Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
1.
J Virol ; 97(2): e0160022, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36757205

ABSTRACT

Infection by Kaposi sarcoma-associated herpesvirus (KSHV) can cause severe consequences, such as cancers and lymphoproliferative diseases. Whole inactivated viruses (WIV) with chemically destroyed genetic materials have been used as antigens in several licensed vaccines. During KSHV productive replication, virus-like vesicles (VLVs) that lack capsids and viral genomes are generated along with virions. Here, we investigated the immunogenicity of KSHV VLVs produced from a viral mutant that was defective in capsid formation and DNA packaging. Mice immunized with adjuvanted VLVs generated KSHV-specific T cell and antibody responses. Neutralization of KSHV infection by the VLV immune serum was low but was markedly enhanced in the presence of the complement system. Complement-enhanced neutralization and complement deposition on KSHV-infected cells was dependent on antibodies targeting viral open reading frame 4 (ORF4). However, limited complement-mediated enhancement was detected in the sera of a small cohort of KSHV-infected humans which contained few neutralizing antibodies. Therefore, vaccination that induces antibody effector functions can potentially improve infection-induced humoral immunity. Overall, our study highlights a potential benefit of engaging complement-mediated antibody functions in future KSHV vaccine development. IMPORTANCE KSHV is a virus that can lead to cancer after infection. A vaccine that prevents KSHV infection or transmission would be helpful in preventing the development of these cancers. We investigated KSHV VLV as an immunogen for vaccination. We determined that antibodies targeting the viral protein ORF4 induced by VLV immunization could engage the complement system and neutralize viral infection. However, ORF4-specific antibodies were seldom detected in the sera of KSHV-infected humans. Moreover, these human sera did not potently trigger complement-mediated neutralization, indicating an improvement that immunization can confer. Our study suggests a new antibody-mediated mechanism to control KSHV infection and underscores the benefit of activating the complement system in a future KSHV vaccine.


Subject(s)
Antibodies, Neutralizing , Herpesvirus 8, Human , Animals , Humans , Mice , Antibodies, Neutralizing/immunology , Herpesviridae Infections , Herpesvirus 8, Human/immunology , Open Reading Frames/immunology , Vaccination , Viral Proteins/immunology
2.
Front Immunol ; 12: 692937, 2021.
Article in English | MEDLINE | ID: mdl-34497604

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) kills thousands of people worldwide every day, thus necessitating rapid development of countermeasures. Immunoinformatics analyses carried out here in search of immunodominant regions in recently identified SARS-CoV-2 unannotated open reading frames (uORFs) have identified eight linear B-cell, one conformational B-cell, 10 CD4+ T-cell, and 12 CD8+ T-cell promising epitopes. Among them, ORF9b B-cell and T-cell epitopes are the most promising followed by M.ext and ORF3c epitopes. ORF9b40-48 (CD8+ T-cell epitope) is found to be highly immunogenic and antigenic with the highest allele coverage. Furthermore, it has overlap with four potent CD4+ T-cell epitopes. Structure-based B-cell epitope prediction has identified ORF9b61-68 to be immunodominant, which partially overlaps with one of the linear B-cell epitopes (ORF9b65-69). ORF3c CD4+ T-cell epitopes (ORF3c2-16, ORF3c3-17, and ORF3c4-18) and linear B-cell epitope (ORF3c14-22) have also been identified as the candidate epitopes. Similarly, M.ext and 7a.iORF1 (overlap with M and ORF7a) proteins have promising immunogenic regions. By considering the level of antigen expression, four ORF9b and five M.ext epitopes are finally shortlisted as potent epitopes. Mutation analysis has further revealed that the shortlisted potent uORF epitopes are resistant to recurrent mutations. Additionally, four N-protein (expressed by canonical ORF) epitopes are found to be potent. Thus, SARS-CoV-2 uORF B-cell and T-cell epitopes identified here along with canonical ORF epitopes may aid in the design of a promising epitope-based polyvalent vaccine (when connected through appropriate linkers) against SARS-CoV-2. Such a vaccine can act as a bulwark against SARS-CoV-2, especially in the scenario of emergence of variants with recurring mutations in the spike protein.


Subject(s)
Antigens, Viral/immunology , COVID-19 Vaccines/immunology , COVID-19/prevention & control , Coronavirus Nucleocapsid Proteins/immunology , SARS-CoV-2/immunology , Amino Acid Sequence/genetics , Antigens, Viral/genetics , COVID-19/immunology , COVID-19/virology , COVID-19 Vaccines/genetics , COVID-19 Vaccines/therapeutic use , Computational Biology , Coronavirus Nucleocapsid Proteins/genetics , Drug Design , Epitope Mapping , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Humans , Open Reading Frames/genetics , Open Reading Frames/immunology , SARS-CoV-2/genetics , Sequence Analysis, Protein , Vaccines, Combined/genetics , Vaccines, Combined/immunology
3.
J Virol ; 95(21): e0094021, 2021 10 13.
Article in English | MEDLINE | ID: mdl-34379494

