Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Publication year range
1.
J Virol ; 94(15)2020 07 16.
Article in English | MEDLINE | ID: mdl-32461318

ABSTRACT

Live oral rotavirus vaccines have been developed by serial passaging in cell culture and found to be safe in infants. However, mechanisms for the adaptation and attenuation of rotavirus vaccines are not fully understood. We prepared a human rotavirus vaccine strain, CDC-9 (G1P[8]), which when grown in MA104 cells to passage 11 or 12 (P11/P12) had no nucleotide or amino acid sequence changes from the original virus in stool. Upon adaptation and passages in Vero cells, the strain underwent five amino acid changes at P28 and one additional change at P44/P45 in the VP4 gene. We performed virologic, immunological, and pathogenic characterization of wild-type CDC-9 virus at P11/P12 and its two mutants at P28 or P44/P45 using in vitro and in vivo model systems. We found that mutants CDC-9 P28 and P44 induced upregulated expression of immunomodulatory cytokines. On the other hand, the two mutant viruses induced lower STAT1 phosphorylation and grew to 2-log-higher titers than wild-type virus in human Caco-2 cells and simian Vero cells. In neonatal rats, CDC-9 P45 showed reduced rotavirus shedding in fecal specimens and did not induce diarrhea compared to wild-type virus and modulated cytokine responses comparably to Rotarix infection. These findings indicate that mutant CDC-9 is attenuated and safe. Our study is the first to provide insight into the possible mechanisms of human rotavirus adaptation and attenuation and supports ongoing efforts to develop CDC-9 as a new generation of rotavirus vaccine for live oral or parenteral administration.IMPORTANCE Mechanisms for in vitro adaptation and in vivo attenuation of human rotavirus vaccines are not known. The present study is the first to comprehensively compare the in vitro growth characteristics, virulence, and host response of a wild-type and an attenuated human rotavirus strain, CDC-9, in Caco-2 cells and neonatal rats. Our study identifies critical sequence changes in the genome that render human rotavirus adapted to growth to high levels in Vero cells and attenuated and safe in neonatal rats; thus, the study supports clinical development of CDC-9 for oral or parenteral vaccination in children.


Subject(s)
Capsid Proteins/metabolism , Mutation, Missense , Rotavirus Vaccines/metabolism , Rotavirus/growth & development , Amino Acid Substitution , Animals , Caco-2 Cells , Capsid Proteins/genetics , Chlorocebus aethiops , Humans , Rotavirus/genetics , Rotavirus Vaccines/genetics , Vaccines, Attenuated/genetics , Vaccines, Attenuated/metabolism , Vero Cells
2.
J Virol ; 93(18)2019 09 15.
Article in English | MEDLINE | ID: mdl-31243129

ABSTRACT

Group A rotaviruses, an important cause of severe diarrhea in children and young animals, initiate infection via interactions of the VP8* domain of the VP4 spike protein with cell surface sialic acids (SAs) or histo-blood group antigens (HBGAs). Although the bovine G6P[5] WC3 strain is an important animal pathogen and is also used in the bovine-human reassortant RotaTeq vaccine, the receptor(s) for the VP8* domain of WC3 and its reassortant strains have not yet been identified. In the present study, HBGA- and saliva-binding assays showed that both G6P[5] WC3 and mono-reassortant G4P[5] strains recognized the αGal HBGA. The infectivity of both P[5]-bearing strains was significantly reduced in αGal-free MA-104 cells by pretreatment with a broadly specific neuraminidase or by coincubation with the α2,6-linked SA-specific Sambucus nigra lectin, but not by the α2,3-linked specific sialidase or by Maackia amurensis lectin. Free NeuAc and the αGal trisaccharide also prevented the infectivity of both strains. This indicated that both P[5]-bearing strains utilize α2,6-linked SA as a ligand on MA104 cells. However, the two strains replicated in differentiated bovine small intestinal enteroids and in their human counterparts that lack α2,6-linked SA or αGal HBGA, suggesting that additional or alternative receptors such as integrins, hsp70, and tight-junction proteins bound directly to the VP5* domain can be used by the P[5]-bearing strains to initiate the infection of human cells. In addition, these data also suggested that P[5]-bearing strains have potential for cross-species transmission.IMPORTANCE Group A rotaviruses initiate infection through the binding of the VP8* domain of the VP4 protein to sialic acids (SAs) or histo-blood group antigens (HBGAs). Although the bovine G6P[5] WC3 strain is an important animal pathogen and is used as the backbone in the bovine-human reassortant RotaTeq vaccine, the receptor(s) for their P[5] VP8* domain has remained elusive. Using a variety of approaches, we demonstrated that the WC3 and bovine-human mono-reassortant G4P[5] vaccine strains recognize both α2,6-linked SA and αGal HBGA as ligands. Neither ligand is expressed on human small intestinal epithelial cells, explaining the absence of natural human infection by P[5]-bearing strains. However, we observed that the P[5]-bearing WC3 and G4P[5] RotaTeq vaccine strains could still infect human intestinal epithelial cells. Thus, the four P[5] RotaTeq vaccine strains potentially binding to additional alternative receptors may be efficient and effective in providing protection against severe rotavirus disease in human.


