1
|
Slein MD, Backes IM, Kelkar NS, Garland CR, Khanwalkar US, Sholukh AM, Johnston CM, Leib DA, Ackerman ME. Improving antibody-mediated protection against HSV infection by eliminating interactions with the viral Fc receptor gE/gI. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.20.624598. [PMID: 39605495 PMCID: PMC11601663 DOI: 10.1101/2024.11.20.624598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Herpes simplex virus (HSV) encodes surface glycoproteins that are host defense evasion molecules, allowing the virus to escape immune clearance. In addition to their role in neuropathogenesis and cell-cell spread, glycoproteins E and I (gE/gI) form a viral Fc receptor (vFcR) for most subclasses and allotypes of human IgG and promote evasion of humoral immune responses. While monoclonal antibodies (mAbs) protect mice from neonatal HSV (nHSV) infections, the impact of the vFcR on mAb-mediated protection by binding to IgG is unknown. Using HSV-1 with intact and ablated gE-mediated IgG Fc binding, and Fc-engineered antibodies with modified ability to interact with gE/gI, we investigated the role of the vFcR in viral pathogenesis and mAb-mediated protection from nHSV. The gD-specific human mAb HSV8 modified to lack binding to gE exhibited enhanced neutralization and in vivo protection compared to its native IgG1 form. This improved protection by the engineered mAbs was dependent on the presence of the vFcR. Human IgG3 allotypes lacking vFcR binding also exhibited enhanced antiviral activity in vivo, suggesting that vaccines that robustly induce IgG3 responses could show enhanced protection. suggesting the value of vaccination strategies that robustly induce this subclass. Lastly, analysis of longitudinal responses to acute primary genital infection in humans raised the possibility that unlike most viruses, HSV may exhibited slow induction of IgG3. In summary, this study demonstrates that mAbs lacking the ability to interact with the vFcR can exhibit improved protection from HSV-offering new prospects for antibody-based interventions.
Collapse
Affiliation(s)
- Matthew D. Slein
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Iara M. Backes
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Natasha S. Kelkar
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Callaghan R. Garland
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | | | | | - Christine M. Johnston
- Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Departments of Medicine and Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104, USA
| | - David A. Leib
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Margaret E. Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| |
Collapse
|
2
|
Wang K, Jordan T, Dowdell K, Herbert R, Moore IN, Koelle DM, Cohen JI. A nonhuman primate model for genital herpes simplex virus 2 infection that results in vaginal vesicular lesions, virus shedding, and seroconversion. PLoS Pathog 2024; 20:e1012477. [PMID: 39226323 PMCID: PMC11371218 DOI: 10.1371/journal.ppat.1012477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/05/2024] [Indexed: 09/05/2024] Open
Abstract
The most commonly used animal models for evaluating the efficacy of HSV-2 candidate vaccines are mice and guinea pigs. While numerous HSV-2 vaccine candidates have been tested in these animals and were effective in reducing disease and mortality, these results did not predict the effectiveness of the vaccines in human trials. Infection of rhesus macaques rarely results in lesions or HSV-2 specific antibody responses. In seeking an animal model that better recapitulates human disease and that might be more predictive of the efficacy of prophylactic vaccines than mice and guinea pigs, we evaluated Cebus apella (C. apella), a New World primate, in an HSV-2 genital infection model. Infectious HSV-2 was cultured from vaginal swabs from all 4 animals for 9-14 days after intravaginal inoculation of HSV-2 seronegative monkeys. Two of 4 monkeys had vesicular lesions in the vagina or vulva. No neurological symptoms were noted. Recurrent lesions and HSV-2 DNA shedding after acute disease resolved was infrequent. UV irradiation of the genital area did not induce recurrent genital lesions or virus shedding. All 4 monkeys developed HSV-2 neutralizing antibodies as well as virus-specific CD4 and CD8 T cell responses. Reinfection of animals 15 to 19 months after primary infection did not result in lesions; animals had reduced virus shedding and a shorter duration of shedding compared with that during primary infection, suggesting that primary infection induced protective immunity. Primary fibroblasts from C. apella monkeys supported the growth of HSV-2 in vitro; in contrast, HSV-2 did not replicate above the titer of the input inoculum in fibroblasts from rhesus macaques. These observations suggest that the C. apella monkey has potential to serve as a model for evaluating the efficacy of prophylactic vaccines, antivirals, or monoclonal antibodies to HSV-2.
Collapse
Affiliation(s)
- Kening Wang
- Medical Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Tristan Jordan
- Medical Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kennichi Dowdell
- Medical Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | | | - Ian N. Moore
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - David M. Koelle
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Global Health, School of Medicine, University of Washington, Seattle, Washington, United States of America
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Benaroya Research Institute, Seattle, Washington, United States of America
| | - Jeffrey I. Cohen
- Medical Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
3
|
Canova PN, Charron AJ, Leib DA. Models of Herpes Simplex Virus Latency. Viruses 2024; 16:747. [PMID: 38793628 PMCID: PMC11125678 DOI: 10.3390/v16050747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 04/29/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
Our current understanding of HSV latency is based on a variety of clinical observations, and in vivo, ex vivo, and in vitro model systems, each with unique advantages and drawbacks. The criteria for authentically modeling HSV latency include the ability to easily manipulate host genetics and biological pathways, as well as mimicking the immune response and viral pathogenesis in human infections. Although realistically modeling HSV latency is necessary when choosing a model, the cost, time requirement, ethical constraints, and reagent availability are also equally important. Presently, there remains a pressing need for in vivo models that more closely recapitulate human HSV infection. While the current in vivo, ex vivo, and in vitro models used to study HSV latency have limitations, they provide further insights that add to our understanding of latency. In vivo models have shed light on natural infection routes and the interplay between the host immune response and the virus during latency, while in vitro models have been invaluable in elucidating molecular pathways involved in latency. Below, we review the relative advantages and disadvantages of current HSV models and highlight insights gained through each.
Collapse
Affiliation(s)
- Paige N. Canova
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA;
- Guarini School of Graduate and Advanced Studies at Dartmouth, Hanover, NH 03755, USA;
| | - Audra J. Charron
- Guarini School of Graduate and Advanced Studies at Dartmouth, Hanover, NH 03755, USA;
| | - David A. Leib
- Guarini School of Graduate and Advanced Studies at Dartmouth, Hanover, NH 03755, USA;
| |
Collapse
|
4
|
Slein MD, Backes IM, Garland CR, Kelkar NS, Leib DA, Ackerman ME. Effector functions are required for broad and potent protection of neonatal mice with antibodies targeting HSV glycoprotein D. Cell Rep Med 2024; 5:101417. [PMID: 38350452 PMCID: PMC10897633 DOI: 10.1016/j.xcrm.2024.101417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/26/2023] [Accepted: 01/17/2024] [Indexed: 02/15/2024]
Abstract
Multiple failed herpes simplex virus (HSV) vaccine candidates induce robust neutralizing antibody (Ab) responses in clinical trials, raising the hypothesis that Fc-domain-dependent effector functions may be critical for protection. While neonatal HSV (nHSV) infection results in mortality and lifelong neurological morbidity in humans, it is uncommon among neonates with a seropositive birthing parent, supporting the hypothesis that Ab-based therapeutics could protect neonates from HSV. We therefore investigated the mechanisms of monoclonal Ab (mAb)-mediated protection in a mouse model of nHSV infection. For a panel of glycoprotein D (gD)-specific mAbs, neutralization and effector functions contributed to nHSV-1 protection. In contrast, effector functions alone were sufficient to protect against nHSV-2, exposing a functional dichotomy between virus types consistent with vaccine trial results. Effector functions are therefore crucial for protection by these gD-specific mAbs, informing effective Ab and vaccine design and demonstrating the potential of polyfunctional Abs as therapeutics for nHSV infections.
Collapse
Affiliation(s)
- Matthew D Slein
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA; Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Iara M Backes
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA; Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Callaghan R Garland
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Natasha S Kelkar
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA; Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - David A Leib
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA.
| | - Margaret E Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA; Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA.
| |
Collapse
|
5
|
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
|
6
|
Horton MS, Minnier M, Cosmi S, Cox K, Galli J, Peters J, Sullivan N, Squadroni B, Tang A, Fridman A, Wang D, Chen Z, Vora KA. Development of a microneutralization assay for HSV-2. J Virol Methods 2021; 297:114268. [PMID: 34437874 DOI: 10.1016/j.jviromet.2021.114268] [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: 04/20/2021] [Revised: 08/17/2021] [Accepted: 08/21/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Plaque Reduction Neutralization Test (PRNT) is the standard assay used for measuring neutralizing antibody responses to Herpes simplex virus type-2 (HSV-2). The PRNT is a cumbersome, time-consuming and laborious assay. The development of a faster, high throughput microneutralization assay (MNA) for HSV-2 viruses carried out in a 96-well format will allow for rapid testing of large numbers of samples for drug and vaccine development. METHODS We describe the generation of a MNA that utilizes a pair of anti-HSV human monoclonal antibodies (mAbs) for virus detection in HSV-2 infected Vero cells. Antibodies were generated by B-cell cloning from PBMC's isolated from HSV-1 negative/HSV-2 positive donors. We describe the selection and characterization of the antibodies used for virus detection by ELISA with purified, recombinant anti-HSV glycoproteins, antibody binding in infected cells, and Western Blot. We determine the anti-HSV-2 neutralizing titers of immune sera from mice by MNA and PRNT and compare these results by linear regression analysis. RESULTS We show that neutralization titers for HSV-2, determined by the 96-well MNA correlate with titers determined by a PRNT completed in 24-well plates in both the absence (R2 = 0.8250) and presence (R2 = 0.7075) of complement. CONCLUSIONS We have successfully developed an MNA that can be used in place of the burdensome PRNT to determine anti-HSV-2 neutralizing activity in serum. This MNA has much greater throughput than the PRNT, allowing many more samples to be processed in a shorter time saving ∼90 % of the time required by the laboratory scientist to complete the task as compared to the traditional PRNT.
