Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Vaccine ; 41(2): 304-314, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36587961

ABSTRACT

All seasonal influenza vaccines for 2021-2022 in the US were quadrivalent and the market continues to be dominated by intramuscular delivery of non-adjuvanted, virion-derived antigens grown in chicken eggs. Up to four new egg-adapted production influenza vaccine strains must be generated each year. The introduction in 2012 of Flucelvax®, which is grown in mammalian suspension cell culture and uses vaccine production strains without adaptive mutations for efficient growth in eggs, represented a major advance in vaccine production technology. Here we demonstrate that Flucelvax can be reformulated and combined with a liposomal adjuvant containing QS-21 (Verndari Adjuvant System 1.1, VAS1.1) or QS-21 and 3D-PHAD (VAS1.2) for intradermal administration using a painless skin patch, VaxiPatch™. VAS1.2 is similar to AS01B, the adjuvant system used in Shingrix® and Mosquirix™. We show that Flucelvax, when reformulated and concentrated using tangential flow filtration (TFF), maintains hemagglutination and single radial immunodiffusion (SRID) potency. Loading the reformulated Flucelvax material onto VaxiPatch arrays conferred high levels of resistance to heat stress and room temperature stability. TFF enriched vaccine antigens were combined with VAS1.1 or VAS1.2 and dispensed in 10nL drops into the pockets of 36 (total 360 nL) stainless steel microneedles arranged in a microarray 1.2 cm in diameter. Using VaxiPatch delivery of 2 µg of antigen, we demonstrated intramusuclar-comparable IgG and hemagglutination inhibition (HAI) immune responses in Sprague Dawley® rats. With addition of VAS1.2, antigen-specific IgG titers were increased as much as 68-fold (47-fold for VAS1.1) with improvements in seroconversion for three of four strains (all four were improved by VAS1.1). TFF-reformulated antigens combined with VAS1.1 or VAS1.2 and delivered by VaxiPatch showed only minor skin reactogenicity after 1 h and no skin reactogenicity after 24 h. These data indicate that VaxiPatch and the VAS system have the potential to be transformative for vaccine delivery.


Subject(s)
Influenza Vaccines , Influenza, Human , Rats , Animals , Humans , Seasons , Rats, Sprague-Dawley , Influenza, Human/prevention & control , Adjuvants, Immunologic , Vaccination , Hemagglutination Inhibition Tests , Vaccines, Combined , Antibodies, Viral , Immunoglobulin G , Injections, Intradermal , Mammals
2.
Vaccine ; 38(43): 6839-6848, 2020 10 07.
Article in English | MEDLINE | ID: mdl-32741668

ABSTRACT

This work introduces VaxiPatch, a novel vaccination system comprised of subunit glycoprotein vaccine antigens, adjuvants and dermal delivery. For this study, rHA of influenza virus B/Colorado/06/2017 was incorporated into synthetic virosomes, and adjuvant liposomes were formed with QS-21 from Saponaria quillaja, with or without the synthetic TLR4 agonist 3D - (6-acyl) PHAD. These components were concentrated and co-formulated into trehalose with dye. Dermal delivery was achieved using an economical 37-point stainless steel microneedle array, designed for automated fill/finish by microfluidic dispensers used for mass production of immunodiagnostics. Vaccine and adjuvant are deposited to form a sugar glass in a pocket on the side of each of the tips, allowing skin penetration to be performed directly by the rigid steel structure. In this study, Sprague Dawley rats (n = 6 per group) were vaccinated by VaxiPatches containing 0.3 µg of rHA, 0.5 µg QS-21 and 0.2 µg 3D - (6-acyl) PHAD and dye, resulting in antigen-specific IgG titers 100-fold higher than 4.5 µg of FluBlok (p = 0.001) delivered intramuscularly. Similarly, hemagglutination inhibition titers in these animals were 14-fold higher than FluBlok controls (p = 0.01). Non-adjuvanted VaxiPatches were also compared with rHA virosomes injected intramuscularly. Accelerated shelf life studies further suggest that formulated virosomal antigens retain activity for at least two months at 60° C. Further, co-formulation of a dye could provide a visible verification of delivery based on the temporary pattern on the skin. A room-temperature-stable vaccination kit such as VaxiPatch has the potential to increase vaccine use and compliance globally.


