viral infection causes antiviral reaction in the host, HSV has multiple mechanisms to evade those responses. Powerful anti-tumor effect can thus be achieved via genetic manipulation of the HSV genes involved in this evading mechanism, namely deletions or mutations that adapt its function towards a tumor microenvironment. Currently, oncolytic HSV (oHSV) is widely use in clinical; moreover, there's hope that its curative effect will be further enhanced through the combination of oHSV with both traditional and emerging therapeutics. RESULTS In this review, we provide a summary of the HSV host antiviral response evasion mechanism, HSV expresses immune evasion genes such as ICP34.5, ICP0, Us3, which are involved in inducing and activating host responses, so that the virus can evade the immune system and establish effective long-term latent infection; we outlined details of the oHSV strains generated by removing genes critical to viral replication such as ICP34.5, ICP0, and inserting therapeutic genes such as LacZ, granulocyte macrophage colony-stimulating factor (GM-CSF); security and limitation of some oHSV such G207, 1716, OncoVEX, NV1020, HF10, G47 in clinical application; and the achievements of oHSV combined with immunotherapy and chemotherapy. CONCLUSION We reviewed the immunotherapy mechanism of the oHSV and provided a series of cases. We also pointed out that an in-depth study of the application of oHSV in cancer treatment will potentially benefits cancer patients more.
Collapse
Affiliation(s)
- Wenqing Ma
- Ruminant Diseases Research Center, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, 250014, China
| | - Hongbin He
- Ruminant Diseases Research Center, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, 250014, China.
| | - Hongmei Wang
- Ruminant Diseases Research Center, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, 250014, China.
| |
Collapse
|
6
|
Boesch AW, Kappel JH, Mahan AE, Chu TH, Crowley AR, Osei-Owusu NY, Alter G, Ackerman ME. Enrichment of high affinity subclasses and glycoforms from serum-derived IgG using FcγRs as affinity ligands. Biotechnol Bioeng 2018; 115:1265-1278. [PMID: 29315477 DOI: 10.1002/bit.26545] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/18/2017] [Accepted: 01/03/2018] [Indexed: 12/26/2022]
Abstract
As antibodies continue to gain predominance in drug discovery and development pipelines, efforts to control and optimize their activity in vivo have matured to incorporate sophisticated abilities to manipulate engagement of specific Fc binding partners. Such efforts to promote diverse functional outcomes include modulating IgG-Fc affinity for FcγRs to alternatively potentiate or reduce effector functions, such as antibody-dependent cellular cytotoxicity and phagocytosis. While a number of natural and engineered Fc features capable of eliciting variable effector functions have been demonstrated in vitro and in vivo, elucidation of these important functional relationships has taken significant effort through use of diverse genetic, cellular and enzymatic techniques. As an orthogonal approach, we demonstrate use of FcγR as chromatographic affinity ligands to enrich and therefore simultaneously identify favored binding species from a complex mixture of serum-derived pooled polycloncal human IgG, a load material that contains the natural repertoire of Fc variants and post-translational modifications. The FcγR-enriched IgG was characterized for subclass and glycoform composition and the impact of this bioseparation step on antibody activity was measured in cell-based effector function assays including Natural Killer cell activation and monocyte phagocytosis. This work demonstrates a tractable means to rapidly distinguish complex functional relationships between two or more interacting biological agents by leveraging affinity chromatography followed by secondary analysis with high-resolution biophysical and functional assays and emphasizes a platform capable of surveying diverse natural post-translational modifications that may not be easily produced with high purity or easily accessible with recombinant expression techniques.
Collapse
Affiliation(s)
- Austin W Boesch
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire
- Zepteon, Inc., Boston, Massachusetts
| | - James H Kappel
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire
| | - Alison E Mahan
- Ragon Institute of MGH, MIT, and Harvard University, Cambridge, Massachusetts
| | - Thach H Chu
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire
| | - Andrew R Crowley
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire
| | - Nana Y Osei-Owusu
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard University, Cambridge, Massachusetts
| | - Margaret E Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire
| |
Collapse
|
7
|
Döhner K, Ramos-Nascimento A, Bialy D, Anderson F, Hickford-Martinez A, Rother F, Koithan T, Rudolph K, Buch A, Prank U, Binz A, Hügel S, Lebbink RJ, Hoeben RC, Hartmann E, Bader M, Bauerfeind R, Sodeik B. Importin α1 is required for nuclear import of herpes simplex virus proteins and capsid assembly in fibroblasts and neurons. PLoS Pathog 2018; 14:e1006823. [PMID: 29304174 PMCID: PMC5773220 DOI: 10.1371/journal.ppat.1006823] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 01/18/2018] [Accepted: 12/16/2017] [Indexed: 01/28/2023] Open
Abstract
Herpesviruses are large DNA viruses which depend on many nuclear functions, and therefore on host transport factors to ensure specific nuclear import of viral and host components. While some import cargoes bind directly to certain transport factors, most recruit importin β1 via importin α. We identified importin α1 in a small targeted siRNA screen to be important for herpes simplex virus (HSV-1) gene expression. Production of infectious virions was delayed in the absence of importin α1, but not in cells lacking importin α3 or importin α4. While nuclear targeting of the incoming capsids, of the HSV-1 transcription activator VP16, and of the viral genomes were not affected, the nuclear import of the HSV-1 proteins ICP4 and ICP0, required for efficient viral transcription, and of ICP8 and pUL42, necessary for DNA replication, were reduced. Furthermore, quantitative electron microscopy showed that fibroblasts lacking importin α1 contained overall fewer nuclear capsids, but an increased proportion of mature nuclear capsids indicating that capsid formation and capsid egress into the cytoplasm were impaired. In neurons, importin α1 was also not required for nuclear targeting of incoming capsids, but for nuclear import of ICP4 and for the formation of nuclear capsid assembly compartments. Our data suggest that importin α1 is specifically required for the nuclear localization of several important HSV1 proteins, capsid assembly, and capsid egress into the cytoplasm, and may become rate limiting in situ upon infection at low multiplicity or in terminally differentiated cells such as neurons. Nuclear pore complexes are highly selective gateways that penetrate the nuclear envelope for bidirectional trafficking between the cytoplasm and the nucleoplasm. Viral and host cargoes have to engage specific transport factors to achieve active nuclear import and export. Like many human and animal DNA viruses, herpesviruses are critically dependent on many functions of the host cell nucleus. Alphaherpesviruses such as herpes simplex virus (HSV) cause many diseases upon productive infection in epithelial cells, fibroblasts and neurons. Here, we asked which nuclear transport factors of the host cells help HSV-1 to translocate viral components into the nucleus for viral gene expression, nuclear capsid assembly, capsid egress into the cytoplasm, and production of infectious virions. Our data show that HSV-1 requires the nuclear import factor importin α1 for efficient replication and virus assembly in fibroblasts and in mature neurons. To our knowledge this is the first time that a specific importin α isoform is shown to be required for herpesvirus infection. Our study fosters our understanding on how the different but highly homologous importin α isoforms could fulfill specific functions in vivo which are only understood for a very limited number of host and viral cargos.
Collapse
Affiliation(s)
- Katinka Döhner
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | | | - Dagmara Bialy
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Fenja Anderson
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | | | - Franziska Rother
- Max-Delbrück Center for Molecular Medicine, Berlin-Buch, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
| | - Thalea Koithan
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Kathrin Rudolph
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Anna Buch
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Ute Prank
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Anne Binz
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Stefanie Hügel
- Max-Delbrück Center for Molecular Medicine, Berlin-Buch, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
| | - Robert Jan Lebbink
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rob C. Hoeben
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Enno Hartmann
- Institute for Biology, University of Lübeck, Lübeck, Germany
| | - Michael Bader
- Max-Delbrück Center for Molecular Medicine, Berlin-Buch, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
| | - Rudolf Bauerfeind
- Research Core Unit Laser Microscopy, Hannover Medical School, Hannover, Germany
| | - Beate Sodeik
- Institute of Virology, Hannover Medical School, Hannover, Germany
- * E-mail:
| |
Collapse
|
8
|
Saha D, Wakimoto H, Rabkin SD. Oncolytic herpes simplex virus interactions with the host immune system. Curr Opin Virol 2016; 21:26-34. [PMID: 27497296 DOI: 10.1016/j.coviro.2016.07.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 07/11/2016] [Accepted: 07/13/2016] [Indexed: 12/28/2022]
Abstract
Oncolytic viruses (OVs), like oncolytic herpes simplex virus (oHSV), are genetically engineered to selectively replicate in and kill cancer cells, while sparing normal cells. Initial OV infection, cell death, and subsequent OV propagation within the tumor microenvironment leads to a cascade of host responses (innate and adaptive), reflective of natural anti-viral immune responses. These host-virus interactions are critical to the balance between OV activities, anti-viral immune responses limiting OV, and induction of anti-tumor immunity. The host response against oHSV is complex, multifaceted, and modulated by the tumor microenvironment and immunosuppression. As a successful pathogen, HSV has multiple mechanisms to evade such host responses. In this review, we will discuss these mechanisms and HSV evasion, and how they impact oHSV therapy.
Collapse
Affiliation(s)
- Dipongkor Saha
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Hiroaki Wakimoto
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Samuel D Rabkin
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
9
|
Characterization of Antibody Bipolar Bridging Mediated by the Human Cytomegalovirus Fc Receptor gp68. J Virol 2016; 90:3262-7. [PMID: 26739053 DOI: 10.1128/jvi.02855-15] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 12/30/2015] [Indexed: 01/27/2023] Open
Abstract
The human cytomegalovirus glycoprotein gp68 functions as an Fc receptor for host IgGs and can form antibody bipolar bridging (ABB) complexes in which gp68 binds the Fc region of an antigen-bound IgG. Here we show that gp68-mediated endocytosis transports ABB complexes into endosomes, after which the complex is routed to lysosomes, presumably for degradation. These results suggest gp68 contributes to evasion of IgG-mediated immune responses by mediating destruction of host IgG and viral antigens.