Subject(s)
Capsid Proteins/immunology , Rotavirus/immunology , Rotavirus/metabolism , Amino Acid Sequence/genetics , Animals , Blood Group Antigens/metabolism , Capsid Proteins/metabolism , Cattle/immunology , Epitopes/metabolism , Humans , N-Acetylneuraminic Acid/metabolism , Receptors, Virus/metabolism , Rotavirus Infections/virology , Rotavirus Vaccines/genetics , Rotavirus Vaccines/metabolism , Vaccines, Attenuated/genetics , Vaccines, Attenuated/metabolism , Viral Nonstructural Proteins/metabolism , Virus Attachment , alpha-Galactosidase/metabolism
3.
Mol Biotechnol ; 54(3): 1010-20, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23430460

ABSTRACT

The aim of this study was to evaluate the usefulness of replicating but non disseminating adenovirus vectors (AdVs) as vaccine vector using human rotavirus (HRV) as a model pathogen. HRV VP7, VP4, or VP4Δ (N-terminal 336 amino acids of VP4) structural proteins as well as the VP4Δ::VP7 chimeric fusion protein were expressed in mammalian cells when delivered with the AdVs. A preliminary experiment demonstrated that VP4Δ was able to induce a HRV-specific IgG response in BALB/c mice inoculated intramuscularly with AdVs expressing the rotaviral protein. Moreover, an AdV-prime/plasmid DNA-boost regimen of vectors resulted in VP4Δ-specific antibody (Ab) titers ~4 times higher than those obtained from mice immunized with AdVs alone. Subsequently, the various HRV protein-encoding AdVs were compared using the AdV-prime/plasmid DNA-boost regimen. Higher IgG and IgA responses to HRV were obtained when VP4Δ::VP7 fusion protein was used as an immunogen as compared to VP7 or VP4 alone or to a mix of both proteins delivered independently by AdVs. A synergetic effect in terms of Ab was obtained with VP4Δ::VP7. In conclusion, this study demonstrated for the first time the suitability of using replicating but non disseminating AdVs as vaccine vector and the VP4Δ::VP7 fusion protein as an immunogen for vaccination against HRV.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/genetics , Recombinant Fusion Proteins/immunology , Rotavirus Infections/immunology , Rotavirus Vaccines/immunology , Rotavirus/immunology , Analysis of Variance , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antigens, Viral/chemistry , Antigens, Viral/genetics , Antigens, Viral/immunology , Antigens, Viral/metabolism , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/immunology , Capsid Proteins/metabolism , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rotavirus/genetics , Rotavirus Infections/prevention & control , Rotavirus Infections/virology , Rotavirus Vaccines/chemistry , Rotavirus Vaccines/genetics , Rotavirus Vaccines/metabolism , Transfection
4.
Microb Cell Fact ; 10: 33, 2011 May 14.
Article in English | MEDLINE | ID: mdl-21569612