Subject(s)
Influenza Vaccines , Influenza, Human , Adjuvants, Immunologic , Animals , Antibodies, Viral , Hemagglutination Inhibition Tests , Rats , Rats, Sprague-Dawley , Vaccination
3.
PLoS One ; 9(11): e111502, 2014.
Article in English | MEDLINE | ID: mdl-25402328

ABSTRACT

The availability of reliable recombinant reporter virus systems has been a great boon to the study of Kaposi's sarcoma-associated herpesvirus (KSHV/HHV-8). Unexpectedly, we found that expression of the ostensibly constitutive green fluorescent protein (GFP) marker was progressively lost during unselected passage in primary rat mesenchymal precursor cells (MM), despite efficient maintenance of latent viral gene expression and episomal partitioning. This repression of EF1-α promoter-driven GFP expression appeared to be passage-dependent, however, since functionally immortalized MM cells derived from long serial passage retained stable expression of GFP following rKSHV.219 infection. Chromatin analysis of cultures that we had infected in parallel demonstrated an increase in repressive H3K27 tri-methylation across the viral episome with the exception of the LANA control region in MM cells infected at early rather than late passage post-isolation. The silencing of GFP expression in the MM cells was reversible in a dose-dependent fashion by the histone deacetylase inhibitor valproic acid, further implicating cellular silencing on incoming viral genomes, and underscoring potential differences in viral gene regulation between primary and functionally immortalized cells. Furthermore, using multispectral imaging flow cytometry, we also determined that the extent of GFP expression per cell among those that were positive did not correlate with the number of LANA dots per nucleus nor the extent of overall LANA expression per cell. This suggests a more complex mode of local gene regulation, rather than one that simply reflects the relative intracellular viral copy number. In sum, we have demonstrated the significant potential for false-negative data when using a constitutive marker gene as a sole means of evaluating herpesviral infection, especially in primary cells.


Subject(s)
Gene Silencing , Genes, Reporter , Genetic Vectors/genetics , Herpesviridae/genetics , Plasmids/genetics , Transduction, Genetic , Animals , Antigens, Viral/metabolism , Chromatin/metabolism , Chromatin Assembly and Disassembly/drug effects , Epigenesis, Genetic/drug effects , Gene Expression , Gene Silencing/drug effects , Herpesvirus 8, Human/genetics , Histones/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/virology , Nuclear Proteins/metabolism , Primary Cell Culture , Rats , Valproic Acid/pharmacology
4.
J Clin Invest ; 121(2): 752-68, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21245574

ABSTRACT

Kaposi sarcoma-associated herpesvirus (KSHV; also known as HHV8) is the causative agent of two B cell tumors, multicentric Castleman disease (MCD) and primary effusion lymphoma (PEL). However, little is known about the nature of the specific B cell subtype(s) most susceptible to infection. Identifying these cells would provide direct insight into KSHV transmission and virus-induced transformation. To identify this subset and to determine whether infection alters its cellular phenotype, we exposed human tonsillar cells to KSHV and characterized infected cells using high-throughput multispectral imaging flow cytometry (MIFC). Stable expression of the virally encoded latency-associated nuclear antigen (LANA), a marker of latent KSHV infection, was observed predominantly in cells expressing the l light chain of the B cell receptor. These LANA+ B cells proliferated and exhibited similarities to the cells characteristic of MCD (IgMl-expressing plasmablasts), including blasting morphology with elevated expression of Ki67, variable expression of CD27, and high levels of IgM and IL-6 receptor. Furthermore, the proportion of infected cells showing a blasting phenotype increased upon addition of exogenous IL-6. Our data lead us to propose that oral transmission of KSHV involves the latent infection of a subset of tonsillar IgMl-expressing B cells, which then proliferate as they acquire the plasmablast phenotype characteristic of MCD.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Proliferation , Herpesvirus 8, Human/immunology , Palatine Tonsil/cytology , Plasma Cells/immunology , Animals , Antigens, Viral/immunology , B-Lymphocytes/cytology , Castleman Disease/immunology , Castleman Disease/pathology , Castleman Disease/virology , Cells, Cultured , Disease Transmission, Infectious , Genome, Viral , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/pathogenicity , Humans , Lymphoma, Primary Effusion/immunology , Lymphoma, Primary Effusion/pathology , Lymphoma, Primary Effusion/virology , Nuclear Proteins/immunology , Palatine Tonsil/immunology , Plasma Cells/cytology
5.
J Biol Chem ; 285(8): 5266-73, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20034935