Collapse
|
10
|
Krawczyk A, Dirks M, Kasper M, Buch A, Dittmer U, Giebel B, Wildschütz L, Busch M, Goergens A, Schneweis KE, Eis-Hübinger AM, Sodeik B, Heiligenhaus A, Roggendorf M, Bauer D. Prevention of herpes simplex virus induced stromal keratitis by a glycoprotein B-specific monoclonal antibody. PLoS One 2015; 10:e0116800. [PMID: 25587898 PMCID: PMC4294644 DOI: 10.1371/journal.pone.0116800] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 12/15/2014] [Indexed: 10/24/2022] Open
Abstract
The increasing incidence of acyclovir (ACV) and multidrug-resistant strains in patients with corneal HSV-1 infections leading to Herpetic Stromal Keratitis (HSK) is a major health problem in industrialized countries and often results in blindness. To overcome this obstacle, we have previously developed an HSV-gB-specific monoclonal antibody (mAb 2c) that proved to be highly protective in immunodeficient NOD/SCID-mice towards genital infections. In the present study, we examined the effectivity of mAb 2c in preventing the immunopathological disease HSK in the HSK BALB/c mouse model. Therefore, mice were inoculated with HSV-1 strain KOS on the scarified cornea to induce HSK and subsequently either systemically or topically treated with mAb 2c. Systemic treatment was performed by intravenous administration of mAb 2c 24 h prior to infection (pre-exposure prophylaxis) or 24, 40, and 56 hours after infection (post-exposure immunotherapy). Topical treatment was performed by periodical inoculations (5 times per day) of antibody-containing eye drops as control, starting at 24 h post infection. Systemic antibody treatment markedly reduced viral loads at the site of infection and completely protected mice from developing HSK. The administration of the antiviral antibody prior or post infection was equally effective. Topical treatment had no improving effect on the severity of HSK. In conclusion, our data demonstrate that mAb 2c proved to be an excellent drug for the treatment of corneal HSV-infections and for prevention of HSK and blindness. Moreover, the humanized counterpart (mAb hu2c) was equally effective in protecting mice from HSV-induced HSK when compared to the parental mouse antibody. These results warrant the future development of this antibody as a novel approach for the treatment of corneal HSV-infections in humans.
Collapse
Affiliation(s)
- Adalbert Krawczyk
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- * E-mail:
| | - Miriam Dirks
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Maren Kasper
- Ophtha-Lab, Department of Ophthalmology at St. Franziskus Hospital, Muenster, Germany
| | - Anna Buch
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Ulf Dittmer
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| | - Lena Wildschütz
- Ophtha-Lab, Department of Ophthalmology at St. Franziskus Hospital, Muenster, Germany
| | - Martin Busch
- Ophtha-Lab, Department of Ophthalmology at St. Franziskus Hospital, Muenster, Germany
| | - Andre Goergens
- Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| | - Karl E. Schneweis
- Institute of Virology, University Medical Center Bonn, Bonn, Germany
| | | | - Beate Sodeik
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Arnd Heiligenhaus
- Ophtha-Lab, Department of Ophthalmology at St. Franziskus Hospital, Muenster, Germany
| | - Michael Roggendorf
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Dirk Bauer
- Ophtha-Lab, Department of Ophthalmology at St. Franziskus Hospital, Muenster, Germany
| |
Collapse
|
11
|
Herpes simplex virus internalization into epithelial cells requires Na+/H+ exchangers and p21-activated kinases but neither clathrin- nor caveolin-mediated endocytosis. J Virol 2014; 88:13378-95. [PMID: 25210183 DOI: 10.1128/jvi.03631-13] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
UNLABELLED Herpes simplex virus 1 (HSV-1) is an alphaherpesvirus that has been reported to infect some epithelial cell types by fusion at the plasma membrane but others by endocytosis. To determine the molecular mechanisms of productive HSV-1 cell entry, we perturbed key endocytosis host factors using specific inhibitors, RNA interference (RNAi), or overexpression of dominant negative proteins and investigated their effects on HSV-1 infection in the permissive epithelial cell lines Vero, HeLa, HEp-2, and PtK2. HSV-1 internalization required neither endosomal acidification nor clathrin- or caveolin-mediated endocytosis. In contrast, HSV-1 gene expression and internalization were significantly reduced after treatment with 5-(N-ethyl-N-isopropyl)amiloride (EIPA). EIPA blocks the activity of Na(+)/H(+) exchangers, which are plasma membrane proteins implicated in all forms of macropinocytosis. HSV-1 internalization furthermore required the function of p21-activated kinases that contribute to macropinosome formation. However, in contrast to some forms of macropinocytosis, HSV-1 did not enlist the activities of protein kinase C (PKC), tyrosine kinases, C-terminal binding protein 1, or dynamin to activate its internalization. These data suggest that HSV-1 depends on Na(+)/H(+) exchangers and p21-activated kinases either for macropinocytosis or for local actin rearrangements required for fusion at the plasma membrane or subsequent passage through the actin cortex underneath the plasma membrane. IMPORTANCE After initial replication in epithelial cells, herpes simplex viruses (HSVs) establish latent infections in neurons innervating these regions. Upon primary infection and reactivation from latency, HSVs cause many human skin and neurological diseases, particularly in immunocompromised hosts, despite the availability of effective antiviral drugs. Many viruses use macropinocytosis for virus internalization, and many host factors mediating this entry route have been identified, although the specific perturbation profiles vary for different host and viral cargo. In addition to an established entry pathway via acidic endosomes, we show here that HSV-1 internalization depended on sodium-proton exchangers at the plasma membrane and p21-activated kinases. These results suggest that HSV-1 requires a reorganization of the cortical actin cytoskeleton, either for productive cell entry via pH-independent fusion from macropinosomes or for fusion at the plasma membrane, and subsequent cytosolic passage to microtubules that mediate capsid transport to the nucleus for genome uncoating and replication.
Collapse
|
12
|
Cortese M, Calò S, D'Aurizio R, Lilja A, Pacchiani N, Merola M. Recombinant human cytomegalovirus (HCMV) RL13 binds human immunoglobulin G Fc. PLoS One 2012; 7:e50166. [PMID: 23226246 PMCID: PMC3511460 DOI: 10.1371/journal.pone.0050166] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 10/22/2012] [Indexed: 01/05/2023] Open
Abstract
The human cytomegalovirus (HCMV) protein RL13 has recently been described to be present in all primary isolates but rapidly mutated in culture adapted viruses. Although these data suggest a crucial role for this gene product in HCMV primary infection, no function has so far been assigned to this protein. Working with RL13 expressed in isolation in transfected human epithelial cells, we demonstrated that recombinant RL13 from the clinical HCMV isolates TR and Merlin have selective human immunoglobulin (Ig)-binding properties towards IgG1 and IgG2 subtypes. An additional Fc binding protein, RL12, was also identified as an IgG1 and IgG2 binding protein but not further characterized. The glycoprotein RL13 trafficked to the plasma membrane where it bound and internalized exogenous IgG or its constant fragment (Fcγ). Analysis of RL13 ectodomain mutants suggested that the RL13 Ig-like domain is responsible for the Fc binding activity. Ligand-dependent internalization relied on a YxxL endocytic motif located in the C-terminal tail of RL13. Additionally, we showed that the tyrosine residue could be replaced by phenylalanine but not by alanine, indicating that the internalization signal was independent from phosphorylation events. In sum, RL13 binds human IgG and may contribute to HCMV immune evasion in the infected host, but this function does not readily explain the instability of the RL13 gene during viral propagation in cultured cells.
Collapse
Affiliation(s)
| | | | | | - Anders Lilja
- Novartis Vaccines and Diagnostics, Cambridge, Massachusetts, United States of America
| | | | - Marcello Merola
- Novartis Vaccines and Diagnostics, Siena, Italy
- Department of Structural and Functional Biology, University of Naples “Federico II”, Naples, Italy
- * E-mail:
| |
Collapse
|
13
|
The C terminus of the large tegument protein pUL36 contains multiple capsid binding sites that function differently during assembly and cell entry of herpes simplex virus. J Virol 2012; 86:3682-700. [PMID: 22258258 DOI: 10.1128/jvi.06432-11] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The largest tegument protein of herpes simplex virus type 1 (HSV1), pUL36, is a multivalent cross-linker between the viral capsids and the tegument and associated membrane proteins during assembly that upon subsequent cell entry releases the incoming capsids from the outer tegument and viral envelope. Here we show that pUL36 was recruited to cytosolic progeny capsids that later colocalized with membrane proteins of herpes simplex virus type 1 (HSV1) and the trans-Golgi network. During cell entry, pUL36 dissociated from viral membrane proteins but remained associated with cytosolic capsids until arrival at the nucleus. HSV1 UL36 mutants lacking C-terminal portions of increasing size expressed truncated pUL36 but could not form plaques. Cytosolic capsids of mutants lacking the C-terminal 735 of the 3,164 amino acid residues accumulated in the cytosol but did not recruit pUL36 or associate with membranes. In contrast, pUL36 lacking only the 167 C-terminal residues bound to cytosolic capsids and subsequently colocalized with viral and host membrane proteins. Progeny virions fused with neighboring cells, but incoming capsids did not retain pUL36, nor could they target the nucleus or initiate HSV1 gene expression. Our data suggest that residues 2430 to 2893 of HSV1 pUL36, containing one binding site for the capsid protein pUL25, are sufficient to recruit pUL36 onto cytosolic capsids during assembly for secondary envelopment, whereas the 167 residues of the very C terminus with the second pUL25 binding site are crucial to maintain pUL36 on incoming capsids during cell entry. Capsids lacking pUL36 are targeted neither to membranes for virus assembly nor to nuclear pores for genome uncoating.
Collapse
|
14
|
The herpes simplex virus 1 IgG fc receptor blocks antibody-mediated complement activation and antibody-dependent cellular cytotoxicity in vivo. J Virol 2011; 85:3239-49. [PMID: 21228231 DOI: 10.1128/jvi.02509-10] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Herpes simplex virus 1 (HSV-1) glycoprotein E (gE) mediates cell-to-cell spread and functions as an IgG Fc receptor (FcγR) that blocks the Fc domain of antibody targeting the virus or infected cell. Efforts to assess the functions of the HSV-1 FcγR in vivo have been hampered by difficulties in preparing an FcγR-negative strain that is relatively intact for spread. Here we report the FcγR and spread phenotypes of NS-gE264, which is a mutant strain that has four amino acids inserted after gE residue 264. The virus is defective in IgG Fc binding yet causes zosteriform disease in the mouse flank model that is only minimally reduced compared with wild-type and the rescue strains. The presence of zosteriform disease suggests that NS-gE264 spread functions are well maintained. The HSV-1 FcγR binds the Fc domain of human, but not murine IgG; therefore, to assess FcγR functions in vivo, mice were passively immunized with human IgG antibody to HSV. When antibody was inoculated intraperitoneally 20 h prior to infection or shortly after virus reached the dorsal root ganglia, disease severity was significantly reduced in mice infected with NS-gE264, but not in mice infected with wild-type or rescue virus. Studies of C3 knockout mice and natural killer cell-depleted mice demonstrated that the HSV-1 FcγR blocked both IgG Fc-mediated complement activation and antibody-dependent cellular cytotoxicity. Therefore, the HSV-1 FcγR promotes immune evasion from IgG Fc-mediated activities and likely contributes to virulence at times when antibody is present, such as during recurrent infections.