Collapse
Affiliation(s)
- Melanie S Horton
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, PA, USA.
| | | | - Scott Cosmi
- Eurofins Lancaster Laboratories Professional Scientific Service, Lancaster, PA, USA
| | - Kara Cox
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, PA, USA
| | - Jennifer Galli
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, PA, USA
| | - Jessica Peters
- Eurofins Lancaster Laboratories Professional Scientific Service, Lancaster, PA, USA
| | - Nicole Sullivan
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, PA, USA
| | - Brian Squadroni
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, PA, USA
| | - Aimin Tang
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, PA, USA
| | - Arthur Fridman
- Scientific Informatics, Merck & Co., Inc., Rahway, NJ, USA
| | - Dai Wang
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, PA, USA
| | - Zhifeng Chen
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, PA, USA
| | - Kalpit A Vora
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, PA, USA
| |
Collapse
|
7
|
Kozel BA, Barak B, Ae Kim C, Mervis CB, Osborne LR, Porter M, Pober BR. Williams syndrome. Nat Rev Dis Primers 2021; 7:42. [PMID: 34140529 PMCID: PMC9437774 DOI: 10.1038/s41572-021-00276-z] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/13/2021] [Indexed: 11/09/2022]
Abstract
Williams syndrome (WS) is a relatively rare microdeletion disorder that occurs in as many as 1:7,500 individuals. WS arises due to the mispairing of low-copy DNA repetitive elements at meiosis. The deletion size is similar across most individuals with WS and leads to the loss of one copy of 25-27 genes on chromosome 7q11.23. The resulting unique disorder affects multiple systems, with cardinal features including but not limited to cardiovascular disease (characteristically stenosis of the great arteries and most notably supravalvar aortic stenosis), a distinctive craniofacial appearance, and a specific cognitive and behavioural profile that includes intellectual disability and hypersociability. Genotype-phenotype evidence is strongest for ELN, the gene encoding elastin, which is responsible for the vascular and connective tissue features of WS, and for the transcription factor genes GTF2I and GTF2IRD1, which are known to affect intellectual ability, social functioning and anxiety. Mounting evidence also ascribes phenotypic consequences to the deletion of BAZ1B, LIMK1, STX1A and MLXIPL, but more work is needed to understand the mechanism by which these deletions contribute to clinical outcomes. The age of diagnosis has fallen in regions of the world where technological advances, such as chromosomal microarray, enable clinicians to make the diagnosis of WS without formally suspecting it, allowing earlier intervention by medical and developmental specialists. Phenotypic variability is considerable for all cardinal features of WS but the specific sources of this variability remain unknown. Further investigation to identify the factors responsible for these differences may lead to mechanism-based rather than symptom-based therapies and should therefore be a high research priority.
Collapse
Affiliation(s)
- Beth A. Kozel
- Translational Vascular Medicine Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, USA
| | - Boaz Barak
- The Sagol School of Neuroscience and The School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Chong Ae Kim
- Department of Pediatrics, Universidade de São Paulo, São Paulo, Brazil
| | - Carolyn B. Mervis
- Department of Psychological and Brain Sciences, University of Louisville, Louisville, USA
| | - Lucy R. Osborne
- Department of Medicine, University of Toronto, Ontario, Canada
| | - Melanie Porter
- Department of Psychology, Macquarie University, Sydney, Australia
| | - Barbara R. Pober
- Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| |
Collapse
|
8
|
Characterization of the Herpes Simplex Virus (HSV) Tegument Proteins That Bind to gE/gI and US9, Which Promote Assembly of HSV and Transport into Neuronal Axons. J Virol 2020; 94:JVI.01113-20. [PMID: 32938770 DOI: 10.1128/jvi.01113-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/11/2020] [Indexed: 01/14/2023] Open
Abstract
The herpes simplex virus (HSV) heterodimer gE/gI and another membrane protein, US9, which has neuron-specific effects, promote the anterograde transport of virus particles in neuronal axons. Deletion of both HSV gE and US9 blocks the assembly of enveloped particles in the neuronal cytoplasm, which explains why HSV virions do not enter axons. Cytoplasmic envelopment depends upon interactions between viral membrane proteins and tegument proteins that encrust capsids. We report that tegument protein UL16 is unstable, i.e., rapidly degraded, in neurons infected with a gE-/US9- double mutant. Immunoprecipitation experiments with lysates of HSV-infected neurons showed that UL16 and three other tegument proteins, namely, VP22, UL11, and UL21, bound either to gE or gI. All four of these tegument proteins were also pulled down with US9. In neurons transfected with tegument proteins and gE/gI or US9, there was good evidence that VP22 and UL16 bound directly to US9 and gE/gI. However, there were lower quantities of these tegument proteins that coprecipitated with gE/gI and US9 from transfected cells than those of infected cells. This apparently relates to a matrix of several different tegument proteins formed in infected cells that bind to gE/gI and US9. In cells transfected with individual tegument proteins, this matrix is less prevalent. Similarly, coprecipitation of gE/gI and US9 was observed in HSV-infected cells but not in transfected cells, which argued against direct US9-gE/gI interactions. These studies suggest that gE/gI and US9 binding to these tegument proteins has neuron-specific effects on virus HSV assembly, a process required for axonal transport of enveloped particles.IMPORTANCE Herpes simplex viruses 1 and 2 and varicella-zoster virus cause significant morbidity and mortality. One basic property of these viruses is the capacity to establish latency in the sensory neurons and to reactivate from latency and then cause disease in peripheral tissues, such as skin and mucosal epithelia. The transport of nascent HSV particles from neuron cell bodies into axons and along axons to axon tips in the periphery is an important component of this reactivation and reinfection. Two HSV membrane proteins, gE/gI and US9, play an essential role in these processes. Our studies help elucidate how HSV gE/gI and US9 promote the assembly of virus particles and sorting of these virions into neuronal axons.
Collapse
|
9
|
Hoang HD, Graber TE, Jia JJ, Vaidya N, Gilchrist VH, Xiang X, Li W, Cowan KN, Gkogkas CG, Jaramillo M, Jafarnejad SM, Alain T. Induction of an Alternative mRNA 5' Leader Enhances Translation of the Ciliopathy Gene Inpp5e and Resistance to Oncolytic Virus Infection. Cell Rep 2020; 29:4010-4023.e5. [PMID: 31851930 DOI: 10.1016/j.celrep.2019.11.072] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 10/16/2019] [Accepted: 11/15/2019] [Indexed: 01/10/2023] Open
Abstract
Residual cell-intrinsic innate immunity in cancer cells hampers infection with oncolytic viruses. Translational control of mRNA is an important feature of innate immunity, yet the identity of translationally regulated mRNAs functioning in host defense remains ill-defined. We report the translatomes of resistant murine "4T1" breast cancer cells infected with three of the most clinically advanced oncolytic viruses: herpes simplex virus 1, reovirus, and vaccinia virus. Common among all three infections are translationally de-repressed mRNAs, including Inpp5e, encoding an inositol 5-phosphatase that modifies lipid second messenger signaling. We find that viral infection induces the expression of an Inpp5e mRNA variant that lacks repressive upstream open reading frames (uORFs) within its 5' leader and is efficiently translated. Furthermore, we show that INPP5E contributes to antiviral immunity by altering virus attachment. These findings uncover a role for translational control through alternative 5' leader expression and assign an antiviral function to the ciliopathy gene Inpp5e.
Collapse
Affiliation(s)
- Huy-Dung Hoang
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada; Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Tyson E Graber
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada; Department of Biochemistry and Goodman Cancer Center, McGill University, Montreal, QC H3A 1A3, Canada
| | - Jian-Jun Jia
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
| | - Nasana Vaidya
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
| | - Victoria H Gilchrist
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada; Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Xiao Xiang
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Wencheng Li
- Department of Biochemistry and Molecular Biology, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| | - Kyle N Cowan
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Department of Surgery, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Christos G Gkogkas
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Maritza Jaramillo
- INRS Institut Armand-Frappier Research Centre, Laval, QC H7V 1B7, Canada
| | - Seyed Mehdi Jafarnejad
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Tommy Alain
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada; Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
10
|
Rocha JN, Dangott LJ, Mwangi W, Alaniz RC, Bordin AI, Cywes-Bentley C, Lawhon SD, Pillai SD, Bray JM, Pier GB, Cohen ND. PNAG-specific equine IgG 1 mediates significantly greater opsonization and killing of Prescottella equi (formerly Rhodococcus equi) than does IgG 4/7. Vaccine 2019; 37:1142-1150. [PMID: 30691984 PMCID: PMC8314964 DOI: 10.1016/j.vaccine.2019.01.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 02/04/2023]
Abstract
Prescottella equi (formerly Rhodococcus equi) is a facultative intracellular bacterial pathogen that causes severe pneumonia in foals 1-6 months of age, whereas adult horses are highly resistant to infection. We have shown that vaccinating pregnant mares against the conserved surface polysaccharide capsule, β-1 → 6-linked poly-N-acetyl glucosamine (PNAG), elicits opsonic killing antibody that transfers via colostrum to foals and protects them against experimental infection with virulent. R. equi. We hypothesized that equine IgG1 might be more important than IgG4/7 for mediating protection against R. equi infection in foals. To test this hypothesis, we compared complement component 1 (C1) deposition and polymorphonuclear cell-mediated opsonophagocytic killing (OPK) mediated by IgG1 or IgG4/7 enriched from either PNAG hyperimmune plasma (HIP) or standard plasma. Subclasses IgG1 and IgG4/7 from PNAG HIP and standard plasma were precipitated onto a diethylaminoethyl ion exchange column, then further enriched using a protein G Sepharose column. We determined C1 deposition by enzyme-linked immunosorbent assay (ELISA) and estimated OPK by quantitative microbiologic culture. Anti-PNAG IgG1 deposited significantly (P < 0.05) more C1 onto PNAG than did IgG4/7 from PNAG HIP or subclasses IgG1 and IgG4/7 from standard plasma. In addition, IgG1 from PNAG HIP mediated significantly (P < 0.05) greater OPK than IgG4/7 from PNAG HIP or IgG1 and IgG4/7 from standard plasma. Our findings indicate that anti-PNAG IgG1 is a correlate of protection against R. equi in foals, which has important implications for understanding the immunopathogenesis of R. equi pneumonia, and as a tool for assessing vaccine efficacy and effectiveness when challenge is not feasible.