ABSTRACT

Despite the prevalence and medical significance of human cytomegalovirus (HCMV) infections, a systematic analysis of the targets of T cell recognition in humans that spans the entire genome and includes recently described potential novel open reading frames (ORFs) is not available. Here, we screened a library of epitopes predicted to bind HLA class II that spans over 350 different HCMV ORFs and includes ∼150 previously described and ∼200 recently described potential novel ORFs by using an ex vivo gamma interferon (IFN-γ) FluoroSpot assay. We identified 235 unique HCMV-specific epitopes derived from 100 ORFs, some previously described as immunodominant and others that were not previously described to be immunogenic. Of those, 41 belong to the set of recently reported novel ORFs, thus providing evidence that at least some of these are actually expressed in vivo in humans. These data reveal that the breadth of the human T cell response to HCMV is much greater than previously thought. The ORFs and epitopes identified will help elucidate how T cell immunity relates to HCMV pathogenesis and instruct ongoing HCMV vaccine research. IMPORTANCE To understand the crucial role of adaptive immunity in controlling cytomegalovirus infection and disease, we systematically analyzed the CMV "ORFeome" to identify new CMV epitopes targeted primarily by CD4 T cells in humans. Our study identified >200 new T cell epitopes derived from both canonical and novel ORFs, highlighting the substantial breadth of the anti-CMV T cell response and providing new targets for vaccine design.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Epitopes, T-Lymphocyte/immunology , Open Reading Frames/immunology , Adult , Aged , Aged, 80 and over , Cytomegalovirus Infections/virology , Epitopes, T-Lymphocyte/genetics , Female , Humans , Interferon-gamma , Male , Middle Aged
4.
Viruses ; 13(7)2021 07 16.
Article in English | MEDLINE | ID: mdl-34372591

ABSTRACT

To evaluate the antigenic properties of Hepatitis E Virus (HEV) Open Reading Frame 2 and 3 (ORF2 and ORF3) codified proteins, we expressed different portions of ORF2 and the entire ORF3 in E. coli, a truncated ORF2, was also expressed in baculovirus. A panel of 37 monoclonal antibodies (MAbs) was raised against ORF2 (1-660 amino acids) and MAbs were mapped and characterized using the ORF2 expressed portions. Selected HEV positive and negative swine sera were used to evaluate ORF2 and ORF3 antigens' immunogenicity. The MAbs were clustered in six groups identifying six antigenic regions along the ORF2. Only MAbs binding to the sixth ORF2 antigenic region (394-608 aa) were found to compete with HEV positive sera and efficiently catch the recombinant antigen expressed in baculovirus. The ORF2 portion from 394-608 aa demonstrated to include most immunogenic epitopes with 85% of HEV positive swine sera reacting against the region from 461-544 aa. Only 5% of the selected HEV sera reacted against the ORF3 antigen.


Subject(s)
Antigens, Viral/immunology , Hepatitis E virus/immunology , Open Reading Frames/genetics , Viral Proteins/genetics , Animals , Baculoviridae/genetics , Baculoviridae/immunology , Epitopes/genetics , Epitopes/immunology , Escherichia coli/genetics , Hepatitis Antibodies/blood , Hepatitis E virus/chemistry , Hepatitis E virus/genetics , Mice , Mice, Inbred BALB C , Open Reading Frames/immunology , Swine , Viral Proteins/immunology
5.
J Virol ; 95(17): e0040221, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34133899

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the viral pathogen responsible for the current coronavirus disease 2019 (COVID-19) pandemic. As of 19 May 2021, John Hopkins University's COVID-19 tracking platform reported 3.3 million deaths associated with SARS-CoV-2 infection. Currently, the World Health Organization has granted emergency use listing (EUL) to six COVID-19 vaccine candidates. However, much of the pathogenesis observed during SARS-CoV-2 infection remains elusive. To gain insight into the contribution of individual accessory open reading frame (ORF) proteins in SARS-CoV-2 pathogenesis, we used our recently described reverse-genetics system approach to successfully engineer recombinant SARS-CoV-2 (rSARS-CoV-2) constructs; we removed individual viral ORF3a, -6, -7a, -7b, and -8 proteins from them, and we characterized the resulting recombinant viruses in vitro and in vivo. Our results indicate differences in plaque morphology, with ORF-deficient (ΔORF) viruses producing smaller plaques than those of the wild type (rSARS-CoV-2/WT). However, growth kinetics of ΔORF viruses were like those of rSARS-CoV-2/WT. Interestingly, infection of K18 human angiotensin-converting enzyme 2 (hACE2) transgenic mice with the ΔORF rSARS-CoV-2s identified ORF3a and ORF6 as the major contributors of viral pathogenesis, while ΔORF7a, ΔORF7b, and ΔORF8 rSARS-CoV-2s induced pathology comparable to that of rSARS-CoV-2/WT. This study demonstrates the robustness of our reverse-genetics system to generate rSARS-CoV-2 constructs and the major role for ORF3a and ORF6 in viral pathogenesis, providing important information for the generation of attenuated forms of SARS-CoV-2 for their implementation as live attenuated vaccines for the treatment of SARS-CoV-2 infection and associated COVID-19. IMPORTANCE Despite great efforts put forward worldwide to combat the current coronavirus disease 2019 (COVID-19) pandemic, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to be a human health and socioeconomic threat. Insights into the pathogenesis of SARS-CoV-2 and the contribution of viral proteins to disease outcome remain elusive. Our study aims (i) to determine the contribution of SARS-CoV-2 accessory open reading frame (ORF) proteins to viral pathogenesis and disease outcome and (ii) to develop a synergistic platform combining our robust reverse-genetics system to generate recombinant SARS-CoV-2 constructs with a validated rodent model of infection and disease. We demonstrate that SARS-CoV-2 ORF3a and ORF6 contribute to lung pathology and ultimately disease outcome in K18 hACE2 transgenic mice, while ORF7a, ORF7b, and ORF8 have little impact on disease outcome. Moreover, our combinatory platform serves as a foundation for generating attenuated forms of the virus to develop live attenuated vaccines for the treatment of SARS-CoV-2.


