Measles virus glycoproteins suppress antigen-specific responses in vivo

1997 ◽  
Vol 56 (1-3) ◽  
pp. 355
Author(s):  
S Niewiesk
Keyword(s):  
2014 ◽  
Vol 149 (1) ◽  
pp. 99-108 ◽  
Author(s):  
Yuqi Jing ◽  
Marcela Toro Bejarano ◽  
Julia Zaias ◽  
Jaime R. Merchan

2014 ◽  
Vol 89 (2) ◽  
pp. 1143-1155 ◽  
Author(s):  
C. Mathieu ◽  
D. Huey ◽  
E. Jurgens ◽  
J. C. Welsch ◽  
I. DeVito ◽  
...  

ABSTRACTMeasles virus (MV) infection is undergoing resurgence and remains one of the leading causes of death among young children worldwide despite the availability of an effective measles vaccine. MV infects its target cells by coordinated action of the MV H and the fusion (F) envelope glycoprotein; upon receptor engagement by H, the prefusion F undergoes a structural transition, extending and inserting into the target cell membrane and then refolding into a postfusion structure that fuses the viral and cell membranes. By interfering with this structural transition of F, peptides derived from the heptad-repeat (HR) regions of F can potently inhibit MV infection at the entry stage. We show here that specific features of H's interaction with its receptors modulate the susceptibility of MV F to peptide fusion inhibitors. A higher concentration of inhibitory peptides is required to inhibit F-mediated fusion when H is engaged to its nectin-4 receptor than when H is engaged to its CD150 receptor. Peptide inhibition of F may be subverted by continued engagement of receptor by H, a finding that highlights the ongoing role of H-receptor interaction after F has been activated and that helps guide the design of more potent inhibitory peptides. Intranasal administration of these peptides results in peptide accumulation in the airway epithelium with minimal systemic levels of peptide and efficiently prevents MV infectionin vivoin animal models. The results suggest an antiviral strategy for prophylaxis in vulnerable and/or immunocompromised hosts.IMPORTANCEMeasles virus (MV) infection causes an acute illness that may be associated with infection of the central nervous system (CNS) and severe neurological disease. No specific treatment is available. We have shown that parenterally delivered fusion-inhibitory peptides protect mice from lethal CNS MV disease. Here we show, using established small-animal models of MV infection, that fusion-inhibitory peptides delivered intranasally provide effective prophylaxis against MV infection. Since the fusion inhibitors are stable at room temperature, this intranasal strategy is feasible even outside health care settings, could be used to protect individuals and communities in case of MV outbreaks, and could complement global efforts to control measles.


2005 ◽  
Vol 79 (18) ◽  
pp. 11824-11836 ◽  
Author(s):  
Mingzhou Chen ◽  
Jean-Claude Cortay ◽  
Ian R. Logan ◽  
Vasileia Sapountzi ◽  
Craig N. Robson ◽  
...  

ABSTRACT Using a C-terminal domain (PCT) of the measles virus (MV) phosphoprotein (P protein) as bait in a yeast two-hybrid screen, a cDNA identical to the recently described human p53-induced-RING-H2 (hPIRH2) cDNA was isolated. A glutathione S-transferase-hPIRH2 fusion protein expressed in bacteria was able to pull down P protein when mixed with an extract from P-expressing HeLa cells in vitro, and myc-tagged hPIRH2 could be reciprocally coimmunoprecipitated with MV P protein from human cells. Additionally, immunoprecipitation experiments demonstrated that hPIRH2-myc, MV P, and nucleocapsid (N) proteins form a ternary complex. The hPIRH2 binding site was mapped to the C-terminal X domain region of the P protein by using a yeast two-hybrid assay. The PCT binding site was mapped on hPIRH2 by using a novel yeast two-hybrid tagged PCR approach and by coimmunoprecipitation of hPIRH2 cysteine mutants and mouse/human PIRH2 chimeras. The hPIRH2 C terminus could mediate the interaction with MV P which was favored by the RING-H2 motif. When coexpressed with an enhanced green fluorescent protein-tagged hPIRH2 protein, MV P alone or in a complex with MV N was able to redistribute hPIRH2 to outside the nucleus, within intracellular aggregates. Finally, MV P efficiently stabilized hPIRH2-myc expression and prevented its ubiquitination in vivo but had no effect on the stability or ubiquitination of an alternative ubiquitin E3 ligase, Mdm2. Thus, MV P protein is the first protein from a pathogen that is able to specifically interact with and stabilize the ubiquitin E3 ligase hPIRH2 by preventing its ubiquitination.


2009 ◽  
Vol 83 (11) ◽  
pp. 5544-5555 ◽  
Author(s):  
Thomas Carsillo ◽  
Mary Carsillo ◽  
Zachary Traylor ◽  
Päivi Rajala-Schultz ◽  
Phillip Popovich ◽  
...  

