1
|
Ebenig A, Lange MV, Mühlebach MD. Versatility of live-attenuated measles viruses as platform technology for recombinant vaccines. NPJ Vaccines 2022; 7:119. [PMID: 36243743 PMCID: PMC9568972 DOI: 10.1038/s41541-022-00543-4] [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: 04/08/2022] [Accepted: 09/22/2022] [Indexed: 11/09/2022] Open
Abstract
Live-attenuated measles virus (MeV) has been extraordinarily effective in preventing measles infections and their often deadly sequelae, accompanied by remarkable safety and stability since their first licensing in 1963. The advent of recombinant DNA technologies, combined with systems to generate infectious negative-strand RNA viruses on the basis of viral genomes encoded on plasmid DNA in the 1990s, paved the way to generate recombinant, vaccine strain-derived MeVs. These live-attenuated vaccine constructs can encode and express additional foreign antigens during transient virus replication following immunization. Effective humoral and cellular immune responses are induced not only against the MeV vector, but also against the foreign antigen cargo in immunized individuals, which can protect against the associated pathogen. This review aims to present an overview of the versatility of this vaccine vector as platform technology to target various diseases, as well as current research and developmental stages, with one vaccine candidate ready to enter phase III clinical trials to gain marketing authorization, MV-CHIK.
Collapse
Affiliation(s)
- Aileen Ebenig
- Division of Veterinary Medicine, Paul-Ehrlich-Institut, D-63225, Langen, Germany
| | - Mona V Lange
- Division of Veterinary Medicine, Paul-Ehrlich-Institut, D-63225, Langen, Germany
| | - Michael D Mühlebach
- Division of Veterinary Medicine, Paul-Ehrlich-Institut, D-63225, Langen, Germany.
| |
Collapse
|
2
|
Multiple Receptors Involved in Invasion and Neuropathogenicity of Canine Distemper Virus: A Review. Viruses 2022; 14:v14071520. [PMID: 35891500 PMCID: PMC9317347 DOI: 10.3390/v14071520] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 07/06/2022] [Accepted: 07/09/2022] [Indexed: 12/04/2022] Open
Abstract
The canine distemper virus (CDV) is a morbillivirus that infects a broad range of terrestrial carnivores, predominantly canines, and is associated with high mortality. Similar to another morbillivirus, measles virus, which infects humans and nonhuman primates, CDV transmission from an infected host to a naïve host depends on two cellular receptors, namely, the signaling lymphocyte activation molecule (SLAM or CD150) and the adherens junction protein nectin-4 (also known as PVRL4). CDV can also invade the central nervous system by anterograde spread through olfactory nerves or in infected lymphocytes through the circulation, thus causing chronic progressive or relapsing demyelination of the brain. However, the absence of the two receptors in the white matter, primary cultured astrocytes, and neurons in the brain was recently demonstrated. Furthermore, a SLAM/nectin-4-blind recombinant CDV exhibits full cell-to-cell transmission in primary astrocytes. This strongly suggests the existence of a third CDV receptor expressed in neural cells, possibly glial cells. In this review, we summarize the recent progress in the study of CDV receptors, highlighting the unidentified glial receptor and its contribution to pathogenicity in the host nervous system. The reviewed studies focus on CDV neuropathogenesis, and neural receptors may provide promising directions for the treatment of neurological diseases caused by CDV. We also present an overview of other neurotropic viruses to promote further research and identification of CDV neural receptors.
Collapse
|
3
|
Rajaraman S, Canjuga D, Ghosh M, Codrea MC, Sieger R, Wedekink F, Tatagiba M, Koch M, Lauer UM, Nahnsen S, Rammensee HG, Mühlebach MD, Stevanovic S, Tabatabai G. Measles Virus-Based Treatments Trigger a Pro-inflammatory Cascade and a Distinctive Immunopeptidome in Glioblastoma. MOLECULAR THERAPY-ONCOLYTICS 2018; 12:147-161. [PMID: 30775418 PMCID: PMC6365369 DOI: 10.1016/j.omto.2018.12.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 12/23/2018] [Indexed: 12/26/2022]
Abstract
Glioblastoma is an aggressive primary brain tumor with bad prognosis. On the other hand, oncolytic measles virus (MeV) therapy is an experimental glioma treatment strategy with clinical safety and first evidence of anti-tumoral efficacy. Therefore, we investigated the combination of MeV with conventional therapies by cytotoxic survival assays in long-term glioma cell lines LN229, LNZ308, and glioma stem-like GS8 cells, as well as the basal viral infectivity in primary glioblastoma cultures T81/16, T1094/17, and T708/16. We employed Chou-Talalay analysis to identify the synergistic treatment sequence chemotherapy, virotherapy, and finally radiotherapy (CT-VT-RT). RNA sequencing and immunopeptidome analyses were used to delineate treatment-induced molecular and immunological profiles. CT-VT-RT displayed synergistic anti-glioma activity and initiated a type 1 interferon response, along with canonical Janus kinase-signal transducers and activators of transcription (JAK-STAT) signaling, and downstream interferon-stimulated genes were induced, resulting in apoptotic cascades. Furthermore, antigen presentation along with immunostimulatory chemokines was increased in CT-VT-RT-treated glioma cells, indicating a treatment-induced pro-inflammatory phenotype. We identified novel treatment-induced viral and tumor-associated peptides through HLA ligandome analysis. Our data delineate an actionable treatment-induced molecular and immunological signature of CT-VT-RT, and they could be exploited for the design of novel tailored treatment strategies involving virotherapy and immunotherapy.