Subject(s)
Angiotensin-Converting Enzyme 2/immunology , Open Reading Frames/immunology , SARS-CoV-2 , Viral Proteins , A549 Cells , Angiotensin-Converting Enzyme 2/genetics , Animals , COVID-19 Vaccines/genetics , COVID-19 Vaccines/immunology , Chlorocebus aethiops , HEK293 Cells , Humans , Mice , Mice, Transgenic , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vero Cells , Viral Proteins/genetics , Viral Proteins/immunology
6.
BMC Cancer ; 21(1): 628, 2021 May 27.
Article in English | MEDLINE | ID: mdl-34044801

ABSTRACT

BACKGROUND: Oral Squamous Cell Carcinoma (OSCC) results from a series of genetic alteration in squamous cells. This particular type of cancer considers one of the most aggressive malignancies to control because of its frequent local invasions to the regional lymph node. Although several biomarkers have been reported, the key marker used to predict the behavior of the disease is largely unknown. Here we report Long INterpersed Element-1 (LINE1 or L1) retrotransposon activity in post-operative oral cancer samples. L1 is the only active retrotransposon occupying around 17% of the human genome with an estimated 500,000 copies. An active L1 encodes two proteins (L1ORF1p and L1ORF2p); both of which are critical in the process of retrotransposition. Several studies report that the L1 retrotransposon is highly active in many cancers. L1 activity is generally determined by assaying L1ORF1p because of its high expression and availability of the antibody. However, due to its lower expression and unavailability of a robust antibody, detection of L1ORF2p has been limited. L1ORF2p is the crucial protein in the process of retrotransposition as it provides endonuclease and reverse transcriptase (RT) activity. METHODS: Immunohistochemistry and Western blotting were performed on the post-operative oral cancer samples and murine tissues. RESULTS: Using in house novel antibodies against both the L1 proteins (L1ORF1p and L1ORF2p), we found L1 retrotransposon is extremely active in post-operative oral cancer tissues. Here, we report a novel human L1ORF2p antibody generated using an 80-amino-acid stretch from the RT domain, which is highly conserved among different species. The antibody detects significant L1ORF2p expression in human oral squamous cell carcinoma (OSCC) samples and murine germ tissues. CONCLUSIONS: We report exceptionally high L1ORF1p and L1ORF2p expression in post-operative oral cancer samples. The novel L1ORF2p antibody reported in this study will serve as a useful tool to understand why L1 activity is deregulated in OSCC and how it contributes to the progression of this particular cancer. Cross-species reactivity of L1ORF2p antibody due to the conserved epitope will be useful to study the retrotransposon biology in mice and rat germ tissues.


Subject(s)
Antigens, Neoplasm/immunology , Long Interspersed Nucleotide Elements/genetics , Mouth Neoplasms/genetics , Open Reading Frames/immunology , Squamous Cell Carcinoma of Head and Neck/genetics , Amino Acid Sequence/genetics , Animals , Antigens, Neoplasm/genetics , HEK293 Cells , Humans , Mice , Mouth Mucosa/immunology , Mouth Mucosa/pathology , Mouth Mucosa/surgery , Mouth Neoplasms/immunology , Mouth Neoplasms/pathology , Mouth Neoplasms/surgery , Open Reading Frames/genetics , Rats , Sequence Alignment , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/surgery
7.
Sci Immunol ; 6(57)2021 03 25.
Article in English | MEDLINE | ID: mdl-33766849

ABSTRACT

Simian immunodeficiency virus (SIV) insert-expressing, 68-1 rhesus cytomegalovirus (RhCMV/SIV) vectors elicit major histocompatibility complex E (MHC-E)- and MHC-II-restricted, SIV-specific CD8+ T cell responses, but the basis of these unconventional responses and their contribution to demonstrated vaccine efficacy against SIV challenge in the rhesus monkeys (RMs) have not been characterized. We show that these unconventional responses resulted from a chance genetic rearrangement in 68-1 RhCMV that abrogated the function of eight distinct immunomodulatory gene products encoded in two RhCMV genomic regions (Rh157.5/Rh157.4 and Rh158-161), revealing three patterns of unconventional response inhibition. Differential repair of these genes with either RhCMV-derived or orthologous human CMV (HCMV)-derived sequences (UL128/UL130; UL146/UL147) leads to either of two distinct CD8+ T cell response types-MHC-Ia-restricted only or a mix of MHC-II- and MHC-Ia-restricted CD8+ T cells. Response magnitude and functional differentiation are similar to RhCMV 68-1, but neither alternative response type mediated protection against SIV challenge. These findings implicate MHC-E-restricted CD8+ T cell responses as mediators of anti-SIV efficacy and indicate that translation of RhCMV/SIV vector efficacy to humans will likely require deletion of all genes that inhibit these responses from the HCMV/HIV vector.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cellular Reprogramming/immunology , Cytomegalovirus Infections/veterinary , Cytomegalovirus/immunology , Monkey Diseases/prevention & control , Simian Acquired Immunodeficiency Syndrome/immunology , Viral Vaccines/immunology , Animals , Antigens, Viral/genetics , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/metabolism , Cellular Reprogramming/genetics , Genetic Engineering/methods , Genetic Vectors/genetics , Immunogenicity, Vaccine , Immunologic Memory , Macaca mulatta , Monkey Diseases/immunology , Monkey Diseases/virology , Open Reading Frames/genetics , Open Reading Frames/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Vaccine Efficacy
8.
Viruses ; 12(4)2020 04 14.
Article in English | MEDLINE | ID: mdl-32295124

