Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
J Biol Chem ; 298(6): 102012, 2022 06.
Article in English | MEDLINE | ID: mdl-35525271

ABSTRACT

Constitutive activation of the canonical NF-κB signaling pathway is a major factor in Kaposi's sarcoma-associated herpes virus pathogenesis where it is essential for the survival of primary effusion lymphoma. Central to this process is persistent upregulation of the inhibitor of κB kinase (IKK) complex by the virally encoded oncoprotein vFLIP. Although the physical interaction between vFLIP and the IKK kinase regulatory component essential for persistent activation, IKKγ, has been well characterized, it remains unclear how the kinase subunits are rendered active mechanistically. Using a combination of cell-based assays, biophysical techniques, and structural biology, we demonstrate here that vFLIP alone is sufficient to activate the IKK kinase complex. Furthermore, we identify weakly stabilized, high molecular weight vFLIP-IKKγ assemblies that are key to the activation process. Taken together, our results are the first to reveal that vFLIP-induced NF-κB activation pivots on the formation of structurally specific vFLIP-IKKγ multimers which have an important role in rendering the kinase subunits active through a process of autophosphorylation. This mechanism of NF-κB activation is in contrast to those utilized by endogenous cytokines and cellular FLIP homologues.


Subject(s)
Herpesvirus 8, Human , Sarcoma, Kaposi , Enzyme Activation/genetics , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/metabolism , Humans , I-kappa B Kinase/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Oncogene Proteins/metabolism , Sarcoma, Kaposi/enzymology , Sarcoma, Kaposi/virology , Viral Proteins/metabolism
2.
J Virol ; 92(23)2018 12 01.
Article in English | MEDLINE | ID: mdl-30232190

ABSTRACT

Vesicular stomatitis virus Indiana strain G protein (VSVind.G) is the most commonly used envelope glycoprotein to pseudotype lentiviral vectors (LV) for experimental and clinical applications. Recently, G proteins derived from other vesiculoviruses (VesG), for example, Cocal virus, have been proposed as alternative LV envelopes with possible advantages over VSVind.G. Well-characterized antibodies that recognize VesG will be useful for vesiculovirus research, development of G protein-containing advanced therapy medicinal products (ATMPs), and deployment of VSVind-based vaccine vectors. Here, we show that one commercially available monoclonal antibody, 8G5F11, binds to and neutralizes G proteins from three strains of VSV, as well as Cocal and Maraba viruses, whereas the other commercially available monoclonal anti-VSVind.G antibody, IE9F9, binds to and neutralizes only VSVind.G. Using a combination of G protein chimeras and site-directed mutations, we mapped the binding epitopes of IE9F9 and 8G5F11 on VSVind.G. IE9F9 binds close to the receptor binding site and competes with soluble low-density lipoprotein receptor (LDLR) for binding to VSVind.G, explaining its mechanism of neutralization. In contrast, 8G5F11 binds close to a region known to undergo conformational changes when the G protein moves to its postfusion structure, and we propose that 8G5F11 cross-neutralizes VesGs by inhibiting this.IMPORTANCE VSVind.G is currently regarded as the gold-standard envelope glycoprotein to pseudotype lentiviral vectors. However, recently other G proteins derived from vesiculoviruses have been proposed as alternative envelopes. Here, we investigated two commercially available anti-VSVind.G monoclonal antibodies for their ability to cross-react with other vesiculovirus G proteins, identified the epitopes they recognize, and explored their neutralization activity. We have identified 8G5F11, for the first time, as a cross-neutralizing antibody against several vesiculovirus G proteins. Furthermore, we elucidated the two different neutralization mechanisms employed by these two monoclonal antibodies. Understanding how cross-neutralizing antibodies interact with other G proteins may be of interest in the context of host-pathogen interaction and coevolution, as well as providing the opportunity to modify the G proteins and improve G protein-containing medicinal products and vaccine vectors.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antigens, Viral/immunology , Epitopes/immunology , Membrane Glycoproteins/immunology , Vesicular Stomatitis/immunology , Vesicular stomatitis Indiana virus/immunology , Viral Envelope Proteins/immunology , Amino Acid Sequence , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/metabolism , Antigens, Viral/genetics , Antigens, Viral/metabolism , Cross Reactions , Epitopes/metabolism , HEK293 Cells , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Neutralization Tests , Phylogeny , Sequence Homology , Vesicular Stomatitis/metabolism , Vesicular Stomatitis/virology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
3.
J Virol ; 91(23)2017 12 01.
Article in English | MEDLINE | ID: mdl-28931678