Collapse
Affiliation(s)
- Joana N Rocha
- College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, 660 Raymond Stotzer Pkwy, College Station, TX 77843-4475, United States.
| | - Lawrence J Dangott
- Protein Chemistry Laboratory, Texas A&M University, 300 Olsen Blvd, College Station, TX 77843, United States.
| | - Waithaka Mwangi
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, 1800 Denison Ave, Manhattan, KS 66506, United States.
| | - Robert C Alaniz
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health and Science Center, 206 Olsen Blvd, College Station, TX 77845, United States.
| | - Angela I Bordin
- College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, 660 Raymond Stotzer Pkwy, College Station, TX 77843-4475, United States.
| | - Colette Cywes-Bentley
- Harvard Medical School, Brigham & Women's Hospital, 181 Longwood Ave, Boston, MA 02115, United States.
| | - Sara D Lawhon
- College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, 660 Raymond Stotzer Pkwy, College Station, TX 77843-4475, United States.
| | - Suresh D Pillai
- National Center for Electron Beam Research-IAEA Collaborative Centre for Electron Beam Technology, Texas A&M University, 400 Discovery Dr, College Station, TX 77845, United States.
| | - Jocelyne M Bray
- College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, 660 Raymond Stotzer Pkwy, College Station, TX 77843-4475, United States
| | - Gerald B Pier
- Harvard Medical School, Brigham & Women's Hospital, 181 Longwood Ave, Boston, MA 02115, United States.
| | - Noah D Cohen
- College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, 660 Raymond Stotzer Pkwy, College Station, TX 77843-4475, United States.
| |
Collapse
|
11
|
Alt M, Falk J, Eis-Hübinger AM, Kropff B, Sinzger C, Krawczyk A. Detection of antibody-secreting cells specific for the cytomegalovirus and herpes simplex virus surface antigens. J Immunol Methods 2018; 462:13-22. [PMID: 30056033 PMCID: PMC7094464 DOI: 10.1016/j.jim.2018.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 06/29/2018] [Accepted: 07/25/2018] [Indexed: 11/24/2022]
Abstract
Infections with the herpes simplex virus (HSV) and the human cytomegalovirus (HCMV) can lead to life-threatening diseases, particularly in immunosuppressed patients. Furthermore, HSV infections at birth (herpes neonatorum) can result in a disseminated disease associated with a fatal multiorgan failure. Congenital HCMV infections can result in miscarriage, serious birth defects or developmental disabilities. Antibody-based interventions with hyperimmunoglobulins showed encouraging results in clinical studies, but clearly need to be improved. The isolation of highly neutralizing monoclonal antibodies is a promising strategy to establish potent therapy options against HSV and HCMV infections. Monoclonal antibodies are commonly isolated from hybridomas or EBV-immortalized B-cell clones. The screening procedure to identify virus-specific cells from a cell mixture is a challenging step, since most of the highly neutralizing antibodies target complex conformational epitopes on the virus surface. Conventional assays such as ELISA are based on purified viral proteins and inappropriate to display complex epitopes. To overcome this obstacle, we have established two full-virus based methods that allow screening for cells and antibodies targeting complex conformational epitopes on viral surface antigens. The methods are suitable to detect surface antigen-specific cells from a cell mixture and may facilitate the isolation of highly neutralizing antibodies against HSV and HCMV.
Collapse
Affiliation(s)
- Mira Alt
- Institute for Virology, University Hospital of Essen, 45147 Essen, Germany
| | - Jessica Falk
- Institute for Virology, University Hospital of Ulm, 89081 Ulm, Germany
| | | | - Barbara Kropff
- Institute of Clinical and Molecular Virology, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | - Christian Sinzger
- Institute for Virology, University Hospital of Ulm, 89081 Ulm, Germany
| | - Adalbert Krawczyk
- Institute for Virology, University Hospital of Essen, 45147 Essen, Germany.
| |
Collapse
|
12
|
Maternal Antiviral Immunoglobulin Accumulates in Neural Tissue of Neonates To Prevent HSV Neurological Disease. mBio 2017; 8:mBio.00678-17. [PMID: 28679745 PMCID: PMC5573671 DOI: 10.1128/mbio.00678-17] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
While antibody responses to neurovirulent pathogens are critical for clearance, the extent to which antibodies access the nervous system to ameliorate infection is poorly understood. In this study on herpes simplex virus 1 (HSV-1), we demonstrate that HSV-specific antibodies are present during HSV-1 latency in the nervous systems of both mice and humans. We show that antibody-secreting cells entered the trigeminal ganglion (TG), a key site of HSV infection, and persisted long after the establishment of latent infection. We also demonstrate the ability of passively administered IgG to enter the TG independently of infection, showing that the naive TG is accessible to antibodies. The translational implication of this finding is that human fetal neural tissue could contain HSV-specific maternally derived antibodies. Exploring this possibility, we observed HSV-specific IgG in HSV DNA-negative human fetal TG, suggesting passive transfer of maternal immunity into the prenatal nervous system. To further investigate the role of maternal antibodies in the neonatal nervous system, we established a murine model to demonstrate that maternal IgG can access and persist in neonatal TG. This maternal antibody not only prevented disseminated infection but also completely protected the neonate from neurological disease and death following HSV challenge. Maternal antibodies therefore have a potent protective role in the neonatal nervous system against HSV infection. These findings strongly support the concept that prevention of prenatal and neonatal neurotropic infections can be achieved through maternal immunization. Herpes simplex virus 1 is a common infection of the nervous system that causes devastating neonatal disease. Using mouse and human tissue, we discovered that antiviral antibodies accumulate in neural tissue after HSV-1 infection in adults. Similarly, these antibodies pass to the offspring during pregnancy. We found that antiviral maternal antibodies can readily access neural tissue of the fetus and neonate. These maternal antibodies then protect neonatal mice against HSV-1 neurological infection and death. These results underscore the previously unappreciated role of maternal antibodies in protecting fetal and newborn nervous systems against infection. These data suggest that maternal immunization would be efficacious at preventing fetal/neonatal neurological infections.