Collapse
Affiliation(s)
- Srinath Rajaraman
- Interdisciplinary Division of Neuro-Oncology, Hertie Institute for Clinical Brain Research, Departments of Neurology and Neurosurgery, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen 72076, Germany
| | - Denis Canjuga
- Interdisciplinary Division of Neuro-Oncology, Hertie Institute for Clinical Brain Research, Departments of Neurology and Neurosurgery, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen 72076, Germany
| | - Michael Ghosh
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, Tübingen 72076, Germany
| | - Marius Cosmin Codrea
- Quantitative Biology Center (QBiC), Eberhard Karls University Tübingen, Tübingen 72076, Germany
| | - Raika Sieger
- Interdisciplinary Division of Neuro-Oncology, Hertie Institute for Clinical Brain Research, Departments of Neurology and Neurosurgery, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen 72076, Germany
| | - Florian Wedekink
- Interdisciplinary Division of Neuro-Oncology, Hertie Institute for Clinical Brain Research, Departments of Neurology and Neurosurgery, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen 72076, Germany
| | - Marcos Tatagiba
- Interdisciplinary Division of Neuro-Oncology, Hertie Institute for Clinical Brain Research, Departments of Neurology and Neurosurgery, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen 72076, Germany
| | - Marilin Koch
- Interdisciplinary Division of Neuro-Oncology, Hertie Institute for Clinical Brain Research, Departments of Neurology and Neurosurgery, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen 72076, Germany
| | - Ulrich M Lauer
- Department of Internal Medicine VIII, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen 72076, Germany.,German Translational Cancer Consortium (DKTK), DKFZ partner site Tübingen, Germany
| | - Sven Nahnsen
- Quantitative Biology Center (QBiC), Eberhard Karls University Tübingen, Tübingen 72076, Germany
| | - Hans-Georg Rammensee
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, Tübingen 72076, Germany.,German Translational Cancer Consortium (DKTK), DKFZ partner site Tübingen, Germany
| | - Michael D Mühlebach
- Division of Veterinary Medicine, Paul-Ehrlich-Institut, Langen 63225, Germany
| | - Stefan Stevanovic
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, Tübingen 72076, Germany.,German Translational Cancer Consortium (DKTK), DKFZ partner site Tübingen, Germany
| | - Ghazaleh Tabatabai
- Interdisciplinary Division of Neuro-Oncology, Hertie Institute for Clinical Brain Research, Departments of Neurology and Neurosurgery, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen 72076, Germany.,German Translational Cancer Consortium (DKTK), DKFZ partner site Tübingen, Germany
| |
Collapse
|
4
|
Ha MN, Delpeut S, Noyce RS, Sisson G, Black KM, Lin LT, Bilimoria D, Plemper RK, Privé GG, Richardson CD. Mutations in the Fusion Protein of Measles Virus That Confer Resistance to the Membrane Fusion Inhibitors Carbobenzoxy-d-Phe-l-Phe-Gly and 4-Nitro-2-Phenylacetyl Amino-Benzamide. J Virol 2017; 91:e01026-17. [PMID: 28904193 PMCID: PMC5686717 DOI: 10.1128/jvi.01026-17] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 08/27/2017] [Indexed: 11/20/2022] Open
Abstract
The inhibitors carbobenzoxy (Z)-d-Phe-l-Phe-Gly (fusion inhibitor peptide [FIP]) and 4-nitro-2-phenylacetyl amino-benzamide (AS-48) have similar efficacies in blocking membrane fusion and syncytium formation mediated by measles virus (MeV). Other homologues, such as Z-d-Phe, are less effective but may act through the same mechanism. In an attempt to map the site of action of these inhibitors, we generated mutant viruses that were resistant to the inhibitory effects of Z-d-Phe-l-Phe-Gly. These 10 mutations were localized to the heptad repeat B (HRB) region of the fusion protein, and no changes were observed in the viral hemagglutinin, which is the receptor attachment protein. Mutations were validated in a luciferase-based membrane fusion assay, using transfected fusion and hemagglutinin expression plasmids or with syncytium-based assays in Vero, Vero-SLAM, and Vero-Nectin 4 cell lines. The changes I452T, D458N, D458G/V459A, N462K, N462H, G464E, and I483R conferred resistance to both FIP and AS-48 without compromising membrane fusion. The inhibitors did not block hemagglutinin protein-mediated binding to the target cell. Edmonston vaccine/laboratory and IC323 wild-type strains were equally affected by the inhibitors. Escape mutations were mapped upon a three-dimensional (3D) structure modeled from the published crystal structure of parainfluenzavirus 5 fusion protein. The most effective mutations were situated in a region located near the base of the globular head and its junction with the alpha-helical stalk of the prefusion protein. We hypothesize that the fusion inhibitors could interfere with the structural changes that occur between the prefusion and postfusion conformations of the fusion protein.IMPORTANCE Due to lapses in vaccination worldwide that have caused localized outbreaks, measles virus (MeV) has regained importance as a pathogen. Antiviral agents against measles virus are not commercially available but could be useful in conjunction with MeV eradication vaccine programs and as a safeguard in oncolytic viral therapy. Three decades ago, the small hydrophobic peptide Z-d-Phe-l-Phe-Gly (FIP) was shown to block MeV infections and syncytium formation in monkey kidney cell lines. The exact mechanism of its action has yet to be determined, but it does appear to have properties similar to those of another chemical inhibitor, AS-48, which appears to interfere with the conformational change in the viral F protein that is required to elicit membrane fusion. Escape mutations were used to map the site of action for FIP. Knowledge gained from these studies could help in the design of new inhibitors against morbilliviruses and provide additional knowledge concerning the mechanism of virus-mediated membrane fusion.
Collapse
Affiliation(s)
- Michael N Ha
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Department of Pediatrics, IWK Health Centre, Canadian Center for Vaccinology, Goldbloom Pavilion, Halifax, Nova Scotia, Canada
| | - Sébastien Delpeut
- Department of Pediatrics, IWK Health Centre, Canadian Center for Vaccinology, Goldbloom Pavilion, Halifax, Nova Scotia, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Ryan S Noyce
- Department of Pediatrics, IWK Health Centre, Canadian Center for Vaccinology, Goldbloom Pavilion, Halifax, Nova Scotia, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Microbiology and Immunology, University of Alberta, Edmonton, Canada
| | - Gary Sisson
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Karen M Black
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Liang-Tzung Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Darius Bilimoria
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Vertex Pharmaceuticals (Canada) Incorporated, Laval, Quebec, Canada
| | - Richard K Plemper
- Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Gilbert G Privé
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Christopher D Richardson
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Department of Pediatrics, IWK Health Centre, Canadian Center for Vaccinology, Goldbloom Pavilion, Halifax, Nova Scotia, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
5
|
Lin LT, Richardson CD. The Host Cell Receptors for Measles Virus and Their Interaction with the Viral Hemagglutinin (H) Protein. Viruses 2016; 8:v8090250. [PMID: 27657109 PMCID: PMC5035964 DOI: 10.3390/v8090250] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/29/2016] [Accepted: 09/02/2016] [Indexed: 12/14/2022] Open
Abstract
The hemagglutinin (H) protein of measles virus (MeV) interacts with a cellular receptor which constitutes the initial stage of infection. Binding of H to this host cell receptor subsequently triggers the F protein to activate fusion between virus and host plasma membranes. The search for MeV receptors began with vaccine/laboratory virus strains and evolved to more relevant receptors used by wild-type MeV. Vaccine or laboratory strains of measles virus have been adapted to grow in common cell lines such as Vero and HeLa cells, and were found to use membrane cofactor protein (CD46) as a receptor. CD46 is a regulator that normally prevents cells from complement-mediated self-destruction, and is found on the surface of all human cells, with the exception of erythrocytes. Mutations in the H protein, which occur during adaptation and allow the virus to use CD46 as a receptor, have been identified. Wild-type isolates of measles virus cannot use the CD46 receptor. However, both vaccine/laboratory and wild-type strains can use an immune cell receptor called signaling lymphocyte activation molecule family member 1 (SLAMF1; also called CD150) and a recently discovered epithelial receptor known as Nectin-4. SLAMF1 is found on activated B, T, dendritic, and monocyte cells, and is the initial target for infections by measles virus. Nectin-4 is an adherens junction protein found at the basal surfaces of many polarized epithelial cells, including those of the airways. It is also over-expressed on the apical and basal surfaces of many adenocarcinomas, and is a cancer marker for metastasis and tumor survival. Nectin-4 is a secondary exit receptor which allows measles virus to replicate and amplify in the airways, where the virus is expelled from the body in aerosol droplets. The amino acid residues of H protein that are involved in binding to each of the receptors have been identified through X-ray crystallography and site-specific mutagenesis. Recombinant measles “blind” to each of these receptors have been constructed, allowing the virus to selectively infect receptor specific cell lines. Finally, the observations that SLAMF1 is found on lymphomas and that Nectin-4 is expressed on the cell surfaces of many adenocarcinomas highlight the potential of measles virus for oncolytic therapy. Although CD46 is also upregulated on many tumors, it is less useful as a target for cancer therapy, since normal human cells express this protein on their surfaces.
Collapse
Affiliation(s)
- Liang-Tzung Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Christopher D Richardson
- Department of Microbiology and Immunology, Dalhousie University, 5850 College St., Halifax, NS B3H 4R2, Canada.