ABSTRACT

Human norovirus (HuNoV) is a principal cause of acute gastroenteritis worldwide, particularly in developing countries. Its global prevalence is underscored by more serious morbidity and some mortality in the young (<5 years) and the elderly. To date, there are no licensed vaccines or approved therapeutics for HuNoV, mostly because there are limited cell culture systems and small animal models available. Recently described cell culture systems are not ideal substrates for HuNoV vaccine development because they are not clonal or only support a single strain. In this study, we show Vero cell-based replication of two pandemic GII.4 HuNoV strains and one GII.3 strain and confirm exosome-mediated HuNoV infection in Vero cells. Lastly, we show that trypsin addition to virus cultures or disruption of Vero cell host genes can modestly increase HuNoV replication. These data provide support for Vero cells as a cell culture model for HuNoV.


Subject(s)
Norovirus/physiology , Virus Replication , Animals , Antigens, Viral , Caliciviridae Infections/immunology , Caliciviridae Infections/virology , Cell Line , Cells, Cultured , Chlorocebus aethiops , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Editing , Gene Knockdown Techniques , Genome, Viral , Humans , Open Reading Frames/genetics , Open Reading Frames/immunology , RNA, Small Interfering/genetics , Vero Cells , Viral Tropism
9.
Fish Shellfish Immunol ; 98: 342-353, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31978531

ABSTRACT

Cyprinid Herpesvirus 3 (CyHV-3), also known as Koi Herpesvirus (KHV), causes Koi Herpesvirus Disease (KHVD) which leads to serious economic losses worldwide. To exploit DNA/subunit vaccine candidates, CyHV-3 ORF131 gene and cDNA was cloned and analyzed in the present study. Major B cell epitopes of deduced CyHV-3 pORF131 was also predicted. Then the complete CDS of CyHV-3 ORF131 was inserted into pEGFP-N1 vector and a modified pYD1/EBY100 system to construct the DNA and subunit vaccine, respectively. Subsequently, carp were immunized with homologous and heterologous prime-boost regimens relying on the constructed DNA and oral subunit vaccines. Then the protective immunity generated from different vaccines and regimens as well as the capacity of yeast (Saccharomyces cerevisiae) as an oral vaccine vehicle was evaluated. Our study confirmed that CyHV-3 ORF131 gene consisted of 2 introns and 3 exons encoding a 428 amino acids peptide. Further analysis indicated that four fragments of CyHV-3 pORF131 contained the major B cell epitopes (Cys20~Val140, Ser169~Tyr245, Thr258~Pro390, Phe414~Gln428), which could be linked and expressed in E. coli (BL21) as a truncated pORF131. The expression of full-length CyHV-3 pORF131 by pEGFP-N1 and yeast surface display was verified by In vitro assays before vaccination. Immunization of carp with CyHV-3 ORF131 DNA and subunit vaccines could evoke the activation of immune-related genes such as CXCa, CXCR1, IL-1ß, TNF-α, INF-a1, Mx-1, IgM, IgT1 and production of specific serum IgM measured by ELISA. RPS (relative percent of survival) ranging from 53.33% to 66.67% was acquired post challenge test. Moreover, flow cytometry analysis illustrated the delivery of surface-displayed CyHV-3 pORF131 to midgut after oral gavage. Thus, our findings suggest that CyHV-3 ORF131 can serve as DNA/subunit vaccines candidate and the yeast as an ideal oral vaccine vehicle.


Subject(s)
Carps , Fish Diseases/prevention & control , Herpesviridae Infections/veterinary , Herpesviridae/immunology , Herpesvirus Vaccines/immunology , Open Reading Frames/immunology , Vaccination/veterinary , Administration, Oral , Animals , Antibodies, Viral/blood , Carps/immunology , Carps/virology , Cell Surface Display Techniques , Epitopes, B-Lymphocyte , Escherichia coli/genetics , Fish Diseases/virology , Gene Expression Regulation/immunology , Herpesviridae Infections/prevention & control , Herpesvirus Vaccines/administration & dosage , Immunization Schedule , Open Reading Frames/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Survival Analysis , Vaccination/methods , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
10.
Comp Immunol Microbiol Infect Dis ; 69: 101423, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31972500