ABSTRACT

Primary effusion lymphoma (PEL) is a lymphogenic disorder associated with Kaposi's sarcoma-associated herpesvirus (KSHV) infection. Key to the survival and proliferation of PEL is the canonical NF-κB pathway, which becomes constitutively activated following overexpression of the viral oncoprotein KSHV vFLIP (ks-vFLIP). This arises from its capacity to form a complex with the modulatory subunit of the IκB kinase (IKK) kinase, IKKγ (or NEMO), resulting in the overproduction of proteins that promote cellular survival and prevent apoptosis, both of which are important drivers of tumorigenesis. Using a combination of cell-based and biophysical assays together with structural techniques, we showed that the observed resistance to cell death is largely independent of autophagy or major death receptor signaling pathways and demonstrated that direct targeting of the ks-vFLIP-IKKγ interaction both in cells and in vitro can be achieved using IKKγ-mimetic peptides. Our results further reveal that these peptides not only induce cell killing but also potently sensitize PEL to the proapoptotic agents tumor necrosis factor alpha and etoposide and are the first to confirm ks-vFLIP as a tractable target for the treatment of PEL and related disorders.IMPORTANCE KSHV vFLIP (ks-vFLIP) has been shown to have a crucial role in cellular transformation, in which it is vital for the survival and proliferation of primary effusion lymphoma (PEL), an aggressive malignancy associated with infection that is resistant to the majority of chemotherapeutic drugs. It operates via subversion of the canonical NF-κB pathway, which requires a physical interaction between ks-vFLIP and the IKK kinase modulatory subunit IKKγ. While this interaction has been directly linked to protection against apoptosis, it is unclear whether the suppression of other cell death pathways implicated in ks-vFLIP pathogenesis is an additional contributor. We demonstrate that the interaction between ks-vFLIP and IKKγ is pivotal in conferring resistance to apoptosis. Additionally, we show that the ks-vFLIP-IKKγ complex can be disrupted using peptides leading to direct killing and the sensitization of PEL cells to proapoptotic agents. Our studies thus provide a framework for future therapeutic interventions.


Subject(s)
Apoptosis , Herpesvirus 8, Human/physiology , I-kappa B Kinase/chemistry , Peptides/metabolism , Peptides/pharmacology , Sarcoma, Kaposi/virology , Autophagy , Etoposide/pharmacology , Herpesvirus 8, Human/chemistry , Humans , I-kappa B Kinase/metabolism , Jurkat Cells , Molecular Mimicry , Peptides/chemistry , Protein Binding , Sarcoma, Kaposi/physiopathology , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Viral Proteins/metabolism
4.
J Biol Chem ; 291(14): 7608-20, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26865630

ABSTRACT

The viral FLICE-like inhibitory protein (FLIP) protein from Kaposi sarcoma-associated herpesvirus activates the NF-κB pathway by forming a stable complex with a central region (amino acids 150-272) of the inhibitor of NF-κB kinase (IKK) γ subunits, thereby activating IKK. Cellular FLIP (cFLIP) forms are also known to activate the NF-κB pathway via IKK activation. Here we demonstrate that cFLIPL, cFLIPS, and their proteolytic product p22-FLIP all require the C-terminal region of NEMO/IKKγ (amino acids 272-419) and its ubiquitin binding function for activation of the IKK kinase (or kinase complex), but none form a stable complex with IKKγ. Our results further reveal that cFLIPLrequires the linear ubiquitin chain assembly complex and the kinase TAK1 for activation of the IKK kinase. Similarly, cFLIPSand p22-FLIP also require TAK1 but do not require LUBAC. In contrast, these isoforms are both components of complexes that incorporate Fas-associated death domain and RIP1, which appear essential for kinase activation. This conservation of IKK activation among the cFLIP family using different mechanisms suggests that the mechanism plays a critical role in their function.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , I-kappa B Kinase/metabolism , NF-kappa B/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Enzyme Activation/physiology , HEK293 Cells , Humans , I-kappa B Kinase/genetics , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , NF-kappa B/genetics , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism
5.
Mol Ther ; 24(11): 1965-1973, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27562586