- Department of Pediatrics and Canadian Center for Vaccinology, Izaak Walton Killam Health Centre, Halifax, NS B3K 6R8, Canada.
| |
Collapse
|
6
|
Liu YP, Russell SP, Ayala-Breton C, Russell SJ, Peng KW. Ablation of nectin4 binding compromises CD46 usage by a hybrid vesicular stomatitis virus/measles virus. J Virol 2014; 88:2195-204. [PMID: 24335299 PMCID: PMC3911550 DOI: 10.1128/jvi.02628-13] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 12/03/2013] [Indexed: 01/18/2023] Open
Abstract
Measles virus (MV) immunosuppression is due to infection of SLAM-positive immune cells, whereas respiratory shedding and virus transmission are due to infection of nectin4-positive airway epithelial cells. The vaccine lineage MV strain Edmonston (MV-Edm) acquired an additional tropism for CD46 which is the basis of its oncolytic specificity. VSVFH is a vesicular stomatitis virus (VSV) encoding the MV-Edm F and H entry proteins in place of G. The virus spreads faster than MV-Edm and is highly fusogenic and a potent oncolytic. To determine whether ablating nectin4 tropism from VSVFH might prevent shedding, increasing its safety profile as an oncolytic, or might have any effect on CD46 binding, we generated VSVFH viruses with H mutations that disrupt attachment to SLAM and/or nectin4. Disruption of nectin4 binding reduced release of VSVFH from the basolateral side of differentiated airway epithelia composed of Calu-3 cells. However, because nectin4 and CD46 have substantially overlapping receptor binding surfaces on H, disruption of nectin4 binding compromised CD46 binding and greatly diminished the oncolytic potency of these viruses on human cancer cells. Thus, our results support continued preclinical development of VSVFH without ablation of nectin4 binding.
Collapse
Affiliation(s)
- Yu-Ping Liu
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Samuel P. Russell
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
- University of Wisconsin—Madison, Madison, Wisconsin, USA
| | | | - Stephen J. Russell
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Department of Internal Medicine, Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | - Kah-Whye Peng
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
7
|
Cuthill JH, Charleston MA. A simple model explains the dynamics of preferential host switching among mammal RNA viruses. Evolution 2013; 67:980-90. [PMID: 23550750 PMCID: PMC7202234 DOI: 10.1111/evo.12064] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A growing number of studies support a tendency toward preferential host switching, by parasites and pathogens, over relatively short phylogenetic distances. This suggests that a host switch is more probable if a potential host is closely related to the original host than if it is a more distant relative. However, despite its importance for the health of humans, livestock, and wildlife, the detailed dynamics of preferential host switching have, so far, been little studied. We present an empirical test of two theoretical models of preferential host switching, using observed phylogenetic distributions of host species for RNA viruses of three mammal orders (primates, carnivores, and ungulates). The analysis focuses on multihost RNA virus species, because their presence on multiple hosts and their estimated ages of origin indicate recent host switching. Approximate Bayesian computation was used to compare observed phylogenetic distances between hosts with those simulated under the theoretical models. The results support a decreasing sigmoidal model of preferential host switching, with a strong effect from increasing phylogenetic distance, on all three studied host phylogenies. This suggests that the dynamics of host switching are fundamentally similar for RNA viruses of different mammal orders and, potentially, a wider range of coevolutionary systems.
Collapse
Affiliation(s)
- Jennifer Hoyal Cuthill
- School of Information Technologies, University of Sydney, Sydney, New South Wales 2006, Australia.
| | | |
Collapse
|
8
|
Mok H, Cheng X, Xu Q, Zengel JR, Parhy B, Zhao J, Wang CK, Jin H. Evaluation of Measles Vaccine Virus as a Vector to Deliver Respiratory Syncytial Virus Fusion Protein or Epstein-Barr Virus Glycoprotein gp350. Open Virol J 2012; 6:12-22. [PMID: 22383906 PMCID: PMC3286841 DOI: 10.2174/1874357901206010012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 01/03/2012] [Accepted: 01/16/2012] [Indexed: 01/31/2023] Open
Abstract
Live attenuated recombinant measles vaccine virus (MV) Edmonston-Zagreb (EZ) strain was evaluated as a viral vector to express the ectodomains of fusion protein of respiratory syncytial virus (RSV F) or glycoprotein 350 of Epstein-Barr virus (EBV gp350) as candidate vaccines for prophylaxis of RSV and EBV. The glycoprotein gene was inserted at the 1st or the 3rd position of the measles virus genome and the recombinant viruses were generated. Insertion of the foreign gene at the 3rd position had a minimal impact on viral replication in vitro. RSV F or EBV gp350 protein was secreted from infected cells. In cotton rats, EZ-RSV F and EZ-EBV gp350 induced MV- and insert-specific antibody responses. In addition, both vaccines also induced insert specific interferon gamma (IFN-γ) secreting T cell response. EZ-RSV F protected cotton rats from pulmonary replication of RSV A2 challenge infection. In rhesus macaques, although both EZ-RSV F and EZ-EBV gp350 induced MV specific neutralizing antibody responses, only RSV F specific antibody response was detected. Thus, the immunogenicity of the foreign antigens delivered by measles vaccine virus is dependent on the nature of the insert and the animal models used for vaccine evaluation.
Collapse
Affiliation(s)
- Hoyin Mok
- MedImmune LLC., 319 North Bernardo Ave, Mountain View, California, USA
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Abstract
Progress in vector design and an increased knowledge of mechanisms underlying tumor-induced immune suppression have led to a new and promising generation of Adenovirus (Ad)-based immunotherapies, which are discussed in this review. As vaccine vehicles Ad vectors (AdVs) have been clinically evaluated and proven safe, but a major limitation of the commonly used Ad5 serotype is neutralization by preexistent or rapidly induced immune responses. Genetic modifications in the Ad capsid can reduce intrinsic immunogenicity and facilitate escape from antibody-mediated neutralization. Further modification of the Ad hexon and fiber allows for liver and scavenger detargeting and selective targeting of, for example, dendritic cells. These next-generation Ad vaccines with enhanced efficacy are now becoming available for testing as tumor vaccines. In addition, AdVs encoding immune-modulating products may be used to convert the tumor microenvironment from immune-suppressive and proinvasive to proinflammatory, thus facilitating cell-mediated effector functions that can keep tumor growth and invasion in check. Oncolytic AdVs, that selectively replicate in tumor cells and induce an immunogenic form of cell death, can also be armed with immune-activating transgenes to amplify primed antitumor immune responses. These novel immunotherapy strategies, employing highly efficacious AdVs in optimized configurations, show great promise and warrant clinical exploration.
Collapse
|
10
|
Jabara HH, Angelini F, Brodeur SR, Geha RS. Ligation of CD46 to CD40 inhibits CD40 signaling in B cells. Int Immunol 2011; 23:215-21. [PMID: 21393637 DOI: 10.1093/intimm/dxq474] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
CD40 induces B cells to switch to IgE in the presence of IL-4 and up-regulates their expression of the low-affinity receptor for IgE, CD23, which promotes the immune response to allergen complexed with IgE antibody. CD40 binds to CD40L and to the C4b-binding protein (C4BP) using distinct sites. CD46 is a receptor for the product of activated complement C4b. Some microbial antigens bind both C4BP and CD46, potentially bridging CD40 to CD46. In addition, immune complexes containing both C4b and C4BP may cross-link CD40 to CD46. We demonstrate that cross-linking CD46 to CD40 on B cells inhibits CD40-mediated up-regulation of surface CD23 expression and induction of IL-4-dependent IgE isotype switching. This was associated with inhibition of induction of Cε germ line transcripts and of activation-induced cytidine deaminase mRNA expression. Furthermore, co-ligation of CD46 to CD40 blocked CD40-mediated NF-κB activation. These observations suggest that complement components may play an important role in regulating CD40 activation of B cells and the allergic response.