Collapse
|
15
|
Nuclear egress and envelopment of herpes simplex virus capsids analyzed with dual-color fluorescence HSV1(17+). J Virol 2007; 82:3109-24. [PMID: 18160444 DOI: 10.1128/jvi.02124-07] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
To analyze the assembly of herpes simplex virus type 1 (HSV1) by triple-label fluorescence microscopy, we generated a bacterial artificial chromosome (BAC) and inserted eukaryotic Cre recombinase, as well as beta-galactosidase expression cassettes. When the BAC pHSV1(17(+))blueLox was transfected back into eukaryotic cells, the Cre recombinase excised the BAC sequences, which had been flanked with loxP sites, from the viral genome, leading to HSV1(17(+))blueLox. We then tagged the capsid protein VP26 and the envelope protein glycoprotein D (gD) with fluorescent protein domains to obtain HSV1(17(+))blueLox-GFPVP26-gDRFP and -RFPVP26-gDGFP. All HSV1 BACs had variations in the a-sequences and lost the oriL but were fully infectious. The tagged proteins behaved as their corresponding wild type, and were incorporated into virions. Fluorescent gD first accumulated in cytoplasmic membranes but was later also detected in the endoplasmic reticulum and the plasma membrane. Initially, cytoplasmic capsids did not colocalize with viral glycoproteins, indicating that they were naked, cytosolic capsids. As the infection progressed, they were enveloped and colocalized with the viral membrane proteins. We then analyzed the subcellular distribution of capsids, envelope proteins, and nuclear pores during a synchronous infection. Although the nuclear pore network had changed in ca. 20% of the cells, an HSV1-induced reorganization of the nuclear pore architecture was not required for efficient nuclear egress of capsids. Our data are consistent with an HSV1 assembly model involving primary envelopment of nuclear capsids at the inner nuclear membrane and primary fusion to transfer capsids into the cytosol, followed by their secondary envelopment on cytoplasmic membranes.
Collapse
|
16
|
Norberg P, Olofsson S, Tarp MA, Clausen H, Bergström T, Liljeqvist JÅ. Glycoprotein I of herpes simplex virus type 1 contains a unique polymorphic tandem-repeated mucin region. J Gen Virol 2007; 88:1683-1688. [PMID: 17485527 DOI: 10.1099/vir.0.82500-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Glycoprotein I (gI) of herpes simplex virus type 1 (HSV-1) contains a tandem repeat (TR) region including the amino acids serine and threonine, residues that can be utilized for O-glycosylation. The length of this TR region was determined for 82 clinical HSV-1 isolates and the results revealed a polymorphic distribution of two to six or eight repeated blocks with a majority harbouring between two and four repeats. Assessment of the O-glycosylation capacity of an acceptor peptide (STPSTTTSTPSTTT), representing two of the gI blocks, showed that the peptide was a universal substrate for O-glycosylation not only for the two most commonly expressed N-acetyl-d-galactosamine (GalNAc)-T1 and -T2 transferases, but also for the GalNAc-T3, -T4 and -T11 transferases. Immunoblotting of virus-infected cells showed that gI was exclusively O-glycosylated with GalNAc monosaccharides (Tn antigen). A polymorphic mucin region has not been described previously for HSV-1 and is a unique finding, as repeated blocks within gI homologues are lacking in other alphaherpesviruses.
Collapse
Affiliation(s)
- Peter Norberg
- Department of Virology, University of Göteborg, Guldhedsgatan 10B, S-413 46 Göteborg, Sweden
| | - Sigvard Olofsson
- Department of Virology, University of Göteborg, Guldhedsgatan 10B, S-413 46 Göteborg, Sweden
| | - Mads Agervig Tarp
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Henrik Clausen
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Tomas Bergström
- Department of Virology, University of Göteborg, Guldhedsgatan 10B, S-413 46 Göteborg, Sweden
| | - Jan Åke Liljeqvist
- Department of Virology, University of Göteborg, Guldhedsgatan 10B, S-413 46 Göteborg, Sweden
| |
Collapse
|
17
|
Sprague ER, Wang C, Baker D, Bjorkman PJ. Crystal structure of the HSV-1 Fc receptor bound to Fc reveals a mechanism for antibody bipolar bridging. PLoS Biol 2006; 4:e148. [PMID: 16646632 PMCID: PMC1450327 DOI: 10.1371/journal.pbio.0040148] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2006] [Accepted: 03/07/2006] [Indexed: 12/13/2022] Open
Abstract
Herpes simplex virus type-1 expresses a heterodimeric Fc receptor, gE-gI, on the surfaces of virions and infected cells that binds the Fc region of host immunoglobulin G and is implicated in the cell-to-cell spread of virus. gE-gI binds immunoglobulin G at the basic pH of the cell surface and releases it at the acidic pH of lysosomes, consistent with a role in facilitating the degradation of antiviral antibodies. Here we identify the C-terminal domain of the gE ectodomain (CgE) as the minimal Fc-binding domain and present a 1.78-angstroms CgE structure. A 5-angstroms gE-gI/Fc crystal structure, which was independently verified by a theoretical prediction method, reveals that CgE binds Fc at the C(H)2-C(H)3 interface, the binding site for several mammalian and bacterial Fc-binding proteins. The structure identifies interface histidines that may confer pH-dependent binding and regions of CgE implicated in cell-to-cell spread of virus. The ternary organization of the gE-gI/Fc complex is compatible with antibody bipolar bridging, which can interfere with the antiviral immune response.
Collapse
Affiliation(s)
- Elizabeth R Sprague
- 1Division of Biology, California Institute of Technology, Pasadena, California, United States of America
| | - Chu Wang
- 2Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
| | - David Baker
- 2Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
- 3Howard Hughes Medical Institute, University of Washington, Seattle, Washington, United States of America
| | - Pamela J Bjorkman
- 1Division of Biology, California Institute of Technology, Pasadena, California, United States of America
- 4Howard Hughes Medical Institute, California Institute of Technology, Pasadena, California United States of America
| |
Collapse
|
18
|
Hook LM, Lubinski JM, Jiang M, Pangburn MK, Friedman HM. Herpes simplex virus type 1 and 2 glycoprotein C prevents complement-mediated neutralization induced by natural immunoglobulin M antibody. J Virol 2006; 80:4038-46. [PMID: 16571820 PMCID: PMC1440426 DOI: 10.1128/jvi.80.8.4038-4046.2006] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Glycoprotein C (gC) of herpes simplex virus type 1 (HSV-1) and type 2 (HSV-2) binds complement component C3b and protects virus from complement-mediated neutralization. Differences in complement interacting domains exist between gC of HSV-1 (gC1) and HSV-2 (gC2), since the amino terminus of gC1 blocks complement C5 from binding to C3b, while gC2 fails to interfere with this activity. We previously reported that neutralization of HSV-1 gC-null virus by HSV antibody-negative human serum requires activation of C5 but not of downstream components of the classical complement pathway. In this report, we evaluated whether activation of C5 is sufficient to neutralize HSV-2 gC-null virus, or whether formation of the membrane attack complex by C6 to C9 is required for neutralization. We found that activation of the classical complement pathway up to C5 was sufficient to neutralize HSV-2 gC-null virus by HSV antibody-negative human serum. We evaluated the mechanisms by which complement activation occurred in seronegative human serum. Interestingly, natural immunoglobulin M antibodies bound to virus, which triggered activation of C1q and the classical complement pathway. HSV antibody-negative sera obtained from four individuals differed over an approximately 10-fold range in their potency for complement-mediated virus neutralization. These findings indicate that humans differ in the ability of their innate immune systems to neutralize HSV-1 or HSV-2 gC-null virus and that a critical function of gC1 and gC2 is to prevent C5 activation.
Collapse
Affiliation(s)
- Lauren M Hook
- Infectious Disease Division, Department of Medicine, 502 Johnson Pavilion, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-6073, USA
| | | | | | | | | |
Collapse
|
19
|
Wang F, Tang W, McGraw HM, Bennett J, Enquist LW, Friedman HM. Herpes simplex virus type 1 glycoprotein e is required for axonal localization of capsid, tegument, and membrane glycoproteins. J Virol 2005; 79:13362-72. [PMID: 16227258 PMCID: PMC1262596 DOI: 10.1128/jvi.79.21.13362-13372.2005] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Herpes simplex virus type 1 (HSV-1) glycoprotein E (gE) promotes cell-to-cell spread at basolateral surfaces of epithelial cells, but its activity in neurons is less clear. We used the mouse retina infection model and neuronal cell cultures to define the spread phenotype of gE mutant viruses. Wild-type (WT) and gE-null (NS-gEnull) viruses both infected retina ganglion cell neurons; however, NS-gEnull viral antigens failed to reach the optic nerve, which indicates a defect in axonal localization. We evaluated two Fc receptor-negative gE mutant viruses containing four amino acid inserts in the gE ectodomain. One mutant virus failed to spread from the retina into the optic nerve, while the other spread normally. Therefore, the gE ectodomain is involved in axonal localization, and the Fc receptor and neuronal spread are mediated by overlapping but distinct gE domains. In the retina infection model, virus can travel to the brain via the optic nerve from presynaptic to postsynaptic neurons (anterograde direction) or via nerves that innervate the iris and ciliary body from postsynaptic to presynaptic neurons (retrograde direction). WT virus infected the brain by anterograde and retrograde routes, whereas NS-gEnull virus failed to travel by either pathway. The site of the defect in retrograde spread remains to be determined; however, infection of rat superior cervical ganglia neurons in vitro indicates that gE is required to target virion components to the axon initial segment. The requirement for gE in axonal targeting and retrograde spread highlights intriguing similarities and differences between HSV-1 and pseudorabies virus gE.
Collapse
Affiliation(s)
- Fushan Wang
- Department of Medicine, Division of Infectious Diseases, 502 Johnson Pavilion, University of Pennsylvania, Philadelphia, PA 19104-6073, USA
| | | | | | | | | | | |
Collapse
|
20
|
Lin X, Lubinski JM, Friedman HM. Immunization strategies to block the herpes simplex virus type 1 immunoglobulin G Fc receptor. J Virol 2004; 78:2562-71. [PMID: 14963159 PMCID: PMC369259 DOI: 10.1128/jvi.78.5.2562-2571.2004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) glycoprotein gE functions as an immunoglobulin G (IgG) Fc receptor (FcgammaR) that promotes immune evasion. When an IgG antibody binds by the F(ab')(2) domain to an HSV antigen, the Fc domain of some of the same antibody molecules binds to the FcgammaR, which blocks Fc-mediated functions. gE is a type 1 membrane glycoprotein with a large ectodomain that is expressed on the virion envelope and infected-cell surface. Our goal was to determine if immunizing with gE protein fragments could produce antibodies that bind by the F(ab')(2) domain to gE and block the FcgammaR, as measured by competitively inhibiting nonimmune human IgG binding to the FcgammaR. Three gE peptides were constructed in baculovirus spanning almost the entire ectodomain and used to immunize mice and rabbits. Two fragments were highly effective at producing antibodies that bind by the F(ab')(2) domain and block the FcgammaR. The most potent of these two antibodies was far more effective at blocking the FcgammaR than antibodies that are only capable of binding by the Fc domains to the FcgammaR, including anti-gC, anti-gD, and nonimmune IgG. These results suggest that immunizing with gE fragments has potential for preventing immune evasion by blocking activities mediated by the HSV-1 FcgammaR.