ABSTRACT

Sumoylation has emerged as a major post-translational modification of cellular proteins, affecting a variety of cellular processes. Viruses have exploited the sumoylation pathway to advance their own replication by evolving several ways to perturb the host sumoylation apparatus. However, there has been no report of virally encoded enzymes directly involved in catalyzing the sumoylation reaction. Here, we report that the K-bZIP protein encoded by Kaposi's sarcoma-associated herpesvirus (KSHV) is a SUMO E3 ligase with specificity toward SUMO2/3. K-bZIP is a nuclear factor that functions to modulate viral gene expression and to prolong the G1 phase, allowing viral transcription and translation to proceed at the early stage of infection. In addition to functioning as a transcriptional factor, we show that K-bZIP carries a SIM (SUMO-interacting motif), which specifically binds to SUMO-2/3 but not SUMO-1. K-bZIP catalyzes its own SUMO modification as well as that of its interacting partners such as the cellular tumor suppressor proteins p53 and Rb, both in vitro and in vivo. This reaction depends on an intact SIM. Sumoylation of p53 leads to its activation and K-bZIP is recruited to several p53 target chromatin sites in a SIM-dependent manner. In addition to the identification of a viral SUMO-2/3 E3 ligase, our results provide additional insights into the mechanisms whereby K-bZIP induces cell cycle arrest.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Herpesvirus 8, Human/enzymology , Repressor Proteins/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitins/metabolism , Viral Proteins/metabolism , Amino Acid Motifs/physiology , Basic-Leucine Zipper Transcription Factors/genetics , Cell Line , G1 Phase/genetics , Gene Expression Regulation, Viral/physiology , Herpesvirus 8, Human/genetics , Humans , Protein Processing, Post-Translational/genetics , Repressor Proteins/genetics , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Small Ubiquitin-Related Modifier Proteins/genetics , Substrate Specificity/physiology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitins/genetics , Viral Proteins/genetics
6.
Cancer Res ; 69(14): 5681-9, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19584288

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) has been linked to the development of Kaposi's sarcoma, a major AIDS-associated malignancy, and to hematologic malignancies, including primary effusion lymphoma and multicentric Castleman's disease. Like other herpesviruses, KSHV is capable of both latent and lytic replication. Understanding the molecular details associated with this transition from latency to lytic replication is key to controlling virus spread and can affect the development of intervention strategies. Here, we report that Kruppel-associated box domain-associated protein-1 (KAP-1)/transcriptional intermediary factor 1beta, a cellular transcriptional repressor that controls chromosomal remodeling, participates in the process of switching viral latency to lytic replication. Knockdown of KAP-1 by small interfering RNA leads to KSHV reactivation mediated by K-Rta, a key transcriptional regulator. In cells harboring latent KSHV, KAP-1 was associated with the majority of viral lytic-gene promoters. K-Rta overexpression induced the viral lytic cycle with concomitant reduction of KAP-1 binding to viral promoters. Association of KAP-1 with heterochromatin was modulated by both sumoylation and phosphorylation. During lytic replication of KSHV, KAP-1 was phosphorylated at Ser(824). Several lines of evidence directly linked the viral protein kinase to this post-translational modification. Additional studies showed that this phosphorylation of KAP-1 produced a decrease in its sumoylation, consequently decreasing the ability of KAP-1 to condense chromatin on viral promoters. In summary, the cellular transcriptional repressor KAP-1 plays a role in regulating KSHV latency, and viral protein kinase modulates the chromatin remodeling function of this repressor.