Collapse
|
13
|
Awasthi S, Huang J, Shaw C, Friedman HM. Blocking herpes simplex virus 2 glycoprotein E immune evasion as an approach to enhance efficacy of a trivalent subunit antigen vaccine for genital herpes. J Virol 2014; 88:8421-8432. [PMID: 24829358 PMCID: PMC4135967 DOI: 10.1128/jvi.01130-14] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Accepted: 05/07/2014] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Herpes simplex virus 2 (HSV-2) subunit antigen vaccines targeting virus entry molecules have failed to prevent genital herpes in human trials. Our approach is to include a virus entry molecule and add antigens that block HSV-2 immune evasion. HSV-2 glycoprotein C (gC2) is an immune evasion molecule that inhibits complement. We previously reported that adding gC2 to gD2 improved vaccine efficacy compared to the efficacy of either antigen alone in mice and guinea pigs. Here we demonstrate that HSV-2 glycoprotein E (gE2) functions as an immune evasion molecule by binding the IgG Fc domain. HSV-2 gE2 is synergistic with gC2 in protecting the virus from antibody and complement neutralization. Antibodies produced by immunization with gE2 blocked gE2-mediated IgG Fc binding and cell-to-cell spread. Mice immunized with gE2 were only partially protected against HSV-2 vaginal challenge in mice; however, when gE2 was added to gC2/gD2 to form a trivalent vaccine, neutralizing antibody titers with and without complement were significantly higher than those produced by gD2 alone. Importantly, the trivalent vaccine protected the dorsal root ganglia (DRG) of 32/33 (97%) mice between days 2 and 7 postchallenge, compared with 27/33 (82%) in the gD2 group. The HSV-2 DNA copy number was significantly lower in mice immunized with the trivalent vaccine than in those immunized with gD2 alone. The extent of DRG protection using the trivalent vaccine was better than what we previously reported for gC2/gD2 immunization. Therefore, gE2 is a candidate antigen for inclusion in a multivalent subunit vaccine that attempts to block HSV-2 immune evasion. IMPORTANCE Herpes simplex virus is the most common cause of genital ulcer disease worldwide. Infection results in emotional distress for infected individuals and their partners, is life threatening for infants exposed to herpes during childbirth, and greatly increases the risk of individuals acquiring and transmitting HIV infection. A vaccine that prevents genital herpes infection will have major public health benefits. Our vaccine approach includes strategies to prevent the virus from evading immune attack. Mice were immunized with a trivalent vaccine containing an antigen that induces antibodies to block virus entry and two antigens that induce antibodies that block immune evasion from antibody and complement. Immunized mice demonstrated no genital disease, and 32/33 (97%) animals had no evidence of infection of dorsal root ganglia, suggesting that the vaccine may prevent the establishment of latency and recurrent infections.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- DNA, Viral/analysis
- DNA, Viral/genetics
- Disease Models, Animal
- Female
- Herpes Genitalis/immunology
- Herpes Genitalis/prevention & control
- Herpesvirus 2, Human/immunology
- Herpesvirus Vaccines/administration & dosage
- Herpesvirus Vaccines/genetics
- Herpesvirus Vaccines/immunology
- Immune Evasion
- Mice
- Mice, Inbred BALB C
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/genetics
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Viral Envelope Proteins/immunology
- Viral Envelope Proteins/metabolism
- Viral Load
Collapse
Affiliation(s)
- Sita Awasthi
- Infectious Disease Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jialing Huang
- Infectious Disease Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Carolyn Shaw
- Infectious Disease Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Harvey M Friedman
- Infectious Disease Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
14
|
The herpes virus Fc receptor gE-gI mediates antibody bipolar bridging to clear viral antigens from the cell surface. PLoS Pathog 2014; 10:e1003961. [PMID: 24604090 PMCID: PMC3946383 DOI: 10.1371/journal.ppat.1003961] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 01/16/2014] [Indexed: 11/19/2022] Open
Abstract
The Herpes Simplex Virus 1 (HSV-1) glycoprotein gE-gI is a transmembrane Fc receptor found on the surface of infected cells and virions that binds human immunoglobulin G (hIgG). gE-gI can also participate in antibody bipolar bridging (ABB), a process by which the antigen-binding fragments (Fabs) of the IgG bind a viral antigen while the Fc binds to gE-gI. IgG Fc binds gE-gI at basic, but not acidic, pH, suggesting that IgG bound at extracellular pH by cell surface gE-gI would dissociate and be degraded in acidic endosomes/lysosomes if endocytosed. The fate of viral antigens associated with gE-gI-bound IgG had been unknown: they could remain at the cell surface or be endocytosed with IgG. Here, we developed an in vitro model system for ABB and investigated the trafficking of ABB complexes using 4-D confocal fluorescence imaging of ABB complexes with transferrin or epidermal growth factor, well-characterized intracellular trafficking markers. Our data showed that cells expressing gE-gI and the viral antigen HSV-1 gD endocytosed anti-gD IgG and gD in a gE-gI-dependent process, resulting in lysosomal localization. These results suggest that gE-gI can mediate clearance of infected cell surfaces of anti-viral host IgG and viral antigens to evade IgG-mediated responses, representing a general mechanism for viral Fc receptors in immune evasion and viral pathogenesis.
Collapse
|
15
|
Lin AE, Greco TM, Döhner K, Sodeik B, Cristea IM. A proteomic perspective of inbuilt viral protein regulation: pUL46 tegument protein is targeted for degradation by ICP0 during herpes simplex virus type 1 infection. Mol Cell Proteomics 2013; 12:3237-52. [PMID: 23938468 DOI: 10.1074/mcp.m113.030866] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Much like the host cells they infect, viruses must also regulate their life cycles. Herpes simples virus type 1 (HSV-1), a prominent human pathogen, uses a promoter-rich genome in conjunction with multiple viral trans-activating factors. Following entry into host cells, the virion-associated outer tegument proteins pUL46 and pUL47 act to increase expression of viral immediate-early (α) genes, thereby helping initiate the infection life cycle. Because pUL46 has gone largely unstudied, we employed a hybrid mass spectrometry-based approach to determine how pUL46 exerts its functions during early stages of infection. For a spatio-temporal characterization of pUL46, time-lapse microscopy was performed in live cells to define its dynamic localization from 2 to 24 h postinfection. Next, pUL46-containing protein complexes were immunoaffinity purified during infection of human fibroblasts and analyzed by mass spectrometry to investigate virus-virus and virus-host interactions, as well as post-translational modifications. We demonstrated that pUL46 is heavily phosphorylated in at least 23 sites. One phosphorylation site matched the consensus 14-3-3 phospho-binding motif, consistent with our identification of 14-3-3 proteins and host and viral kinases as specific pUL46 interactions. Moreover, we determined that pUL46 specifically interacts with the viral E3 ubiquitin ligase ICP0. We demonstrated that pUL46 is partially degraded in a proteasome-mediated manner during infection, and that the catalytic activity of ICP0 is responsible for this degradation. This is the first evidence of a viral protein being targeted for degradation by another viral protein during HSV-1 infection. Together, these data indicate that pUL46 levels are tightly controlled and important for the temporal regulation of viral gene expression throughout the virus life cycle. The concept of a structural virion protein, pUL46, performing nonstructural roles is likely to reflect a theme common to many viruses, and a better understanding of these functions will be important for developing therapeutics.
Collapse
Affiliation(s)
- Aaron E Lin
- Department of Molecular Biology, Princeton University, Princeton, New Jersey
| | | | | | | | | |
Collapse
|
16
|
Abstract
HSV infections are prevalent worldwide. A vaccine to prevent genital herpes would have a significant impact on this disease. Several vaccines have shown promise in animal models; however, so far these have not been successful in human clinical studies. Prophylactic HSV vaccines to prevent HSV infection or disease have focused primarily on eliciting antibody responses. Potent antibody responses are needed to result in sufficiently high levels of virus-specific antibody in the genital tract. Therapeutic vaccines that reduce recurrences need to induce potent T-cell responses at the site of infection. With the increasing incidence of HSV-1 genital herpes, an effective herpes vaccine should protect against both HSV-1 and HSV-2. Novel HSV vaccines, such as replication-defective or attenuated viruses, have elicited humoral and cellular immune responses in preclinical studies. These vaccines and others hold promise in future clinical studies.
Collapse
Affiliation(s)
- Lesia K Dropulic
- Medical Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | | |
Collapse
|
17
|
Pelli A, Castellano LR, Cardoso MRS, Vasconcelos LAS, Domingues MA, Ferreira MB, Rodrigues V. Differential reactivity of serum immunoglobulins from Brazilian wild mammals to staphylococcal A and streptococcal G proteins. J Vet Diagn Invest 2012; 24:148-52. [DOI: 10.1177/1040638711434322] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Human pathogens have evolved to infect vertebrate hosts other than human beings without causing symptoms of the disease, thus permitting them to complete their life cycle and to develop into infectious forms. The identification and management of infected animals are alternatives to control dissemination of the disease and to prevent human illness. In the current study, the potential use of staphylococcal A or streptococcal G proteins was evaluated with enzyme-linked immunosorbent assays (ELISAs) for seroepidemiological studies. Sera were collected from animals that were representative of 23 different Brazilian wild mammals. A high protein A binding rate was observed in all animals, except for the orders Didelphimorphia, Artiodactyla, and Rodentia, in which affinity was medium or low. Affinity for streptococcal G protein was higher in animals of the order Artiodactyla, whereas no streptococcal G protein binding was observed in samples obtained from felines (order Carnivora). Bacterial protein binding to mammalian immunoglobulins was confirmed by immunoblotting. The results suggest that secondary detection systems should be better investigated in ELISA protocols before their implementation in seroepidemiological studies involving wild mammals.