ABSTRACT

BACKGROUND: Virus-like particles (VLP) have an increasing range of applications including vaccination, drug delivery, diagnostics, gene therapy and nanotechnology. These developments require large quantities of particles that need to be obtained in efficient and economic processes. Production of VLP in yeast is attractive, as it is a low-cost protein producer able to assemble viral structural proteins into VLP. However, to date only single-layered VLP with simple architecture have been produced in this system. In this work, the first steps required for the production of rotavirus-like particles (RLP) in S. cerevisiae were implemented and improved, in order to obtain the recombinant protein concentrations required for VLP assembly. RESULTS: The genes of the rotavirus structural proteins VP2, VP6 and VP7 were cloned in four Saccharomyces cerevisiae strains using different plasmid and promoter combinations to express one or three proteins in the same cell. Performance of the best constructs was evaluated in batch and fed-batch cultures using a complete synthetic media supplemented with leucine, glutamate and succinate. The strain used had an important effect on recombinant protein concentration, while the type of plasmid, centromeric (YCp) or episomal (YEp), did not affect protein yields. Fed-batch culture of the PD.U-267 strain resulted in the highest concentration of rotavirus proteins. Volumetric and specific productivities increased 28.5- and 11-fold, respectively, in comparison with batch cultures. Expression of the three rotavirus proteins was confirmed by immunoblotting and RLP were detected using transmission electron microscopy. CONCLUSIONS: We present for the first time the use of yeast as a platform to express multilayered rotavirus-like particles. The present study shows that the combined use of molecular and bioprocess tools allowed the production of triple-layered rotavirus RLP. Production of VLP with complex architecture in yeasts could lead to the development of new vaccine candidates with reduced restrictions by regulatory agencies, using the successful experience with other yeast-based VLP vaccines commercialized worldwide.


Subject(s)
Rotavirus Vaccines/metabolism , Saccharomyces cerevisiae/metabolism , Vaccines, Virus-Like Particle/metabolism , Cesium/chemistry , Chlorides/chemistry , Kinetics , Plasmids/chemistry , Plasmids/metabolism , Promoter Regions, Genetic , Rotavirus/genetics , Rotavirus/immunology , Rotavirus Vaccines/genetics , Rotavirus Vaccines/immunology , Saccharomyces cerevisiae/growth & development , Vaccines, Virus-Like Particle/genetics , Vaccines, Virus-Like Particle/immunology
6.
Virology ; 356(1-2): 171-8, 2006.
Article in English | MEDLINE | ID: mdl-16956640

ABSTRACT

Major efforts have been put forth for the development of effective rotavirus vaccines including transgenic plant vaccines. Previous studies have reported that rotavirus VP7 maintains its neutralizing immunity when it is transformed into the potato genome. The present study was aimed at investigating the hereditary stability of VP7-transformed potatoes over fifty generations. The VP7 gene was stably transcribed and expressed in potato cells as detected by RT-PCR and Western blotting. Humeral and mucosal responses were successfully induced in BALB/c mice fed with the fiftieth generation transformed potato tubers. There were no significant differences in serum IgG and fecal IgA between the mice fed with the first and fiftieth generation potatoes (P>0.05). Profiles of cytokines such as IFN-gamma, IL-2, IL-4, IL-5 and TGF-beta in immunized mice showed a naive T-cells bias to Th1 and Th3 polarization. Moreover, specific CTL responses were also detected in C57BL/6 mice fed with transformed potatoes. This research represents a significant step towards the development of rotavirus vaccines derived from a transgenic plant that can be obtained by long-term and large-scale vegetative reproduction. To our knowledge, this is the first finding regarding vaccines derived from plants that can be propagated for many generations.


Subject(s)
Antigens, Viral/genetics , Capsid Proteins/genetics , Plants, Genetically Modified/genetics , Rotavirus Vaccines/immunology , Solanum tuberosum/genetics , Vaccines, Edible/immunology , Animals , Antigens, Viral/metabolism , Capsid Proteins/metabolism , Cytokines/metabolism , Immunoglobulin A, Secretory/analysis , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plants, Genetically Modified/metabolism , Rotavirus , Rotavirus Infections/immunology , Rotavirus Infections/prevention & control , Rotavirus Infections/virology , Rotavirus Vaccines/administration & dosage , Rotavirus Vaccines/genetics , Rotavirus Vaccines/metabolism , Solanum tuberosum/metabolism , T-Lymphocytes, Cytotoxic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...