Subject(s)
Herpesvirus 8, Human/physiology , Protein Kinases/metabolism , Repressor Proteins/metabolism , Viral Proteins/metabolism , Blotting, Western , Cell Line , Cell Line, Tumor , Chromatin Immunoprecipitation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Herpesvirus 8, Human/genetics , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Phosphorylation , Protein Binding , Protein Kinases/genetics , RNA Interference , Repressor Proteins/genetics , Serine/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Tripartite Motif-Containing Protein 28 , Viral Proteins/genetics , Virus Activation , Virus Latency , Virus Replication
7.
Virology ; 387(1): 76-88, 2009 Apr 25.
Article in English | MEDLINE | ID: mdl-19269659

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiologic agent of Kaposi's sarcoma. K-Rta and K-bZIP are two major viral transcription factors that control reactivation of this virus. Here we report a genome-wide analysis of transcriptional capacity by evaluation of a comprehensive library of 83 putative KSHV promoters. In reporter assays, 34 viral promoters were activated by K-Rta, whereas K-bZIP activated 21 promoters. When K-Rta and K-bZIP were combined, 3 K-Rta responsive promoters were repressed by K-bZIP. The occupancy of K-Rta and K-bZIP across KSHV promoters was analyzed by chromatin immunoprecipitation with a viral promoter-chip in BCBL-1 cells. In addition, acetylation of local histones was examined to determine accessibility of promoters during latency and reactivation. Finally, 10 promoters were selected to study the dynamics of transcription factor recruitment. This study provides a comprehensive overview of the responsiveness of KSHV promoters to K-Rta and K-bZIP, and describes key chromatin changes during viral reactivation.


Subject(s)
Herpesvirus 8, Human/physiology , Immediate-Early Proteins/metabolism , Promoter Regions, Genetic/physiology , Trans-Activators/physiology , Virus Activation , Basic-Leucine Zipper Transcription Factors , Cell Line , DNA, Viral/genetics , Gene Expression Regulation, Viral , Herpesvirus 8, Human/genetics , Humans , Trans-Activators/chemistry , Trans-Activators/metabolism , Transcriptional Activation , Virus Replication
8.
J Virol ; 83(9): 4435-46, 2009 May.
Article in English | MEDLINE | ID: mdl-19244329

ABSTRACT

Successful viral replication is dependent on a conducive cellular environment; thus, viruses must be sensitive to the state of their host cells. We examined the idea that an interplay between viral and cellular regulatory factors determines the switch from Kaposi's sarcoma-associated herpesvirus (KSHV) latency to lytic replication. The immediate-early gene product K-Rta is the first viral protein expressed and an essential factor in reactivation; accordingly, this viral protein is in a key position to serve as a viral sensor of cellular physiology. Our approach aimed to define a host transcription factor, i.e., host sensor, which modulates K-Rta activity on viral promoters. To this end, we developed a panel of reporter plasmids containing all 83 putative viral promoters for a comprehensive survey of the response to both K-Rta and cellular transcription factors. Interestingly, members of the NF-kappaB family were shown to be strong negative regulators of K-Rta transactivation for all but two viral promoters (Ori-RNA and K12). Recruitment of K-Rta to the ORF57 and K-bZIP promoters, but not the K12 promoter, was significantly impaired when NF-kappaB expression was induced. Many K-Rta-responsive promoters modulated by NF-kappaB contain the sequence of the RBP-Jkappa binding site, a major coactivator which anchors K-Rta to target promoters via consensus motifs which overlap with that of NF-kappaB. Gel shift assays demonstrated that NF-kappaB inhibits the binding of RBP-Jkappa and forms a complex with RBP-Jkappa. Our results support a model in which a balance between K-Rta/RBP-Jkappa and NF-kappaB activities determines KSHV reactivation. An important feature of this model is that the interplay between RBP-Jkappa and NF-kappaB on viral promoters controls viral gene expression mediated by K-Rta.