Collapse
Affiliation(s)
- Xiaoqing Lin
- Department of Medicine, Division of Infectious Diseases, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | | | |
Collapse
|
21
|
Sprague ER, Martin WL, Bjorkman PJ. pH dependence and stoichiometry of binding to the Fc region of IgG by the herpes simplex virus Fc receptor gE-gI. J Biol Chem 2004; 279:14184-93. [PMID: 14734541 DOI: 10.1074/jbc.m313281200] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Herpes simplex virus type 1 encodes two glycoproteins, gE and gI, that form a heterodimer on the surface of virions and infected cells. The gE-gI heterodimer has been implicated in cell-to-cell spread of virus and is a receptor for the Fc fragment of IgG. Previous studies localized the gE-gI-binding site on human IgG to a region near the interface between the C(H)2 and C(H)3 domains of Fc, which also serves as the binding site for bacterial and mammalian Fc receptors. Although there are two potential gE-gI-binding sites per Fc homodimer, only one gE-gI heterodimer binds per IgG in gel filtration experiments. Here we report production of recombinant human Fc molecules that contain zero, one, or two potential gE-gI-binding sites and use them in analytical ultracentrifugation experiments to show that two gE-gI heterodimers can bind to each Fc. Further characterization of the gE-gI interaction with Fc reveals a sharp pH dependence of binding, with K(D) values of approximately 340 and approximately 930 nm for the first and second binding events, respectively, at the slightly basic pH of the cell surface (pH 7.4), but undetectable binding at pH 6.0. This strongly pH-dependent interaction suggests a physiological role for gE-gI dissociation from IgG within acidic intracellular compartments, consistent with a mechanism whereby herpes simplex virus promotes intracellular degradation of anti-viral antibodies.
Collapse
Affiliation(s)
- Elizabeth R Sprague
- Division of Biology, Howard Hughes Medical Institute, California Institute of Technology, Pasadena, California 91125, USA
| | | | | |
Collapse
|
22
|
Favoreel HW, Van de Walle GR, Nauwynck HJ, Pensaert MB. Virus complement evasion strategies. J Gen Virol 2003; 84:1-15. [PMID: 12533696 DOI: 10.1099/vir.0.18709-0] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The immune system has a variety of tools at its disposal to combat virus infections. These can be subdivided roughly into two categories: 'first line defence', consisting of the non-specific, innate immune system, and 'adaptive immune response', acquired over time following virus infection or vaccination. During evolution, viruses have developed numerous, and often very ingenious, strategies to counteract efficient recognition of virions or virus-infected cells by both innate and adaptive immunity. This review will focus on the different strategies that viruses use to avoid recognition by one of the components of the immune system: the complement system. Complement evasion is of particular importance for viruses, since complement activation is a crucial component of innate immunity (alternative and mannan-binding lectin activation pathway) as well as of adaptive immunity (classical, antibody-dependent complement activation).
Collapse
Affiliation(s)
- Herman W Favoreel
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - Gerlinde R Van de Walle
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - Hans J Nauwynck
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - Maurice B Pensaert
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| |
Collapse
|
23
|
Rizvi SM, Raghavan M. An N-terminal domain of herpes simplex virus type Ig E is capable of forming stable complexes with gI. J Virol 2001; 75:11897-901. [PMID: 11689673 PMCID: PMC114778 DOI: 10.1128/jvi.75.23.11897-11901.2001] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Using limited proteolytic analyses, we show that gE present in soluble herpes simplex virus type 1 gE-gI complexes is cleaved into a C-terminal (CgE) and an N-terminal (NgE) domain. The domain boundary is in the vicinity of residue 188 of mature gE. NgE, but not CgE, forms a stable complex with soluble gI.
Collapse
Affiliation(s)
- S M Rizvi
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan 48109-0620, USA
| | | |
Collapse
|
24
|
Antonsson A, Johansson PJH. Binding of human and animal immunoglobulins to the IgG Fc receptor induced by human cytomegalovirus. J Gen Virol 2001; 82:1137-1145. [PMID: 11297688 DOI: 10.1099/0022-1317-82-5-1137] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Human cytomegalovirus (HCMV)-infected cells express a virus-encoded receptor that is able to bind the Fc part of IGG: Some basic binding properties of this Fc receptor (FcR) have been examined. The affinity constant (K(a)) for human IgG Fc fragment in its interaction with acetone-fixed, HCMV-infected human embryonic lung fibroblasts was estimated to be around 2 x 10(8) M(-1) and the number of binding sites was estimated to be around 2 x 10(6) per cell. Of the human IgG, IgA, IgM and IgD classes, only IgG reacted with the receptor, and all four of the IgG subclasses were reactive. IgG from rabbit, hamster, cat, swine and horse exhibited binding to the HCMV FcR, in contrast to IgG from mouse, rat, guinea pig, dog, sheep, goat, cow and chicken. Immunoglobulins with and without HCMV IgG FcR-binding properties, like IgG from rabbit and mouse, can be of value in revealing the functional importance of the receptor. When the immunoglobulins were tested against herpes simplex virus type 1-induced FcR, both similarities and differences in immunoreactivity were seen relative to the HCMV FcR, which makes it unlikely that the binding sites for these two herpesvirus FcRs on the IgG molecule are identical.
Collapse
Affiliation(s)
- Annika Antonsson
- Section of Virology, Department of Infectious Diseases and Medical Microbiology, University of Lund, Sölvegatan 23, S-221 85 Lund, Sweden1
| | - P J Hugo Johansson
- Section of Virology, Department of Infectious Diseases and Medical Microbiology, University of Lund, Sölvegatan 23, S-221 85 Lund, Sweden1
| |
Collapse
|
25
|
Bryant HE, Matthews DA, Wadd S, Scott JE, Kean J, Graham S, Russell WC, Clements JB. Interaction between herpes simplex virus type 1 IE63 protein and cellular protein p32. J Virol 2000; 74:11322-8. [PMID: 11070032 PMCID: PMC113237 DOI: 10.1128/jvi.74.23.11322-11328.2000] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The herpes simplex virus type 1 (HSV-1) immediate-early gene IE63 (ICP27), the only HSV-1 regulatory gene with a homologue in every mammalian and avian herpesvirus sequenced so far, is a multifunctional protein which regulates transcriptional and posttranscriptional processes. One of its posttranscriptional effects is the inhibition of splicing of viral and cellular transcripts. We previously identified heterogeneous nuclear ribonucleoprotein (hnRNP) K and casein kinase 2 (CK2) as two protein partners of IE63 (H. Bryant et al., J. Biol. Chem. 274:28991-28998, 1999). Here, using a yeast two-hybrid assay, we identify another partner of IE63, the cellular protein p32. Confirmation of this interaction was provided by coimmunoprecipitation from virus-infected cells and recombinant p32 binding assays. A p32-hnRNP K-CK2 complex, which required IE63 to form, was isolated from HSV-1-infected cells, and coimmunoprecipitating p32 was phosphorylated by CK2. Expression of IE63 altered the cytoplasmic distribution of p32, with some now colocalizing with IE63 in the nuclei of infected and transfected cells. As p32 copurifies with splicing factors and can inhibit splicing, we propose that IE63 together with p32, possibly with other IE63 partner proteins, acts to disrupt or regulate pre-mRNA splicing. As well as contributing to host cell shutoff, this effect could facilitate splicing-independent nuclear export of viral transcripts.
Collapse
Affiliation(s)
- H E Bryant
- Division of Virology, Institute of Biomedical and Life Sciences, University of Glasgow, Glasgow G11 5JR, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Saldanha CE, Lubinski J, Martin C, Nagashunmugam T, Wang L, van Der Keyl H, Tal-Singer R, Friedman HM. Herpes simplex virus type 1 glycoprotein E domains involved in virus spread and disease. J Virol 2000; 74:6712-9. [PMID: 10888608 PMCID: PMC112186 DOI: 10.1128/jvi.74.15.6712-6719.2000] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) glycoprotein E (gE) functions as an immunoglobulin G (IgG) Fc binding protein and is involved in virus spread. Previously we studied a gE mutant virus that was impaired for IgG Fc binding but intact for spread and another that was normal for both activities. To further evaluate the role of gE in spread, two additional mutant viruses were constructed by introducing linker insertion mutations either outside the IgG Fc binding domain at gE position 210 or within the IgG Fc binding domain at position 380. Both mutant viruses were impaired for spread in epidermal cells in vitro; however, the 380 mutant virus was significantly more impaired and was as defective as gE null virus. gE mutant viruses were inoculated into the murine flank to measure epidermal disease at the inoculation site, travel of virus to dorsal root ganglia, and spread of virus from ganglia back to skin to produce zosteriform lesions. Disease at the inoculation and zosteriform sites was reduced for both mutant viruses, but more so for the 380 mutant virus. Moreover, the 380 mutant virus was highly impaired in its ability to reach the ganglia, as demonstrated by virus culture and real-time quantitative PCR. The results indicate that the domain surrounding amino acid 380 is important for both spread and IgG Fc binding and suggest that this domain is a potential target for antiviral therapy or vaccines.
Collapse
MESH Headings
- Animals
- Blotting, Southern
- Blotting, Western
- Cell Line
- Chlorocebus aethiops
- DNA, Viral/analysis
- Female
- Ganglia, Spinal/virology
- Herpes Simplex/pathology
- Herpes Simplex/virology
- Herpesvirus 1, Human/genetics
- Herpesvirus 1, Human/pathogenicity
- Herpesvirus 1, Human/physiology
- Humans
- Immunoglobulin Fc Fragments/genetics
- Immunoglobulin Fc Fragments/metabolism
- Immunoglobulin G/genetics
- Immunoglobulin G/metabolism
- Mice
- Mice, Inbred BALB C
- Mutagenesis, Insertional
- Protein Structure, Tertiary
- RNA, Viral/analysis
- Rosette Formation
- Vero Cells
- Viral Envelope Proteins/chemistry
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/metabolism
Collapse
Affiliation(s)
- C E Saldanha
- Division of Infectious Diseases, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-6073, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Tirabassi RS, Enquist LW. Role of the pseudorabies virus gI cytoplasmic domain in neuroinvasion, virulence, and posttranslational N-linked glycosylation. J Virol 2000; 74:3505-16. [PMID: 10729124 PMCID: PMC111858 DOI: 10.1128/jvi.74.8.3505-3516.2000] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The glycoproteins I and E of pseudorabies virus are important mediators of cell-to-cell spread and virulence in all animal models tested. Although these two proteins form a complex with one another, ascribing any function to the individual proteins has been difficult. We have shown previously, using nonsense mutations, that the N-terminal ectodomain of the gE protein is sufficient for gE-mediated transsynaptic spread whereas the cytoplasmic domain of the protein is required for full expression of virulence. These same studies demonstrated that the cytoplasmic domain of gE is also required for endocytosis of the protein. In this report, we describe the construction of viruses with nonsense mutations in gI that allowed us to determine the contributions of the gI cytoplasmic domain to protein expression as well as virus neuroinvasion and virulence after infection of the rat eye. We also constructed double mutants with nonsense mutations in both gE and gI so that the contributions of both the gE and gI cytoplasmic domains could be determined. We observed that the gI cytoplasmic domain is required for efficient posttranslational modification of the gI protein. The gE cytoplasmic domain has no effect on gE posttranslational glycosylation. In addition, we found that infection of all gE-gI-dependent anterograde circuits projecting from the rat retina requires both ectodomains and at least one of the cytoplasmic domains of the proteins. The gI cytoplasmic domain promotes transsynaptic spread of virus better than the gE cytoplasmic domain. Interestingly, both gE and gI cytoplasmic tails are required for virulence; lack of either one or both results in an attenuated infection. These data suggest that gE and gI play differential roles in mediating directional neuroinvasion of the rat; however, the gE and gI cytoplasmic domains most likely function together to promote virulence.