Collapse
Affiliation(s)
- Haifa H Jabara
- Division of Immunology, Children's Hospital, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
11
|
Niculescu-Duvaz D, Negoita-Giras G, Niculescu-Duvaz I, Hedley D, Springer CJ. Directed Enzyme Prodrug Therapies. PRODRUGS AND TARGETED DELIVERY 2011. [DOI: 10.1002/9783527633166.ch12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
12
|
Runkler N, Dietzel E, Carsillo M, Niewiesk S, Maisner A. Sorting signals in the measles virus wild-type glycoproteins differently influence virus spread in polarized epithelia and lymphocytes. J Gen Virol 2009; 90:2474-2482. [PMID: 19570960 DOI: 10.1099/vir.0.012575-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The spread of virus infection within an organism is partially dictated by the receptor usage of the virus and can be influenced by sorting signals present in the viral glycoproteins expressed in infected cells. In previous studies, we have shown that the haemagglutinin (H) and fusion protein (F) of the measles virus (MV) vaccine strain MV(Edm) harbour tyrosine-dependent sorting signals which influence virus spread in both lymphocytes and epithelial cells to a similar degree. In contrast with the vaccine strain, MV wild-type virus does not use CD46 but CD150/SLAM and a not clearly identified molecule on epithelial cells as receptors. To determine differences in viral spread between vaccine and wild-type virus, we generated recombinant MV expressing glycoproteins of both the wild-type strain WTFb and the corresponding tyrosine mutants. In contrast with observations based on vaccine virus glycoproteins, mutations in wild-type virus H and F differently influenced cell-to-cell fusion and replication in polarized epithelia and lymphocytes. For wild-type H, our data suggest a key role of the cytoplasmic tyrosine signal for virus dissemination in vivo. It seems to be important for efficient virus spread between lymphocytes, while the tyrosine signal in the F protein gains importance in epithelial cells as both signals have to be intact to allow efficient spread of infection within epithelia.
Collapse
Affiliation(s)
- Nicole Runkler
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Str. 2, D-35043 Marburg, Germany
| | - Erik Dietzel
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Str. 2, D-35043 Marburg, Germany
| | - Mary Carsillo
- Department of Veterinary Biosciences, College of Veterinary Medicine, Ohio State University, Columbus, OH 43210-1093, USA
| | - Stefan Niewiesk
- Department of Veterinary Biosciences, College of Veterinary Medicine, Ohio State University, Columbus, OH 43210-1093, USA
| | - Andrea Maisner
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Str. 2, D-35043 Marburg, Germany
| |
Collapse
|
13
|
Künzi V, Oberholzer PA, Heinzerling L, Dummer R, Naim HY. Recombinant Measles Virus Induces Cytolysis of Cutaneous T-Cell Lymphoma In Vitro and In Vivo. J Invest Dermatol 2006; 126:2525-32. [PMID: 16960554 DOI: 10.1038/sj.jid.5700529] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Measles virus (MV) has shown promise as an oncolytic virus in the treatment of different tumor models for human B-cell lymphoma, multiple myeloma, ovarian cancer, and glioma. We have shown that, in a phase I clinical trial, MV vaccine induces tumor regression in cutaneous T-cell lymphoma (CTCL) patients. Here, we investigated in detail, the effect of recombinant MV (rMV) vaccine strain in CTCL cell cultures, and in vivo in established CTCL xenografts in nude mice. The susceptibility of three CTCL cell lines, originating from patients, to rMV was tested by determination of cell surface expression of MV receptors. All cell lines expressed the receptors CD150 and CD46 and were easily infected by rMV and induced complete cell lysis. The cytoreductive activity was apparent in cells forming aggregates, indicating a cell-to-cell spread of MV and cytolysis owing to virus infection. Intratumoral (i.t.) injection of rMV, expressing enhanced green fluorescent protein induced complete regression of large established human CTCL tumors in nude mice, whereas tumors with control treatment progressed exponentially. Immunohistochemical analysis of tumor biopsies, after i.t. treatment, for MV-NP protein complex demonstrated replication of MV within the tumors. The data demonstrate the potential of MV as a therapeutic agent against CTCL.
Collapse
|
14
|
Abstract
Tumor angiogenesis is crucial for the progression and metastasis of cancer. The vasculature of tumor tissue is different from normal vasculature. Therefore, tumor vascular targeting therapy could represent an effective therapeutic strategy with which to suppress both primary tumor growth and tumor metastasis. The use of viral vectors for tumor vascular targeting therapy is a promising strategy based on the unique properties of viral vectors. In order to circumvent the potential problems of antiviral neutralizing antibodies, poor access to extravascular tumor tissue, and toxicities to normal tissue, viral vectors need to be modified to target the tumor endothelial cells. Viral vectors that could be used for tumor vascular targeting therapy include adenoviral vectors, adeno-associated viral vectors, retroviral vectors, lentiviral vectors, measles virus, and herpes simplex viral vectors. In this review, we will summarize the strategies available for targeting viral vectors for tumor vascular targeting therapy.
Collapse
Affiliation(s)
- Yanzheng Liu
- Sidney Kimmel Cancer Center, 10835 Road to the Cure, San Diego, CA 92121, USA
| | | |
Collapse
|
15
|
Hallak LK, Merchan JR, Storgard CM, Loftus JC, Russell SJ. Targeted Measles Virus Vector Displaying Echistatin Infects Endothelial Cells via αvβ3 and Leads to Tumor Regression. Cancer Res 2005; 65:5292-300. [PMID: 15958576 DOI: 10.1158/0008-5472.can-04-2879] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Targeting tumor-associated vascular endothelium by replication-competent viral vectors is a promising strategy for cancer gene therapy. Here we describe the development of a viral vector based on the Edmonston vaccine strain of measles virus targeted to integrin alpha(v)beta3, which is expressed abundantly on activated but not quiescent vascular endothelium. We displayed a disintegrin, M28L echistatin that binds with a high affinity to integrin alpha(v)beta3 on the COOH terminus of the viral attachment (H) protein and rescued the replication-competent recombinant virus by reverse genetics. The new targeted virus was named measles virus echistatin vector (MV-ERV). Its native binding to CD46 was purposefully retained to allow virus infection of tumor cells expressing this receptor. MV-ERV correctly displayed echistatin on the outer surface of its envelope and produced interesting ring formation phenomena due to cell detachment upon infection of susceptible Vero cells in vitro. MV-ERV grew to 10(6) plaque-forming units/mL, slightly lower than the parental Edmonston strain of measles virus (MV-Edm), but it selectively infected Chinese hamster ovary cells expressing integrin alpha(v)beta3. It also selectively infected both bovine and human endothelial cells on matrigels and unlike MV-Edm, MV-ERV infected newly formed blood vessels in chorioallantoic membrane assays. In animal models, MV-ERV but not the control MV-Edm caused the regression of s.c. xenografts of resistant multiple myeloma tumors (MM1) in severe combined immunodeficient mice. The tumors were either completely eradicated or their growth was significantly retarded. The specificity, potency, and feasibility of MV-ERV infection clearly show the potential use of MV-ERV in gene therapy for targeting tumor-associated vasculature for the treatment of solid tumors.
Collapse
Affiliation(s)
- Louay K Hallak
- Molecular Medicine Program, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | |
Collapse
|
16
|
Schneider-Schaulies S, ter Meulen V. Triggering of and interference with immune activation: interactions of measles virus with monocytes and dendritic cells. Viral Immunol 2003; 15:417-28. [PMID: 12479392 DOI: 10.1089/088282402760312304] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
17
|
Riley RC, Kemper C, Leung M, Atkinson JP. Characterization of human membrane cofactor protein (MCP; CD46) on spermatozoa. Mol Reprod Dev 2002; 62:534-46. [PMID: 12112588 DOI: 10.1002/mrd.10144] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Membrane cofactor protein (MCP; CD46) is a complement regulator widely expressed as four isoforms that arise via alternative splicing. On human spermatozoa, MCP is expressed on the inner acrosomal membrane and alterations of spermatozoa MCP may be associated with infertility. In rodents, expression of MCP is largely restricted to the testes. MCP on human spermatozoa has a unique M(r) pattern that we have investigated. We also characterized MCP expression in mice transgenic (tg) for human MCP. Human MCP expression in the tg mice mimics the human pattern in that it is located on the inner acrosomal membrane and has a faster M(r) than MCP expressed elsewhere. Sequencing of RT-PCR products from the testis indicates that there is not a unique male reproductive tissue specific cytoplasmic tail. Instead, human spermatozoa express MCP bearing cytoplasmic tail two, which is also utilized in most other tissues and contains several signaling motifs. Further, using N-glycosidases, we demonstrate that the unique lower molecular weight of MCP on spermatozoa is secondary to a modification in the N-linked sugars. Specifically, as the spermatozoa mature, but before they reach the epididymis, the three N-linked sugars of MCP are trimmed to less complex structures. While the purpose of this deglycosylation is unknown, we propose that it is a common feature of proteins expressed on the plasma and inner acrosomal membranes of spermatozoa and hypothesize that it is a spermatozoa specific event critical for facilitating sperm-egg interactions.
