Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.477
Filter
1.
Arch Virol ; 169(6): 131, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38819530

ABSTRACT

Noroviruses (NoVs) are the chief cause of acute viral gastroenteritis worldwide. By employing the major capsid protein VP1 of a GII.6 NoV strain as an immunogen, we generated two monoclonal antibodies (mAbs) with wide-spectrum binding activities against NoV genogroup II (GII) VP1 proteins. One mAb (10G7) could bind to native and denatured GII-specific VP1 proteins. The other mAb (10F2) could bind to all tested native GII VP1 proteins, but not to denatured GII.3, GII.4, GII.7, or GII.17 VP1 proteins. Using GII.6/GII.4 fusion proteins, the mAb 10F2 binding region was confirmed to be located in the C-terminal P1 domain. An enzyme-linked immunosorbent assay based on peptides covering the P domain did not detect any binding. Using a panel of VP1 proteins with swapped regions, deletions, and mutations, the mAb 10F2 binding region was determined to be located between residues 496 and 513. However, the residue(s) responsible for its varied binding affinity for different denatured GII VP1 proteins remain to be identified. In summary, two NoV GII-specific cross-reactive mAbs were generated, and their binding regions were determined. Our results might facilitate the detection and immunogenic study of NoVs.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , Capsid Proteins , Epitopes , Norovirus , Norovirus/genetics , Norovirus/immunology , Antibodies, Monoclonal/immunology , Capsid Proteins/immunology , Capsid Proteins/genetics , Capsid Proteins/chemistry , Epitopes/immunology , Epitopes/genetics , Antibodies, Viral/immunology , Animals , Antigens, Viral/immunology , Antigens, Viral/genetics , Mice , Humans , Caliciviridae Infections/virology , Caliciviridae Infections/immunology , Mice, Inbred BALB C , Epitope Mapping , Cross Reactions
2.
Methods Mol Biol ; 2786: 289-300, 2024.
Article in English | MEDLINE | ID: mdl-38814400

ABSTRACT

In this protocol, we outline how to produce a chimeric viral vaccine in a biosafety level 1 (BSL1) environment. An animal viral vector RNA encapsidated with tobacco mosaic virus (TMV) coat protein can be fully assembled in planta. Agrobacterium cultures containing each component are inoculated together into tobacco leaves and the self-assembled hybrid chimeric viral vaccine is harvested 4 days later and purified with a simple PEG precipitation. The viral RNA delivery vector is derived from the BSL1 insect virus, Flock House virus (FHV), and replicates in human and animal cells but does not spread systemically. A polyethylene glycol purification protocol is also provided to collect and purify these vaccines for immunological tests. In this update, we also provide a protocol for in trans co-inoculation of a modified FHV protein A, which significantly increased the yield of in planta chimeric viral vaccine.


Subject(s)
Nicotiana , Replicon , Tobacco Mosaic Virus , Viral Vaccines , Nicotiana/genetics , Viral Vaccines/immunology , Viral Vaccines/genetics , Animals , Tobacco Mosaic Virus/genetics , Tobacco Mosaic Virus/immunology , Replicon/genetics , RNA, Viral/genetics , Genetic Vectors/genetics , Nodaviridae/genetics , Nodaviridae/immunology , Plants, Genetically Modified/genetics , Capsid Proteins/genetics , Capsid Proteins/immunology , Agrobacterium/genetics , Humans
3.
Virulence ; 15(1): 2360133, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38803081

ABSTRACT

Norovirus (NV) infection causes acute gastroenteritis in children and adults. Upon infection with NV, specific CD8+ T cells, which play an important role in anti-infective immunity, are activated in the host. Owing to the NV's wide genotypic variability, it is challenging to develop vaccines with cross-protective abilities against infection. To aid effective vaccine development, we examined specific CD8+ T-cell responses towards viral-structural protein (VP) epitopes, which enable binding to host susceptibility receptors. We isolated peripheral blood mononuclear cells from 196 participants to screen and identify predominant core peptides towards NV main and small envelope proteins using ex vivo and in vitro intracellular cytokine staining assays. Human leukocyte antigen (HLA) restriction characteristics were detected using next-generation sequencing. Three conservative immunodominant VP-derived CD8+ T-cell epitopes, VP294-102 (TDAARGAIN), VP2153-161 (RGPSNKSSN), and VP1141-148 (FPHIIVDV), were identified and restrictively presented by HLA-Cw * 0102, HLA-Cw * 0702, and HLA-A *1101 alleles, separately. Our findings provide useful insights into the development of future vaccines and treatments for NV infection.