ABSTRACT

This study compared the efficacy, in terms of reproductive performance, of a porcine reproductive and respiratory syndrome virus (PRRSV)-1 or PRRSV-2 modified-live virus (MLV) vaccine against a dual heterologous PRRSV-1 and PRRSV-2 challenge. Gilts were administered either the PRRSV-1 or PRRSV-2 MLV vaccine at 21 days prior to breeding and were challenged intranasally with both PRRSV species at day 93 of gestation. Vaccination of gilts with PRRSV-2 MLV vaccine resulted in improved reproductive performance in sows (e.g. duration of pregnancy) and piglet health and overall viability (e.g. increase of the number of live-born and weaned pigs, and decrease of stillborn). Vaccination of gilts with PRRSV-1 MLV vaccine was able to reduce only PRRSV-1 viremia in contrast, PRRSV-2 MLV vaccine was able to reduce both PRRSV-1 and PRRSV-2 viremia. Vaccination of gilts with PRRSV-2 MLV induced higher numbers of PRRSV-2 specific interferon-γ secreting cells (IFN-γ-SC) compared to the PRRSV-1 MLV while there was no difference in the number of PRRSV-1 specific IFN-γ-SC between the two vaccines. Taken together, the results presented here suggest that vaccination of gilts with the PRRSV-2 MLV vaccine is more efficacious against dual heterologous PRRSV-1 and PRRSV-2 challenge compared to the PRRSV-1 MLV vaccine.


Subject(s)
Porcine Reproductive and Respiratory Syndrome/prevention & control , Porcine respiratory and reproductive syndrome virus/immunology , Pregnancy Complications, Infectious/prevention & control , Vaccines, Attenuated/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antigens, Viral/immunology , Female , Interferon-gamma/biosynthesis , Open Reading Frames/immunology , Porcine respiratory and reproductive syndrome virus/classification , Pregnancy , Seroconversion , Stillbirth , Swine , Vaccination/veterinary , Vaccines, Attenuated/administration & dosage , Viral Proteins/immunology , Viral Vaccines/administration & dosage
11.
J Virol ; 94(2)2020 01 06.
Article in English | MEDLINE | ID: mdl-31666376

ABSTRACT

We subjected various open reading frames (ORFs) in the genome of respiratory syncytial virus (RSV) to codon pair optimization (CPO) by increasing the content of codon pairs that are overrepresented in the human genome without changing overall codon usage and amino acid sequences. CPO has the potential to increase the expression of the encoded protein(s). Four viruses were made: Max A (with CPO of NS1, NS2, N, P, M, and SH ORFs), Max B (with CPO of G and F), Max L (with CPO of L), and Max FLC (with CPO of all ORFs except M2-1 and M2-2). Because of the possibility of increased viral replication, each CPO virus was attenuated by the inclusion of a codon deletion mutation (Δ1313) and a missense mutation (I1314L) in the L polymerase. CPO had no effect on multicycle virus replication in vitro, temperature sensitivity, or specific infectivity. Max A and L, which in common had CPO of one or more ORFs of proteins of the polymerase complex, exhibited global increases in viral protein synthesis. Max B alone exhibited decreased protein synthesis, and it alone had reduced single-cycle virus replication in vitro All CPO RSVs exhibited marginal reductions in replication in mice and hamsters. Surprisingly, the CPO RSVs induced lower levels of serum RSV-neutralizing antibodies in hamsters. This reduced immunogenicity might reflect reduced viral replication and possibly also the decrease in CpG and UpA dinucleotides as immune stimulators. Overall, our study describes paradoxical effects of CPO of an RNA virus on viral replication and the adaptive humoral immune response.IMPORTANCE Using computer algorithms and large-scale DNA synthesis, one or more ORFs of a microbial pathogen can be recoded by different strategies that involve the introduction of up to thousands of nucleotide changes without affecting amino acid coding. This approach has been used mostly to generate deoptimized viruses used as vaccine candidates. However, the effects of the converse approach of generating optimized viruses are still largely unknown. Here, various ORFs in the genome of respiratory syncytial virus (RSV) were codon pair optimized (CPO) by increasing the content of codon pairs that are overrepresented in the human genome. CPO did not affect RSV replication in multicycle replication experiments in vitro. However, replication was marginally reduced in two rodents models. In hamsters, CPO RSVs induced lower levels of serum RSV-neutralizing antibodies. Thus, CPO of an RNA virus for a mammalian host has paradoxical effects on virus replication and the adaptive humoral immune response.


Subject(s)
Codon Usage , Genome, Viral/immunology , Open Reading Frames/immunology , Respiratory Syncytial Virus Infections , Respiratory Syncytial Viruses/physiology , Virus Replication , A549 Cells , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Chlorocebus aethiops , Cricetinae , Humans , Mesocricetus , Mice , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , Vero Cells , Virus Replication/genetics , Virus Replication/immunology
12.
J Virol ; 93(14)2019 07 15.
Article in English | MEDLINE | ID: mdl-31043533

ABSTRACT

Human herpesvirus 6 (HHV-6) and cytomegalovirus (CMV) are population-prevalent betaherpesviruses with intermittent lytic replication that can be pathogenic in immunocompromised hosts. Elucidation of the adaptive immune response is valuable for understanding pathogenesis and designing novel treatments. Knowledge of T-cell antigens has reached the genome-wide level for CMV and other human herpesviruses, but study of HHV-6 is at an earlier stage. Using rare-cell enrichment combined with an HLA-agnostic, proteome-wide approach, we queried HHV-6B-specific CD4 T cells from 18 healthy donors with each known HHV-6B protein. We detected a low abundance of HHV-6-specific CD4 T cells in blood; however, the within-person CD4 T-cell response is quite broad: the median number of open reading frame (ORF) products recognized was nine per person. Overall, the data expand the number of documented HHV-6B CD4 T-cell antigens from approximately 11 to 60. Epitopes in the proteins encoded by U14, U90, and U95 were mapped with synthetic peptides, and HLA restriction was defined for some responses. Intriguingly, CD4 T-cell antigens newly described in this report are among the most population prevalent, including U73, U72, U95, and U30. Our results indicate that selection of HHV-6B ORFs for immunotherapy should consider this expanded panel of HHV-6B antigens.IMPORTANCE Human herpesvirus 6 is highly prevalent and maintains chronic infection in immunocompetent individuals, with the potential to replicate widely in settings of immunosuppression, leading to clinical disease. Antiviral compounds may be ineffective and/or pose dose-limiting toxicity, and therefore, immune-based therapies have garnered increased interest in recent years. Attempts at addressing this unmet medical need begin with understanding the cellular response to HHV-6 at the individual and population levels. The present study provides a comprehensive assessment of HHV-6-specific T-cell responses that may inform the development of cell-based therapies directed at this virus.


Subject(s)
Antigens, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Herpesvirus 6, Human/immunology , Open Reading Frames/immunology , Roseolovirus Infections/immunology , Antigens, Viral/genetics , Cell Line , Epitope Mapping , Epitopes, T-Lymphocyte/genetics , Genome-Wide Association Study , Herpesvirus 6, Human/genetics , Humans , Roseolovirus Infections/genetics
13.
Sci Rep ; 9(1): 6577, 2019 04 29.
Article in English | MEDLINE | ID: mdl-31036835

ABSTRACT

Somatic mutations in cancer can result in neoantigens against which patients can be vaccinated. The quest for tumor specific neoantigens has yielded no targets that are common to all tumors, yet foreign to healthy cells. Single base pair substitutions (SNVs) at best can alter 1 amino acid which can result in a neoantigen; with the exception of rare site-specific oncogenic driver mutations (such as RAS) such mutations are private. Here, we describe a source of common neoantigens induced by frame shift mutations, based on analysis of 10,186 TCGA tumor samples. We find that these frame shift mutations can produce long neoantigens. These are completely new to the body, and indeed recent evidence suggests that frame shifts can be highly immunogenic. We report that many different frame shift mutations converge to the same small set of 3' neo open reading frame peptides (NOPs), all encoded by the Neo-ORFeome. We find that a fixed set of only 1,244 neo-peptides in as much as 30% of all TCGA cancer patients. For some tumor classes this is higher; e.g. for colon and cervical cancer, peptides derived from only ten genes (saturated at 90 peptides) can be applied to 39% of all patients. 50% of all TCGA patients can be achieved at saturation (using all those peptides in the library found more than once). A pre-fabricated library of vaccines (peptide, RNA or DNA) based on this set can provide off the shelf, quality certified, 'personalized' vaccines within hours, saving months of vaccine preparation. This is crucial for critically ill cancer patients with short average survival expectancy after diagnosis.


Subject(s)
Antigens, Neoplasm/immunology , Neoplasms/immunology , Open Reading Frames/immunology , Peptides/immunology , Amino Acid Substitution/genetics , Antigens, Neoplasm/genetics , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Humans , Mutation/genetics , Neoplasms/drug therapy , Open Reading Frames/genetics , Peptides/genetics , Peptides/therapeutic use
14.
Dev Comp Immunol ; 96: 51-57, 2019 07.
Article in English | MEDLINE | ID: mdl-30822453

ABSTRACT

Bombyx mori cypovirus (BmCPV) is one of the most infectious pathogen in sericulture and a member of the family Reoviridae. It specifically infects the midgut of silkworm. The BmCPV genome consists of 10 dsRNAs segments (S1-S10), which have generally been assumed to be monocistronic. In this study, a small open reading frame encoding the peptide S5-sORF, containing 27 amino acid residues, was predicted in a region of the negative (-) strand of BmCPV segment S5. An immunofluorescence assay detected S5-sORF in the cytoplasm and nuclei of BmCPV-infected cells, and it was also detected in the virion with western blotting, suggesting that S5-sORF may be assembled into the BmCPV virion. Viral gene expression was inhibited by overexpressed S5-sORF, and viral multiplication was dose-dependently suppressed by the S5-sORF peptide. A viable recombinant virus, BmCPV-S5-sORFmut, in which the start codon (ATG) of S5-sORF was mutated to a stop codon (TGA), was generated with reverse genetics. The proliferation of BmCPV was increased by the abolition of S5-sORF expression. Furthermore, the RNA transcript of S5-sORF and small peptide of S5-sORF were involved in BmCPV replication. The expression of genes related to the innate immune pathways and apoptosis in the silkworm were not significantly affected by S5-sORF overexpression. Our results suggest that a viral nucleotide sequence is utilized by the host to generate an antiviral peptide, which may be a novel strategy protecting the host from viral infection.


Subject(s)
Arthropod Proteins/immunology , Bombyx/immunology , Host-Pathogen Interactions/immunology , Peptides/immunology , Reoviridae/genetics , Animals , Arthropod Proteins/genetics , Bombyx/genetics , Bombyx/virology , Cell Line , Host-Pathogen Interactions/genetics , Open Reading Frames/genetics , Open Reading Frames/immunology , Peptides/genetics , RNA, Double-Stranded/genetics , RNA, Viral/genetics , Reoviridae/pathogenicity , Viral Proteins/genetics , Virion/genetics , Virus Replication/immunology
15.
Vaccine ; 37(14): 1928-1937, 2019 03 28.
Article in English | MEDLINE | ID: mdl-30824359

ABSTRACT

Porcine Circovirus Type 2 (PCV2) is one of the most important pathogens in pigs around the world. PCV2 is a non-enveloped virus and its capsid is formed by a single protein known as open reading frame 2 (ORF2). The aim of this study was to evaluate the antigenicity and immunogenicity of genetically-encoded protein nanoparticles (NPs) containing ORF2 from PCV2 fused to the first 110 amino acids of the N-terminus of polyhedrin from the insect virus Autographa californica nucleopolyhedrovirus (PH(1 -1 1 0)). Our group has previously described that some polyhedrin fragments self-aggregate forming polyhedra-like particles. We identified a self-aggregating signal within the first 110 amino acids from polyhedrin (PH(1 -1 1 0)). Fusing the ORF2 from PCV2 to the carboxyl terminus from PH(1 -1 1 0) results in the formation of NPs which incorporate the antigen of interest. Using this system we synthesized NPs containing PH(1 -1 1 0) fused to ORF2 (PH(1 -1 1 0)PCV2) and purify them to immunize pigs and evaluate the humoral immune response generated by these NPs comparing them to a commercially available vaccine. Pigs immunized with PH(1 -1 1 0)PCV2 NPs produced antibodies against ORF2 from PCV2 as indicated by western blot and ELISA analysis. Antibodies obtained with PH(1 -1 1 0)PCV2 NPs were comparable to those obtained using a commercial PCV2 vaccine. These antibodies neutralized the infection of a recombinant PCV2 expressing the green fluorescent protein (GFP). These results together suggest that the self-aggregating peptide PH(1 -1 1 0) can be used for the synthesis of subunit vaccines against PCV2.


Subject(s)
Circoviridae Infections/veterinary , Circovirus/genetics , Circovirus/immunology , Nanoparticles , Open Reading Frames/immunology , Porcine Postweaning Multisystemic Wasting Syndrome/prevention & control , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cell Line , Enzyme-Linked Immunosorbent Assay , Female , Immunization , Male , Open Reading Frames/genetics , Swine , Vaccines, Subunit , Viral Vaccines/chemistry , Viral Vaccines/genetics
17.
Cytokine ; 113: 31-38, 2019 01.
Article in English | MEDLINE | ID: mdl-29885990

ABSTRACT

Human interferon regulatory factor 7 (IRF7) plays an important role in the innate antiviral immune response. To date, the characteristics and functions of waterfowl IRF7 have not been clarified. This study reports the cDNA sequence, tissue distribution, and antiviral function of duck IRF7. The duck IRF7 gene has a 1536-bp open read frame (ORF) and encodes a 511-amino acid polypeptide. IRF7 is highly expressed in the blood and pancreas of 5-day-old ducklings and in the small intestine, large intestine and liver of 60-day-old adult ducks. Indirect immunofluorescence assay (IFA) showed that over-expressed duck IRF7 was located in both the cytoplasm and nucleus of transfected duck embryo fibroblasts (DEFs), which was also observed in poly(I:C)-stimulated or duck Tembusu virus (DTMUV)-infected DEFs. Titres and copies of DTMUV were significantly reduced in DEFs overexpressing IRF7. Moreover, overexpression of duck IRF7 significantly induced IFNα/ß, but not IFNγ, mRNA expression, and transcription of downstream interferon-stimulated genes (ISGs), such as MX, OASL and IL-6, which were significantly induced by poly(I:C) co-stimulation, was enhanced. Additionally, duck IRF7 overexpression can significantly activate the IFNß promoter in DEFs. Collectively, duck IRF7 plays an important role in host anti-DTMUV immune regulation, which depends on type I interferons and associated signal transduction pathway(s).


Subject(s)
Ducks/immunology , Ducks/virology , Flavivirus Infections/immunology , Flavivirus/immunology , Interferon Regulatory Factor-7/immunology , Interferon Type I/immunology , Signal Transduction/immunology , Animals , Cell Nucleus/immunology , Cell Nucleus/virology , Cytoplasm/immunology , Cytoplasm/virology , Fibroblasts/immunology , Fibroblasts/virology , Flavivirus Infections/virology , Open Reading Frames/immunology , Peptides/immunology , Promoter Regions, Genetic/immunology
18.
Front Immunol ; 10: 2984, 2019.
Article in English | MEDLINE | ID: mdl-31921215