Collapse
Affiliation(s)
- Rebecca C Riley
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|
18
|
McQuaid S, Cosby SL. An immunohistochemical study of the distribution of the measles virus receptors, CD46 and SLAM, in normal human tissues and subacute sclerosing panencephalitis. J Transl Med 2002; 82:403-9. [PMID: 11950898 DOI: 10.1038/labinvest.3780434] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
We have compared the expression of the known measles virus (MV) receptors, membrane cofactor protein (CD46) and the signaling lymphocyte-activation molecule (SLAM), using immunohistochemistry, in a range of normal peripheral tissues (known to be infected by MV) as well as in normal and subacute sclerosing panencephalitis (SSPE) brain. To increase our understanding of how these receptors could be utilized by wild-type or vaccine strains in vivo, the results have been considered with regard to the known route of infection and systemic spread of MV. Strong staining for CD46 was observed in endothelial cells lining blood vessels and in epithelial cells and tissue macrophages in a wide range of peripheral tissues, as well as in Langerhans' and squamous cells in the skin. In lymphoid tissues and blood, subsets of cells were positive for SLAM, in comparison to CD46, which stained all nucleated cell types. Strong CD46 staining was observed on cerebral endothelium throughout the brain and also on ependymal cells lining the ventricles and choroid plexus. Comparatively weaker CD46 staining was observed on subsets of neurons and oligodendrocytes. In SSPE brain sections, the areas distant from lesion sites and negative for MV by immunocytochemistry showed the same distribution for CD46 as in normal brain. However, cells in lesions, positive for MV, were negative for CD46. Normal brain showed no staining for SLAM, and in SSPE brain only subsets of leukocytes in inflammatory infiltrates were positive. None of the cell types most commonly infected by MV show detectable expression of SLAM, whereas CD46 is much more widely expressed and could fulfill a receptor function for some wild-type strains. In the case of wild-type stains, which are unable to use CD46, a further as yet unknown receptor(s) would be necessary to fully explain the pathology of MV infection.
Collapse
Affiliation(s)
- Stephen McQuaid
- Neuropathology Laboratory, Royal Victoria Hospital, Queen's University Belfast, Northern Ireland.
| | | |
Collapse
|
19
|
Ohyama M, Fukui T, Tanaka Y, Kato K, Hoshino R, Sugawara T, Yamanaka M, Ijiri R, Sata T, Itani Y. Measles virus infection in the placenta of monozygotic twins. Mod Pathol 2001; 14:1300-3. [PMID: 11743054 DOI: 10.1038/modpathol.3880478] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We report a case of monozygotic twins whose mother was infected with measles at 19 weeks' gestation. One of the twins died in utero at 32 weeks' gestation. The placenta of the stillbirth showed massive fibrin deposition, and some residual trophoblasts contained many inclusion bodies positive for measles virus antigen. Fetal organs and cells other than a few splenic lymphocytes showed no evidence of measles virus infection. The placenta of the surviving infant showed focal intervillous fibrin deposits, and only a few syncytiotrophoblasts were positive for measles virus antigen. At present, 7 months after the delivery, the surviving infant has not developed any sign of measles virus infection. Postpartum course of the mother has been uneventful, although high titers of serum anti-measles virus IgM persisted for 6 months after delivery. This case is informative in the following respects: the villous trophoblasts had diagnostic inclusion bodies and ultrastructural evidence of measles virus infection, the degree of viral involvement within the monochorionic placenta was uneven, both of the twins were virtually free from measles virus infection despite the marked involvement of the placenta, and measles virus infection had persisted in the monochorionic placenta for approximately 13 weeks.
Collapse
Affiliation(s)
- M Ohyama
- Division of Neonatology, Kanagawa Children's Medical Center, Yokahama, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Soldan SS, Fogdell-Hahn A, Brennan MB, Mittleman BB, Ballerini C, Massacesi L, Seya T, McFarland HF, Jacobson S. Elevated serum and cerebrospinal fluid levels of soluble human herpesvirus type 6 cellular receptor, membrane cofactor protein, in patients with multiple sclerosis. Ann Neurol 2001; 50:486-93. [PMID: 11603380 DOI: 10.1002/ana.1135] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Membrane cofactor protein (CD46) is a member of a family of glycoproteins that are regulators of complement and prevent activation of complement on autologous cells. Recently, CD46 has been identified as the cellular receptor for human herpesvirus Type 6 (HHV-6). Elevated levels of soluble CD46 have been described in several autoimmune disorders, and may be implicated in the pathogenesis of these diseases. As several reports have supported an association of HHV-6 and multiple sclerosis, it was of interest to compare levels of soluble CD46 in the sera of multiple sclerosis patients to that of healthy controls, other neurological disease controls, and other inflammatory disease controls. Using an immunoaffinity column comprised of immobilized monoclonal antibodies to CD46, serum levels of soluble CD46 were found to be significantly elevated in multiple sclerosis patients compared with healthy and other neurological disease controls. Moreover, multiple sclerosis patients who tested positive for HHV-6 DNA in serum had significantly elevated levels of soluble CD46 in their serum compared with those who were negative for HHV-6 DNA. A significant increase in soluble CD46 was also found in the serum of other inflammatory disease controls tested compared to healthy controls. Additionally, a significant correlation was demonstrated between levels of soluble CD46 in the serum and cerebrospinal fluid of multiple sclerosis patients. Collectively, these data suggest that elevated levels of soluble CD46 may contribute to the pathogenesis of inflammatory diseases, including multiple sclerosis.
Collapse
Affiliation(s)
- S S Soldan
- Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Kemper C, Leung M, Stephensen CB, Pinkert CA, Liszewski MK, Cattaneo R, Atkinson JP. Membrane cofactor protein (MCP; CD46) expression in transgenic mice. Clin Exp Immunol 2001; 124:180-9. [PMID: 11422193 PMCID: PMC1906059 DOI: 10.1046/j.1365-2249.2001.01458.x] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2000] [Indexed: 11/20/2022] Open
Abstract
Human membrane cofactor protein (MCP; CD46) is a widely distributed complement regulator. In the mouse, expression of MCP is largely restricted to the testis while a related, widely expressed protein (Crry) appears to perform MCP's (CD46) regulatory activity. We have developed two mouse strains transgenic for human MCP (CD46) utilizing an approximately 400 kb YAC clone carrying the complete gene. A third mouse strain was generated using an overlapping YAC clone isolated from a second library. The expression of human MCP (CD46) in these mouse strains was characterized by immunohistochemistry, FACS, Western blotting and RT-PCR. No differences were detected in the isoform pattern or distribution among the three strains, although the expression level varied according to how many copies of the gene were integrated. The expression profile closely mimicked that observed in humans, including the same pattern of isoform expression as the donor. In addition, tissue-specific isoform expression in the kidney, salivary gland and brain paralleled that observed in man. The transgenic mice expressed low levels of MCP (CD46) on their E, in contrast to humans but in line with most other primates. These mice should be a useful tool to analyse tissue-specific expression, to establish animal models of infections and to characterize the role of MCP (CD46) in reproduction.