Subject(s)
CD8-Positive T-Lymphocytes , Caliciviridae Infections , Capsid Proteins , Epitopes, T-Lymphocyte , Gastroenteritis , Norovirus , Humans , CD8-Positive T-Lymphocytes/immunology , Capsid Proteins/immunology , Capsid Proteins/genetics , Caliciviridae Infections/immunology , Caliciviridae Infections/virology , Norovirus/immunology , Norovirus/genetics , Adult , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Male , Gastroenteritis/virology , Gastroenteritis/immunology , Female , Middle Aged , Young Adult , Child , Adolescent , Leukocytes, Mononuclear/immunology , Immunodominant Epitopes/immunology , Child, Preschool , Aged
5.
ACS Nano ; 18(21): 13755-13767, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38752610

ABSTRACT

The ability to manipulate the self-assembly of proteins is essential to understanding the mechanisms of life and beneficial to fabricating advanced nanomaterials. Here, we report the transformation of the MS2 phage capsid from nanocages to nanotubes and then to nanotube hydrogels through simple point mutations guided by interfacial interaction redesign. We demonstrate that site 70, which lies in the flexible FG loop of the capsid protein (CP), is a "magic" site that can largely dictate the final morphology of assemblies. By varying the amino acid at site 70, with the aid of a cysteine-to-alanine mutation at site 46, we achieved the assembly of double-helical or single-helical nanotubes in addition to nanocages. Furthermore, an additional cysteine substitution on the surface of nanotubes mediated their cross-linking to form hydrogels with reducing agent responsiveness. The hierarchical self-assembly system allowed for the investigation of morphology-related immunogenicity of MS2 CPs, which revealed dramatic differences among nanocages, nanotubes, and nanotube hydrogels in terms of immune response types, antibody levels and T cell functions. This study provides insights into the assembly manipulation of protein nanomaterials and the customized design of nanovaccines and drug delivery systems.


Subject(s)
Capsid Proteins , Capsid , Hydrogels , Nanotubes , Hydrogels/chemistry , Nanotubes/chemistry , Capsid Proteins/chemistry , Capsid Proteins/immunology , Capsid Proteins/genetics , Capsid/chemistry , Capsid/immunology , Levivirus/chemistry , Levivirus/immunology , Levivirus/genetics , Animals , Nanostructures/chemistry , Mice , Models, Molecular
6.
Dev Comp Immunol ; 157: 105189, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38692524

ABSTRACT

Grass carp, one of the major freshwater aquaculture species in China, is susceptible to grass carp reovirus (GCRV). GCRV is a non-enveloped RNA virus and has a double-layered capsid, causing hemorrhagic disease and high mortalities in infected fish. However, the tropism of GCRV infection has not been investigated. In this study, monoclonal antibodies against recombinant VP35 protein were generated in mice and characterized. The antibodies exhibited specific binding to the N terminal region (1-155 aa) of the recombinant VP35 protein expressed in the HEK293 cells, and native VP35 protein in the GCRV-II infected CIK cells. Immunofluorescent staining revealed that viruses aggregated in the cytoplasm of infected cells. In vivo challenge experiments showed that high levels of GCRV-II viruses were present in the gills, intestine, spleen and liver, indicating that they are the major sites for virus infection. Our study showed that the VP35 antibodies generated in this study exhibited high specificity, and are valuable for the development of diagnostic tools for GCRV-II infection.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , Carps , Fish Diseases , Reoviridae Infections , Reoviridae , Animals , Carps/immunology , Carps/virology , Reoviridae Infections/immunology , Reoviridae Infections/veterinary , Reoviridae Infections/virology , Reoviridae/immunology , Reoviridae/physiology , Fish Diseases/immunology , Fish Diseases/virology , Mice , Humans , HEK293 Cells , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Viral Tropism , Capsid Proteins/immunology , Capsid Proteins/metabolism , Mice, Inbred BALB C , China
7.
Vaccine ; 42(17): 3733-3743, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38705805

ABSTRACT

Hand, foot, and mouth disease (HFMD) poses a significant public health threat primarily caused by four major enteroviruses: enterovirus 71 (EV71), coxsackieviruses A16, A10, and A6. Broadly protective immune responses are essential for complete protection against these major enteroviruses. In this study, we designed a new tetravalent immunogen for HFMD, validated it in silico, in vivo evaluated the immunogenicity of the DNA-based tetravalent vaccine in mice, and identified immunogenic B-cell and T-cell epitopes. A new tetravalent immunogen, VP1me, was designed based on the chimeric protein and epitope-based vaccine principles. It contains a complete EV71 VP1 protein and six reported neutralizing B-cell epitopes derived from the four major enteroviruses causing HFMD. In silico validation using multiple immunoinformatic tools indicated good attributes of the VP1me immunogen suitable for vaccine development. The VP1me-based DNA vaccine efficiently induced both humoral and cellular immune responses in BALB/cAJcl mice. A combination of in silico prediction and immunoassays enabled the identification of immunogenic linear B-cell and CD8 T-cell epitopes within the VP1me immunogen. Immunodominant linear B-cell epitopes were identified in six regions of VP1me, with one epitope located at the N-terminus of the VP1 protein (aa 9-23) regarded as a novel epitope. Interestingly, some B-cell epitopes could also induce the CD8 T-cell response, suggesting their dual functions in immune stimulation. These results lay the groundwork for further development of VP1me as a new vaccine candidate.