ABSTRACT

Lymph node stromal cells play a role in self-tolerance by presenting tissue antigens to T cells. Yet, immunomodulatory properties of lymphoid tissue stroma, particularly toward CD4+ T cells, remain insufficiently characterized by lack of tools to target antigens for presentation by stromal cells. A lentiviral vector was therefore designed for antigen delivery to MHC class II+ cells of nonhematopoietic origin. Following intravenous vector delivery, the transgene was detected in lymph node gp38+ stromal cells which were CD45- MHCII+ and partly positive for CD86 and CTLA4 or B7-H4. The transgene was not detected in classical dendritic cells of lymph nodes or spleen. Transgene-specific CD4+ and CD8+ T cell responses were primed and regulatory T cells were also induced but effector T cell response did not develop, even after a peptide boost. Antigen-specific CD8+ T cells were not cytolytic in vivo. Thus, expressing a neo-antigen in MHC-II+ lymph node stroma seems to trigger blunt CD4 T cell responses leading to antigen-specific CD8+ T cell anergy. These results open up new perspectives to further characterize lymph node stromal cell functional properties and to develop gene transfer protocols targeting lymph node stroma to induce peripheral tolerance.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Lymph Nodes/immunology , Stromal Cells/metabolism , Animals , Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , Clonal Anergy , Female , Genes, MHC Class II , Lentivirus/genetics , Male , Mice , Organ Specificity
6.
J Biol Chem ; 290(27): 16539-49, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-25979343

ABSTRACT

Viral flice-interacting protein (vFLIP), encoded by the oncogenic Kaposi sarcoma-associated herpes virus (KSHV), constitutively activates the canonical nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) pathway. This is achieved through subversion of the IκB kinase (IKK) complex (or signalosome), which involves a physical interaction between vFLIP and the modulatory subunit IKKγ. Although this interaction has been examined both in vivo and in vitro, the mechanism by which vFLIP activates the kinase remains to be determined. Because IKKγ functions as a scaffold, recruiting both vFLIP and the IKKα/ß subunits, it has been proposed that binding of vFLIP could trigger a structural rearrangement in IKKγ conducive to activation. To investigate this hypothesis we engineered a series of mutants along the length of the IKKγ molecule that could be individually modified with nitroxide spin labels. Subsequent distance measurements using electron paramagnetic resonance spectroscopy combined with molecular modeling and molecular dynamics simulations revealed that IKKγ is a parallel coiled-coil whose response to binding of vFLIP or IKKß is localized twisting/stiffening and not large-scale rearrangements. The coiled-coil comprises N- and C-terminal regions with distinct registers accommodated by a twist: this structural motif is exploited by vFLIP, allowing it to bind and subsequently activate the NF-κB pathway. In vivo assays confirm that NF-κB activation by vFLIP only requires the N-terminal region up to the transition between the registers, which is located directly C-terminal of the vFLIP binding site.


Subject(s)
Herpesvirus 8, Human/metabolism , I-kappa B Kinase/chemistry , I-kappa B Kinase/metabolism , Sarcoma, Kaposi/enzymology , Viral Proteins/metabolism , Amino Acid Motifs , Binding Sites , Electron Spin Resonance Spectroscopy , Herpesvirus 8, Human/chemistry , Herpesvirus 8, Human/genetics , Humans , I-kappa B Kinase/genetics , Protein Binding , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/virology , Viral Proteins/chemistry , Viral Proteins/genetics
7.
J Immunol ; 193(10): 5056-64, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25305314