Collapse
Affiliation(s)
- C Kemper
- Division of Rheumatology, Washington University School of Medicine, St. Louis 63110-1093, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
The closely related bacterial pathogens Neisseria gonorrhoeae (gonococci, GC) and N. meningitidis (meningococci, MC) initiate infection at human mucosal epithelia. Colonization begins at apical epithelial surfaces with a multistep adhesion cascade, followed by invasion of the host cell, intracellular persistence, transcytosis, and exit. These activities are modulated by the interaction of a panoply of virulence factors with their cognate host cell receptors, and signals are sent from pathogen to host and host to pathogen at multiple stages of the adhesion cascade. Recent advances place us on the verge of understanding the colonization process at a molecular level of detail. In this review we describe the Neisseria virulence factors in the context of epithelial cell biology, placing special emphasis on the signaling functions of type IV pili, pilus-based twitching motility, and the Opa and Opc outermembrane adhesin/invasin proteins. We also summarize what is known about bacterial intracellular trafficking and growth. With the accelerated integration of tools from cell biology, biochemistry, biophysics, and genomics, experimentation in the next few years should bring unprecedented insights into the interactions of Neisseriae with their host.
Collapse
Affiliation(s)
- A J Merz
- Department of Molecular Microbiology & Immunology, L220, Oregon Health Sciences University, Portland, Oregon 97201-3098, USA
| | | |
Collapse
|
23
|
Affiliation(s)
- J Schneider-Schaulies
- Institut für Virologie und Immunbiologie, Universität Würzburg, Verbacher Str. 7, 97078 Würzburg, Germany.
| |
Collapse
|
24
|
Wang G, Liszewski MK, Chan AC, Atkinson JP. Membrane cofactor protein (MCP; CD46): isoform-specific tyrosine phosphorylation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:1839-46. [PMID: 10657632 DOI: 10.4049/jimmunol.164.4.1839] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Membrane cofactor protein (MCP; CD46) is a widely expressed type 1 transmembrane glycoprotein that inhibits complement activation on host cells. It also is a receptor for several pathogens including measles virus, Streptococcus pyogenes, Neisseria gonorrhea, and Neisseria meningitidis. That MCP may have signaling capability was suggested by its microbial interactions. That is, binding of MCP on human monocytes by measles virus hemagglutinin or cross-linking by an anti-MCP Ab resulted in IL-12 down-regulation, while binding to MCP by Neisseria on epithelial cells produced a calcium flux. Through alternative splicing, MCP is expressed on most cells with two distinct cytoplasmic tails of 16 (CYT-1) or 23 (CYT-2) amino acids. These play pivotal roles in intracellular precursor processing and basolateral localization. We investigated the putative signal transduction pathway mediated by MCP and demonstrate that CYT-2, but not CYT-1, is phosphorylated on tyrosine. We examined MCP tail peptides and performed Ab cross-linking experiments on several human cell lines and MCP isoform transfectants. We found an MCP peptide of CYT-2 was phosphorylated by a src kinase system. Western blots of the cells lines demonstrated that cells bearing CYT-2 were also phosphorylated on tyrosine. Additionally, we provide genetic and biochemical evidence that the src family of kinases is responsible for the latter phosphorylation events. In particular, the src kinase, Lck, is required for phosphorylation of MCP in the Jurkat T cell line. Taken together, these studies suggest a src family-dependent pathway for signaling through MCP.
Collapse
Affiliation(s)
- G Wang
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
25
|
Affiliation(s)
- M J Goddard
- Department of Histopathology, Papworth Hospital, Cambridge, UK
| | | | | |
Collapse
|
26
|
Santoro F, Kennedy PE, Locatelli G, Malnati MS, Berger EA, Lusso P. CD46 is a cellular receptor for human herpesvirus 6. Cell 1999; 99:817-27. [PMID: 10619434 DOI: 10.1016/s0092-8674(00)81678-5] [Citation(s) in RCA: 368] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Human herpesvirus 6 (HHV-6) is the etiologic agent of exanthema subitum, causes opportunistic infections in immunocompromised patients, and has been implicated in multiple sclerosis and in the progression of AIDS. Here, we show that the two major HHV-6 subgroups (A and B) use human CD46 as a cellular receptor. Downregulation of surface CD46 was documented during the course of HHV-6 infection. Both acute infection and cell fusion mediated by HHV-6 were specifically inhibited by a monoclonal antibody to CD46; fusion was also blocked by soluble CD46. Nonhuman cells that were resistant to HHV-6 fusion and entry became susceptible upon expression of recombinant human CD46. The use of a ubiquitous immunoregulatory receptor opens novel perspectives for understanding the tropism and pathogenicity of HHV-6.
Collapse
MESH Headings
- Antibodies, Monoclonal/metabolism
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Cell Fusion/genetics
- Cell Fusion/physiology
- Cells, Cultured
- Herpesviridae Infections/metabolism
- Herpesviridae Infections/virology
- Herpesvirus 6, Human/metabolism
- Herpesvirus 6, Human/pathogenicity
- Humans
- Leukocytes, Mononuclear/cytology
- Leukocytes, Mononuclear/metabolism
- Leukocytes, Mononuclear/virology
- Membrane Cofactor Protein
- Membrane Glycoproteins/immunology
- Membrane Glycoproteins/metabolism
- Receptors, Virus/immunology
- Receptors, Virus/metabolism
- Recombinant Proteins/metabolism
- Transfection
- Transgenes/genetics
- Transgenes/physiology
Collapse
Affiliation(s)
- F Santoro
- Unit of Human Virology, DIBIT, San Raffaele Scientific Institute, Milano, Italy
| | | | | | | | | | | |
Collapse
|
27
|
Hsu EC, Sabatinos S, Hoedemaeker FJ, Rose DR, Richardson CD. Use of site-specific mutagenesis and monoclonal antibodies to map regions of CD46 that interact with measles virus H protein. Virology 1999; 258:314-26. [PMID: 10366568 DOI: 10.1006/viro.1999.9712] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Researchers at our laboratory have been dissecting the binding domains of the receptor for the Edmonston laboratory strain of measles virus (CD46) through site-specific mutagenesis. We initially substituted most of the hydrophilic amino acids in the two external short consensus regions (SCRI and SCRII) of CD46 with the amino acid alanine [Hsu et al. (1997) J. Virol. 71:6144-6154] and found that the glutamic-arginine residues at positions 58 and 59 were particularly sensitive to change. Here we consider the roles of hydrophobic amino acids in the binding between measles virus H protein and CD46. Hydrophobic amino acids in the SCRI and SCRII domains of CD46 were systematically replaced with serine. The effects of these changes were monitored through the interaction of Sf9 insect cells expressing the H protein and mouse OST-7 cells synthesizing the mutant CD46 molecules. Binding was quantified through a colorimetric assay for beta-galactosidase that was also produced by the insect cells. Our results indicate that E45, Y54, 58E/R59, Y68, F69, Y101, I102, R103, D104, and Y117 seem to be critical residues for the binding of CD46 to measles virus H protein. The hydrophilic amino acid R59 in SCR1 and hydrophobic residues Y101, I102, and Y117 in SCR2 seem to be especially important for interaction between H protein and CD46. In addition, we mapped the antigenic epitopes of five monoclonal antibodies that are known to inhibit the binding between H protein and CD46. Three of these antibodies recognized regions in SCR1, and two reacted with amino acids in SCR2. For the most part, the determinants recognized by the monoclonal antibody corresponded to the amino acids that were most sensitive to change in the binding process. The SCR1 and SCR2 domains of CD46 were modeled from an analogous region in another complement regulatory protein, factor H, whose three-dimensional structure has been previously reported. Amino acids implicated in binding seem to lie on one planar face of the SCR1 and SCR2 domains. These studies serve as a prelude to understanding the structural interactions that occur between CD46 and the measles virus H protein.