Subject(s)
Antibodies, Viral , Epitopes, B-Lymphocyte , Hand, Foot and Mouth Disease , Immunodominant Epitopes , Mice, Inbred BALB C , Vaccines, DNA , Viral Vaccines , Animals , Vaccines, DNA/immunology , Epitopes, B-Lymphocyte/immunology , Hand, Foot and Mouth Disease/prevention & control , Hand, Foot and Mouth Disease/immunology , Mice , Viral Vaccines/immunology , Immunodominant Epitopes/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Female , Epitopes, T-Lymphocyte/immunology , Capsid Proteins/immunology , Capsid Proteins/genetics , Enterovirus/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Enterovirus A, Human/immunology , Enterovirus A, Human/genetics , Immunogenicity, Vaccine , Immunity, Cellular , Immunity, Humoral
8.
Appl Microbiol Biotechnol ; 108(1): 350, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38809284

ABSTRACT

The African swine fever virus (ASFV) has the ability to infect pigs and cause a highly contagious acute fever that can result in a mortality rate as high as 100%. Due to the viral epidemic, the pig industry worldwide has suffered significant financial setbacks. The absence of a proven vaccine for ASFV necessitates the development of a sensitive and reliable serological diagnostic method, enabling laboratories to effectively and expeditiously detect ASFV infection. In this study, four strains of monoclonal antibodies (mAbs) against p72, namely, 5A1, 4C4, 8A9, and 5E10, were generated through recombinant expression of p72, the main capsid protein of ASFV, and immunized mice with it. Epitope localization was performed by truncated overlapping polypeptides. The results indicate that 5A1 and 4C4 recognized the amino acid 20-39 aa, 8A9 and 5E10 are recognized at 263-282 aa, which is consistent with the reported 265-280 aa epitopes. Conserved analysis revealed 20-39 aa is a high conservation of the epitopes in the ASFV genotypes. Moreover, a blocking ELISA assay for detection ASFV antibody based on 4C4 monoclonal antibody was developed and assessed. The receiver-operating characteristic (ROC) was performed to identify the best threshold value using 87 negative and 67 positive samples. The established test exhibited an area under the curve (AUC) of 0.9997, with a 95% confidence interval ranging from 99.87 to 100%. Furthermore, the test achieved a diagnostic sensitivity of 100% (with a 95% confidence interval of 95.72 to 100%) and a specificity of 98.51% (with a 95% confidence interval of 92.02 to 99.92%) when the threshold was set at 41.97%. The inter- and intra-batch coefficient of variation were below 10%, demonstrating the exceptional repeatability of the method. This method can detect the positive standard serum at a dilution as high as 1:512. Subsequently, an exceptional blocking ELISA assay was established with high diagnostic sensitivity and specificity, providing a novel tool for detecting ASFV antibodies. KEY POINTS: • Four strains of ASFV monoclonal antibodies against p72 were prepared and their epitopes were identified. • Blocking ELISA method was established based on monoclonal antibody 4C4 with an identified conservative epitope. • The established blocking ELISA method has a good effect on the detection of ASFV antibody.


Subject(s)
African Swine Fever Virus , African Swine Fever , Antibodies, Monoclonal , Antibodies, Viral , Capsid Proteins , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Animals , Antibodies, Monoclonal/immunology , African Swine Fever Virus/immunology , African Swine Fever Virus/genetics , Enzyme-Linked Immunosorbent Assay/methods , Antibodies, Viral/blood , Antibodies, Viral/immunology , Swine , African Swine Fever/diagnosis , African Swine Fever/immunology , African Swine Fever/virology , Mice , Capsid Proteins/immunology , Capsid Proteins/genetics , Mice, Inbred BALB C , Sensitivity and Specificity , Epitopes/immunology
9.
Int J Biol Macromol ; 267(Pt 2): 131446, 2024 May.
Article in English | MEDLINE | ID: mdl-38621561

ABSTRACT

Infection with bovine leukemia virus (BLV) leads to enzootic bovine leukosis, the most prevalent neoplastic disease in cattle. Due to the lack of commercially available vaccines, reliable eradication of the disease can be achieved through the testing and elimination of BLV antibody-positive animals. In this study, we developed a novel competitive ELISA (cELISA) to detect antibodies against BLV capsid protein p24. Recombinant p24 protein expressed by Escherichia coli, in combination with the monoclonal antibody 2G11 exhibiting exceptional performance, was used for the establishment of the cELISA. Receiver-operating characteristic curve analysis showed that the sensitivity and specificity of the assay were 98.85 % and 98.13 %, respectively. Furthermore, the established cELISA was specific for detecting BLV-specific antibodies, without cross-reactivity to antisera for six other bovine viruses. Significantly, experimental infection of cattle and sheep with BLV revealed that the cELISA accurately monitors seroconversion. In a performance evaluation, the established cELISA displayed a high agreement with Western blotting and the commercial BLV gp51 cELISA kit in the detection of 242 clinical samples, respectively. In conclusion, the novel p24 cELISA exhibited the potential to be a reliable and efficient diagnostic tool for BLV serological detection with a broad application prospect.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , Enzootic Bovine Leukosis , Enzyme-Linked Immunosorbent Assay , Leukemia Virus, Bovine , Leukemia Virus, Bovine/immunology , Animals , Enzyme-Linked Immunosorbent Assay/methods , Cattle , Antibodies, Viral/immunology , Antibodies, Monoclonal/immunology , Enzootic Bovine Leukosis/diagnosis , Enzootic Bovine Leukosis/immunology , Capsid Proteins/immunology , Sensitivity and Specificity , Recombinant Proteins/immunology , ROC Curve
10.
Hum Gene Ther ; 35(9-10): 355-364, 2024 May.
Article in English | MEDLINE | ID: mdl-38581431