Collapse
Affiliation(s)
- Afonso Pelli
- Department of Biological Sciences, Triângulo Mineiro Federal University, Minas Gerais, Brazil (Pelli, Castellano, Rodrigues), Jacarandá City Zoo Park, Minas Gerais, Brazil (Domingues)
- Department of Technical School of Health, Paraíba Federal University, Paraíba, Brazil (Castellano), Jacarandá City Zoo Park, Minas Gerais, Brazil (Domingues)
- Department of Furnas Central Electric S.A., Mato Grosso, Brazil (Cardoso, Vasconcelos), Jacarandá City Zoo Park, Minas Gerais, Brazil (Domingues)
- Companhia Energética de Minas Gerais S.A., Minas Gerais, Brazil (Ferreira)
| | - Lucio R. Castellano
- Department of Biological Sciences, Triângulo Mineiro Federal University, Minas Gerais, Brazil (Pelli, Castellano, Rodrigues), Jacarandá City Zoo Park, Minas Gerais, Brazil (Domingues)
- Department of Technical School of Health, Paraíba Federal University, Paraíba, Brazil (Castellano), Jacarandá City Zoo Park, Minas Gerais, Brazil (Domingues)
- Department of Furnas Central Electric S.A., Mato Grosso, Brazil (Cardoso, Vasconcelos), Jacarandá City Zoo Park, Minas Gerais, Brazil (Domingues)
- Companhia Energética de Minas Gerais S.A., Minas Gerais, Brazil (Ferreira)
| | - Marcos R. S. Cardoso
- Department of Biological Sciences, Triângulo Mineiro Federal University, Minas Gerais, Brazil (Pelli, Castellano, Rodrigues), Jacarandá City Zoo Park, Minas Gerais, Brazil (Domingues)
- Department of Technical School of Health, Paraíba Federal University, Paraíba, Brazil (Castellano), Jacarandá City Zoo Park, Minas Gerais, Brazil (Domingues)
- Department of Furnas Central Electric S.A., Mato Grosso, Brazil (Cardoso, Vasconcelos), Jacarandá City Zoo Park, Minas Gerais, Brazil (Domingues)
- Companhia Energética de Minas Gerais S.A., Minas Gerais, Brazil (Ferreira)
| | - Luís A. S. Vasconcelos
- Department of Biological Sciences, Triângulo Mineiro Federal University, Minas Gerais, Brazil (Pelli, Castellano, Rodrigues), Jacarandá City Zoo Park, Minas Gerais, Brazil (Domingues)
- Department of Technical School of Health, Paraíba Federal University, Paraíba, Brazil (Castellano), Jacarandá City Zoo Park, Minas Gerais, Brazil (Domingues)
- Department of Furnas Central Electric S.A., Mato Grosso, Brazil (Cardoso, Vasconcelos), Jacarandá City Zoo Park, Minas Gerais, Brazil (Domingues)
- Companhia Energética de Minas Gerais S.A., Minas Gerais, Brazil (Ferreira)
| | - Marcos A. Domingues
- Department of Biological Sciences, Triângulo Mineiro Federal University, Minas Gerais, Brazil (Pelli, Castellano, Rodrigues), Jacarandá City Zoo Park, Minas Gerais, Brazil (Domingues)
- Department of Technical School of Health, Paraíba Federal University, Paraíba, Brazil (Castellano), Jacarandá City Zoo Park, Minas Gerais, Brazil (Domingues)
- Department of Furnas Central Electric S.A., Mato Grosso, Brazil (Cardoso, Vasconcelos), Jacarandá City Zoo Park, Minas Gerais, Brazil (Domingues)
- Companhia Energética de Minas Gerais S.A., Minas Gerais, Brazil (Ferreira)
| | - Maria B. Ferreira
- Department of Biological Sciences, Triângulo Mineiro Federal University, Minas Gerais, Brazil (Pelli, Castellano, Rodrigues), Jacarandá City Zoo Park, Minas Gerais, Brazil (Domingues)
- Department of Technical School of Health, Paraíba Federal University, Paraíba, Brazil (Castellano), Jacarandá City Zoo Park, Minas Gerais, Brazil (Domingues)
- Department of Furnas Central Electric S.A., Mato Grosso, Brazil (Cardoso, Vasconcelos), Jacarandá City Zoo Park, Minas Gerais, Brazil (Domingues)
- Companhia Energética de Minas Gerais S.A., Minas Gerais, Brazil (Ferreira)
| | - Virmondes Rodrigues
- Department of Biological Sciences, Triângulo Mineiro Federal University, Minas Gerais, Brazil (Pelli, Castellano, Rodrigues), Jacarandá City Zoo Park, Minas Gerais, Brazil (Domingues)
- Department of Technical School of Health, Paraíba Federal University, Paraíba, Brazil (Castellano), Jacarandá City Zoo Park, Minas Gerais, Brazil (Domingues)
- Department of Furnas Central Electric S.A., Mato Grosso, Brazil (Cardoso, Vasconcelos), Jacarandá City Zoo Park, Minas Gerais, Brazil (Domingues)
- Companhia Energética de Minas Gerais S.A., Minas Gerais, Brazil (Ferreira)
| |
Collapse
|
18
|
MacLellan LM, Montgomery J, Sugiyama F, Kitson SM, Thümmler K, Silverman GJ, Beers SA, Nibbs RJB, McInnes IB, Goodyear CS. Co-opting endogenous immunoglobulin for the regulation of inflammation and osteoclastogenesis in humans and mice. ACTA ACUST UNITED AC 2012; 63:3897-907. [PMID: 22127707 DOI: 10.1002/art.30629] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Cells of the monocytic lineage play fundamental roles in the regulation of health, ranging from the initiation and resolution of inflammation to bone homeostasis. In rheumatoid arthritis (RA), the inflamed synovium exhibits characteristic infiltration of macrophages along with local osteoclast maturation, which, together, drive chronic inflammation and downstream articular destruction. The aim of this study was to explore an entirely novel route of immunoglobulin-mediated regulation, involving simultaneous suppression of the inflammatory and erosive processes in the synovium. METHODS Using in vivo and in vitro studies of human cells and a murine model of RA, the ability of staphylococcal protein A (SPA) to interact with and modulate cells of the monocytic lineage was tested. In addition, the efficacy of SPA as a therapeutic agent was evaluated in murine collagen-induced arthritis (CIA). RESULTS SPA showed a capacity to appropriate circulating IgG, by generating small immunoglobulin complexes that interacted with monocytes, macrophages, and preosteoclasts. Formation of these complexes resulted in Fcγ receptor type I-dependent polarization of macrophages to a regulatory phenotype, rendering them unresponsive to activators such as interferon-γ. The antiinflammatory complexes also had the capacity to directly inhibit differentiation of preosteoclasts into osteoclasts in humans. Moreover, administration of SPA in the early stages of disease substantially alleviated the clinical and histologic erosive features of CIA in mice. CONCLUSION These findings demonstrate the overarching utility of immunoglobulin complexes for the prevention and treatment of inflammatory diseases. The results shed light on the interface between immunoglobulin complex-mediated pathways, osteoclastogenesis, and associated pathologic processes. Thus, therapeutic agents designed to harness all of these properties may be an effective treatment for arthritis, by targeting both the innate inflammatory response and prodestructive pathways.
Collapse
MESH Headings
- Animals
- Antigen-Antibody Complex/pharmacology
- Antigen-Antibody Complex/therapeutic use
- Antirheumatic Agents/pharmacology
- Antirheumatic Agents/therapeutic use
- Arthritis, Experimental/drug therapy
- Arthritis, Experimental/immunology
- Arthritis, Experimental/physiopathology
- Cell Differentiation/physiology
- Cell Proliferation
- Cells, Cultured
- Cytokines/physiology
- Disease Models, Animal
- Humans
- Immunoglobulins/physiology
- Immunoglobulins/therapeutic use
- Inflammation/drug therapy
- Inflammation/physiopathology
- Leukocytes, Mononuclear/cytology
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/physiology
- Macrophages/cytology
- Macrophages/drug effects
- Macrophages/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Knockout
- Osteoclasts/cytology
- Osteoclasts/drug effects
- Osteoclasts/physiology
- Receptors, IgG/genetics
- Receptors, IgG/physiology
- Staphylococcal Protein A/pharmacology
- Staphylococcal Protein A/therapeutic use
- Stem Cells/cytology
- Stem Cells/drug effects
- Stem Cells/physiology
Collapse
|
19
|
Immunization with a vaccine combining herpes simplex virus 2 (HSV-2) glycoprotein C (gC) and gD subunits improves the protection of dorsal root ganglia in mice and reduces the frequency of recurrent vaginal shedding of HSV-2 DNA in guinea pigs compared to immunization with gD alone. J Virol 2011; 85:10472-86. [PMID: 21813597 DOI: 10.1128/jvi.00849-11] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Attempts to develop a vaccine to prevent genital herpes simplex virus 2 (HSV-2) disease have been only marginally successful, suggesting that novel strategies are needed. Immunization with HSV-2 glycoprotein C (gC-2) and gD-2 was evaluated in mice and guinea pigs to determine whether adding gC-2 to a gD-2 subunit vaccine would improve protection by producing antibodies that block gC-2 immune evasion from complement. Antibodies produced by gC-2 immunization blocked the interaction between gC-2 and complement C3b, and passive transfer of gC-2 antibody protected complement-intact mice but not C3 knockout mice against HSV-2 challenge, indicating that gC-2 antibody is effective, at least in part, because it prevents HSV-2 evasion from complement. Immunization with gC-2 also produced neutralizing antibodies that were active in the absence of complement; however, the neutralizing titers were higher when complement was present, with the highest titers in animals immunized with both antigens. Animals immunized with the gC-2-plus-gD-2 combination had robust CD4+ T-cell responses to each immunogen. Multiple disease parameters were evaluated in mice and guinea pigs immunized with gC-2 alone, gD-2 alone, or both antigens. In general, gD-2 outperformed gC-2; however, the gC-2-plus-gD-2 combination outperformed gD-2 alone, particularly in protecting dorsal root ganglia in mice and reducing recurrent vaginal shedding of HSV-2 DNA in guinea pigs. Therefore, the gC-2 subunit antigen enhances a gD-2 subunit vaccine by stimulating a CD4+ T-cell response, by producing neutralizing antibodies that are effective in the absence and presence of complement, and by blocking immune evasion domains that inhibit complement activation.