Collapse
Affiliation(s)
- R S Tirabassi
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | | |
Collapse
|
28
|
Jacobs A, Breakefield XO, Fraefel C. HSV-1-based vectors for gene therapy of neurological diseases and brain tumors: part I. HSV-1 structure, replication and pathogenesis. Neoplasia 1999; 1:387-401. [PMID: 10933054 PMCID: PMC1508113 DOI: 10.1038/sj.neo.7900055] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The design of effective gene therapy strategies for brain tumors and other neurological disorders relies on the understanding of genetic and pathophysiological alterations associated with the disease, on the biological characteristics of the target tissue, and on the development of safe vectors and expression systems to achieve efficient, targeted and regulated, therapeutic gene expression. The herpes simplex virus type 1 (HSV-1) virion is one of the most efficient of all current gene transfer vehicles with regard to nuclear gene delivery in central nervous system-derived cells including brain tumors. HSV-1-related research over the past decades has provided excellent insight into the structure and function of this virus, which, in turn, facilitated the design of innovative vector systems. Here, we review aspects of HSV-1 structure, replication and pathogenesis, which are relevant for the engineering of HSV-1-based vectors.
Collapse
Affiliation(s)
- A Jacobs
- Department of Neurology at the University and MPI for Neurological Research, Cologne, Germany.
| | | | | |
Collapse
|
29
|
Nishikawa Y, Xuan X, Otsuka H. Biosynthesis and interaction of glycoproteins E and I of canine herpesvirus. Virus Res 1999; 61:11-8. [PMID: 10426205 DOI: 10.1016/s0168-1702(99)00020-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
In cells infected with canine herpesvirus (CHV), the mature form of glycoprotein E (gE) had a molecular weight of 94 kDa, and that of glycoprotein I (gI) had a broad range of molecular weights of 55-62 kDa. gE and gI formed a complex like gE and gI of other alphaherpesviruses. When cells were infected with the gI minus mutant of CHV (gI/Z), the mature form of the 94 kDa gE was not formed, but a 76 kDa gE polypeptide was found. Similarly, no mature gI was formed in cells infected with the gE minus mutant of CHV (gE/Z), but a 40 kDa gI polypeptide was formed. When cells were coinfected with gE/Z and gI/Z, the molecular masses of gE and gI were increased from 76 to 94 kDa and from 40 to 55-62 kDa, respectively. We constructed vaccinia virus recombinants which expressed CHV gE or CHV gI. Only when cells were coinfected with both the vaccinia recombinant which expressed gE and the vaccinia recombinant which expressed gI, gE and gI were processed into their mature forms. Our results suggest that the presence of both gE and gI is necessary for efficient processing of the precursors of gE and gI to their mature forms.
Collapse
Affiliation(s)
- Y Nishikawa
- Department of Global Agricultural Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Japan
| | | | | |
Collapse
|
30
|
Chapman TL, You I, Joseph IM, Bjorkman PJ, Morrison SL, Raghavan M. Characterization of the interaction between the herpes simplex virus type I Fc receptor and immunoglobulin G. J Biol Chem 1999; 274:6911-9. [PMID: 10066744 DOI: 10.1074/jbc.274.11.6911] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Herpes simplex virus type I (HSV-1) virions and HSV-1-infected cells bind to human immunoglobulin G (hIgG) via its Fc region. A complex of two surface glycoproteins encoded by HSV-1, gE and gI, is responsible for Fc binding. We have co-expressed soluble truncated forms of gE and gI in Chinese hamster ovary cells. Soluble gE-gI complexes can be purified from transfected cell supernatants using a purification scheme that is based upon the Fc receptor function of gE-gI. Using gel filtration and analytical ultracentrifugation, we determined that soluble gE-gI is a heterodimer composed of one molecule of gE and one molecule of gI and that gE-gI heterodimers bind hIgG with a 1:1 stoichiometry. Biosensor-based studies of the binding of wild type or mutant IgG proteins to soluble gE-gI indicate that histidine 435 at the CH2-CH3 domain interface of IgG is a critical residue for IgG binding to gE-gI. We observe many similarities between the characteristics of IgG binding by gE-gI and by rheumatoid factors and bacterial Fc receptors such as Staphylococcus aureus protein A. These observations support a model for the origin of some rheumatoid factors, in which they represent anti-idiotypic antibodies directed against antibodies to bacterial and viral Fc receptors.
Collapse
Affiliation(s)
- T L Chapman
- Division of Biology 156-29, California Institute of Technology, Pasadena, California 91125, USA
| | | | | | | | | | | |
Collapse
|
31
|
Lubinski JM, Wang L, Soulika AM, Burger R, Wetsel RA, Colten H, Cohen GH, Eisenberg RJ, Lambris JD, Friedman HM. Herpes simplex virus type 1 glycoprotein gC mediates immune evasion in vivo. J Virol 1998; 72:8257-63. [PMID: 9733869 PMCID: PMC110183 DOI: 10.1128/jvi.72.10.8257-8263.1998] [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: 05/12/1998] [Accepted: 06/25/1998] [Indexed: 11/20/2022] Open
Abstract
Many microorganisms encode proteins that interact with molecules involved in host immunity; however, few of these molecules have been proven to promote immune evasion in vivo. Herpes simplex virus type 1 (HSV-1) glycoprotein C (gC) binds complement component C3 and inhibits complement-mediated virus neutralization and lysis of infected cells in vitro. To investigate the importance of the interaction between gC and C3 in vivo, we studied the virulence of a gC-null strain in complement-intact and C3-deficient animals. Using a vaginal infection model in complement-intact guinea pigs, we showed that gC-null virus grows to lower titers and produces less severe vaginitis than wild-type or gC rescued virus, indicating a role for gC in virulence. To determine the importance of complement, studies were performed with C3-deficient guinea pigs; the results demonstrated significant increases in vaginal titers of gC-null virus, while wild-type and gC rescued viruses showed nonsignificant changes in titers. Similar findings were observed for mice where gC null virus produced significantly less disease than gC rescued virus at the skin inoculation site. Proof that C3 is important was provided by studies of C3 knockout mice, where disease scores of gC-null virus were significantly higher than in complement-intact mice. The results indicate that gC-null virus is approximately 100-fold (2 log10) less virulent that wild-type virus in animals and that gC-C3 interactions are involved in pathogenesis.
Collapse
Affiliation(s)
- J M Lubinski
- Departments of Medicine, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Mijnes JD, Lutters BC, Vlot AC, Horzinek MC, Rottier PJ, de Groot RJ. The disulfide-bonded structure of feline herpesvirus glycoprotein I. J Virol 1998; 72:7245-54. [PMID: 9696819 PMCID: PMC109947 DOI: 10.1128/jvi.72.9.7245-7254.1998] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/1998] [Accepted: 06/11/1998] [Indexed: 11/20/2022] Open
Abstract
Alphaherpesvirus glycoproteins E and I (gE and gI, respectively) assemble into a hetero-oligomeric complex which promotes cell-to-cell transmission, a determining factor of virulence. Focusing on gI of feline herpesvirus (FHV), we examined the role of disulfide bonds during its biosynthesis, its interaction with gE, and gE-gI-mediated spread of the infection in vitro. The protein's disulfide linkage pattern was determined by single and pairwise substitutions for the four conserved cysteine residues in the ectodomain. The resulting mutants were coexpressed with gE in the vaccinia virus-based vTF7-3 system, and the formation and endoplasmic reticulum (ER)-to-Golgi transport of the hetero-oligomeric complex were monitored. The results were corroborated biochemically by performing an endoproteinase Lys-C digestion of a [35S]Cys-labeled secretory recombinant form of gI followed by tricine-sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis of the peptides under reducing and nonreducing conditions. We found that (i) gI derivatives lacking Cys79 (C1) and/or Cys223 (C4) still assemble with gE into transport-competent complexes, (ii) mutant proteins lacking Cys91 (C2) and/or Cys102 (C3) bind to gE but are retained in the ER, (iii) radiolabeled endoproteinase Lys-C-generated peptide species containing C1 and C4 are linked through disulfide bonds, and (iv) peptides containing both C2 and C3 are not disulfide linked to any other peptide. From these findings emerges a model in which C1 and C4 as well as C2 and C3 form intramolecular disulfide bridges. Since the cysteines in the ectodomain have been conserved during alphaherpesvirus divergence, we postulate that the model applies for all gI proteins. Analysis of an FHV recombinant with a C1-->S substitution confirmed that the C1-C4 disulfide bond is not essential for the formation of a transport-competent gE-gI complex. The mutation affected the posttranslational modification of gI and caused a slight cold-sensitivity defect in the assembly or the intracellular transport of the gE-gI complex but did not affect plaque size. Thus, C1 and the C1-C4 bond are not essential for gE-gI-mediated cell-to-cell spread, at least not in vitro.