ABSTRACT

Recombinant adeno-associated virus (rAAV) vectors appear, more than ever, to be efficient viral vectors for in vivo gene transfer as illustrated by the approvals of 7 drugs across Europe and the United States. Nevertheless, preexisting immunity to AAV capsid in humans remains one of the major limits for a successful clinical translation. Whereas a preexisting humoral response to AAV capsid is well documented, the prevalence of preexisting capsid-specific T cell responses still needs to be studied and characterized. In this study, we investigated the prevalence of AAV-specific circulating T cells toward AAV2, 4, 5, 8, 9, and rh10 in a large cohort of healthy donors using the standard IFNγ ELISpot assay. We observed the highest prevalence of preexisting cellular immunity to AAV9 serotype followed by AAV8, AAV4, AAV2, AAVrh10, and AAV5 independently of the donors' serological status. An in-depth analysis of T cell responses toward the 2 most prevalent serotypes 8 and 9 shows that IFNγ secretion is mainly mediated by CD8 T cells for both serotypes. A polyfunctional analysis reveals different cytokine profiles between AAV8 and AAV9. Surprisingly, no IL-2 secretion was mediated by anti-AAV9 immune cells suggesting that these cells may rather be exhausted or terminally differentiated than cytotoxic T cells. Altogether, these results suggest that preexisting immunity to AAV may vary depending on the serotype and support the necessity of using multiparametric monitoring methods to better characterize anticapsid cellular immunity and foresee its impact in rAAV-mediated clinical trials.


Subject(s)
Capsid Proteins , Dependovirus , Genetic Vectors , Immunity, Cellular , Humans , Dependovirus/genetics , Dependovirus/immunology , Capsid Proteins/immunology , Capsid Proteins/genetics , Genetic Vectors/genetics , Healthy Volunteers , Capsid/immunology , CD8-Positive T-Lymphocytes/immunology , Interferon-gamma/metabolism , Adult , Serogroup , Male , Female , Cytokines/metabolism , T-Lymphocytes/immunology
11.
ACS Sens ; 9(5): 2429-2439, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38668680

ABSTRACT

Norovirus (NoV) stands as a significant causative agent of nonbacterial acute gastroenteritis on a global scale, presenting a substantial threat to public health. Hence, the development of simple and rapid analytical techniques for NoV detection holds great importance in preventing and controlling the outbreak of the epidemic. In this work, a self-powered photoelectrochemical (PEC) immunosensor of NoV capsid protein (VP1) was proposed by the π-electron-rich carbon nitride homojunction (ER-CNH) as the photoanode. C4N2 ring derived from π-rich locust bean gum was introduced into the tri-s-triazine structure, creating a large π-delocalized conjugated carbon nitride homojunction. This strategy enhances the C/N atomic ratio, which widens light utilization, narrows the bandgap, and optimizes the electronic band structure of carbon nitride. By introduction of a π-rich conjugated structure, p-type domains were induced within n-type domains to build the internal electric field at the interface, thus forming a p-n homojunction to boost carrier separation and transfer. The ER-CNH photoanode exhibited excellent photoelectric performance and water oxidation capacity. Since VP1 inhibits the water oxidation of the ER-CNH photoanode, the open-circuit potential of the as-prepared PEC immunosensor system was reduced for detecting NoV VP1. The self-powered PEC immunosensor achieved a remarkably low detection limit (∼5 fg mL-1) and displayed high stability and applicability for actual stool samples. This research serves as a foundation concept for constructing immunosensors to detect other viruses and promotes the application of self-powered systems for life safety.