Collapse
|
20
|
Cheng SB, Ferland P, Webster P, Bearer EL. Herpes simplex virus dances with amyloid precursor protein while exiting the cell. PLoS One 2011; 6:e17966. [PMID: 21483850 PMCID: PMC3069030 DOI: 10.1371/journal.pone.0017966] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2010] [Accepted: 02/19/2011] [Indexed: 11/18/2022] Open
Abstract
Herpes simplex type 1 (HSV1) replicates in epithelial cells and secondarily enters local sensory neuronal processes, traveling retrograde to the neuronal nucleus to enter latency. Upon reawakening newly synthesized viral particles travel anterograde back to the epithelial cells of the lip, causing the recurrent cold sore. HSV1 co-purifies with amyloid precursor protein (APP), a cellular transmembrane glycoprotein and receptor for anterograde transport machinery that when proteolyzed produces A-beta, the major component of senile plaques. Here we focus on transport inside epithelial cells of newly synthesized virus during its transit to the cell surface. We hypothesize that HSV1 recruits cellular APP during transport. We explore this with quantitative immuno-fluorescence, immuno-gold electron-microscopy and live cell confocal imaging. After synchronous infection most nascent VP26-GFP-labeled viral particles in the cytoplasm co-localize with APP (72.8+/−6.7%) and travel together with APP inside living cells (81.1+/−28.9%). This interaction has functional consequences: HSV1 infection decreases the average velocity of APP particles (from 1.1+/−0.2 to 0.3+/−0.1 µm/s) and results in APP mal-distribution in infected cells, while interplay with APP-particles increases the frequency (from 10% to 81% motile) and velocity (from 0.3+/−0.1 to 0.4+/−0.1 µm/s) of VP26-GFP transport. In cells infected with HSV1 lacking the viral Fc receptor, gE, an envelope glycoprotein also involved in viral axonal transport, APP-capsid interactions are preserved while the distribution and dynamics of dual-label particles differ from wild-type by both immuno-fluorescence and live imaging. Knock-down of APP with siRNA eliminates APP staining, confirming specificity. Our results indicate that most intracellular HSV1 particles undergo frequent dynamic interplay with APP in a manner that facilitates viral transport and interferes with normal APP transport and distribution. Such dynamic interactions between APP and HSV1 suggest a mechanistic basis for the observed clinical relationship between HSV1 seropositivity and risk of Alzheimer's disease.
Collapse
Affiliation(s)
- Shi-Bin Cheng
- Department of Pathology and Laboratory Medicine, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Paulette Ferland
- Department of Pathology and Laboratory Medicine, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Paul Webster
- House Ear Institute, Los Angeles, California, United States of America
| | - Elaine L. Bearer
- Department of Pathology and Laboratory Medicine, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
- Departments of Pathology and of Neurosurgery, University of New Mexico School of Medicine, Albuquerque, New Mexico, United States of America
- * E-mail:
| |
Collapse
|
21
|
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: 4.2] [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
|
22
|
Identification of rep-associated factors in herpes simplex virus type 1-induced adeno-associated virus type 2 replication compartments. J Virol 2010; 84:8871-87. [PMID: 20573815 DOI: 10.1128/jvi.00725-10] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Adeno-associated virus (AAV) is a human parvovirus that replicates only in cells coinfected with a helper virus, such as adenovirus or herpes simplex virus type 1 (HSV-1). We previously showed that nine HSV-1 factors are able to support AAV rep gene expression and genome replication. To elucidate the strategy of AAV replication in the presence of HSV-1, we undertook a proteomic analysis of cellular and HSV-1 factors associated with Rep proteins and thus potentially recruited within AAV replication compartments (AAV RCs). This study resulted in the identification of approximately 60 cellular proteins, among which factors involved in DNA and RNA metabolism represented the largest functional categories. Validation analyses indicated that the cellular DNA replication enzymes RPA, RFC, and PCNA were recruited within HSV-1-induced AAV RCs. Polymerase delta was not identified but subsequently was shown to colocalize with Rep within AAV RCs even in the presence of the HSV-1 polymerase complex. In addition, we found that AAV replication is associated with the recruitment of components of the Mre11/Rad50/Nbs1 complex, Ku70 and -86, and the mismatch repair proteins MSH2, -3, and -6. Finally, several HSV-1 factors were also found to be associated with Rep, including UL12. We demonstrated for the first time that this protein plays a role during AAV replication by enhancing the resolution of AAV replicative forms and AAV particle production. Altogether, these analyses provide the basis to understand how AAV adapts its replication strategy to the nuclear environment induced by the helper virus.
Collapse
|
23
|
Wright DE, Colaco S, Colaco C, Stevenson PG. Antibody limits in vivo murid herpesvirus-4 replication by IgG Fc receptor-dependent functions. J Gen Virol 2009; 90:2592-2603. [PMID: 19625459 PMCID: PMC2885036 DOI: 10.1099/vir.0.014266-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2009] [Accepted: 07/17/2009] [Indexed: 12/11/2022] Open
Abstract
Antibody is an important antiviral defence. However, it is considered to do little against human gamma-herpesviruses, which establish predominantly latent infections regulated by T cells. One limitation on analysing these infections has been that latency is already well-established at clinical presentation; early infection may still be accessible to antibody. Here, using murid herpesvirus-4 (MuHV-4), we tested the impact of adoptively transferred antibody on early gamma-herpesvirus infection. Immune sera and neutralizing and non-neutralizing monoclonal antibodies (mAbs) all reduced acute lytic MuHV-4 replication. The reductions, even by neutralizing mAbs, were largely or completely dependent on host IgG Fc receptors. Therefore, passive antibody can blunt acute gamma-herpesvirus lytic infection, and does this principally by IgG Fc-dependent functions rather than by neutralization.
Collapse
Affiliation(s)
- Debbie E. Wright
- Division of Virology, Department of Pathology, University of Cambridge, UK
| | - Susanna Colaco
- Division of Virology, Department of Pathology, University of Cambridge, UK
| | - Camilo Colaco
- Immunobiology Ltd, Babraham Research Campus, Cambridge, UK
| | | |
Collapse
|
24
|
Awasthi S, Lubinski JM, Friedman HM. Immunization with HSV-1 glycoprotein C prevents immune evasion from complement and enhances the efficacy of an HSV-1 glycoprotein D subunit vaccine. Vaccine 2009; 27:6845-53. [PMID: 19761834 DOI: 10.1016/j.vaccine.2009.09.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2009] [Revised: 09/01/2009] [Accepted: 09/02/2009] [Indexed: 11/19/2022]
Abstract
Herpes simplex virus type 1 (HSV-1) glycoprotein C (gC-1) binds complement component C3b and inhibits complement-mediated immunity. HSV-1 glycoprotein D (gD-1) is a potent immunogen and a candidate antigen for a subunit vaccine. We evaluated whether combined immunization with gD-1 and gC-1 provides better protection against challenge than gD-1 alone based on antibodies to gC-1 preventing HSV-1-mediated immune evasion. IgG purified from mice immunized with gC-1 blocked C3b binding to gC-1 and greatly increased neutralization by gD-1 IgG in the presence of complement. Passive transfer of gC-1 IgG protected complement intact mice against HSV-1 challenge but not C3 knockout mice, indicating that gC-1 antibody activity in vivo is complement-dependent. Immunizing mice with gD-1 and gC-1 provided better protection than gD-1 alone in preventing zosteriform disease and infection of dorsal root ganglia. Therefore, gC-1 immunization prevents HSV-1 evasion from complement and enhances the protection provided by gD-1 immunization.
Collapse
Affiliation(s)
- Sita Awasthi
- Infectious Disease Division, Department of Medicine, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6073, United States.
| | | | | |
Collapse
|
25
|
Caprine herpesvirus-1-specific IgG subclasses in naturally and experimentally infected goats. Vet Microbiol 2009; 138:266-72. [PMID: 19411146 DOI: 10.1016/j.vetmic.2009.04.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2008] [Revised: 03/16/2009] [Accepted: 04/03/2009] [Indexed: 11/23/2022]
|
26
|
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: 3.5] [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
|
27
|
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: 2.3] [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
|
28
|
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: 1.1] [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
|
29
|
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: 1.7] [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
|
30
|
Friedman HM. Immune evasion by herpes simplex virus type 1, strategies for virus survival. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2003; 114:103-112. [PMID: 12813914 PMCID: PMC2194497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Many viruses capable of persistent or recurrent infections have evolved strategies to evade host immunity. Viral evasion molecules target components of innate and acquired immunity, including complement proteins, natural killer cells, MHC Class I or Class II molecules and antibody. Our work focuses on HSV-1 glycoproteins gC and gE that impair antibody and complement responses. gC inhibits complement activation by binding C3b and blocking activities mediated by this pivotal complement protein, while gE binds the IgG Fc domain, blocking Fc-mediated activities, including complement activation and antibody-dependent cellular cytotoxicity. HSV-1 mutant viruses that lack the ability to bind C3b, IgG Fc, or both are much less virulent than wild-type virus in a murine model. These HSV-1 immunoevasins help explain the virus' ability to produce recurrent infections despite intact immunity. Strategies to prevent immune evasion may be required to develop successful HSV vaccines.
Collapse
|
31
|
Rizvi SM, Raghavan M. Responses of herpes simplex virus type 1-infected cells to the presence of extracellular antibodies: gE-dependent glycoprotein capping and enhancement in cell-to-cell spread. J Virol 2003; 77:701-8. [PMID: 12477873 PMCID: PMC140612 DOI: 10.1128/jvi.77.1.701-708.2003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Binding of anti-herpes simplex virus (HSV) immunoglobulin G (IgG) to HSV type 1 (HSV-1)-infected HEL and HEp-2 cells causes changes in surface viral glycoprotein distribution, resulting in a capping of all viral glycoproteins towards one pole of the cell. This occurs in a gE-dependent manner. In HEL cells, low concentrations of anti-HSV IgG also enhance cell-to-cell spread of wild-type HSV-1 but not of gE deletion mutant HSV-1. These observations raised the possibility that gE-dependent mechanisms exist that allow some HSV-1-infected cells to respond to the presence of extracellular antibodies by enhancing the antibody-resistant mode of virus transmission.