Collapse
Affiliation(s)
- J D Mijnes
- Virology Unit, Department of Infectious Diseases and Immunology, Veterinary Faculty, Utrecht University, 3584 CL Utrecht, The Netherlands
| | | | | | | | | | | |
Collapse
|
33
|
Nishikawa Y, Xuan X, Otsuka H. Identification and characterization of the glycoprotein E and I genes of canine herpesvirus. Virus Res 1998; 56:77-92. [PMID: 9784067 DOI: 10.1016/s0168-1702(98)00062-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We have determined the sequence of the gE and gI genes of canine herpesvirus (CHV), DFD-6 strain. The gE ORF codes for a 522 a.a. polypeptide with a signal sequence at the amino-terminus and a trans-membrane domain at the carboxy-terminus. The gI ORF codes for a 259 a.a. polypeptide with a signal sequence but no trans-membrane domain. Comparison with another line of CHV indicated that the DFD-6 gI gene underwent a frame-shift mutation which caused the loss of the trans-membrane domain. Antibodies against the gE and gI polypeptides detected a 94 kDa gE and a broad band of gI (55-62 kDa) in DFD-6 infected cells, respectively. The precursor of DFD-6 gE is modified to the mature form by N-linked glycosylation only in the presence of gI. Together with the fact that the gI- mutant of DFD-6 produced smaller plaques, it is suggested that the truncated DFD-6 gI is functional. The precursor of DFD-6 gI is modified to the mature form by N-linked glycosylation only in the presence of gE.
Collapse
Affiliation(s)
- Y Nishikawa
- Department of Global Agricultural Sciences, Graduate School of Agricultural and Life Science, The University of Tokyo, Japan
| | | | | |
Collapse
|
34
|
Nagashunmugam T, Lubinski J, Wang L, Goldstein LT, Weeks BS, Sundaresan P, Kang EH, Dubin G, Friedman HM. In vivo immune evasion mediated by the herpes simplex virus type 1 immunoglobulin G Fc receptor. J Virol 1998; 72:5351-9. [PMID: 9620988 PMCID: PMC110157 DOI: 10.1128/jvi.72.7.5351-5359.1998] [Citation(s) in RCA: 95] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Herpes simplex virus (HSV) glycoproteins gE and gI form an immunoglobulin G (IgG) Fc receptor (FcgammaR) that binds the Fc domain of human anti-HSV IgG and inhibits Fc-mediated immune functions in vitro. gE or gI deletion mutant viruses are avirulent, probably because gE and gI are also involved in cell-to-cell spread. In an effort to modify FcgammaR activity without affecting other gE functions, we constructed a mutant virus, NS-gE339, that has four amino acids inserted into gE within the domain homologous to mammalian IgG FcgammaRs. NS-gE339 expresses gE and gI, is FcgammaR-, and does not participate in antibody bipolar bridging since it does not block activities mediated by the Fc domain of anti-HSV IgG. In vivo studies were performed with mice because the HSV-1 FcgammaR does not bind murine IgG; therefore, the absence of an FcgammaR should not affect virulence in mice. NS-gE339 causes disease at the skin inoculation site comparably to wild-type and rescued viruses, indicating that the FcgammaR- mutant virus is pathogenic in animals. Mice were passively immunized with human anti-HSV IgG and then infected with mutant or wild-type virus. We postulated that the HSV-1 FcgammaR should protect wild-type virus from antibody attack. Human anti-HSV IgG greatly reduced viral titers and disease severity in NS-gE339-infected animals while having little effect on wild-type or rescued virus. We conclude that the HSV-1 FcgammaR enables the virus to evade antibody attack in vivo, which likely explains why antibodies are relatively ineffective against HSV infection.
Collapse
Affiliation(s)
- T Nagashunmugam
- Infectious Diseases Division, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6073, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Ng TI, Ogle WO, Roizman B. UL13 protein kinase of herpes simplex virus 1 complexes with glycoprotein E and mediates the phosphorylation of the viral Fc receptor: glycoproteins E and I. Virology 1998; 241:37-48. [PMID: 9454715 DOI: 10.1006/viro.1997.8963] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Herpes simplex virus 1 encodes a Fc receptor consisting of glycoproteins E (gE) and I (gI) and two protein kinases specified by UL13 and US3, respectively. We report the following: (i) Antibody to UL13 formed immune complexes containing gE and gI in addition to UL13 protein. Immune complexes formed by monoclonal antibody to gE, but not those formed by monoclonal antibody to gI, also contained the UL13 protein. This association may reflect direct interaction between gE and UL13 inasmuch as IgG in preimmune rabbit serum and an antiserum made against another viral protein which does not react with the UL13 protein directly also bound gE and UL13. (ii) In cells infected with the wild-type virus, gE formed two sharp bands and a diffuse, slower migrating band. The slower sharp band was undetectable, and the diffuse slower migrating forms of gE were diminished in lysates of cells infected with a mutant virus lacking the UL13 gene (DeltaUL13). (iii) Both gE and gI were labeled with 32Pi in cells infected with wild-type or the DeltaUL13 virus, but the labeling was significantly stronger in cells infected with the wild-type virus than in those infected with the DeltaUL13 virus. (iv) In an in vitro protein kinase assay, UL13 immunoprecipitated from cells infected with wild-type virus labeled gE in the presence of [gamma-32P]ATP. This activity was absent in precipitates from cells infected with DeltaUL13 virus. The labeled gE comigrated with the slower, sharp band of gE. (v) gI present in the UL13 immune complex was also phosphorylated in the in vitro kinase assay. (vi) The cytoplasmic domain of gE contains recognition sequences for phosphorylation by casein kinase II (CKII). Exogenous CKII phosphorylated gE in immune complexes from lysates of cells infected with the DeltaUL13 mutant or in immune complexes from lysates of cells infected with wild-type virus that had been heated to inactivate all endogenous kinase activity including that of UL13. In both instances, CKII phosphorylated gE in both the slow and fast migrating sharp bands. We conclude that UL13 physically associates with gE and mediates the phosphorylation of gE and gI. UL13 may also be a determinant in posttranslational processing of gE.
Collapse
Affiliation(s)
- T I Ng
- The Marjorie B. Kovler Viral Oncology Laboratories, The University of Chicago, 910 East 58th Street, Chicago, Illinois, 60637, USA
| | | | | |
Collapse
|
36
|
Kimura H, Straus SE, Williams RK. Varicella-zoster virus glycoproteins E and I expressed in insect cells form a heterodimer that requires the N-terminal domain of glycoprotein I. Virology 1997; 233:382-91. [PMID: 9217061 DOI: 10.1006/viro.1997.8625] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Varicella-zoster virus (VZV) glycoproteins E and I (gE and gI), which are major components of the virion envelope, form a noncovalently linked complex. To understand their properties and functions, we expressed and purified soluble forms of gE and gI in the baculovirus system. Extracellular domains of gE and gI were cloned into baculoviruses, using either native or insect-derived signal peptides. Each recombinant virus yielded soluble protein in culture medium although a higher level of secretion was achieved with insect-derived signal peptides in recombinant gE baculoviruses. A soluble gE-gI complex was formed by co-infecting insect cells with recombinant gE and gI baculoviruses and detected by immunoprecipitation followed by Western blotting analyses. By gel filtration and cross-linking studies, we showed that the VZV gE-gI complex expressed in insect cells is a heterodimer. Interestingly, two recombinant gI proteins in which signal peptides were replaced with insect-derived signal peptides did not associate with gE. Amino-terminal sequencing and site-specific mutational studies showed that the replacement of only the signal peptides did not prevent complex formation but alterations in the processed amino-terminus of gI abrogated its ability to complex with gE. These findings indicate that the mature amino-terminus of gI is required for gE-gI complex formation by the external domains of VZV gE and gI.
Collapse
Affiliation(s)
- H Kimura
- Laboratory of Clinical Investigation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | |
Collapse
|
37
|
Weeks BS, Sundaresan P, Nagashunmugam T, Kang E, Friedman HM. The herpes simplex virus-1 glycoprotein E (gE) mediates IgG binding and cell-to-cell spread through distinct gE domains. Biochem Biophys Res Commun 1997; 235:31-5. [PMID: 9196030 DOI: 10.1006/bbrc.1997.6720] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Herpes simplex virus-1 (HSV-1) glycoprotein E (gE) is a multifunctional protein capable of both binding the Fc portion of IgG and mediating cell-to-cell spread of HSV-1. Here we report that the domain on gE involved in IgG binding is distinct from the domain involved in mediating cell-to-cell spread. To do this we have used five mutants of the HSV-1 strain NS: NS-gE(null), a gE deletion virus; rNS-gE(null), a gE rescued virus; NS-gE339, a gE mutant virus with a four amino acid insert at position 339; rNS-gE339, a gE rescue of NS-gE339; and NS-gE406, a gE mutant virus with the same four amino acids inserted at position 406. Using IgG coated sheep red blood cells in rosetting assays, we show that the NS-gE339 does not bind IgG, yet retains the ability to mediate normal cell-to-cell spread. These results demonstrate that the gE domain involved in IgG binding differs from the domain involved in cell-to-cell spread.
Collapse
Affiliation(s)
- B S Weeks
- Department of Medicine, University of Pennsylvania, Philadelphia 19104, USA
| | | | | | | | | |
Collapse
|
38
|
Davis-Poynter NJ, Farrell HE. Masters of deception: a review of herpesvirus immune evasion strategies. Immunol Cell Biol 1996; 74:513-22. [PMID: 8989589 DOI: 10.1038/icb.1996.84] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Herpesviruses have acquired a variety of different mechanisms to avoid the damaging effects of host immunity. Frequently, these viruses subvert normal immune regulatory functions utilized by the host. The focus of this review is upon herpesvirus genes encoding known or potential immunomodulatory proteins. Areas covered include inhibition of complement and antibody function, herpesvirus-encoded homologues of cytokines and chemokine receptors, and potential disruption of cellular recognition of virally infected targets.
Collapse
Affiliation(s)
- N J Davis-Poynter
- Department of Microbiology, University of Western Australia, Queen Elizabeth II Medical Centre, Nedlands, Australia.
| | | |
Collapse
|
39
|
Handler CG, Cohen GH, Eisenberg RJ. Cross-linking of glycoprotein oligomers during herpes simplex virus type 1 entry. J Virol 1996; 70:6076-82. [PMID: 8709231 PMCID: PMC190629 DOI: 10.1128/jvi.70.9.6076-6082.1996] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Herpes simplex virus (HSV) has 10 glycoproteins in its envelope. Glycoprotein B (gB), gC, gD, gH, and gL have been implicated in virus entry. We previously used chemical cross-linking to show that these five glycoproteins were close enough to each other to be cross-linked into homodimeric and hetero-oligomeric forms; hetero-oligomers of gB-gC, gC-gD, gD-gB, gH-gL, gC-gL and gD-gL were found in purified virions. To better understand the roles of these glycoproteins in viral entry, we have modified a standard HSV penetration assay to include cross-linkers. This allowed us to examine changes in associations of viral glycoproteins during the entry process. HSV-1(KOS) was adsorbed at 4 degrees C to human neuroblastoma cells (SY5Y). The temperature was raised to 37 degrees C and cells were treated with cross-linker at various times after the temperature shift. Cytoplasmic extracts were examined by Western blotting (immunoblotting) for viral glycoproteins. We found that (i) as in virus alone, the length and concentration of the cross-linking agent affected the number of specific complexes isolated; (ii) the same glycoprotein patterns found in purified virions were also present after attachment of virions to cells; and (iii) the ability to cross-link HSV glycoproteins changed as virus penetration proceeded, e.g., gB and gD complexes which were present during attachment disappeared with increasing time, and their disappearance paralleled the kinetics of penetration. However, this phenomenon appeared to be selective since it was not observed with gC oligomers. In addition, we examined the cross-linking patterns of gB and gD in null viruses K082 and KOSgD beta. Neither of these mutants, which attach but cannot penetrate, showed changes in glycoprotein cross-linking over time. We speculate that these changes are due to conformational changes which preclude cross-linking or spatial alterations which dissociate the glycoprotein interactions during the penetration events.