ABSTRACT In vitro studies show that hsp70 promotes gene expression for multiple viral families, although there are few reports on the in vivo significance of virus-hsp70 interaction. Previously we showed that hsp70-dependent stimulation of Edmonston measles virus (Ed MeV) transcription caused an increased cytopathic effect and mortality in transgenic hsp70-overexpressing C57BL/6 mice (H-2 b ). The response to MeV infection is influenced by the major histocompatibility complex haplotype; H-2 d mice are resistant to brain infection due to robust antiviral immune responses, whereas H-2 b mice are susceptible due to deficiencies in this response. We therefore tested the hypothesis that the outcome of MeV-hsp70 interaction may be dependent upon the host H-2 haplotype. The impact of selective neuronal hsp70 overexpression on Ed MeV brain infection was tested with congenic C57BL/10 H-2 d neonatal mice. In this context, hsp70 overexpression conferred complete protection against virus-induced mortality, compared to >30% mortality in nontransgenic mice. Selective depletion of T-cell populations showed that transgenic mice exhibit a diminished reliance on T cells for protection. Brain transcript analysis indicated enhanced innate immune activation and signaling through Toll-like receptors 2 and 4 at early times postinfection for transgenic infected mice relative to those for nontransgenic infected mice. Collectively, results suggest that hsp70 can enhance innate antiviral immunity through Toll-like receptor signaling, supporting a protective role for physiological responses that enhance tissue levels of hsp70 (e.g., fever), and that the H-2 haplotype determines the effectiveness of this response.


2014 ◽  
Vol 89 (4) ◽  
pp. 2192-2200 ◽  
Author(s):  
Linda J. Rennick ◽  
Rory D. de Vries ◽  
Thomas J. Carsillo ◽  
Ken Lemon ◽  
Geert van Amerongen ◽  
...  

ABSTRACTAlthough live-attenuated measles virus (MV) vaccines have been used successfully for over 50 years, the target cells that sustain virus replicationin vivoare still unknown. We generated a reverse genetics system for the live-attenuated MV vaccine strain Edmonston-Zagreb (EZ), allowing recovery of recombinant (r)MVEZ. Three recombinant viruses were generated that contained the open reading frame encoding enhanced green fluorescent protein (EGFP) within an additional transcriptional unit (ATU) at various positions within the genome. rMVEZEGFP(1), rMVEZEGFP(3), and rMVEZEGFP(6) contained the ATU upstream of the N gene, following the P gene, and following the H gene, respectively. The viruses were comparedin vitroby growth curves, which indicated that rMVEZEGFP(1) was overattenuated. Intratracheal infection of cynomolgus macaques with these recombinant viruses revealed differences in immunogenicity. rMVEZEGFP(1) and rMVEZEGFP(6) did not induce satisfactory serum antibody responses, whereas bothin vitroandin vivorMVEZEGFP(3) was functionally equivalent to the commercial MVEZ-containing vaccine. Intramuscular vaccination of macaques with rMVEZEGFP(3) resulted in the identification of EGFP+cells in the muscle at days 3, 5, and 7 postvaccination. Phenotypic characterization of these cells demonstrated that muscle cells were not infected and that dendritic cells and macrophages were the predominant target cells of live-attenuated MV.IMPORTANCEEven though MV strain Edmonston-Zagreb has long been used as a live-attenuated vaccine (LAV) to protect against measles, nothing is known about the primary cells in which the virus replicatesin vivo. This is vital information given the push to move toward needle-free routes of vaccination, since vaccine virus replication is essential for vaccination efficacy. We have generated a number of recombinant MV strains expressing enhanced green fluorescent protein. The virus that best mimicked the nonrecombinant vaccine virus was formulated according to protocols for production of commercial vaccine virus batches, and was subsequently used to assess viral tropism in nonhuman primates. The virus primarily replicated in professional antigen-presenting cells, which may explain why this LAV is so immunogenic and efficacious.


Virology ◽  
1987 ◽  
Vol 157 (2) ◽  
pp. 497-508 ◽  
Author(s):  
Timothy C. Wong ◽  
Gregory Wipf ◽  
Akiko Hirano

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3757-3757
Author(s):  
Bella Patel ◽  
Dey Aditi ◽  
Ehsan Ghorani ◽  
Yogesh Malam ◽  
Steele Andy ◽  
...  