ABSTRACT

Expression of the costimulatory receptor 4-1BB is induced by TCR recognition of Ag, whereas 4-1BB ligand (4-1BBL) is highly expressed on activated APC. 4-1BB signaling is particularly important for survival of activated and memory CD8(+) T cells. We wished to test whether coexpression of Ag and 4-1BBL by dendritic cells (DC) would be an effective vaccine strategy. Therefore, we constructed lentiviral vectors (LV) coexpressing 4-1BBL and influenza nucleoprotein (NP). Following s.c. immunization of mice, which targets DC, we found superior CD8(+) T cell responses against NP and protection from influenza when 4-1BBL was expressed. However, functionally superior CD8(+) T cell responses were obtained when two LV were coinjected: one expressing 4-1BBL and the other expressing NP. This surprising result suggested that 4-1BBL is more effective when expressed in trans, acting on adjacent DC. Therefore, we investigated the effect of LV expression of 4-1BBL in mouse DC cultures and observed induced maturation of bystander, untransduced cells. Maturation was blocked by anti-4-1BBL Ab, required cell-cell contact, and did not require the cytoplasmic signaling domain of 4-1BBL. Greater maturation of untransduced cells could be explained by LV expression of 4-1BBL, causing downregulation of 4-1BB. These data suggest that coexpression of 4-1BBL and Ag by vaccine vectors that target DC may not be an optimal strategy. However, 4-1BBL LV immunization activates significant numbers of bystander DC in the draining lymph nodes. Therefore, transactivation by 4-1BBL/4-1BB interaction following DC-DC contact may play a role in the immune response to infection or vaccination.


Subject(s)
4-1BB Ligand/immunology , Antigens, Viral/immunology , Dendritic Cells/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae/immunology , Viral Core Proteins/immunology , 4-1BB Ligand/genetics , Animals , Antigens, Viral/genetics , Bystander Effect , CD8-Positive T-Lymphocytes/immunology , Cell Communication , Female , Genetic Vectors , Immunization , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Lentivirus/genetics , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Signal Transduction , Transcriptional Activation , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Viral Core Proteins/genetics
8.
Immunology ; 146(2): 264-70, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26095282

ABSTRACT

Although bacillus Calmette-Guérin (BCG) is an established vaccine with excellent efficacy against disseminated Mycobacterium tuberculosis infection in young children, efficacy in adults suffering from respiratory tuberculosis (TB) is suboptimal. Prime-boost viral vectored vaccines have been shown to induce effective immune responses and lentivectors (LV) have been shown to improve mucosal immunity in the lung. A mucosal boost to induce local immunogenicity is also referred to as a 'pull' in a prime and pull approach, which has been found to be a promising vaccine strategy. The majority of infants worldwide receive BCG immunization through current vaccine protocols. We therefore aimed to investigate the role of a boost (or pull) immunization with an LV vaccine expressing the promising TB antigen (Ag85A). We immunized BALB/c mice subcutaneously with BCG or an LV vaccine expressing a nuclear factor-κB activator vFLIP together with Ag85A (LV vF/85A), then boosted with intranasal LV vF/85A. Prime and pull immunization with LV85A induced significantly enhanced CD8(+) and CD4(+) T-cell responses in the lung, but did not protect against intranasal BCG challenge. In contrast, little T-cell response in the lung was seen when the prime vaccine was BCG, and intranasal vF/85A provided no additional protection against mucosal BCG infection. Our study demonstrates that not all LV prime and pull approaches may be successful against TB in man and careful antigen and immune activator selection is therefore required.


Subject(s)
Acyltransferases/immunology , Antigens, Bacterial/immunology , BCG Vaccine/immunology , Genetic Vectors , Immunization, Secondary , Lentivirus/genetics , Lung/immunology , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/prevention & control , Vaccines, DNA/immunology , Acyltransferases/administration & dosage , Acyltransferases/genetics , Administration, Intranasal , Animals , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/genetics , BCG Vaccine/administration & dosage , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/microbiology , Cells, Cultured , Female , Immunity, Mucosal , Lung/microbiology , Mice, Inbred BALB C , Mice, Transgenic , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/genetics , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
9.
Int J Cancer ; 134(3): 575-86, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-23901045