Subject(s)
Down-Regulation , Herpesvirus 8, Human/metabolism , Immediate-Early Proteins/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/antagonists & inhibitors , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , NF-kappa B/metabolism , Trans-Activators/metabolism , Virus Latency , Cell Line , Gene Expression Regulation, Viral , Herpesvirus 8, Human/genetics , Humans , Immediate-Early Proteins/genetics , NF-kappa B/genetics , Promoter Regions, Genetic/genetics , Trans-Activators/genetics , Virus Activation
9.
J Virol ; 79(15): 9912-25, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16014952

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is a human gammaherpesvirus implicated in AIDS-related neoplasms. Previously, we demonstrated that the early lytic gene product K-bZIP is a transcriptional repressor that affects a subset of viral gene transcriptions mediated by the viral transactivator K-Rta (Y. Izumiya et al. J. Virol. 77:1441-1451, 2003). Sumoylation has emerged as an important posttranslational modification that affects the location and function of cellular and viral proteins and also plays a significant role in transcriptional repression along with Ubc9, the E2 SUMO conjugation enzyme. Here, we provide evidence that K-bZIP is sumoylated at the lysine 158 residue and associates with Ubc9 both in a cell-free system and in virus-infected BCBL-1 cells. Reporter assays showed that the expression of SUMO-specific protease 1 attenuated the transcriptional repression activity of K-bZIP. The expression of a K-bZIPK158R mutant, which was no longer sumoylated, exhibited the reduced transcriptional repression activity. This indicates that sumoylation plays an important part in the transcriptional repression activity of K-bZIP. Finally, chromatin immunoprecipitation experiments demonstrated that K-bZIP interacts with and recruits Ubc9 to specific KSHV promoters. Thus, our data indicate that K-bZIP is a SUMO adaptor, which recruits Ubc9 to specific viral target promoters, thereby exerting its transcriptional repression activity.


Subject(s)
Herpesvirus 8, Human/physiology , SUMO-1 Protein/metabolism , Transcription, Genetic , Basic-Leucine Zipper Transcription Factors , Carrier Proteins , Cell Culture Techniques , Gene Expression Regulation, Viral , Herpesvirus 8, Human/genetics , Humans , Lysine/metabolism , Peptide Hydrolases/metabolism , Promoter Regions, Genetic , Repressor Proteins , Ubiquitin-Conjugating Enzymes/metabolism , Viral Proteins
10.
J Virol ; 77(17): 9652-61, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12915577

ABSTRACT

In order to cope with hostile host environments, many viruses have developed strategies to perturb the cellular machinery to suit their replication needs. Some herpesvirus genes protect cells from undergoing apoptosis to prolong the lives of infected cells, while others, such as Epstein-Barr virus Zta, slow down the G(1)/S transition phase to allow ample opportunity for transcription and translation of viral genes before the onset of cellular genomic replication. In this study, we investigated whether Kaposi's sarcoma-associated herpesvirus (KSHV) K-bZIP, a homologue of the Epstein-Barr virus transcription factor BZLF1 (Zta), plays a role in cell cycle regulation. Here we show that K-bZIP physically associates with cyclin-CDK2 and downmodulates its kinase activity. The association can be detected in the natural environment of KSHV-infected cells without artificial overexpression of either component. With purified protein, it can be shown that the interaction between K-bZIP and cyclin-CDK2 is direct and that K-bZIP alone is sufficient to inhibit CDK2 activity. The interacting domain of K-bZIP has been mapped to the basic region. The result of these associations is a prolonged G(1) phase, accompanied by the induction of p21 and p27 in a naturally infected B-cell line. Thus, in addition to the previously described transcription and genome replication functions, a new role of K-bZIP in KSHV replication is identified in this report.


Subject(s)
CDC2-CDC28 Kinases , Carrier Proteins/physiology , Cell Cycle/physiology , Cyclin-Dependent Kinases/physiology , Herpesvirus 8, Human/physiology , Protein Serine-Threonine Kinases/physiology , Viral Proteins/physiology , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Basic-Leucine Zipper Transcription Factors , Binding Sites , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line , Cyclin-Dependent Kinase 2 , G1 Phase/physiology , Gene Expression , Genes, Viral , HeLa Cells , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/pathogenicity , Humans , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins , Sequence Deletion , Transfection , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Replication/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...