Abstract Abstract 3757 Replicating viruses that selectively lyse transformed cells are attractive agents for cancer therapy. The vaccine strain of measles virus has proven oncolytic activity in various murine models of malignancy including myeloma and lymphoma. These pre-clinical reports of MV efficacy have led to advanced phase clinical trial. In the study here we investigate the anti-tumour potential of MV in 2 novel disease targets:- adult B lineage acute lymphoblastic leukaemia (ALL) and Chronic lymphocytic leukaemia (CLL) using in-vitro and in-vivo models. MV derived from the Edmonston strain genetically engineered to express GFP was used to infect primary ALL (n = 6) and chronic lymphocytic leukaemia (CLL, n = 7) cells. All CLL and ALL cells expressed the MV receptor CD46 and were efficiently infected by MV-GFP as indicated by quantitation of viral nucleocapsid mRNA by RQ-PCR and immunoblotting of viral proteins N and H. Large multinucleated syncytia, characteristic of MV- induced cytopathology, were found in all infected ALL cultures, by contrast syncitium formation was much less prominent in the infected CLL specimens. Despite this, both CLL and ALL cells were efficiently killed by MV-GFP, as characterised by viability assays and immunoblotting for PARP cleavage. To further probe the contribution of cell to cell fusion in MV induced oncolysis we used a relatively non-fusogenic strain of MV:- MV-Moraten to infect CLL and ALL specimens. As expected ALL and CLL cells infected with MV-Moraten lacked the typical features of MV induced cytopathology. Despite this cell viability was markedly reduced in both ALL and CLL cultures infected with MV-Moraten compared to uninfected controls suggesting that intracellular fusion might be dispensable for MV-induced oncolysis in our two models. To test whether MV had therapeutic efficacy in-vivo we established subcutaneous xenografts of pre-B ALL in CB17/SCID mice using the Nalm-6 cell line and administered 1 × 107 pfu of MV (n==12) or UV inactivated MV (n=12) intratumorally on 10 occasions. In vivo MV administration had striking antitumour activity resulting in complete resolution of 11/12 or regression (1/12) of established subcutaneous pre-B ALL tumours by week 4. In contrast, all UV-MV treated tumours progressed. The differences in tumour growth between the MV treated and UV-MV control groups was significantly different (p < 0.0001, Figure 1). To test for MV-induced oncolysis in a model more closely related to ALL in humans we used a disseminated pre-B ALL model established by tail vein injection of 1 × 106 Nalm-6 cells. 1 × 106 pfu of MV or UV-MV was administered by the intravenous route six times. Eleven of twelve mice receiving replication competent MV remain disease free whereas 6/7 mice receiving tail vein administered UV MV had become moribund by 67 days (Figure 2). Bone marrow examination of moribund mice revealed 52 – 99% of CD19+/CD10+ Nalm-6 cells present. Overall, our data suggest that both ALL and CLL are targets for MV-mediated lysis in vitro. The significant antineoplastic activity of MV against both subcutaneous and disseminated ALL xenografts holds great promise towards developing vaccine MV as a therapeutic tool in adult ALL. Figure 1 Regression of Nalm-6 subcutaneous xenografts in SCID mice after intratumoral injection of MV. Figure 1. Regression of Nalm-6 subcutaneous xenografts in SCID mice after intratumoral injection of MV. Figure 2 Prolonged survival of disseminated Nalm-6 SCID xenografts after intravenous injection of MV. Figure 2. Prolonged survival of disseminated Nalm-6 SCID xenografts after intravenous injection of MV. Regression of Nalm-6 subcutaneous xenografts in SCID mice after intratumoral injection of MV. Prolonged survival of disseminated Nalm-6 SCID xenografts after intravenous injection of MV. Disclosures: No relevant conflicts of interest to declare.


2015 ◽  
Vol 35 (4) ◽  
pp. 292-301 ◽  
Author(s):  
Rupak Shivakoti ◽  
Debra Hauer ◽  
Robert J. Adams ◽  
Wen-Hsuan W. Lin ◽  
William Paul Duprex ◽  
...  

2006 ◽  
Vol 87 (10) ◽  
pp. 2767-2779 ◽  
Author(s):  
Yusuke Yanagi ◽  
Makoto Takeda ◽  
Shinji Ohno

Measles virus (MV), a member of the genus Morbillivirus in the family Paramyxoviridae, is an enveloped virus with a non-segmented, negative-strand RNA genome. It has two envelope glycoproteins, the haemagglutinin (H) and fusion proteins, which are responsible for attachment and membrane fusion, respectively. Human signalling lymphocyte activation molecule (SLAM; also called CD150), a membrane glycoprotein of the immunoglobulin superfamily, acts as a cellular receptor for MV. SLAM is expressed on immature thymocytes, activated lymphocytes, macrophages and dendritic cells and regulates production of interleukin (IL)-4 and IL-13 by CD4+ T cells, as well as production of IL-12, tumour necrosis factor alpha and nitric oxide by macrophages. The distribution of SLAM is in accord with the lymphotropism and immunosuppressive nature of MV. Canine distemper virus and Rinderpest virus, other members of the genus Morbillivirus, also use canine and bovine SLAM as receptors, respectively. Laboratory-adapted MV strains may use the ubiquitously expressed CD46, a complement-regulatory molecule, as an alternative receptor through amino acid substitutions in the H protein. Furthermore, MV can infect SLAM− cells, albeit inefficiently, via the SLAM- and CD46-independent pathway, which may account for MV infection of epithelial, endothelial and neuronal cells in vivo. MV infection, however, is not determined entirely by the H protein–receptor interaction, and other MV proteins can also contribute to its efficient growth by facilitating virus replication at post-entry steps. Identification of SLAM as the principal receptor for MV has provided us with an important clue for better understanding of MV tropism and pathogenesis.


Sign in / Sign up

Export Citation Format

Share Document