Collapse
Affiliation(s)
- Syed Monem Rizvi
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor 48109-0620, USA
| | | |
Collapse
|
32
|
Tolba KA, Bowers WJ, Eling DJ, Casey AE, Kipps TJ, Federoff HJ, Rosenblatt JD. HSV amplicon-mediated delivery of LIGHT enhances the antigen-presenting capacity of chronic lymphocytic leukemia. Mol Ther 2002; 6:455-63. [PMID: 12377186 DOI: 10.1006/mthe.2002.0693] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a B lymphocyte malignancy that remains a largely incurable disease. CLL B cells possess the ability to process and present tumor antigens but lack expression of costimulatory molecules, rendering them inefficient effectors of T-cell activation. We previously demonstrated that helper virus-free preparations of herpes simplex virus (HSV) amplicon vectors encoding CD40L efficiently transduce CLL B cells and render them capable of eliciting specific anti-tumor T-cell responses. LIGHT (TNFSF14), a member of the tumor necrosis factor (TNF) superfamily, efficiently activates both T cells and antigen-presenting cells (APCs). We employed an HSV amplicon vector expressing human LIGHT (hf-HSV-LIGHT) to transduce CLL B cells and compared the immunomodulatory function and T-cell activation induced by hf-HSV-LIGHT transduction to that observed with a CD40L-expressing HSV amplicon (hf-HSV-CD40L). hf-HSV-LIGHT transduction induced expression of endogenous B7.1, B7.2, and ICAM.1 on CLL cells, albeit to a lesser degree than that observed in response to transduction with hf-HSV-CD40L. hf-HSV-LIGHT enhanced the antigen-presenting capacity of CLL B cells, as measured by induction of T-cell proliferation in an allogeneic mixed lymphocyte tumor reaction. Finally, hf-HSV-LIGHT-transduced CLL B cells successfully stimulated the outgrowth of autologous cytotoxic T-lymphocytes in vitro. In aggregate, these data suggest that hf-HSV-LIGHT transduction may be useful for induction of immune responses to CLL and other B-cell lymphoid malignancies.
Collapse
Affiliation(s)
- Khaled A Tolba
- James P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
Lubinski JM, Jiang M, Hook L, Chang Y, Sarver C, Mastellos D, Lambris JD, Cohen GH, Eisenberg RJ, Friedman HM. Herpes simplex virus type 1 evades the effects of antibody and complement in vivo. J Virol 2002; 76:9232-41. [PMID: 12186907 PMCID: PMC136467 DOI: 10.1128/jvi.76.18.9232-9241.2002] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.2] [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) encodes a complement-interacting glycoprotein, gC, and an immunoglobulin G (IgG) Fc binding glycoprotein, gE, that mediate immune evasion by affecting multiple aspects of innate and acquired immunity, including interfering with complement components C1q, C3, C5, and properdin and blocking antibody-dependent cellular cytotoxicity. Previous studies evaluated the individual contributions of gC and gE to immune evasion. Experiments in a murine model that examines the combined effects of gC and gE immune evasion on pathogenesis are now reported. Virulence of wild-type HSV-1 is compared with mutant viruses defective in gC-mediated C3 binding, gE-mediated IgG Fc binding, or both immune evasion activities. Eliminating both activities greatly increased susceptibility of HSV-1 to antibody and complement neutralization in vitro and markedly reduced virulence in vivo as measured by disease scores, virus titers, and mortality. Studies with C3 knockout mice indicated that other activities attributed to these glycoproteins, such as gC-mediated virus attachment to heparan sulfate or gE-mediated cell-to-cell spread, do not account for the reduced virulence of mutant viruses. The results support the importance of gC and gE immune evasion in vivo and suggest potential new targets for prevention and treatment of HSV disease.
Collapse
Affiliation(s)
- John M Lubinski
- Department of Medicine, Division of Infectious Diseases, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
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: 0.7] [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
|
35
|
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
|
36
|
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
|
37
|
Favoreel HW, Nauwynck HJ, Van Oostveldt P, Pensaert MB. Role of anti-gB and -gD antibodies in antibody-induced endocytosis of viral and cellular cell surface glycoproteins expressed on pseudorabies virus-infected monocytes. Virology 2000; 267:151-8. [PMID: 10662611 DOI: 10.1006/viro.1999.0132] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The addition of porcine pseudorabies virus (PrV)-specific polyclonal IgG antibodies to PrV-infected monocytes induces internalization of plasma membrane-anchored viral glycoproteins and major histocompatibility complex (MHC) class I. Using PrV deletion strains, it was shown that gB and gD are essential for the process to occur. The purpose of the current study was to evaluate whether antibodies directed against single viral glycoproteins are able to induce endocytosis. It was shown that monoclonal antibodies directed against viral glycoprotein gB and gD, but not against gC and gE, are able to induce internalization of their respective ligand. Adding a combination of monoclonal antibodies against gB and gD resulted in endocytosis levels, comparable to the endocytosis levels observed when adding porcine PrV-specific polyclonal antibodies. The addition of genistein and tyrphostin 25, two inhibitors of tyrosine kinase activity, abolished endocytosis induced by monoclonal anti-gB and -gD antibodies in a concentration-dependent manner. The addition of similar concentrations of tyrphostin 1, an inactive tyrphostin, had no effect on endocytosis. It was also shown that a mixture of polyclonal, but not monoclonal, antibodies against gB and gD is able to induce cointernalization of MHC class I. This indicates that MHC class I cointernalization results from a passive catching of the molecules rather than from a specific interaction of the MHC class I molecules with one or more viral glycoproteins. In conclusion, it can be stated that antibody-induced crosslinking of gB and gD induces the activation of a tyrosine phosphorylation-dependent signal transduction pathway, leading to their endocytosis. Cointernalization of other viral glycoproteins and MHC class I is most likely caused by a passive catching of these molecules in the gB and gD aggregates.
Collapse
Affiliation(s)
- H W Favoreel
- Laboratory of Virology, University of Gent, Gent, Belgium
| | | | | | | |
Collapse
|
38
|
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: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Herpes simplex virus type 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
|
39
|
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: 3.7] [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
|
40
|
Johansson PJ, Ota T, Tsuchiya N, Malone CC, Williams RC. Studies of protein A and herpes simplex virus-1 induced Fc gamma-binding specificities. Different binding patterns for IgG3 from Caucasian and Oriental subjects. Immunology 1994; 83:631-8. [PMID: 7875743 PMCID: PMC1415083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Herpes simplex virus type 1 (HSV-1) expresses a receptor that binds the Fc portion of IgG. This HSV-1 Fc gamma-binding protein is, like protein A of Staphylococcus aureus, known to bind human IgG1, IgG2 and IgG4 but not IgG3 subclasses. However, IgG3 with the allotype Gm(s+)(t+), prominent in the Oriental population, reacts with protein A. This prompted us to investigate the reactivity of Oriental IgG3 monoclonal myeloma proteins of various allotypes with the HSV-1 Fc gamma-binding protein. Of seven Oriental IgG3 myeloma proteins with allotypes Gm(s+)(t+)(u-)(b+)(g-), Gm(s-)(t-)(u+)(b+)(g-) and Gm(s-)(t-)(u+)(b-)(g+), all reacted with the HSV-1 Fc gamma-binding protein. This was in contrast to negative reactions obtained with three IgG3 myeloma proteins of Caucasian origin with Gm(b+)(g-) or Gm(b-)(g+) phenotypes. The same binding pattern, i.e. binding of IgG3 of Oriental but not of Caucasian origin, was found with protein A. The binding of the monoclonal Oriental IgG3 proteins was again independent of the G3m phenotype. These findings support the concept that the HSV-1 Fc gamma-binding protein A have a similar binding site on the IgG molecule. All monoclonal IgG3 proteins derived from Oriental subjects with or without histidine at position 435 bound to HSV Fc gamma-binding protein. This suggests that Oriental IgG3 myeloma proteins with Gm(s-)(t-) phenotypes have additional critical amino acid residue substitutions important for HSV Fc gamma binding different from those already known.
Collapse
Affiliation(s)
- P J Johansson
- Department of Medicine, University of Florida, Gainesville
| | | | | | | | | |
Collapse
|
41
|
Jacobs L. Glycoprotein E of pseudorabies virus and homologous proteins in other alphaherpesvirinae. Arch Virol 1994; 137:209-28. [PMID: 7944945 DOI: 10.1007/bf01309470] [Citation(s) in RCA: 54] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
This paper reviews biological properties of glycoprotein E (gE) of pseudorabies virus (Aujeszky's disease virus) and homologous proteins in other alphaherpesvirinae. It focuses on the gene encoding gE, conserved regions in the gE protein and its homologs, the complex of gE and gI, biological functions of gE in vitro and in vivo, the role of gE in latency and the role of gE in the induction of humoral and cellular immune responses. Special emphasis is placed on the use of gE as a marker protein in the control and eradication of pseudorabies virus.