Collapse
MESH Headings
- Amino Acid Sequence
- Amino Acids
- Animals
- Antibodies, Monoclonal/metabolism
- Antigens, CD/chemistry
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Binding Sites
- Cell Line
- Epitope Mapping
- Epitopes, B-Lymphocyte/immunology
- Flow Cytometry
- Hemagglutinins, Viral/genetics
- Hemagglutinins, Viral/metabolism
- Humans
- Immunoblotting
- Measles virus/metabolism
- Membrane Cofactor Protein
- Membrane Glycoproteins/chemistry
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Mice
- Models, Molecular
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- Protein Conformation
- Receptors, Virus/chemistry
- Receptors, Virus/genetics
- Receptors, Virus/metabolism
- Spodoptera/cytology
Collapse
Affiliation(s)
- E C Hsu
- Department of Medical Biophysics, University of Toronto, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | | | | | | | | |
Collapse
|
28
|
Gans HA, Maldonado Y, Yasukawa LL, Beeler J, Audet S, Rinki MM, DeHovitz R, Arvin AM. IL-12, IFN-γ, and T Cell Proliferation to Measles in Immunized Infants. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.9.5569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
Measles infection in infants is associated with severe complications, and secondary infections are attributed to generalized immunosuppression. Measles binding to its monocyte receptor down-regulates IL-12 which is expected to diminish Th1-like cytokine responses, including IFN-γ. Whether young infants can be immunized effectively against measles is an important public health issue. We evaluated Ag-specific IL-12, IFN-γ, and T cell responses of infants at 6 (n = 60), 9 (n = 46), or 12 mo (n = 56) of age and 29 vaccinated adults. IL-12 and IFN-γ release by PBMC stimulated with measles Ag increased significantly after measles immunization in infants. IL-12 and IFN-γ concentrations were equivalent in younger and older infants, but IL-12 concentrations were significantly lower in infants than in adults (p = 0.04). IL-12 production by monocytes was down-regulated by measles; the addition of recombinant human IL-12 enhanced IFN-γ production by PBMC stimulated with measles Ag, but infant T cells released significantly less IFN-γ than adult T cells under this condition. Of particular interest, the presence of passive Abs to measles had no effect on the specific T cell proliferation or IFN-γ production after measles stimulation. Cellular immunity to measles infection and vaccination may be limited in infants compared with adults as a result of less effective IFN-γ and IL-12 production in response to measles Ags. These effects were not exaggerated in younger infants compared with effects in infants who were immunized at 12 mo. In summary, infant T cells were primed with measles Ag despite the presence of passive Abs, but their adaptive immune responses were limited compared with those of adults.
Collapse
Affiliation(s)
- Hayley A. Gans
- *Infectious Diseases Division, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305
| | - Yvonne Maldonado
- *Infectious Diseases Division, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305
| | - Linda L. Yasukawa
- *Infectious Diseases Division, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305
| | - Judy Beeler
- †Department of Health and Human Services, Division of Viral Products, Office of Vaccine Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Rockville, MD 20867; and
| | - Susette Audet
- †Department of Health and Human Services, Division of Viral Products, Office of Vaccine Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Rockville, MD 20867; and
| | - Mary M. Rinki
- *Infectious Diseases Division, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305
| | - Ross DeHovitz
- ‡Department of Pediatrics, Palo Alto Medical Foundation, Palo Alto, CA 94301
| | - Ann M. Arvin
- *Infectious Diseases Division, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
29
|
Hsu EC, Sarangi F, Iorio C, Sidhu MS, Udem SA, Dillehay DL, Xu W, Rota PA, Bellini WJ, Richardson CD. A single amino acid change in the hemagglutinin protein of measles virus determines its ability to bind CD46 and reveals another receptor on marmoset B cells. J Virol 1998; 72:2905-16. [PMID: 9525611 PMCID: PMC109736 DOI: 10.1128/jvi.72.4.2905-2916.1998] [Citation(s) in RCA: 118] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/1997] [Accepted: 12/08/1997] [Indexed: 02/06/2023] Open
Abstract
This paper provides evidence for a measles virus receptor other than CD46 on transformed marmoset and human B cells. We first showed that most tissues of marmosets are missing the SCR1 domain of CD46, which is essential for the binding of Edmonston measles virus, a laboratory strain that has been propagated in Vero monkey kidney cells. In spite of this deletion, the common marmoset was shown to be susceptible to infections by wild-type isolates of measles virus, although they did not support Edmonston measles virus production. As one would expect from these results, measles virus could not be propagated in owl monkey or marmoset kidney cell lines, but surprisingly, both a wild-type isolate (Montefiore 89) and the Edmonston laboratory strain of measles virus grew efficiently in B95-8 marmoset B cells. In addition, antibodies directed against CD46 had no effect on wild-type infections of marmoset B cells and only partially inhibited the replication of the Edmonston laboratory strain in the same cells. A direct binding assay with insect cells expressing the hemagglutinin (H) proteins of either the Edmonston or Montefiore 89 measles virus strains was used to probe the receptors on these B cells. Insect cells expressing Edmonston H but not the wild-type H bound to rodent cells with CD46 on their surface. On the other hand, both the Montefiore 89 H and Edmonston H proteins adhered to marmoset and human B cells. Most wild-type H proteins have asparagine residues at position 481 and can be converted to a CD46-binding phenotype by replacement of the residue with tyrosine. Similarly, the Edmonston H protein did not bind CD46 when its Tyr481 was converted to asparagine. However, this mutation did not affect the ability of Edmonston H to bind marmoset and human B cells. The preceding results provide evidence, through the use of a direct binding assay, that a second receptor for measles virus is present on primate B cells.
Collapse
Affiliation(s)
- E C Hsu
- Department of Medical Biophysics, University of Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Niewiesk S, Schneider-Schaulies J, Ohnimus H, Jassoy C, Schneider-Schaulies S, Diamond L, Logan JS, ter Meulen V. CD46 expression does not overcome the intracellular block of measles virus replication in transgenic rats. J Virol 1997; 71:7969-73. [PMID: 9311889 PMCID: PMC192156 DOI: 10.1128/jvi.71.10.7969-7973.1997] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The study of measles pathogenesis and the testing of improved vaccine candidates is hampered by the lack of a small animal model which is susceptible to infection by the intranasal route. With the identification of CD46 as a measles virus (MV) receptor, it was feasible to generate transgenic rats to overcome this problem. Although there was widespread expression of CD46 in the transgenic Sprague-Dawley rats, no measles-like disease could be induced after various routes of infection. The expressed transgenic protein was functionally intact since it mediated MV fusion and was downregulated by contact with MV hemagglutinin. In vitro studies revealed that CD46-expressing rat fibroblasts take up MV but do not allow viral replication, which explains the nonpermissiveness of the transgenic rats for in vivo infection.