Subject(s)
Electrochemical Techniques , Feces , Norovirus , Norovirus/immunology , Norovirus/isolation & purification , Norovirus/chemistry , Immunoassay/methods , Humans , Feces/virology , Feces/chemistry , Electrochemical Techniques/methods , Capsid Proteins/chemistry , Capsid Proteins/immunology , Nitriles/chemistry , Biosensing Techniques/methods , Limit of Detection , Electrons
12.
Vet Microbiol ; 293: 110088, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38640639

ABSTRACT

Orf virus (ORFV), a member of the genus Parapoxvirus, possesses an excellent immune activation capability, which makes it a promising immunomodulation agent. In this study, we evaluated ORFV as a novel adjuvant to enhance the immune response of mice to a subunit vaccine using porcine circovirus type 2 (PCV2) capsid (Cap) protein as a model. Our results showed that both inactivated and live attenuated ORFV activated mouse bone marrow-derived dendritic cells and increased expression of immune-related cytokines interleukin (IL)-1ß, IL-6, and TNF-α. Enhanced humoral and cellular immune responses were induced in mice immunized with PCV2 Cap protein combined with inactivated or live attenuated ORFV adjuvant compared with the aluminum adjuvant. Increased secretion of Th1 and Th2 cytokines by splenic lymphocytes in immunized mice further indicated that the ORFV adjuvant promoted a mixed Th1/Th2 immune response. Moreover, addition of the ORFV adjuvant to the PCV2 subunit vaccine significantly reduced the viral load in the spleen and lungs of PCV2-challenged mice and prevented pathological changes in lungs. This study demonstrates that ORFV enhances the immunogenicity of a PCV2 subunit vaccine by improving the adaptive immune response, suggesting the potential application of ORFV as a novel adjuvant.


Subject(s)
Adjuvants, Immunologic , Circoviridae Infections , Circovirus , Cytokines , Orf virus , Vaccines, Subunit , Viral Vaccines , Animals , Circovirus/immunology , Mice , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Circoviridae Infections/prevention & control , Circoviridae Infections/veterinary , Circoviridae Infections/immunology , Circoviridae Infections/virology , Adjuvants, Immunologic/administration & dosage , Cytokines/immunology , Orf virus/immunology , Capsid Proteins/immunology , Female , Immunity, Cellular , Dendritic Cells/immunology , Viral Load , Antibodies, Viral/blood , Immunity, Humoral , Swine , Adjuvants, Vaccine , Mice, Inbred BALB C , Th1 Cells/immunology
13.
Int J Biol Macromol ; 268(Pt 1): 131695, 2024 May.
Article in English | MEDLINE | ID: mdl-38642684

ABSTRACT

Due to the absence of effective vaccine and treatment, African swine fever virus (ASFV) control is entirely dependent on accurate and early diagnosis, along with culling of infected pigs. The B646L/p72 is the major capsid protein of ASFV and is an important target for developing a diagnostic assays and vaccines. Herein, we generated a monoclonal antibody (mAb) (designated as 2F11) against the trimeric p72 protein, and a blocking ELISA (bELISA) was established for the detection of both genotype I and II ASFV antibodies. To evaluate the performance of the diagnostic test, a total of 506 porcine serum samples were tested. The average value of percent of inhibition (PI) of 133 negative pig serum was 8.4 % with standard deviation (SD) 6.5 %. Accordingly, the cut-off value of the newly established method was set at 28 % (mean + 3SD). Similarly, a receiver operating characteristic (ROC) was applied to determine the cut off value and the p72-bELISA exhibited a sensitivity of 100 % and a specificity of 99.33 % when the detection threshold was set at 28 %. The bELISA was also able to specifically recognize anti-ASFV sera without cross-reacting with other positive serums for other major swine pathogens. Moreover, by designing a series of overlapped p72 truncated proteins, the linear B cell epitope recognized by 2F11 mAb was defined to be 283NSHNIQ288. Amino acid sequence comparison revealed that the amino acid sequence 283NSHNIQ288 is highly conserved between different ASFV isolates. Our findings indicate that the newly established mAb based blocking ELISA may have a great potential in improving the detection of ASFV antibodies and provides solid foundation for further studies.


Subject(s)
African Swine Fever Virus , Antibodies, Monoclonal , Antibodies, Viral , Enzyme-Linked Immunosorbent Assay , Epitopes, B-Lymphocyte , Animals , African Swine Fever Virus/immunology , Enzyme-Linked Immunosorbent Assay/methods , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Swine , Epitopes, B-Lymphocyte/immunology , Capsid Proteins/immunology , African Swine Fever/immunology , African Swine Fever/diagnosis , African Swine Fever/virology , Amino Acid Sequence , Epitope Mapping
14.
Viruses ; 16(4)2024 04 17.
Article in English | MEDLINE | ID: mdl-38675963