Collapse
Affiliation(s)
- L Jacobs
- Central Veterinary Institute (CDI-DLO), Lelystad, The Netherlands
| |
Collapse
|
42
|
Dingwell KS, Brunetti CR, Hendricks RL, Tang Q, Tang M, Rainbow AJ, Johnson DC. Herpes simplex virus glycoproteins E and I facilitate cell-to-cell spread in vivo and across junctions of cultured cells. J Virol 1994; 68:834-45. [PMID: 8289387 PMCID: PMC236520 DOI: 10.1128/jvi.68.2.834-845.1994] [Citation(s) in RCA: 257] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Herpes simplex virus (HSV) glycoproteins E and I (gE and gI) can act as a receptor for the Fc domain of immunoglobulin G (IgG). To examine the role of HSV IgG Fc receptor in viral pathogenesis, rabbits and mice were infected by the corneal route with HSV gE- or gI- mutants. Wild-type HSV-1 produced large dendritic lesions in the corneal epithelium and subsequent stromal disease leading to viral encephalitis, whereas gE- and gI- mutant viruses produced microscopic punctate or small dendritic lesions in the epithelium and no corneal disease or encephalitis. These differences were not related to the ability of the gE-gI oligomer to bind IgG because the differences were observed before the appearance of anti-HSV IgG and in mice, in which IgG binds to the Fc receptor poorly or not at all. Mutant viruses produced small plaques on monolayers of normal human fibroblasts and epithelial cells. Replication of gE- and gI- mutant viruses in human fibroblasts were normal, and the rates of entry of mutant and wild-type viruses into fibroblasts were similar; however, spread of gE- and gI- mutant viruses from cell to cell was significantly slower than that of wild-type HSV-1. In experiments in which fibroblast monolayers were infected with low multiplicities of virus and multiple rounds of infection occurred, the presence of neutralizing antibodies in the culture medium caused the yields of mutant viruses to drop dramatically, whereas there was a lesser effect on the production of wild-type HSV. It appears that cell-to-cell transmission of wild-type HSV-1 occurs by at least two mechanisms: (i) release of virus from cells and entry of extracellular virus into a neighboring cell and (ii) transfer of virus across cell junctions in a manner resistant to neutralizing antibodies. Our results suggest that gE- and gI- mutants are defective in the latter mechanism of spread, suggesting the possibility that the gE-gI complex facilitates virus transfer across cell junctions, a mode of spread which may predominate in some tissues. It is ironic that the gE-gI complex, usually considered an IgG Fc receptor, may, through its ability to mediate cell-to-cell spread, actually protect HSV from IgG in a manner different than previously thought.
Collapse
Affiliation(s)
- K S Dingwell
- Department of Pathology, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
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
|
45
|
Dubin G, Fishman NO, Eisenberg RJ, Cohen GH, Friedman HM. The role of herpes simplex virus glycoproteins in immune evasion. Curr Top Microbiol Immunol 1992; 179:111-20. [PMID: 1323450 DOI: 10.1007/978-3-642-77247-4_7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- G Dubin
- Department of Medicine, School of Medicine, University of Pennsylvania, Philadelphia
| | | | | | | | | |
Collapse
|
46
|
Dubin G, Socolof E, Frank I, Friedman HM. Herpes simplex virus type 1 Fc receptor protects infected cells from antibody-dependent cellular cytotoxicity. J Virol 1991; 65:7046-50. [PMID: 1658396 PMCID: PMC250825 DOI: 10.1128/jvi.65.12.7046-7050.1991] [Citation(s) in RCA: 109] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Recent studies indicate that the herpes simplex virus type 1 (HSV-1) Fc receptor (FcR) can bind antiviral immunoglobulin G by participating in antibody bipolar bridging. This occurs when the Fab domain of an immunoglobulin G molecule binds to its antigenic target and the Fc domain binds to the HSV-1 FcR. In experiments comparing cells infected with wild-type HSV-1 (NS) and cells infected with an FcR-deficient mutant (ENS), we demonstrate that participation of the HSV-1 FcR in antibody bipolar bridging reduces the effectiveness of antibody-dependent cellular cytotoxicity.
Collapse
Affiliation(s)
- G Dubin
- Department of Medicine, University of Pennsylvania, Philadelphia
| | | | | | | |
Collapse
|
47
|
Abstract
Recent work has demonstrated that a tripeptide derivative mimicking the active proteinase-binding site of cystatin C, a human cysteine proteinase inhibitor, can block growth of group A streptococci and replication of herpes simplex virus (HSV). In the case of HSV, intact cystatin C was also found to inhibit replication of the virus. Many streptococcal strains and HSV-infected cells produce immunoglobulin (Ig)-binding proteins, and a possible connection between such proteins and proteolytic activity was indicated by the finding that bacterial Ig-binding proteins also show affinity for proteinase inhibitors. The significance of these various observations is not clear, but available data suggest that proteinases play a role in vital microbial functions (e.g. viral replication) and may be utilized as targets for antimicrobial agents. The results discussed here also indicate that peptide derivatives based on the structure of proteinase inhibitors occurring in nature could be used as such agents.
Collapse
Affiliation(s)
- L Björck
- Department of Medical and Physiological Chemistry, University of Lund, Sweden
| |
Collapse
|
48
|
Litwin V, Sandor M, Grose C. Cell surface expression of the varicella-zoster virus glycoproteins and Fc receptor. Virology 1990; 178:263-72. [PMID: 2167554 DOI: 10.1016/0042-6822(90)90402-d] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Varicella-zoster virus (VZV) specifies the synthesis of viral glycoproteins which are important antigens for induction of the host immune response. In this report the technology of laser-activated flow cytometry has been employed to measure the membrane expression of VZV glycoproteins gpI, gpII, gpIII, and gpIV. By use of biotinylated monoclonal antibodies as probes, all four glycoproteins were demonstrated on the infected cell surface. The temporal appearance of the viral glycoproteins was defined in a time course experiment and shown to be maximal about 24 hr postinfection. The issue whether VZV induces the cell surface expression of an Fc receptor (FcR) was investigated with biotinylated nonimmune human IgG, followed by streptavidin-phycoerythrin. By this technique a 10-fold increase in fluorescence intensity was seen in the VZV-infected cells as compared to the mock-infected controls. When the experiment was repeated with purified human Fc fragment rather than whole IgG, a similar degree of binding was seen. Both the VZV glycoproteins and the VZV FcR were exquisitely sensitive to trypsin treatment (1 mg/ml); likewise, the cell surface expression of these VZV products was diminished by treatment of the infected cultures with monensin, an inhibitor of glycoprotein transport. In order to prove that VZV infection was not causing the induction of a cellular Fc gamma R, the VZV-infected and mock-infected cells were stained with monoclonal antibodies directed against each of the three human cellular IgG FcR, but no differences were observed. Therefore, the FcR activity seen in the infected culture was not due to one of the known cellular Fc gamma R.
Collapse
Affiliation(s)
- V Litwin
- Department of Microbiology, University of Iowa College of Medicine, Iowa City 52242
| | | | | |
Collapse
|
49
|
Johansson PJ, Blomberg J. Characterization of herpes simplex virus type 1-induced Fc receptor in its interaction with rabbit immunoglobulin G (IgG). APMIS 1990; 98:685-94. [PMID: 2169770 DOI: 10.1111/j.1699-0463.1990.tb04989.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Herpes simplex virus (HSV) induces a receptor on infected cells that is able to bind the Fc part of immunoglobulin G (IgG). We have examined some basic physicochemical and binding properties of the Fc receptor induced on HSV-1 infected green monkey kidney (GMK) cells in its interaction with rabbit IgG. Fixation of HSV-1 infected cells with glutaraldehyde, formaldehyde, acetone or ethanol did not inhibit the Fc binding ability. The binding specificity of the receptor was not affected by ethanol treatment and all subsequent binding studies were performed with cells treated with ethanol. The receptor was detected within 4 hours of infection and the binding increased until 16 hours post infection. The interaction between ligand and receptor was dependent on pH with a binding optimum around pH 8.0 and 8.5. EDTA, but not EGTA, inhibited receptor binding, suggesting participation of divalent cations in the receptor-ligand interaction. Inhibition of binding was also seen when cells were preincubated for 30 min at 56 degrees, 60 degrees and 100 degrees C in contrast with cells incubated at 37 degrees and 45 degrees C. The number of binding sites on ethanol-treated GMK cells 18 hours after infection was estimated to be around 4 x 10(6)/cell and the affinity constant at approximately 2 x 10(7) M-1.
Collapse
Affiliation(s)
- P J Johansson
- Institute of Medical Microbiology, University of Lund, Sweden
| | | |
Collapse
|
50
|
Liang SM, Liang CM, Chiueh CC. Visualization of interleukin-2-like molecules in MPP(+)-lesioned rat brain. Biochem Biophys Res Commun 1989; 165:1312-8. [PMID: 2610695 DOI: 10.1016/0006-291x(89)92746-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The tissue distribution of interleukin-2 (IL-2) in normal and 1-methyl-4-phenyl-pyridinium (MPP+)-lesioned brains of rats was investigated. Intrastriatal administration of MPP+ caused visible damage in the vicinity of the injected region two weeks after injection. Autoradiography of the tissue section with anti-IL-2 antibodies plus trace amounts of radiolabeled IL-2 showed that the antibodies treatment elicited a selective radiolabeling of the brain tissues localized at the MPP(+)-lesioned region but not at normal cryo-sliced sections. Addition of radiolabeled IL-2 alone or normal rabbit immunoglobulins did not show any labeling effect. These autoradiographic imaging results suggest that there is an accumulation of cells bearing IL-2-like molecules at the MPP(+)-induced lesion sites.
Collapse
Affiliation(s)
- S M Liang
- Division of Cytokine Biology, Food and Drug Administration, Bethesda, MD 20892
| | | | | |
Collapse
|