Collapse
Affiliation(s)
- S Niewiesk
- Institut für Virologie und Immunbiologie, Universität Würzburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Hsu EC, Dörig RE, Sarangi F, Marcil A, Iorio C, Richardson CD. Artificial mutations and natural variations in the CD46 molecules from human and monkey cells define regions important for measles virus binding. J Virol 1997; 71:6144-54. [PMID: 9223509 PMCID: PMC191875 DOI: 10.1128/jvi.71.8.6144-6154.1997] [Citation(s) in RCA: 79] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
CD46 was previously shown to be a primate-specific receptor for the Edmonston strain of measles virus. This receptor consists of four short consensus regions (SCR1 to SCR4) which normally function in complement regulation. Measles virus has recently been shown to interact with SCR1 and SCR2. In this study, receptors on different types of monkey erythrocytes were employed as "natural mutant proteins" to further define the virus binding regions of CD46. Erythrocytes from African green monkeys and rhesus macaques hemagglutinate in the presence of measles virus, while baboon erythrocytes were the least efficient of the Old World monkey cells used in these assays. Subsequent studies demonstrated that the SCR2 domain of baboon CD46 contained an Arg-to-Gln mutation at amino acid position 103 which accounted for reduced hemagglutination activity. Surprisingly, none of the New World monkey erythrocytes hemagglutinated in the presence of virus. Sequencing of cDNAs derived from the lymphocytes of these New World monkeys and analysis of their erythrocytes with SCR1-specific polyclonal antibodies indicated that the SCR1 domain was deleted in these cells. Additional experiments, which used 35 different site-specific mutations inserted into CD46, were performed to complement the preceding studies. The effects of these artificial mutations were documented with a convenient binding assay using insect cells expressing the measles virus hemagglutinin. Mutations which mimicked the change found in baboon CD46 or another which deleted the SCR2 glycosylation site reduced binding substantially. Another mutation which altered GluArg to AlaAla at positions 58 and 59, totally abolished binding. Finally, the epitopes for two monoclonal antibodies which inhibit measles virus attachment were mapped to the same regions implicated by mutagenesis.
Collapse
Affiliation(s)
- E C Hsu
- Department of Medical Biophysics, University of Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
32
|
Stephensen CB, Welter J, Thaker SR, Taylor J, Tartaglia J, Paoletti E. Canine distemper virus (CDV) infection of ferrets as a model for testing Morbillivirus vaccine strategies: NYVAC- and ALVAC-based CDV recombinants protect against symptomatic infection. J Virol 1997; 71:1506-13. [PMID: 8995676 PMCID: PMC191207 DOI: 10.1128/jvi.71.2.1506-1513.1997] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Canine distemper virus (CDV) infection of ferrets causes an acute systemic disease involving multiple organ systems, including the respiratory tract, lymphoid system, and central nervous system (CNS). We have tested candidate CDV vaccines incorporating the fusion (F) and hemagglutinin (HA) proteins in the highly attenuated NYVAC strain of vaccinia virus and in the ALVAC strain of canarypox virus, which does not productively replicate in mammalian hosts. Juvenile ferrets were vaccinated twice with these constructs, or with an attenuated live-virus vaccine, while controls received saline or the NYVAC and ALVAC vectors expressing rabies virus glycoprotein. Control animals did not develop neutralizing antibody and succumbed to distemper after developing fever, weight loss, leukocytopenia, decreased activity, conjunctivitis, an erythematous rash typical of distemper, CNS signs, and viremia in peripheral blood mononuclear cells (as measured by reverse transcription-PCR). All three CDV vaccines elicited neutralizing titers of at least 1:96. All vaccinated ferrets survived, and none developed viremia. Both recombinant vaccines also protected against the development of symptomatic distemper. However, ferrets receiving the live-virus vaccine lost weight, became lymphocytopenic, and developed the erythematous rash typical of CDV. These data show that ferrets are an excellent model for evaluating the ability of CDV vaccines to protect against symptomatic infection. Because the pathogenesis and clinical course of CDV infection of ferrets is quite similar to that of other Morbillivirus infections, including measles, this model will be useful in testing new candidate Morbillivirus vaccines.
Collapse
Affiliation(s)
- C B Stephensen
- Department of International Health, School of Public Health, University of Alabama at Birmingham, 35294-0019, USA.
| | | | | | | | | | | |
Collapse
|
33
|
Maisner A, Alvarez J, Liszewski MK, Atkinson DJ, Atkinson JP, Herrler G. The N-glycan of the SCR 2 region is essential for membrane cofactor protein (CD46) to function as a measles virus receptor. J Virol 1996; 70:4973-7. [PMID: 8764003 PMCID: PMC190450 DOI: 10.1128/jvi.70.8.4973-4977.1996] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Membrane cofactor protein (MCP) (CD46), a complement-regulatory protein, serves as a cellular receptor for measles virus. Its amino-terminal portion is composed of four short consensus repeats (SCR), three of which (SCR1, SCR2, and SCR4) carry an N-linked oligosaccharide. In order to determine the importance of the three N-glycans for the function of MCP as a measles virus receptor, we established Chinese hamster ovary (CHO) cell lines that stably express mutant MCPs lacking one of the three motifs for N glycosylation (NQ1, NQ2, and NQ4). In an additional mutant (NQ1-2), two glycosylation motifs were altered, allowing the addition of an N-linked oligosaccharide only in SCR4. The abilities of the mutant MCPs to function as measles virus receptors were analyzed with three different assays: (i) binding of measles virus hemagglutinin to MCP immobilized on nitrocellulose; (ii) binding of measles virus to CHO cells expressing wild-type or mutant MCP; and (iii) infection of the transfected CHO cells by measles virus. In all three assays, the abilities of the NQ2 and NQ1-2 mutants to serve as measles virus receptors were drastically impaired. The NQ1 and NQ4 mutants were recognized by measles virus almost as efficiently as the wild-type protein. These results indicate that the N-glycan attached to SCR2 is essential for MCP to serve as a measles virus receptor, while the oligosaccharides attached to SCR1 and SCR4 are of only minor importance.
Collapse
Affiliation(s)
- A Maisner
- Institut für Virologie, Philipps-Universität Marburg, Germany
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
The complement system has developed a remarkably simple but elegant manner of regulating itself. It has faced and successfully dealt with how to facilitate activation on a microbe while preventing the same on host tissue. It solved this problem primarily by creating a series of secreted and membrane-regulatory proteins that prevent two highly undesirable events: activation in the fluid phase (no target) and on host tissue (inappropriate target). Also, if not checked, even on an appropriate target, the system would go to exhaustion and have nothing left for the next microbe. Therefore, the complement enzymes have an intrinsic instability and the fluid-phase control proteins play a major role in limiting activation in time. The symmetry of the regulatory process between fluid phase and membrane inhibitors at the C4/C3 step of amplification and convertase formation as well as at the MAC steps are particularly striking features of the self/nonself discrimination system. The use of glycolipid anchored proteins on membranes to decay enzymes and block membrane insertion events is unlikely to be by chance. Finally, it is economical for the cofactor regulatory activity to produce derivatives of C3b that now specifically engage additional receptors. Likewise, C1-Inh leads to C1q remaining on the immune complex to interact with the C1q receptor. Thus the complement system is designed to allow rapid, efficient, unimpeded activation on an appropriate foreign target while regulatory proteins intervene to prevent three undesirable consequences of complement activation: excessive activation on a single target, fluid phase activation, and activation on self.
Collapse
Affiliation(s)
- M K Liszewski
- Department of Medicine, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | | | | | | | | |
Collapse
|
35
|
Gerlier D, Varior-Krishnan G, Devaux P. CD46-mediated measles virus entry: a first key to host-range specificity. Trends Microbiol 1995; 3:338-45. [PMID: 8520886 DOI: 10.1016/s0966-842x(00)88972-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Humans are the sole natural host of measles virus. The identification of CD46 as a virus receptor and of the involvement of moesin sheds some light on the molecular events occurring during virus entry into the cell. Knowledge of the key role of CD46 paves the way to creating transgenic mice sensitive to measles virus infection.
Collapse
Affiliation(s)
- D Gerlier
- Laboratoire Immunité et Infections Virales, I.V.M.C. CNRS-UCBL UMR30, Faculté de Médecine Alexis Carrel, Lyon, France
| | | | | |
Collapse
|