ABSTRACT

There is growing interest in myeloid (my) dendritic cells (DC) as an alternative to monocyte-derived DC (moDC) for immunotherapy. However, in contrast to moDC, little is known regarding the effect of malignancy on the function, abundance or use of intracellular signaling pathways in myDC. Understanding the molecular detail of circulating myDC is therefore important for future use in advanced cancer. Advanced cancer patients had similar numbers of circulating myDC to cancer-free patients and healthy individuals, and secreted similar levels of IL-1ß, IL-6, IL-10, IL-12 and IL-23. However, myDC from some patients failed to secrete the Th1-cytokine IL-12. Surprisingly, inhibiting p38 (p38i) signaling (using BIRB0796 or SB203580) markedly increased IL-12 secretion by myDC. This is in complete contrast to what is established for moDC where inhibiting p38 ablates IL-12. Interestingly, this was specific to IL-12, since IL-10 was suppressed by p38i in both DC types. The opposing effect of p38i on IL-12 was evident at the transcriptional level and in both DC types was mediated through the p38-MK2 pathway but did not involve differential phosphorylation of the distal Rsk kinase. Importantly, where patient myDC did not secrete IL-12 (or after treatment with suppressive melanoma lysate), p38i restored IL-12 to normal levels. In contrast to p38, inhibiting the other MAPK pathways had similar consequences in both DC types. We show for the first time the differential use of a major intracellular signaling pathway by myDC. Importantly, there are sufficient circulating myDC in advanced cancer patients to consider development of adoptive immunotherapy.


Subject(s)
Antigens, CD1/immunology , Dendritic Cells/immunology , Glycoproteins/immunology , Interleukin-10/metabolism , Interleukin-12/metabolism , Neoplasms/immunology , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism , Adult , Aged , Blotting, Western , Case-Control Studies , Female , Humans , Male , Middle Aged , Polymerase Chain Reaction , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
10.
Commun Biol ; 6(1): 368, 2023 04 20.
Article in English | MEDLINE | ID: mdl-37081096

ABSTRACT

COVID-19 mRNA vaccines induce protective adaptive immunity against SARS-CoV-2 in most individuals, but there is wide variation in levels of vaccine-induced antibody and T-cell responses. However, the mechanisms underlying this inter-individual variation remain unclear. Here, using a systems biology approach based on multi-omics analyses of human blood and stool samples, we identified several factors that are associated with COVID-19 vaccine-induced adaptive immune responses. BNT162b2-induced T cell response is positively associated with late monocyte responses and inversely associated with baseline mRNA expression of activation protein 1 (AP-1) transcription factors. Interestingly, the gut microbial fucose/rhamnose degradation pathway is positively correlated with mRNA expression of AP-1, as well as a gene encoding an enzyme producing prostaglandin E2 (PGE2), which promotes AP-1 expression, and inversely correlated with BNT162b2-induced T-cell responses. These results suggest that baseline AP-1 expression, which is affected by commensal microbial activity, is a negative correlate of BNT162b2-induced T-cell responses.


Subject(s)
COVID-19 , Gastrointestinal Microbiome , Humans , COVID-19 Vaccines , BNT162 Vaccine , Transcription Factor AP-1 , COVID-19/prevention & control , SARS-CoV-2/genetics , Antibodies, Viral , RNA, Messenger/genetics
11.
J Gene Med ; 14(3): 151-7, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22262303

ABSTRACT

BACKGROUND: Lung cancer remains a leading cause of cancer mortality, and so the aim of the present study was to develop a therapeutic vaccine protocol. METHODS: We constructed a lentiviral vector (LV) expressing the extracellular domain (ECD) of murine Her1, an antigen associated with poor prognosis in lung cancer. RESULTS: A single LV injection, followed by two Her1 protein boosts, was effective in reducing the metastatic burden of Lewis lung carcinoma in mice. The Her1 LV immunisation generated CD8+ T cells that recognised Her1 ECD presented by dendritic cells, and that also homed to Her1-expressing tumours. Protein boosting further increased the CD8+ T cell response and generated anti-Her1 antibodies; in the antibody response, Her1 LV priming increased Th1-dependent immunoglobulin G2c production. CONCLUSIONS: The ability of this vaccine protocol to break both T cell and B cell tolerance to a self-antigen likely explains its effectiveness.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines , Carcinoma, Lewis Lung/immunology , ErbB Receptors/metabolism , Immunotherapy/methods , Neoplasm Metastasis/prevention & control , Animals , Antibodies/immunology , ErbB Receptors/genetics , Genetic Vectors/genetics , Immune Tolerance/immunology , Lentivirus , Mice
12.
Clin Immunol Commun ; 2: 6-11, 2022 Dec.
Article in English | MEDLINE | ID: mdl-38621014