ABSTRACT

Southern Africa Territories 2 (SAT2) foot-and-mouth disease (FMD) has crossed long-standing regional boundaries in recent years and entered the Middle East. However, the existing vaccines offer poor cross-protection against the circulating strains in the field. Therefore, there is an urgent need for an alternative design approach for vaccines in anticipation of a pandemic of SAT2 Foot-and-mouth disease virus (FMDV). The porcine parvovirus (PPV) VP2 protein can embed exogenous epitopes into the four loops on its surface, assemble into virus-like particles (VLPs), and induce antibodies and cytokines to PPV and the exogenous epitope. In this study, chimeric porcine parvovirus VP2 VLPs (chimeric PPV-SAT2-VLPs) expressing the T-and/or B-cell epitopes of the structural protein VP1 of FMDV SAT2 were produced using the recombinant pFastBac™ Dual vector of baculoviruses in Sf9 and HF cells We used the Bac-to-Bac system to construct the recombinant baculoviruses. The VP2-VLP--SAT2 chimeras displayed chimeric T-cell epitope (amino acids 21-40 of VP1) and/or the B-cell epitope (amino acids 135-174) of SAT FMDV VP1 by substitution of the corresponding regions at the N terminus (amino acids 2-23) and/or loop 2 and/or loop 4 of the PPV VP2 protein, respectively. In mice, the chimeric PPV-SAT2-VLPs induced specific antibodies against PPV and the VP1 protein of SAT2 FMDV. The VP2-VLP-SAT2 chimeras induced specific antibodies to PPV and the VP1 protein specific epitopes of FMDV SAT2. In this study, as a proof-of-concept, successfully generated chimeric PPV-VP2 VLPs expressing epitopes of the structural protein VP1 of FMDV SAT2 that has a potential to prevent FMDV SAT2 and PPV infection in pigs.


Subject(s)
Antibodies, Viral , Antigens, Viral , Capsid Proteins , Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Parvovirus, Porcine , Vaccines, Virus-Like Particle , Viral Vaccines , Animals , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease Virus/genetics , Mice , Foot-and-Mouth Disease/immunology , Foot-and-Mouth Disease/prevention & control , Foot-and-Mouth Disease/virology , Capsid Proteins/immunology , Capsid Proteins/genetics , Parvovirus, Porcine/immunology , Parvovirus, Porcine/genetics , Antibodies, Viral/immunology , Antibodies, Viral/blood , Viral Vaccines/immunology , Viral Vaccines/genetics , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/genetics , Swine , Immunity, Humoral , Immunity, Cellular , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Serogroup , Mice, Inbred BALB C , Female , Epitopes/immunology , Epitopes/genetics , Sf9 Cells , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood
15.
Vet Microbiol ; 293: 110094, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38636175

ABSTRACT

Infectious bursa disease (IBD) is an acute, highly contactable, lethal, immunosuppressive infectious disease caused by the Infectious bursa disease virus (IBDV). Currently, the emerged novel variant IBDV (nVarIBDV) and the sustainedly prevalent very virulent IBDV (vvIBDV) are the two most prevalent strains of IBDV in China. The antigenic properties of the two prevalent strains differed significantly, which led to the escape of nVarIBDV from the immune protection provided by the existing vvIBDV vaccine. However, the molecular basis of the nVarIBDV immune escape remains unclear. In this study, we demonstrated, for the first time, that residues 252, 254, and 256 in the PDE of VP2 are involved in the immune escape of the emerging nVarIBDV. Firstly, the IFA-mediated antigen-antibody affinity assay showed that PBC and PDE of VP2 could affect the affinity of vvIBDV antiserum to VP2, of which PDE was more significant. The key amino acids of PDE influencing the antigen-antibody affinity were also identified, with G254N being the most significant, followed by V252I and I256V. Then the mutated virus with point or combined mutations was rescued by reverse genetics. it was further demonstrated that mutations of V252I, G254N, and I256V in PDE could individually or collaboratively reduce antigen-antibody affinity and interfere with antiserum neutralization, with G254N being the most significant. This study revealed the reasons for the widespread prevalence of nVarIBDV in immunized chicken flocks and provided innovative ideas for designing novel vaccines that match the antigen of the epidemic strain.


Subject(s)
Birnaviridae Infections , Capsid Proteins , Chickens , Immune Evasion , Infectious bursal disease virus , Poultry Diseases , Infectious bursal disease virus/genetics , Infectious bursal disease virus/immunology , Animals , Chickens/virology , Capsid Proteins/genetics , Capsid Proteins/immunology , Poultry Diseases/virology , Poultry Diseases/immunology , Birnaviridae Infections/veterinary , Birnaviridae Infections/virology , Birnaviridae Infections/immunology , China , Antibodies, Viral/immunology , Mutation , Viral Vaccines/immunology , Viral Structural Proteins
16.
Vaccine ; 42(15): 3514-3521, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38670845