ABSTRACT

Gammaherpesviruses (γHV) are important pathogens causing persistent infections which lead to several malignancies in immunocompromised patients. Murine γHV 68 (MHV-68), a homolog to human EBV and KSHV, has been employed as a classical pathogen to investigate the molecular pathogenicity of γHV infections. γHV express distinct antigens during lytic or latent infection and antigen-specific T cells have a significant role in controlling the acute and latent viral infection, although the quality of anti-viral T cell responses required for protective immunity is not well-understood. We have generated recombinant modified vaccinia virus Ankara (recMVA) vaccines via MVA-BAC homologous recombination technology expressing MHV-68 ORF6 and ORF61 antigens encoding both MHC class I and II-restricted epitopes. After vaccination, we examined T cell responses before and after MHV-68 infection to determine their involvement in latent virus control. We show recognition of recMVA- and MHV-68-infected APC by ORF6 and ORF61 epitope-specific T cell lines in vitro. The recMVA vaccines efficiently induced MHV-68-specific CD8+ and CD4+ T cell responses after a single immunization and more pronounced after homologous prime/boost vaccination in mice. Moreover, we exhibit protective capacity of prophylactic recMVA vaccination during early latency at day 17 after intranasal challenge with MHV-68, but failed to protect from latency at day 45. Further T cell analysis indicated that T cell exhaustion was not responsible for the lack of protection by recMVA vaccination in long-term latency at day 45. The data support further efforts aiming at improved vaccine development against γHV infections with special focus on targeting protective CD4+ T cell responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Herpesviridae Infections , Open Reading Frames , Rhadinovirus/physiology , Vaccinia virus , Viral Proteins , Viral Vaccines , Virus Latency , Animals , HeLa Cells , Herpesviridae Infections/genetics , Herpesviridae Infections/immunology , Herpesviridae Infections/prevention & control , Humans , Mice , NIH 3T3 Cells , Open Reading Frames/genetics , Open Reading Frames/immunology , Vaccines, DNA , Vaccinia virus/genetics , Vaccinia virus/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Viral Vaccines/genetics , Viral Vaccines/immunology , Virus Latency/genetics , Virus Latency/immunology
19.
J Infect Dis ; 219(7): 1058-1066, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30383234

ABSTRACT

BACKGROUND: Orolabial herpes simplex virus type 1 (HSV-1) infection has a wide spectrum of severity in immunocompetent persons. To study the role of viral genotype and host immunity, we characterized oral HSV-1 shedding rates and host cellular response, and genotyped viral strains, in monozygotic (MZ) and dizygotic (DZ) twins. METHODS: A total of 29 MZ and 22 DZ HSV-1-seropositive twin pairs were evaluated for oral HSV-1 shedding for 60 days. HSV-1 strains from twins were genotyped as identical or different. CD4+ T-cell responses to HSV-1 proteins were studied. RESULTS: The median per person oral HSV shedding rate was 9% of days that a swab was obtained (mean, 10.2% of days). A positive correlation between shedding rates was observed within all twin pairs, and in the MZ and DZ twins. In twin subsets with sufficient HSV-1 DNA to genotype, 15 had the same strain and 14 had different strains. Viral shedding rates were correlated for those with the same but not different strains. The median number of HSV-1 open reading frames recognized per person was 16. The agreement in the CD4+ T-cell response to specific HSV-1 open reading frames was greater between MZ twins than between unrelated persons (P = .002). CONCLUSION: Viral strain characteristics likely contribute to oral HSV-1 shedding rates.


Subject(s)
Herpes Labialis/immunology , Herpes Labialis/virology , Herpesvirus 1, Human/genetics , Virus Shedding/genetics , Adult , Aged , CD4-Positive T-Lymphocytes/immunology , Female , Genotype , Herpes Labialis/classification , Herpesvirus 1, Human/physiology , Humans , Male , Middle Aged , Mouth/virology , Open Reading Frames/genetics , Open Reading Frames/immunology , Phylogeny , Twins, Dizygotic , Twins, Monozygotic , Young Adult
20.
Dev Comp Immunol ; 87: 188-192, 2018 10.
Article in English | MEDLINE | ID: mdl-29944898

ABSTRACT

Bombyx mori bidensovirus (BmBDV) causes fatal flacherie disease leading to severe economic losses in sericultures. The BmDNV-Z genome contains two single-stranded DNA molecules, VD1 and VD2. For generating silkworm lines with antiviral properties, two transgenic RNA interference (RNAi) vectors were constructed. Open reading frames (ORFs) 1-4 of VD1 were knockdown by vector pb-BDV1 while ORF1a, ORF1b, and ORF3 of VD2 were knockdown by vector pb-BDV2. Transgenic silkworm lines BDV1-I and BDV2-I were generated via RNAi microinjection. Mortality rates of BDV1-I and BDV2-I were reduced by 45% and 39%, respectively, and quantitative PCR showed that VD1 and VD2 contents in BDV1-I and BDV2-I were significantly lower than in the non-transgenic line. However, economic traits showed no obvious differences. Thus, knockdown of multiple BmDNV-Z genes provides strong resistance to BDV1-I and BDV2-I lines, and these can be used in sericulture without hampering silk production.


Subject(s)
Bombyx/immunology , Genes, Viral/immunology , Insect Viruses/immunology , RNA Interference/immunology , Animals , Animals, Genetically Modified , Bombyx/genetics , Bombyx/virology , Disease Resistance/genetics , Disease Resistance/immunology , Genes, Viral/genetics , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Insect Viruses/genetics , Insect Viruses/physiology , Open Reading Frames/genetics , Open Reading Frames/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...