Collapse
Affiliation(s)
- C G Handler
- School of Dental Medicine, University of Pennsylvania, Philadelphia 19104, USA.
| | | | | |
Collapse
|
40
|
Colle CF, Tarbet EB, Grafton WD, Jennings SR, O'Callaghan DJ. Equine herpesvirus-1 strain KyA, a candidate vaccine strain, reduces viral titers in mice challenged with a pathogenic strain, RacL. Virus Res 1996; 43:111-24. [PMID: 8864201 DOI: 10.1016/0168-1702(96)01324-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The equine herpesvirus type-1 (EHV-1) strain Kentucky A (KyA) has a long history of repeated passage either in vivo in the Syrian hamster or in vitro in mouse L-M fibroblast tissue culture. This repeated passage in cells other than those of the natural host has caused genomic alterations of the KyA chromosome resulting in deletion of several genes or portions of open reading frames (ORFs). This report presents in vivo data from a mouse model of EHV-1 infection demonstrating the attenuated nature of EHV-1 strain KyA and that intranasal infection with KyA protects animals from subsequent challenge with a pathogenic strain, RacL, by reducing RacL viral titers in the lungs of the challenged animals. Mice infected with KyA exhibit no clinical manifestations of EHV-1 disease and do not experience the wasting that occurs with RacL infection. KyA-infected mice clear virus from the lung by day 5 post-infection (p.i.), whereas RacL infected mice have substantial virus titers (5 x 10(5) pfu/lung) at this time point. Intranasal infection with KyA followed by a challenge with RacL 4 weeks post-KyA infection resulted in a significant (P = 0.0079) reduction in the lung titers of the RacL virus. RacL was identified as the virus present in the lungs of the challenged mice by a PCR assay employing primers to amplify the EUS4 gene which differs in size by 1.2 kilobase pairs (kbp) in the two strains. Importantly, the protection afforded by KyA is long lasting in that challenge with RacL 15 months after KyA infection, results in reduced virus titers and viral clearance by day 5 post-challenge. These results support the further consideration of EHV-1 KyA as a live virus vaccine.
Collapse
Affiliation(s)
- C F Colle
- Department of Microbiology and Immunology, Louisiana State University Medical Center, Shreveport 71130-3932, USA
| | | | | | | | | |
Collapse
|
41
|
Mijnes JD, van der Horst LM, van Anken E, Horzinek MC, Rottier PJ, de Groot RJ. Biosynthesis of glycoproteins E and I of feline herpesvirus: gE-gI interaction is required for intracellular transport. J Virol 1996; 70:5466-75. [PMID: 8764058 PMCID: PMC190504 DOI: 10.1128/jvi.70.8.5466-5475.1996] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The biosynthesis of glycoproteins E and I of feline herpesvirus was studied by using the vaccinia virus vTF7-3 expression system. gE and gI were synthesized as N-glycosylated, endoglycosidase H (EndoH)-sensitive precursors with Mrs of 83,000 and 67,000, respectively. When coexpressed, gE and gI formed sodium dodecyl sulfate-sensitive hetero-oligomeric complexes that were readily transported from the endoplasmic reticulum (ER). Concomitantly, the glycoproteins acquired extensive posttranslational modifications, including O glycosylation, leading to an increase in their apparent molecular weights to 95,000 and 80,000 to 100,000 for gE and gI, respectively. In the absence of gE, most gI remained EndoH sensitive. Only a minor population became EndoH resistant, but these molecules were processed aberrantly as indicated by their Mrs (100,000 to 120,000). By immunofluorescence microscopy, gI was detected primarily in the ER but also at the plasma membrane. gE, when expressed by itself, remained EndoH sensitive and was found only in the ER and the nuclear envelope. These results were corroborated by studying the biosynthesis of gE in feline herpesvirus (FHV)-infected cells. In cells infected with wild-type FHV, gE acquired the same co- and posttranslational modifications as during vTF7-3-driven expression. However, an FHV mutant lacking gI failed to produce mature gE. We conclude that gE is retained in the ER, presumably by associating with molecular chaperones, and becomes transport competent only when in a complex with gI.
Collapse
Affiliation(s)
- J D Mijnes
- Virology Unit, Department of Infectious Diseases, Utrecht University, The Netherlands
| | | | | | | | | | | |
Collapse
|
42
|
Rux AH, Moore WT, Lambris JD, Abrams WR, Peng C, Friedman HM, Cohen GH, Eisenberg RJ. Disulfide bond structure determination and biochemical analysis of glycoprotein C from herpes simplex virus. J Virol 1996; 70:5455-65. [PMID: 8764057 PMCID: PMC190503 DOI: 10.1128/jvi.70.8.5455-5465.1996] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
A biochemical analysis of glycoprotein C (gC of herpes simplex virus was undertaken to further characterize the structure of the glycoprotein and to determine its disulfide bond arrangement. We used three recombinant forms of gC, gC1(457t), gC1(delta33-123t), and gC2(426t), each truncated prior to the transmembrane region. The proteins were expressed and secreted by using a baculovirus expression system and have been shown to bind to monoclonal antibodies which recognize discontinuous epitopes and to complement component C3b in a dose-dependent manner. We confirmed the N-terminal residues of each mature protein by Edman degradation and confirmed the internal deletion in gC1(delta33-123t). The molecular weight and extent of glycosylation of gC1 (457t), gC1(delta33-123t), and gC2(426t) were determined by treating each protein with endoglycosidases and then subjecting it to sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and mass spectrometric analysis. The data indicate that eight to nine of the predicted N-linked oligosaccharide sites on gC1(457t) are occupied by glycans of approximately 1,000 Da. In addition, O-linked oligosaccharides are present on gC1(457t), primarily localized to the N-terminal region (amino acids [aa] 33 to 123) of the protein. gC2(426t) contains N-linked oligosaccharides, but no O-linked oligosaccharides were detected. To determine the disulfide bond arrangement of the eight cysteines of gC1(457t),the protein was cleaved with cyanogen bromide. SDS-PAGE analysis followed by Edman degradation identified three cysteine-containing fragments which are not connected by disulfide linkages. Chemical modification of cysteines combined with matrix-assisted laser desorption ionization mass spectrometry identified disulfide bonds between cysteine 1 (aa 127) and cysteine 2 (aa 144) and between cysteine 3 (aa 286) and cysteine 4 (aa 347). Further proteolysis of the cyanogen bromide-generated fragment containing cysteine 5 through cysteine 8, combined with mass spectrometry and Edman degradation, showed that disulfide bonds link cysteine 5 (aa 386) to cysteine 8 (aa 442) and cysteine 6 (aa 390) to cysteine 7 (aa 419). A similar disulfide bond arrangement is postulated to exist in gC homologs from other herpesviruses.
Collapse
Affiliation(s)
- A H Rux
- Department of Microbiology, University of Pennsylvania, Philadelphia 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Dubin G, Basu S, Mallory DL, Basu M, Tal-Singer R, Friedman HM. Characterization of domains of herpes simplex virus type 1 glycoprotein E involved in Fc binding activity for immunoglobulin G aggregates. J Virol 1994; 68:2478-85. [PMID: 7511171 PMCID: PMC236725 DOI: 10.1128/jvi.68.4.2478-2485.1994] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Herpes simplex virus type 1 glycoproteins gE and gI form receptors for the Fc domain of immunoglobulin G (IgG) which are expressed on the surface of infected cells and on the virion envelope and which protect the virus from immune attack. Glycoprotein gE-1 is a low-affinity Fc receptor (FcR) that binds IgG aggregates, while gE-1 and gI-1 form a complex which serves as a higher-affinity FcR capable of binding IgG monomers. In this study, we describe two approaches used to map an Fc binding domain on gE-1 for IgG aggregates. First, we constructed nine plasmids encoding gE-1/gD-1 fusions proteins, each containing a large gE-1 peptide inserted into the ectodomain of gD-1. Fusion proteins were tested for FcR activity with IgG-sensitized erythrocytes in a rosetting assay. Three of the fusion proteins containing overlapping gE-1 peptides demonstrated FcR activity; the smallest peptide that retained Fc binding activity includes gE-1 amino acids 183 to 402. These results indicate that an Fc binding domain is located between gE-1 amino acids 183 and 402. To more precisely map the Fc binding domain, we tested a panel of 21 gE-1 linker insertion mutants. Ten mutants with insertions between gE-1 amino acids 235 and 380 failed to bind IgG-sensitized erythrocytes, while each of the remaining mutants demonstrated wild-type Fc binding activity. Taken together, these results indicate that the region of gE-1 between amino acids 235 and 380 forms an FcR domain. A computer-assisted analysis of the amino acid sequence of gE-1 demonstrates an immunoglobulin-like domain contained within this region (residues 322 to 359) which shares homology with mammalian FcRs.
Collapse
Affiliation(s)
- G Dubin
- Department of Medicine, School of Medicine, University of Pennsylvania, Philadelphia 19104-6073
| | | | | | | | | | | |
Collapse
|
44
|
Ghiasi H, Kaiwar R, Nesburn AB, Slanina S, Wechsler SL. Expression of seven herpes simplex virus type 1 glycoproteins (gB, gC, gD, gE, gG, gH, and gI): comparative protection against lethal challenge in mice. J Virol 1994; 68:2118-26. [PMID: 8138996 PMCID: PMC236686 DOI: 10.1128/jvi.68.4.2118-2126.1994] [Citation(s) in RCA: 124] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
We have constructed recombinant baculoviruses individually expressing seven of the herpes simplex virus type 1 (HSV-1) glycoproteins (gB, gC, gD, gE, gG, gH, and gI). Vaccination of mice with gB, gC, gD, gE, or gI resulted in production of high neutralizing antibody titers to HSV-1 and protection against intraperitoneal and ocular challenge with lethal doses of HSV-1. This protection was statistically significant and similar to the protection provided by vaccination with live nonvirulent HSV-1 (90 to 100% survival). In contrast, vaccination with gH produced low neutralizing antibody titers and no protection against lethal HSV-1 challenge. Vaccination with gG produced no significant neutralizing antibody titer and no protection against ocular challenge. However, gG did provide modest, but statistically significant, protection against lethal intraperitoneal challenge (75% protection). Compared with the other glycoproteins, gG and gH were also inefficient in preventing the establishment of latency. Delayed-type hypersensitivity responses to HSV-1 at day 3 were highest in gG-, gH-, and gE-vaccinated mice, while on day 6 mice vaccinated with gC, gE, and gI had the highest delayed-type hypersensitivity responses. All seven glycoproteins produced lymphocyte proliferation responses, with the highest response being seen with gG. The same five glycoproteins (gB, gC, gD, gE, and gI) that induced the highest neutralization titers and protection against lethal challenge also induced some killer cell activity. The results reported here therefore suggest that in the mouse protection against lethal HSV-1 challenge and the establishment of latency correlate best with high preexisting neutralizing antibody titers, although there may also be a correlation with killer cell activity.
Collapse
Affiliation(s)
- H Ghiasi
- Cedars-Sinai Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048
| | | | | | | | | |
Collapse
|
45
|
Van Vliet KE, De Graaf-Miltenburg LA, Verhoef J, Van Strijp JA. A flow cytometric rosetting assay for the analysis of Fc receptors and C3 receptors on HSV-infected cells. J Immunol Methods 1993; 157:57-64. [PMID: 8423374 DOI: 10.1016/0022-1759(93)90070-n] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
A sensitive and reproducible flow cytometric assay was developed for the analysis of Fc gamma and C3b(i) receptors on HSV-infected cells. The method is based on a rosette technique using fluorochrome-labeled erythrocytes sensitized with IgG or C3b(i). A comparison of flow cytometric and microscopic quantitation demonstrated that the binding of EIgG, EC3b(i) to HSV-infected cells were correlated. Flow cytometric analysis provides the opportunity to study simultaneously the distribution of E per HSV-infected cell and the total binding of E to the whole population of HSV-infected cells. Receptor activity and HSV glycoprotein cell surface expression were shown to be correlated in a linear fashion. The assay could be applied to other Fc gamma R- and C3b(i)R-bearing cells.
Collapse
Affiliation(s)
- K E Van Vliet
- Eijkman-Winkler Laboratory of Medical Microbiology, Utrecht University, Netherlands
| | | | | | | |
Collapse
|
46
|
Van Vliet KE, De Graaf-Miltenburg LA, Verhoef J, Van Strijp JA. Direct evidence for antibody bipolar bridging on herpes simplex virus-infected cells. Immunol Suppl 1992; 77:109-15. [PMID: 1328043 PMCID: PMC1421581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cells infected with herpes simplex virus type 1 (HSV-1) express a cell-surface receptor able to bind the Fc portion of immunoglobulin G (IgG). In this study we provide direct evidence that bipolar bridging of antibodies, bound to the surface antigens on HSV-infected cells and to the Fc-receptor through the Fc part, offers the virus a survival advantage. Evidence was obtained by comparing the binding of FITC-labelled protein A, which has a similar binding site on IgG as the HSV-FcR, to cell-bound antibodies on HSV-infected cells and non-infected cells. The effectiveness of antibody bipolar bridging was dependent on the concentration of cell-bound IgG. At low concentrations of serum (0.1%) an 80% reduction in protein A-FITC binding to HSV-infected cells compared to non-infected cells was found. Even at higher concentrations of serum, antibody bipolar bridging resulted in a 40% reduction in the number of 'free' available Fc parts on HSV-infected cells compared to non-infected cells. Furthermore, these findings could be confirmed in a functional assay. The Fc-mediated attachment of granulocytes was significantly lower in HSV-infected cells compared to non-infected cells. From this study we conclude that HSV-FcR, by binding immune IgG in a bipolar fashion, provides the virus with an effective defence mechanism.
Collapse
Affiliation(s)
- K E Van Vliet
- Eijkman-Winkler Laboratory of Medical Microbiology, Utrecht University, The Netherlands
| | | | | | | |
Collapse
|
47
|
Tsuchiya N, Williams RC. Molecular mimicry--hypothesis or reality? West J Med 1992; 157:133-8. [PMID: 1279899 PMCID: PMC1011230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A number of observations support molecular mimicry as a possible pathogenetic mechanism in diseases such as acute rheumatic fever, reactive arthritis after enteric infection or associated with Reiter's syndrome, myasthenia gravis, or even in rheumatoid arthritis. Molecular mimicry can be defined as a sharing of epitopes in linear or 3-dimensional presentation on disparate proteins from entirely different sources--for instance, group A streptococcal membranes and human cardiac myosin. How exposure to or infection with organisms sharing molecular similarity with antigens of the human host can evade tolerance and actually induce a self-reacting humoral or cellular immune response is still not clear; however, a large body of evidence has now been accumulated that documents apparent molecular mimicry mechanisms in these disorders. In some diseases, the molecular mimicry appears to involve human target organs and specific components of the infectious organism, whereas in others the host HLA cell surface molecules appear to share antigens with presumed bacterial or viral initiators of disease.
Collapse
Affiliation(s)
- N Tsuchiya
- Department of Medicine and Physical Therapy, University of Tokyo School of Medicine, Japan
| | | |
Collapse
|
48
|
Hung SL, Srinivasan S, Friedman HM, Eisenberg RJ, Cohen GH. Structural basis of C3b binding by glycoprotein C of herpes simplex virus. J Virol 1992; 66:4013-27. [PMID: 1602532 PMCID: PMC241204 DOI: 10.1128/jvi.66.7.4013-4027.1992] [Citation(s) in RCA: 51] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Glycoproteins C (gC) from herpes simplex virus type 1 (HSV-1) and HSV-2, gC-1 and gC-2, bind the human complement fragment C3b, although the two glycoproteins differ in their abilities to act as C3b receptors on infected cells and in their effects on the alternative complement pathway. Previously, we identified three regions of gC-2 (I, II, and III) which are important for C3b binding. In this study, our goal was to identify C3b-binding sites on gC-1 and to continue our analysis of gC-2. We constructed a large panel of mutants by using the cloned gC-1 and gC-2 genes. Most of the mutant proteins were transported to the surface of transiently transfected L cells and reacted with one or more monoclonal antibodies to discontinuous epitopes. By using 31 linker insertion mutants spread across the coding region of gC-1, we identified four regions in the ectodomain of gC-1 which are important for C3b binding, three of which are similar in position to C3b-binding regions I, II, and III of gC-2. Region III shares some similarities with the short consensus repeat found in CR1, the human complement receptor. These were, in part, the targets for construction of 20 single amino acid changes in region III of gC-1 and gC-2. These mutants identified similarities and differences in the C3b-binding properties of gC-1 and gC-2 and suggest that the amino half of region III is more important for C3b binding. However, our results do not support the concept of a structural relationship between the short consensus repeat of CR1 and gC, since mutations of some of the conserved residues, including three of four cysteines in region III, had no effect on C3b binding. Finally, we constructed four deletion mutants of gC-1, including one which lacked residues 33 to 123, as well as residues 367 to 449. This severely truncated molecule, lacking four cysteines and five potential N-linked glycosylation sites, was transported to the cell surface and retained its ability to bind monoclonal antibodies as well as C3b. Thus, the four distinct C3b-binding regions of gC-1 and several epitopes within two different antigenic sites are localized within residues 124 to 366.
Collapse
Affiliation(s)
- S L Hung
- Department of Microbiology, University of Pennsylvania, Philadelphia 19104-6003
| | | | | | | | | |
Collapse
|
49
|
Litwin V, Jackson W, Grose C. Receptor properties of two varicella-zoster virus glycoproteins, gpI and gpIV, homologous to herpes simplex virus gE and gI. J Virol 1992; 66:3643-51. [PMID: 1316474 PMCID: PMC241147 DOI: 10.1128/jvi.66.6.3643-3651.1992] [Citation(s) in RCA: 99] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The varicella-zoster virus (VZV) genome contains 70 reading frames (ORF), 5 of which encode the glycoproteins gpI, gpII, gpIII, gpIV, and gpV. ORF 67 and 68 lie adjacent to each other in the unique short region of the VZV genome and code for gpIV and gpI, respectively. These two genes, which are contained within the HindIII C fragment of the VZV genome, were subcloned in the correct orientation downstream from the promoter regions of the eukaryotic expression vectors pCMV5 and pBJ. After transfection, 5 to 20% of the Cos cells bound antibody specific for the given glycoprotein. In this study, it was shown that only the cells transfected with the gpI construct bound to the Fc fragment of human immunoglobulin G. Neither the transfected gpIV gene product nor the vector only bound to the Fc fragment. Thus, VZV gpI is confirmed to be the VZV-encoded Fc-binding glycoprotein. Like the wild-type form of gpI expressed in VZV-infected cells, gpI precipitated from transfected cells contained both N-linked and O-linked glycans and was heavily sialated. In addition, the transfected gpI gene product was phosphorylated both in cell culture and in protein kinase assays by mammalian casein kinases I and II. Extensive computer-assisted analyses of the VZV gpI sequence, as well as those of alphaherpesviral homolog glycoproteins, disclosed properties similar to those of other cell surface receptors; these included (i) exocytoplasmic regions rich in cysteine residues, (ii) membrane-proximal regions with potential O-linked glycosylation sites, and (iii) cytoplasmic domains with consensus phosphorylation sites.
Collapse
Affiliation(s)
- V Litwin
- Department of Microbiology, University of Iowa College of Medicine, Iowa City 52242
| | | | | |
Collapse
|
50
|
Ghiasi H, Kaiwar R, Nesburn AB, Slanina S, Wechsler SL. Baculovirus-expressed glycoprotein E (gE) of herpes simplex virus type-1 (HSV-1) protects mice against lethal intraperitoneal and lethal ocular HSV-1 challenge. Virology 1992; 188:469-76. [PMID: 1585630 DOI: 10.1016/0042-6822(92)90500-o] [Citation(s) in RCA: 30] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We have constructed a recombinant baculovirus expressing high levels of the herpes simplex virus type 1 (HSV-1) glycoprotein E (gE) in Sf9 cells. The expressed gE migrated on gels as a double band with apparent molecular weights of 68 and 70 kDa. The recombinant gE was glycosylated based on its susceptibility to tunicamycin treatment and was transported to the membrane of Sf9 cells based on indirect immunofluorescence. Mice vaccinated with gE developed high serum titers of HSV-1-neutralizing antibodies based on plaque reduction assays. gE vaccination also induced a strong delayed type hypersensitivity (DTH) response to HSV-1. In addition, mice vaccinated with the recombinant gE were protected from both intraperitoneal and ocular lethal HSV-1 challenge. To our knowledge, this is the first report in which vaccination with gE was shown to induce high neutralizing antibody titers, a DTH response, or protection against lethal HSV-1 challenge.
Collapse
Affiliation(s)
- H Ghiasi
- Ophthalmology Research, Cedars-Sinai Medical Center, Los Angeles, California 90048
| | | | | | | | | |
Collapse
|