ABSTRACT

Group A rotavirus (RVA) is the primary etiological agent of acute gastroenteritis (AGE) in children under 5 years of age. Despite the global implementation of vaccines, rotavirus infections continue to cause over 120,000 deaths annually, with a majority occurring in developing nations. Among infants, the P[8] rotavirus strain is the most prevalent and can be categorized into four distinct lineages. In this investigation, we expressed five VP4(aa26-476) proteins from different P[8] lineages of human rotavirus in E. coli and assessed their immunogenicity in rabbits. Among the different P[8] strains, the Wa-VP4 protein, derived from the MT025868.1 strain of the P[8]-1 lineage, exhibited successful purification in a highly homogeneous form and significantly elicited higher levels of neutralizing antibodies (nAbs) against both homologous and heterologous rotaviruses compared to other VP4 proteins derived from different P[8] lineages in rabbits. Furthermore, we assessed the immunogenicity of the Wa-VP4 protein in mice, pigs, and cynomolgus monkeys, observing that it induced robust production of nAbs in all animals. Interestingly, there was no significant difference between in nAb titers against homologous and heterologous rotaviruses in pigs and mankeys. Collectively, these findings suggest that the Wa-VP4* protein may serve as a potential candidate for a rotavirus vaccine.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Capsid Proteins , Macaca fascicularis , Rotavirus Infections , Rotavirus Vaccines , Rotavirus , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Rotavirus Vaccines/immunology , Rotavirus Vaccines/administration & dosage , Antibodies, Viral/immunology , Antibodies, Viral/blood , Swine , Rabbits , Mice , Rotavirus/immunology , Rotavirus/genetics , Capsid Proteins/immunology , Capsid Proteins/genetics , Rotavirus Infections/prevention & control , Rotavirus Infections/immunology , Female , Mice, Inbred BALB C , Humans , Immunogenicity, Vaccine , Viral Nonstructural Proteins/immunology , Viral Nonstructural Proteins/genetics
17.
Genes (Basel) ; 15(4)2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38674401

ABSTRACT

The variable domain of a heavy-chain antibody (VHH) has the potential to be used to redirect the cell tropism of adenoviral vectors. Here, we attempted to establish platforms to simplify the screening of VHHs for their specific targeting function when being incorporated into the fiber of adenovirus. Both fowl adenovirus 4 (FAdV-4) and simian adenovirus 1 (SAdV-1) have two types of fiber, one of which is dispensable for virus propagation and is a proper site for VHH display. An intermediate plasmid, pMD-FAV4Fs, was constructed as the start plasmid for FAdV-4 fiber2 modification. Foldon from phage T4 fibritin, a trigger for trimerization, was employed to bridge the tail/shaft domain of fiber2 and VHHs against human CD16A, a key membrane marker of natural killer (NK) cells. Through one step of restriction-assembly, the modified fiber2 was transferred to the adenoviral plasmid, which was linearized and transfected to packaging cells. Five FAdV-4 viruses carrying the GFP gene were finally rescued and amplified, with three VHHs being displayed. One recombinant virus, FAdV4FC21-EG, could hardly transduce human 293 or Jurkat cells. In contrast, when it was used at a multiplicity of infection of 1000 viral particles per cell, the transduction efficiency reached 51% or 34% for 293 or Jurkat cells expressing exogenous CD16A. Such a strategy of fiber modification was transplanted to the SAdV-1 vector to construct SAdV1FC28H-EG, which moderately transduced primary human NK cells while the parental virus transduced none. Collectively, we reformed the strategy of integrating VHH to fiber and established novel platforms for screening VHHs to construct adenoviral vectors with a specific tropism.


Subject(s)
Genetic Vectors , Viral Tropism , Humans , Genetic Vectors/genetics , HEK293 Cells , Immunoglobulin Heavy Chains/genetics , Aviadenovirus/genetics , Aviadenovirus/immunology , Animals , Capsid Proteins/genetics , Capsid Proteins/immunology , Capsid Proteins/metabolism
18.
Int Immunopharmacol ; 133: 112079, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38615376

ABSTRACT

Porcine rotavirus (PoRV) poses a threat to the development of animal husbandry and human health, leading to substantial economic losses. VP6 protein is the most abundant component in virus particles and also the core structural protein of the virus. Firstly, this study developed an antibiotic-resistance-free, environmentally friendly expression vector, named asd-araC-PBAD-alr (AAPA). Then Recombinant Lactiplantibacillus plantarum (L. plantarum) strains induced by arabinose to express VP6 and VP6-pFc fusion proteins was constructed. Subsequently, This paper discovered that NC8/Δalr-pCXa-VP6-S and NC8/Δalr-pCXa-VP6-pFc-S could enhance host immunity and prevent rotavirus infection in neonatal mice and piglets. The novel recombinant L. plantarum strains constructed in this study can serve as oral vaccines to boost host immunity, offering a new strategy to prevent PoRV infection.


Subject(s)
Capsid Proteins , Lactobacillus plantarum , Rotavirus Infections , Swine Diseases , Animals , Mice , Animals, Newborn , Antigens, Viral/immunology , Capsid Proteins/immunology , Capsid Proteins/genetics , Lactobacillus plantarum/immunology , Mice, Inbred BALB C , Rotavirus/immunology , Rotavirus Infections/prevention & control , Rotavirus Infections/immunology , Rotavirus Infections/virology , Swine , Swine Diseases/prevention & control , Swine Diseases/virology , Swine Diseases/microbiology , Swine Diseases/immunology
19.
J Virol ; 98(5): e0021224, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38591886

ABSTRACT

Porcine rotaviruses (PoRVs) cause severe economic losses in the swine industry. P[7] and P[23] are the predominant genotypes circulating on farms, but no vaccine is yet available. Here, we developed a bivalent subunit PoRV vaccine using truncated versions (VP4*) of the VP4 proteins from P[7] and P[23]. The vaccination of mice with the bivalent subunit vaccine elicited more robust neutralizing antibodies (NAbs) and cellular immune responses than its components, even at high doses. The bivalent subunit vaccine and inactivated bivalent vaccine prepared from strains PoRVs G9P[7] and G9P[23] were used to examine their protective efficacy in sows and suckling piglets after passive immunization. The immunized sows showed significantly elevated NAbs in the serum and colostrum, and the suckling piglets acquired high levels of sIgA antibodies from the colostrum. Challenging subunit-vaccinated or inactivated-vaccinated piglets with homologous virulent strains did not induce diarrhea, except in one or two piglets, which had mild diarrhea. Immunization with the bivalent subunit vaccine and inactivated vaccine also alleviated the microscopic lesions in the intestinal tissues caused by the challenge with the corresponding homologous virulent strain. However, all the piglets in the challenged group displayed mild to watery diarrhea and high levels of viral shedding, whereas the feces and intestines of the piglets in the bivalent subunit vaccine and inactivated vaccine groups had lower viral loads. In summary, our data show for the first time that a bivalent subunit vaccine combining VP4*P[7] and VP4*P[23] effectively protects piglets against the diarrhea caused by homologous virulent strains.IMPORTANCEPoRVs are the main causes of diarrhea in piglets worldwide. The multisegmented genome of PoRVs allows the reassortment of VP4 and VP7 genes from different RV species and strains. The P[7] and P[23] are the predominant genotypes circulating in pig farms, but no vaccine is available at present in China. Subunit vaccines, as nonreplicating vaccines, are an option to cope with variable genotypes. Here, we have developed a bivalent subunit candidate vaccine based on a truncated VP4 protein, which induced robust humoral and cellular immune responses and protected piglets against challenge with homologous PoRV. It also appears to be safe. These data show that the truncated VP4-protein-based subunit vaccine is a promising candidate for the prevention of PoRV diarrhea.


Subject(s)
Rotavirus Vaccines , Vaccines, Subunit , Animals , Female , Mice , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Capsid Proteins/immunology , Capsid Proteins/genetics , Diarrhea/prevention & control , Diarrhea/virology , Diarrhea/veterinary , Diarrhea/immunology , Genotype , Immunity, Cellular , Mice, Inbred BALB C , Rotavirus/immunology , Rotavirus Infections/prevention & control , Rotavirus Infections/veterinary , Rotavirus Infections/immunology , Rotavirus Infections/virology , Rotavirus Vaccines/immunology , Rotavirus Vaccines/administration & dosage , Swine , Swine Diseases/prevention & control , Swine Diseases/virology , Swine Diseases/immunology , Vaccination , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage
20.
J Virol ; 98(5): e0019724, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38593321

ABSTRACT

Noroviruses are major causative agents of acute nonbacterial gastroenteritis in humans. There are neither antiviral therapeutic agents nor vaccines for noroviruses at this time. To evaluate the potential usefulness of two previously isolated human monoclonal antibody fragments, CV-1A1 and CV-2F5, we first conducted a single-particle analysis to determine the cryo-electron microscopy structure of virus-like particles (VLPs) from the genogroup I genotype 4 (GI.4) Chiba strain uniformly coated with CV-1A1 fragments. The results revealed that the GI.4-specific CV-1A1 antibody bound to the P2 subdomain, in which amino acids are less conserved and variable. Interestingly, a part of the CV-1A1 intrudes into the histo-blood group antigen-binding site, suggesting that this antibody might exert neutralizing activity. Next, we determined the crystal structure of the protruding (P) domain of the capsid protein in the complex form with the CV-2F5 antibody fragment. Consistent with the cross-reactivity, the CV-2F5 bound to the P1 subdomain, which is rich in amino acids conserved among the GI strains, and moreover induced a disruption of Chiba VLPs. These results suggest that the broadly reactive CV-2F5 antibody can be used as both a universal detection reagent and an antiviral drug for GI noroviruses. IMPORTANCE: We conducted the structural analyses of the VP1 protein from the GI.4 Chiba norovirus to identify the binding sites of the previously isolated human monoclonal antibodies CV-1A1 and CV-2F5. The cryo-electron microscopy of the Chiba virus-like particles (VLPs) complexed with the Fv-clasp forms of GI.4-specific CV-1A1 revealed that this antibody binds to the highly variable P2 subdomain, suggesting that this antibody may have neutralizing ability against the GI.4 strains. X-ray crystallography revealed that the CV-2F5 antibody bound to the P1 subdomain, which is rich in conserved amino acids. This result is consistent with the ability of the CV-2F5 antibody to react with a wide variety of GI norovirus strains. It is also found that the CV-2F5 antibody caused a disruption of VLPs. Our findings, together with previous reports on the structures of VP1 proteins and VLPs, are expected to open a path for the structure-based development of antivirals and vaccines against norovirus disease.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , Norovirus , Humans , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Binding Sites , Capsid Proteins/immunology , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Cryoelectron Microscopy/methods , Crystallography, X-Ray , Models, Molecular , Norovirus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...