ABSTRACT

Pre-existing SARS-CoV-2-specific T cells, but not antibodies, have been detected in some unexposed individuals. This may account for some of the diversity in clinical outcomes ranging from asymptomatic infection to severe COVID-19. Although age is a risk factor for COVID-19, how age affects SARS-CoV-2-specific T cell responses remains unknown. We found that pre-existing T cell responses to specific SARS-CoV-2 proteins, Spike (S) and Nucleoprotein (N), were significantly lower in elderly donors (>70 years old) than in young donors. However, substantial pre-existing T cell responses to the viral membrane (M) protein were detected in both young and elderly donors. In contrast, young and elderly donors exhibited comparable T cell responses to S, N, and M proteins after infection with SARS-CoV-2. These data suggest that although SARS-CoV-2 infection can induce T cell responses specific to various viral antigens regardless of age, diversity of target antigen repertoire for long-lived memory T cells specific for SARS-CoV-2 may decline with age; however, memory T cell responses can be maintained by T cells reactive to specific viral proteins such as M. A better understanding of the role of pre-existing SARS-CoV-2-specific T cells that are less susceptible to age-related loss may contribute to development of more effective vaccines for elderly people.

14.
J Gene Med ; 13(3): 181-7, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21394858

ABSTRACT

BACKGROUND: Lentiviral vectors (LV) are promising vaccines because they transduce dendritic cells (DC) in vivo. To translate LV vaccines into clinical trials, bulk production will be necessary. The present study aimed to find a suitable envelope for LV vaccine production from stable packaging cells because the commonly used vesicular stomatitis virus envelope (VSV-G) is cytotoxic. METHODS: The envelope from Ross river virus (RRV) was selected. It can infect mouse and human cells, allowing testing in animals before clinical translation. We used VSV-G for comparison. Vectors produced with each envelope were titred on human 293T cells and mouse 3T3 cells. RESULTS: RRV-pseudotyped LV (RRV-LV) infected mouse myeloid DC in culture and immunized mice. An approximately 50-fold higher dose of RRV-LV than VSV-G-LV was required to generate a similar T cell response. The RRV-LV could also be used to infect human mDC and to prime a human T cell immune response. CONCLUSIONS: RRV envelope is a suitable candidate to be used for the construction of an LV producer cell line. LV vaccines with RRV envelope can be tested in mice and in human immune cell cultures. The higher dose of RRV-LV required for vaccine efficacy compared to VSV-G-LV will partly be offset by ease of production.


Subject(s)
Genetic Vectors/metabolism , Lentivirus/genetics , Ross River virus/metabolism , Vaccines, Synthetic/biosynthesis , Viral Envelope Proteins/metabolism , 3T3 Cells , Animals , Cell Line , Dendritic Cells/metabolism , Enzyme-Linked Immunospot Assay , Gene Transfer Techniques , Humans , Mice , Mice, Inbred C57BL , Ross River virus/genetics , T-Lymphocytes/metabolism , Transduction, Genetic , Vaccines, Synthetic/immunology , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/metabolism , Viral Envelope Proteins/genetics , Viral Envelope Proteins/toxicity
15.
Cancer Immunol Immunother ; 60(8): 1127-35, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21479639

ABSTRACT

Although cancer immunology has made vigorous progress over the last decade, its future remains uncertain. Tumors have clearly proved subject to immune surveillance, leading to antigenic editing, and means of activating both T and B arms of the immune system have been devised. Therapeutic vaccination and monoclonal antibody therapy have so far proved disappointing, because tumors prove adept at evasion from immune control. Dual targeting could well counteract evasion, provided that the two targets are independent and are attacked simultaneously. This stage has nearly but not quite been reached in several forms of immunotherapy, particularly of B-cell cancers, although such treatment also carries hazards.


Subject(s)
B-Lymphocytes/immunology , Cancer Vaccines , Immunotherapy , T-Lymphocytes/immunology , Tumor Escape , Animals , Antigens, Neoplasm/immunology , Humans , Immunodominant Epitopes/immunology , Immunologic Surveillance , Molecular Targeted Therapy , Neoplasms
16.
J Virol ; 83(4): 1555-62, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19036811

ABSTRACT

Lentiviral vectors deliver antigens to dendritic cells (DCs) in vivo, but they do not trigger DC maturation. We therefore expressed a viral protein that constitutively activates NF-kappaB, vFLIP from Kaposi's sarcoma-associated herpesvirus (KSHV), in a lentivector to mature DCs. vFLIP activated NF-kappaB in mouse bone marrow-derived DCs in vitro and matured these DCs to a similar extent as lipopolysaccharide; costimulatory markers CD80, CD86, CD40, and ICAM-1 were upregulated and tumor necrosis factor alpha and interleukin-12 secreted. The vFLIP-expressing lentivector also matured DCs in vivo. When we coexpressed vFLIP in a lentivector with ovalbumin (Ova), we found an increased immune response to Ova; up to 10 times more Ova-specific CD8(+) T cells secreting gamma interferon were detected in the spleens of vFLIP_Ova-immunized mice than in the spleens of mice immunized with GFP_Ova. Furthermore, this increased CD8(+) T-cell response correlated with improved tumor-free survival in a tumor therapy model. A single immunization with vFLIP_Ova also reduced the parasite load when mice were challenged with OVA-Leishmania donovani. In conclusion, vFLIP from KSHV is a DC activator, maturing DCs in vitro and in vivo. This demonstrates that NF-kappaB activation is sufficient to induce many aspects of DC maturation and that expression of a constitutive NF-kappaB activator can improve the efficacy of a vaccine vector.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/virology , Lentivirus/genetics , NF-kappa B/biosynthesis , Viral Proteins/immunology , Viral Vaccines/genetics , Animals , Cancer Vaccines/immunology , Cytokines/biosynthesis , Leishmania donovani/immunology , Leishmaniasis/prevention & control , Lentivirus/immunology , Mice , Neoplasms/immunology , Ovalbumin/immunology , Receptors, Immunologic/biosynthesis , Spleen/immunology , Survival Analysis , Viral Proteins/genetics , Viral Vaccines/immunology
17.
J Virol ; 83(1): 283-94, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18945765

ABSTRACT

Gammaretroviral and lentiviral vectors are promising tools for gene therapy, but they can be oncogenic. The development of safer vectors depends on a quantitative assay for insertional mutagenesis. Here we report a rapid, inexpensive, and reproducible assay which uses a murine cell line to measure the frequency of interleukin-3 (IL-3)-independent mutants. Lentiviral and gammaretroviral vectors cause insertional mutagenesis at similar frequencies; however, they use different mechanisms. Human immunodeficiency virus (HIV)-based vectors generate mutants by insertion only into the growth hormone receptor (Ghr) locus. The HIV enhancer/promoter is active in the absence of the HIV Tat protein in this locus, and an HIV/Ghr spliced transcript expresses GHR and cells respond to GH. Deletion of the enhancer/promoter in a self-inactivating HIV-based vector prevents this mechanism of insertional mutagenesis. In contrast, gammaretroviral vectors insert into other loci, including IL-3 and genes identified as common insertion sites in the Retroviral Tagged Cancer Gene Database (RTCGD).


Subject(s)
Gammaretrovirus/genetics , Genetic Therapy/adverse effects , Genetic Vectors/adverse effects , Lentivirus/genetics , Mutagenesis, Insertional , Animals , Cell Line , Mice
18.
J Virol ; 83(7): 3094-103, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19176629

ABSTRACT

Lentiviral vectors (lentivectors) are effective for stimulation of cell-mediated and humoral immunity following subcutaneous and intramuscular immunization. However, lentivector genome integration carries a risk of perturbation of host gene expression. Here, we demonstrate that lentivectors with multiple mutations that prevent integration are also effective immunogens. First, systemic CD8(+) T-cell responses to the model antigen ovalbumin were detected following subcutaneous injection of nonintegrating lentivectors. Transfer of transgenic OT1 T cells demonstrated that antigen presentation persisted for at least 30 days. Furthermore, an enhanced CD8(+) T-cell response, peaking at 7 days, was stimulated by coexpression of p38 MAP kinase or an NF-kappaB activator from the same vector. Second, we demonstrated systemic CD8(+) T-cell and antibody responses to the secreted hepatitis B virus (HBV) surface antigen expressed from a nonintegrating lentivector injected intramuscularly. The induction, specificity, and kinetics of antibody production closely mimicked those of natural HBV infection. In this case, both the vector genome and the immune response were maintained for at least 2 months. Together, our data indicate that nonintegrating lentivectors can be employed to generate effective vaccines.


Subject(s)
Antibodies, Neoplasm/blood , Antibodies, Viral/blood , Cancer Vaccines/immunology , Genetic Vectors , Hepatitis B Vaccines/immunology , Lentivirus/genetics , T-Lymphocytes/immunology , Animals , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasms/pathology , Neoplasms/prevention & control , Virus Integration
19.
Mol Ther ; 17(9): 1643-50, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19584812

ABSTRACT

Lentiviral vectors (LVs) are tools for in vivo gene delivery, to correct genetic defects or to deliver antigens for vaccination. It was reported that systemic injection of LVs in mice transduced cells in liver and spleen. Here we describe the reasons for, and consequences of, persistent gene expression in spleen. After 5 days of intravenous injection, a green fluorescence protein (GFP)-expressing LV was detected in lymphocytes, macrophages and all subsets of dendritic cells (DCs) in spleen. In the case of macrophages and DCs, the percentage of transduced cells increased between 5 and 30 days after injection. We used bromodeoxyuridine (BrdU) incorporation to show that the macrophages were largely nondividing, whereas the transduced DCs arose from dividing precursor cells and could be detected in spleen 2 months after injection. Expression of ovalbumin (OVA) in the LV reduced the number of transduced DCs in spleen after 30 days. However, the remaining transduced cells stimulated proliferation and activation of OVA-specific CD8(+) T cells transferred 2 months after LV injection. The mice also maintained cytolytic activity against OVA-pulsed targets. These results show that LVs transduce DC precursors, which maintain transduced DCs in spleen for at least 2 months, leading to prolonged antigen presentation and effective T-cell memory.


Subject(s)
Antigen Presentation/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Genetic Vectors/genetics , Immunization/methods , Lentivirus/genetics , Transduction, Genetic/methods , Animals , Antigen Presentation/genetics , Cell Differentiation , Dendritic Cells/cytology , Lymphocytes/cytology , Lymphocytes/immunology , Lymphocytes/metabolism , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Polymerase Chain Reaction , Spleen/cytology , Spleen/metabolism
20.
J Virol ; 82(19): 9789-94, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18667519

ABSTRACT

The env gene of gammaretroviruses encodes a glycoprotein conserved among diverse retroviruses, except for the domains involved in receptor binding. Here we show that pairs of gammaretrovirus envelope proteins (from Friend virus and GALV or xenotropic viruses) assemble into heteromers when coexpressed. This assembly results in a strong inhibition of infectivity. An unrelated envelope protein does not assemble in heteromers with the gammaretrovirus glycoproteins tested and does not affect their infectivity, demonstrating the specificity of the mechanism we describe. We propose that the numerous copies of endogenous retroviral env genes conserved within mammalian genomes act as restriction factors against infectious retroviruses.


Subject(s)
Gammaretrovirus/metabolism , Gene Products, env/genetics , Viral Envelope Proteins/chemistry , Amino Acid Sequence , Cell Line , Genes, env , Glycoproteins/chemistry , Humans , Models, Genetic , Molecular Sequence Data , Receptors, Virus/chemistry , Retroviridae/genetics , Sequence Homology, Amino Acid , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL