Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Mol Ther ; 23(3): 578-90, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25492563

ABSTRACT

Current therapies for the hepatitis B virus (HBV), a major cause of severe liver disease, suppress viral replication but replication rebounds if therapy is withdrawn. It is widely accepted that immune activation is needed to control replication off-therapy. To specifically activate T cells crossreactive between the hepatitis B core and e antigens (HBcAg/HBeAg) in chronically infected patients, we developed a therapeutic vaccine candidate. The vaccine encompass codon-optimized HBcAg and IL-12 expressing plasmids delivered using targeted high-pressure injection combined with in vivo electroporation. One dose of the vaccine primed a B-cell-independent polyfunctional T-cell response, in wild-type, and in HBeAg-transgenic mice with an impaired ability to respond to HBc/eAg. The response peaked at 2 weeks and contracted at week 6 after vaccination. Coadministration of IL-12 improved antibody levels, and T-cell expansion and functionality. The vaccine primed T cells that, 2 weeks after a single dose, cleared hepatocytes transiently expressing HBcAg in vaccinated wild-type and HBeAg-transgenic mice. However, 4 weeks later, these functional responses were lost. Booster doses after 8-12 weeks effectively restored function and expansion of the rapidly contracting T cells. Thus, this vaccine strategy primes functional HBcAg-specific T cells in a host with dysfunctional response to HBV.


Subject(s)
Hepatitis B Antibodies/biosynthesis , Hepatitis B Vaccines/immunology , Hepatitis B virus/immunology , Hepatitis B, Chronic/prevention & control , T-Lymphocytes/immunology , Vaccines, DNA/immunology , Animals , Cell Proliferation , Electroporation , Gene Expression , Hepatitis B Core Antigens/genetics , Hepatitis B Core Antigens/immunology , Hepatitis B Vaccines/administration & dosage , Hepatitis B Vaccines/genetics , Hepatitis B e Antigens/genetics , Hepatitis B e Antigens/immunology , Hepatitis B virus/genetics , Hepatitis B, Chronic/genetics , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/virology , Hepatocytes/immunology , Hepatocytes/virology , Humans , Interleukin-12/genetics , Interleukin-12/immunology , Liver/immunology , Liver/virology , Mice , Mice, Transgenic , Plasmids/chemistry , Plasmids/metabolism , T-Lymphocytes/virology , Vaccination , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
2.
Int J Mol Sci ; 15(3): 4747-79, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24646914

ABSTRACT

The hepatitis C virus (HCV) is a global health problem affecting more than 170 million people. A chronic HCV infection is associated with liver fibrosis, liver cirrhosis and hepatocellular carcinoma. To enable viral persistence, HCV has developed mechanisms to modulate both innate and adaptive immunity. The recruitment of antiviral immune cells in the liver is mainly dependent on the release of specific chemokines. Thus, the modulation of their expression could represent an efficient viral escape mechanism to hamper specific immune cell migration to the liver during the acute phase of the infection. HCV-mediated changes in hepatic immune cell chemotaxis during the chronic phase of the infection are significantly affecting antiviral immunity and tissue damage and thus influence survival of both the host and the virus. This review summarizes our current understanding of the HCV-mediated modulation of chemokine expression and of its impact on the development of liver disease. A profound knowledge of the strategies used by HCV to interfere with the host's immune response and the pro-fibrotic and pro-carcinogenic activities of HCV is essential to be able to design effective immunotherapies against HCV and HCV-mediated liver diseases.


Subject(s)
Chemokines/immunology , Hepacivirus/immunology , Hepatitis C/immunology , Liver Diseases/immunology , Adaptive Immunity/immunology , Chemokines/physiology , Hepacivirus/physiology , Hepatitis C/physiopathology , Hepatitis C/virology , Host-Pathogen Interactions/immunology , Humans , Immune System/cytology , Immune System/immunology , Immunity, Innate/immunology , Liver Diseases/physiopathology , Liver Diseases/virology , Models, Immunological
3.
J Immunol ; 192(4): 1671-80, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24442435

ABSTRACT

Ribavirin has proven to be a key component of hepatitis C therapies both involving IFNs and new direct-acting antivirals. The hepatitis C virus-mediated interference with intrahepatic immunity by cleavage of mitochondrial antiviral signaling protein (MAVS) and T cell protein tyrosine phosphatase (TCPTP) suggests an avenue for compounds that may counteract these effects. We therefore studied the effects of ribavirin, with or without inhibition of the nonstructural (NS)3/4A protease, on intrahepatic immunity. The intrahepatic immunity of wild-type and NS3/4A-transgenic mice was determined by Western blot, ELISA, flow cytometry, and survival analysis. Various MAVS or TCPTP constructs were injected hydrodynamically to study their relevance. Ribavirin pretreatment was performed in mice expressing a functional or inhibited NS3/4A protease to analyze its effect on NS3/4A-mediated changes. Intrahepatic NS3/4A expression made mice resistant to TNF-α-induced liver damage and caused an alteration of the intrahepatic cytokine (IFN-γ and IL-10) and chemokine (CCL3, CCL17, CCL22, CXCL9, and CXCL11) profiles toward an anti-inflammatory state. Consistent with this, the number of intrahepatic Th1 cells and IFN-γ(+) T cells in NS3/4A-transgenic mice decreased, whereas the amount of Th2 cells increased. These effects could be reversed by injection of uncleavable TCPTP but not uncleavable MAVS and were absent in a mouse expressing a nonfunctional NS3/4A protease. Importantly, the NS3/4A-mediated effects were reversed by ribavirin treatment. Thus, cleavage of TCPTP by NS3/4A induces a shift of the intrahepatic immune response toward a nonantiviral Th2-dominated immunity. These effects are reversed by ribavirin, supporting that ribavirin complements the effects of direct-acting antivirals as an immunomodulatory compound.


Subject(s)
Hepacivirus/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Ribavirin/pharmacology , Viral Nonstructural Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antiviral Agents/pharmacology , Cell Differentiation/drug effects , Chemokine CCL17/biosynthesis , Chemokine CCL22/biosynthesis , Chemokine CCL3/biosynthesis , Chemokine CXCL11/biosynthesis , Chemokine CXCL9/biosynthesis , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/metabolism , Hepatitis C, Chronic/virology , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Liver/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Th1 Cells , Th2 Cells , Tumor Necrosis Factor-alpha/metabolism , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/genetics
4.
Drug Resist Updat ; 16(3-5): 60-7, 2013.
Article in English | MEDLINE | ID: mdl-23911647

ABSTRACT

Presently the development of new therapies for hepatitis C virus (HCV) is rapidly moving forward. Almost every week new data appear on how direct acting antivirals (DAAs) succeed or fail in clinical trials. Despite the potency of many of the DAA combinations, the effect exerted by ribavirin (RBV) is still needed for an effective therapy in many new DAA combinations. Due to the strong antiviral effect of DAAs, it is likely that a major complementary therapeutic effect exerted by RBV is immune modulation resulting in an increased barrier to development of resistance. For HCV genotype 1a infections elimination of pegylated interferon, is not possible in many DAA combinations without jeopardizing the results. The host immune response is thus likely to play a key role even during DAA-based therapies. Hence, T cells may recognize and eliminate viral variants with resistance to the DAAs. We herein show several examples where this may be the case, supporting the rationale of including the host response also in the new therapeutic regimens. This review will describe the potential benefits of combining various DAAs with means to activate the specific immune response against HCV.


Subject(s)
Antiviral Agents/therapeutic use , Hepacivirus/drug effects , Hepatitis C, Chronic/therapy , Immunomodulation/drug effects , Models, Biological , Animals , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , Combined Modality Therapy , Drug Resistance, Viral , Hepacivirus/immunology , Hepacivirus/metabolism , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/metabolism , Hepatitis C, Chronic/virology , Humans , Ligands , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Ribavirin/therapeutic use , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism , Viral Hepatitis Vaccines/therapeutic use
5.
J Infect Dis ; 208(6): 1008-19, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23776192

ABSTRACT

BACKGROUND: We explored the concept of heterologous prime/boost vaccination using 2 therapeutic vaccines currently in clinical development aimed at treating chronically infected hepatitis C virus (HCV) patients: prime with a DNA-based vaccine expressing HCV genotype 1a NS3/4A proteins (ChronVac-C) and boost with a modified vaccinia virus Ankara vaccine expressing genotype 1b NS3/4/5B proteins (MVATG16643). METHODS: Two ChronVac-C immunizations 4 weeks apart were delivered intramuscularly in combination with in vivo electroporation and subsequently 5 or 12 weeks later boosted by 3 weekly subcutaneous injections of MVATG16643. Two mouse strains were used, and we evaluated quality, magnitude, and functionality of the T cells induced. RESULTS: DNA prime/MVA boost regimen induced significantly higher levels of interferon γ (IFN-γ) or interleukin 2 (IL-2) ELISpot responses compared with each vaccine alone, independent of the time of analysis and the time interval between vaccinations. Both CD8⁺ and CD4⁺ T-cell responses as well as the spectrum of epitopes recognized was improved. A significant increase in polyfunctional IFN-γ/tumor necrosis factor α (TNF-α)/CD107a⁺ CD8⁺ T cells was detected following ChronVac-C/MVATG16643 vaccination (from 3% to 25%), and prime/boost was the only regimen that activated quadrifunctional T cells (IFN-γ/TNF-α/CD107a/IL-2). In vivo functional protective capacity of DNA prime/MVA boost was demonstrated in a Listeria-NS3-1a challenge model. CONCLUSIONS: We provide a proof-of-concept that immunogenicity of 2 HCV therapeutic vaccines can be improved using their combination, which merits further clinical development.


Subject(s)
Antibody Formation , Hepatitis C/prevention & control , Vaccination/methods , Viral Hepatitis Vaccines/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Genotype , Hepacivirus , Hepatitis C/immunology , Immunization, Secondary , Interferon-gamma/blood , Interleukin-2/blood , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tumor Necrosis Factor-alpha/blood , Vaccines, DNA/immunology , Viral Hepatitis Vaccines/genetics
6.
J Immunol ; 190(3): 1113-24, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23284053

ABSTRACT

The hepatitis C virus (HCV) nonstructural (NS) 5A protein has been shown to promote viral persistence by interfering with both innate and adaptive immunity. At the same time, the HCV NS5A protein has been suggested as a target for antiviral therapy. In this study, we performed a detailed characterization of HCV NS5A immunogenicity in wild-type (wt) and immune tolerant HCV NS5A-transgenic (Tg) C57BL/6J mice. We evaluated how efficiently HCV NS5A-based genetic vaccines could activate strong T cell responses. Truncated and full-length wt and synthetic codon-optimized NS5A genotype 1b genes were cloned into eukaryotic expression plasmids, and the immunogenicity was determined after i.m. immunization in combination with in vivo electroporation. The NS5A-based genetic vaccines primed high Ab levels, with IgG titers of >10(4) postimmunization. With respect to CD8(+) T cell responses, the coNS5A gene primed more potent IFN-γ-producing and lytic cytotoxic T cells in wt mice compared with NS5A-Tg mice. In addition, high frequencies of NS5A-specific CD8(+) T cells were found in wt mice after a single immunization. To test the functionality of the CTL responses, the ability to inhibit growth of NS5A-expressing tumor cells in vivo was analyzed after immunization. A single dose of coNS5A primed tumor-inhibiting responses in both wt and NS5A-Tg mice. Finally, immunization with the coNS5A gene primed polyfunctional NS5A-specific CD8(+) T cell responses. Thus, the coNS5A gene is a promising therapeutic vaccine candidate for chronic HCV infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , DNA, Viral/immunology , Hepacivirus/immunology , Vaccines, DNA/immunology , Viral Hepatitis Vaccines/immunology , Viral Nonstructural Proteins/immunology , Animals , Antibody Specificity , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines , Codon/genetics , Cytotoxicity, Immunologic , DNA, Viral/chemical synthesis , DNA, Viral/genetics , Genes, Synthetic , H-2 Antigens/immunology , Hepacivirus/genetics , Hepatitis C Antibodies/biosynthesis , Hepatitis C Antibodies/genetics , Hepatitis C Antibodies/immunology , Immunization , Immunoglobulin G/biosynthesis , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Lymphocyte Activation , Lymphokines/metabolism , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/genetics , Peptide Fragments/immunology , T-Cell Antigen Receptor Specificity , T-Lymphocytes, Cytotoxic/immunology , Viral Hepatitis Vaccines/therapeutic use , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics
7.
J Immunol ; 189(9): 4510-9, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23024278

ABSTRACT

Virus-specific CTL with high levels of functional avidity have been associated with viral clearance in hepatitis C virus (HCV) infection and with enhanced protective immunity. In chronic HCV infection, lack of antiviral CTL is frequently observed. In this study, we aim to investigate novel HCV TCRs that differ in Ag specificity. This involved isolating new HCV-specific murine TCRs that recognize a conserved HLA-A2-restricted CTL epitope within the nonstructural protein (NS) 5A viral protein and comparing them with TCRs recognizing another conserved CTL target in the NS3 viral protein. This was done by expressing the TCRs in human T cells and analyzing the function of the resulting TCR-transduced T cells. Our result indicates that these TCRs are efficiently assembled in transduced human T cells. They recognize peptide-loaded targets and demonstrate polyfunctional features such as IL-2, IFN-γ, and TNF-α secretion. However, in contrast to NS3-specific TCRs, the NS5A TCR-transduced T cells consist of a smaller proportion of polyfunctional T cells and require more peptide ligands to trigger the effector functions, including degranulation. Despite the differences, NS5A TCRs show effective inhibition of HCV replication in human hepatoma cells with persistent HCV RNA replication. Moreover, cellular injury demonstrated by aspartate aminotransferase release and cell death is less significant in the hepatoma cells following coincubation with NS5A TCR-transduced T cells, which is a property consistent with noncytotoxic antiviral CTLs. Our results suggest that HCV TCR-transduced T cells may be promising for the treatment of patients with chronic HCV infections.


Subject(s)
Antiviral Agents/pharmacology , Cytotoxicity, Immunologic , Epitopes, T-Lymphocyte/physiology , Hepacivirus/immunology , Hepacivirus/pathogenicity , Receptors, Antigen, T-Cell/physiology , Virus Replication/immunology , Amino Acid Sequence , Animals , Antiviral Agents/toxicity , Cell Line , Cell Line, Tumor , Cytotoxicity, Immunologic/genetics , Epitopes, T-Lymphocyte/toxicity , Female , Gene Transfer Techniques , Humans , Leukocytes, Mononuclear/immunology , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Receptors, Antigen, T-Cell/genetics , Transduction, Genetic/methods , Virus Replication/genetics
8.
Scand J Infect Dis ; 44(1): 55-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21933033

ABSTRACT

A major problem in chronic hepatitis B virus (HBV) infection is that treatment with specific antivirals is life-long since they rarely induce a sustained response. An attractive option is therefore to combine antiviral therapy with some type of immune stimulator, such as a therapeutic vaccine. Several lines of evidence suggest that a key target for the cellular immune response is the HBV core antigen (HBcAg). However, it may also be of advantage to simultaneously improve the neutralizing antibody response to the surface (S) region of HBV. We therefore generated chimeric HBcAg particles expressing preS1 residues 1-42 at the tip of the spike region. We could show that this chimeric HBcAg-preS1 protein primed both HBcAg-specific T cells and antibodies to preS1. This strongly suggests that this may be a viable approach to develop an effective bi-functional therapeutic vaccine as an add-on for the treatment of chronic HBV infections.


Subject(s)
Hepatitis B Core Antigens/therapeutic use , Hepatitis B Surface Antigens/therapeutic use , Hepatitis B virus/immunology , Hepatitis B, Chronic/drug therapy , T-Lymphocytes/immunology , Vaccination/methods , Animals , Antibodies, Neutralizing , Female , Hepatitis B Core Antigens/immunology , Hepatitis B Surface Antigens/immunology , Hepatitis B, Chronic/immunology , Immunity, Cellular/immunology , Mice , Mice, Inbred C57BL , Vaccines, Synthetic/immunology , Vaccines, Synthetic/therapeutic use
9.
J Gen Virol ; 93(Pt 2): 247-258, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22071510

ABSTRACT

Hepatitis C virus (HCV) is a major cause of severe liver disease, and one major contributing factor is thought to involve a dysfunction of virus-specific T-cells. T-cell receptor (TCR) gene therapy with HCV-specific TCRs would increase the number of effector T-cells to promote virus clearance. We therefore took advantage of HLA-A2 transgenic mice to generate multiple TCR candidates against HCV using DNA vaccination followed by generation of stable T-cell-BW (T-BW) tumour hybrid cells. Using this approach, large numbers of non-structural protein 3 (NS3)-specific functional T-BW hybrids can be generated efficiently. These predominantly target the genetically stable HCV genotype 1 NS3(1073-1081) CTL epitope, frequently associated with clearance of HCV in humans. These T-BW hybrid clones recognized the NS3(1073) peptide with a high avidity. The hybridoma effectively recognized virus variants and targeted cells with low HLA-A2 expression, which has not been reported previously. Importantly, high-avidity murine TCRs effectively redirected human non-HCV-specific T-lymphocytes to recognize human hepatoma cells with HCV RNA replication driven by a subgenomic HCV replicon. Taken together, TCR candidates with a range of functional avidities, which can be used to study immune recognition of HCV-positive targets, have been generated. This has implications for TCR-related immunotherapy against HCV.


Subject(s)
Hepacivirus/immunology , Immunodominant Epitopes/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , Viral Nonstructural Proteins/immunology , Animals , Blood Donors , Cells, Cultured , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Viral Nonstructural Proteins/genetics
10.
Gut ; 61(4): 589-96, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21813471

ABSTRACT

BACKGROUND: The non-structural (NS) 3/4A protease/helicase of the hepatitis C virus is known to modulate signalling pathways in the infected hepatocyte by cleaving CARD adaptor inducing IFNß (Cardif), T-cell protein tyrosine phosphatase (TC-PTP) and TIR domain-containing adaptor inducing IFNß (TRIF), but the effects of NS3/4A in vivo still remain unclear. AIM: To investigate the influence of NS3/4A on intracellular and intercellular signalling in vivo by analysing the intrahepatic inflammatory response of naïve, lipopolysaccharide (LPS)/D-galactosamine (D-galN) or tumour necrosis factor-α (TNFα)/D-galN-treated NS3/4A-transgenic (Tg) mice. METHODS: The intrahepatic immunity of naïve and LPS/D-galN- or TNFα/D-galN-treated NS3/4A-Tg mice was determined using western blot, ELISA, real-time PCR, flow cytometry and survival monitoring. The injection of cytokines or antibodies against signalling components was performed to analyse the relevance of the respective pathways for the investigated issues. A Tg mouse lineage expressing an inactivated NS3/4A protease (NS3/4A(Ile1073Ala)-Tgs) was generated to examine if protective effects were NS3/4A protease dependent. RESULTS: The activation of hepatic signal transducer and activator of transcription 1 and 2 was impaired in NS3/4A-Tg mice after treatment with LPS/D-galN or TNFα/D-galN. This was paralleled by a reduction in hepatic interferon-γ (IFNγ). Reconstitution of IFNγ reverted the resistance to LPS/TNFα in NS3/4A-Tg mice. Subsequently, blocking IFNγ in vivo rendered wild-type mice resistant against treatment with LPS/TNFα. A new Tg mouse expressing an inactivated NS3/4A protease had the same phenotype as wild-type mice with respect to hepatic IFNγ levels and sensitivity to LPS/d-galN. Finally, the chemokine profile was altered in the NS3/4A-Tg mice towards an anti-inflammatory state, which helps to explain the altered immune cell subsets and reduction in hepatic IFNγ production. CONCLUSIONS: Our data demonstrate that the NS3/4A protease reduces the intrahepatic production of IFNγ and alters TNFα-mediated effects, thereby impairing the hepatic inflammatory response. This may contribute to viral persistence.


Subject(s)
Carrier Proteins/physiology , Hepacivirus/metabolism , Interferon-gamma/biosynthesis , Liver/immunology , Viral Nonstructural Proteins/physiology , Animals , Case-Control Studies , Female , Hepatitis C/immunology , Humans , Interferon-alpha/metabolism , Interferon-gamma/pharmacology , Intracellular Signaling Peptides and Proteins , Lipopolysaccharides/pharmacology , Lymphocyte Subsets/immunology , Male , Mice , Mice, Transgenic , Middle Aged , STAT1 Transcription Factor/metabolism , Signal Transduction
11.
J Immunol ; 186(9): 5107-18, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21430225

ABSTRACT

The hepatitis C virus (HCV)-specific T cell response in patients with chronic HCV is dysfunctional. In this study, we aimed at restoring immunological function through therapeutic vaccination in a transgenic mouse model with impaired HCV-specific T cell responses due to a persistent presence of hepatic HCV nonstructural (NS)3/4A Ags. The HCV-specific T cells have an actively maintained dysfunction reflected in reduced frequency, impaired cytokine production, and impaired effector function in vivo, which can be partially restored by blocking regulatory T cells or programmed cell death ligand 1. We hypothesized that the impairment could be corrected by including sequences that created a normal priming environment by recruiting "healthy" heterologous T cells and by activating innate signaling. Endogenously expressed hepatitis B core Ag (HBcAg) can recruit heterologous T cells and activate TLR (TLR7) signaling. Hence, by combining HCV NS3/4A with different forms of HBcAg we found that heterologous sequences somewhat improved activation and expansion of NS3/4A-specific T cells in a wild-type host. Importantly, the signals provided by HBcAg effectively restored the activation of HCV-specific T cells in a tolerant NS3/4A-transgenic mouse model. The adjuvant effect could also be transferred to the priming of dysfunctional HLA-A2-restricted NS3-specific T cells in vivo. Thus, recruiting healthy heterologous T cells to the site of priming may also help restore HCV-specific responses present in a chronically infected host.


Subject(s)
Hepatitis C, Chronic/immunology , T-Lymphocytes/immunology , Vaccination/methods , Viral Hepatitis Vaccines/immunology , Viral Nonstructural Proteins/immunology , Animals , Animals, Genetically Modified , Antigens, Viral/immunology , Antigens, Viral/therapeutic use , Blotting, Western , Cell Line, Tumor , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Hepacivirus/immunology , Hepatitis B Core Antigens/immunology , Hepatitis B Core Antigens/therapeutic use , Hepatitis C, Chronic/therapy , Humans , Immunoprecipitation , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic
12.
Drug News Perspect ; 23(10): 647-53, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21180650

ABSTRACT

The without a doubt major obstacle for making DNA vaccines a commercial success is delivery. If delivery cannot be made simple, cheap and effective, DNA vaccines may not become a viable option for human use. Numerous clinical trials have confirmed that a standard needle and syringe simply do not do the job, i.e., delivering the DNA payload inside the cell. Having recognized this shortcoming, investigators have developed several new approaches for DNA vaccine delivery. In particular, new types of delivery devices, originally intended for in vitro use, have been applied for in vivo delivery. These include particle bombardment or biolistic delivery, and in vivo electroporation (EP). Importantly, both techniques seem to overcome the size barrier, meaning that they work in both mice and larger animals. In vivo EP has the key features of improved DNA uptake, increased antigen expression and a local inflammation. These factors are essential to make DNA vaccines effective in a larger host. Early data from clinical trials with DNA vaccines delivered by in vivo EP are cautiously promising. Thus, we may be entering a new era of DNA vaccination where we start to see clinical effects in humans; however, these may also be accompanied by side effects, as the vaccines become more effective.


Subject(s)
Drug Delivery Systems , Electroporation/methods , Vaccines, DNA/administration & dosage , Animals , Clinical Trials as Topic , Drug Design , Humans , Mice , Technology, Pharmaceutical/methods , Vaccines, DNA/adverse effects , Vaccines, DNA/immunology
13.
J Infect Dis ; 201(12): 1867-79, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20446851

ABSTRACT

Hepatitis B virus core antigen (HBcAg) is thought to be a major target for specific cytotoxic T cells (CTLs) in hepatitis B virus infections. A single dose of hepatitis C virus nonstructural 3/4A DNA (<5 microg) effectively primes functional specific CTLs, independently of CD4(+) T helper cells and by different routes of immunization. In contrast, HBcAg-specific CTL priming was T helper cell dependent and highly sensitive to the dose and route of delivery. Although CTL priming was improved 10-fold by codon optimization and in vivo electroporation, low levels of DNA still failed to prime CTLs effectively. Only high doses (5 microg) of codon-optimized HBcAg delivered by in vivo electroporation primed in vivo lytic and polyfunctional CTLs. The ability of endogenous HBcAg to prime CTLs is surprisingly inefficient and differs from that of nonstructural 3/4A. This has important implications for the design of HBcAg-based therapeutic vaccines in humans.


Subject(s)
DNA, Viral/immunology , Hepatitis B Core Antigens/immunology , Lymphocyte Activation , T-Lymphocytes, Cytotoxic/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , DNA, Viral/genetics , Dose-Response Relationship, Immunologic , Electroporation , Hepacivirus/genetics , Hepacivirus/immunology , Hepatitis B Antibodies/blood , Hepatitis B Core Antigens/genetics , Hepatitis B virus/genetics , Hepatitis B virus/immunology , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Helper-Inducer/immunology , Transfection , Viral Core Proteins/genetics , Viral Core Proteins/immunology
14.
J Immunol ; 183(1): 578-92, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19542469

ABSTRACT

Extracellular NAD induces the ATP-independent activation of the ionotropic P2X(7) purinergic receptor (P2X(7)R) in murine T lymphocytes via a novel covalent pathway involving ADP-ribosylation of arginine residues on the P2X(7)R ectodomain. This modification is catalyzed by ART2.2, a GPI-anchored ADP-ribosyltransferase (ART) that is constitutively expressed in murine T cells. We previously reported that ART2.1, a related ecto-ART, is up-regulated in inflammatory murine macrophages that constitutively express P2X(7)R. Thus, we tested the hypothesis that extracellular NAD acts via ART2.1 to regulate P2X(7)R function in murine macrophages. Coexpression of the cloned murine P2X(7)R with ART2.1 or ART2.2 in HEK293 cells verified that P2X(7)R is an equivalent substrate for ADP-ribosylation by either ART2.1 or ART2.2. However, in contrast with T cells, the stimulation of macrophages or HEK293 cells with NAD alone did not activate the P2X(7)R. Rather, NAD potentiated ATP-dependent P2X(7)R activation as indicated by a left shift in the ATP dose-response relationship. Thus, extracellular NAD regulates the P2X(7)R in both macrophages and T cells but via distinct mechanisms. Although ADP-ribosylation is sufficient to gate a P2X(7)R channel opening in T cells, this P2X(7)R modification in macrophages does not gate the channel but decreases the threshold for gating in response to ATP binding. These findings indicate that extracellular NAD and ATP can act synergistically to regulate P2X(7)R signaling in murine macrophages and also suggest that the cellular context in which P2X(7)R signaling occurs differs between myeloid and lymphoid leukocytes.


Subject(s)
ADP Ribose Transferases/physiology , Macrophages/immunology , NAD/physiology , Receptors, Purinergic P2/metabolism , T-Lymphocytes/immunology , ADP Ribose Transferases/biosynthesis , ADP Ribose Transferases/genetics , Animals , Cell Line , Cells, Cultured , Dose-Response Relationship, Immunologic , Extracellular Space/enzymology , Extracellular Space/immunology , Extracellular Space/metabolism , Humans , Inflammation Mediators/physiology , Macrophages/enzymology , Macrophages/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NZB , Mice, Knockout , Protein Structure, Tertiary , Receptors, Purinergic P2/biosynthesis , Receptors, Purinergic P2/deficiency , Receptors, Purinergic P2/physiology , Receptors, Purinergic P2X7 , Signal Transduction/genetics , Signal Transduction/immunology , Substrate Specificity/genetics , Substrate Specificity/immunology , T-Lymphocytes/enzymology , T-Lymphocytes/metabolism
15.
Purinergic Signal ; 5(3): 369-83, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19404775

ABSTRACT

ADP-ribosylation of cell surface proteins in mammalian cells is a post-translational modification by which ecto-ADP-ribosyltransferases (ARTs) transfer ADP-ribose from extracellular NAD to protein targets. The ART2 locus at murine chromosome 7 encompasses the tandem Art2a and Art2b genes that encode the distinct ART2.1 and ART2.2 proteins. Although both ecto-enzymes share 80% sequence identity, ART2.1 activity is uniquely regulated by an allosteric disulfide bond that is reducible in the presence of extracellular thiols, such as cysteine and glutathione, that accumulate in hypoxic and ischemic tissues. Previous studies have characterized the expression of ART2.1 and ART2.2 in murine T lymphocytes but not in other major classes of lymphoid and myeloid leukocytes. Here, we describe the expression of ART2.1 activity in a wide range of freshly isolated or tissue-cultured murine myeloid and lymphoid leukocytes. Spleen-derived macrophages, dendritic cells (DC), and B cells constitutively express ART2.1 as their predominant ART while spleen T cells express both ART2.1 and the thiol-independent ART2.2 isoform. Although bone-marrow-derived macrophages (BMDM) and dendritic cells (BMDC) constitutively express ART2.1 at low levels, it is markedly up-regulated when these cells are stimulated in vitro with IFNbeta or IFNgamma. ART2.1 expression and activity in splenic B cells is modestly up-regulated during incubation in vitro for 24 h, a condition that promotes B cell apoptosis. This increase in ART2.1 is attenuated by IL-4 (a B cell survival factor), but is not affected by IFNbeta/gamma, suggesting a possible induction of ART2.1 as an ancillary response to B cell apoptosis. In contrast, ART2.1 and ART2.2, which are highly expressed in freshly isolated splenic T cells, are markedly down-regulated when purified T cells are incubated in vitro for 12-24 h. Studies with the BW5147 mouse thymocyte line verified basal expression of ART2.1 and ART2.2, as in primary spleen T cells, and demonstrated that both isoforms can be up-regulated when T cells are maintained in the presence of IFNs. Comparison of the surface proteins which are ADP-ribosylated by ART2.1 in the different leukocyte subtypes indicated both shared and cell-specific proteins as ART2.1 substrates. The LFA-1 integrin, a major target for ART2.2 in T cells, is also ADP-ribosylated by the ART2.1 expressed in macrophages. Thus, ART2.1, in contrast to ART2.2, is expressed in a broad range of myeloid and lymphoid leukocytes. The thiol redox-sensitive nature of this ecto-enzyme suggests an involvement in purinergic signaling that occurs in the combined context of inflammation and hypoxia/ischemia.

16.
J Immunol ; 179(9): 6215-27, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17947697

ABSTRACT

Nicotinamide adenosine dinucleotide (NAD) can act as a modulator of multiple immune and inflammatory responses when released into extracellular compartments. These actions of extracellular NAD are largely mediated by a family of mammalian ecto-ADP-ribosyltransferases (ARTs) that covalently modify target extracellular or cell surface proteins by transferring ADP-ribose to arginine or cysteine residues. In this study, we report that bone marrow-derived macrophages (BMDM) from BALB/c mice lack constitutive expression of any of the six murine ecto-ART subtypes, but selectively up-regulate ART2.1 in response to multiple proinflammatory mediators including agonists for TLR and type I and type II IFN. Stimulation of BMDM with LPS, IFN-beta, or IFN-gamma induced high expression of ART2.1, but not ART2.2, as a GPI-anchored cell surface ectoenzyme. ART2.1 expression in response to LPS was potentiated by inhibition of ERK1/2 signaling, but inhibited by blockade of the NF-kappaB, PI3K, and JAK-STAT pathways or the presence of neutralizing anti-IFN-beta. The catalytic function of the induced cell surface ART2.1 was strictly dependent on the presence of extracellular thiol-reducing cofactors, suggesting that in vivo activity of ART2.1-expressing macrophages may be potentiated in hypoxic or ischemic compartments. Consistent with the mutated art2a gene in C57BL/6 mice, LPS- or IFN-stimulated BMDM from this strain lacked expression of cell surface ART2 activity in the presence or absence of extracellular thiol reductants. Collectively, these studies identify ART2.1 as a new candidate for linking autocrine/paracrine activation of inflammatory macrophages to the release of NAD, a critical intracellular metabolite.


Subject(s)
ADP Ribose Transferases/metabolism , Interferon-beta/pharmacology , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/enzymology , Sulfhydryl Compounds/metabolism , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/genetics , Animals , Bone Marrow Cells/cytology , Cell Differentiation , Cell Membrane/drug effects , Cell Membrane/enzymology , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Enzymologic , Inflammation/enzymology , Inflammation/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Macrophages/cytology , Mice , Models, Molecular , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Structure, Tertiary , Rats , Sensitivity and Specificity , Signal Transduction
17.
Purinergic Signal ; 3(1-2): 71-81, 2007 Mar.
Article in English | MEDLINE | ID: mdl-18404420

ABSTRACT

Extracellular NAD and ATP exert multiple, partially overlapping effects on immune cells. Catabolism of both nucleotides by extracellular enzymes keeps extracellular concentrations low under steady-state conditions and generates metabolites that are themselves signal transducers. ATP and its metabolites signal through purinergic P2 and P1 receptors, whereas extracellular NAD exerts its effects by serving as a substrate for ADP-ribosyltransferases (ARTs) and NAD glycohydrolases/ADPR cyclases like CD38 and CD157. Both nucleotides activate the P2X7 purinoceptor, although by different mechanisms and with different characteristics. While ATP activates P2X7 directly as a soluble ligand, activation via NAD occurs by ART-dependent ADP-ribosylation of cell surface proteins, providing an immobilised ligand. P2X7 activation by either route leads to phosphatidylserine exposure, shedding of CD62L, and ultimately to cell death. Activation by ATP requires high micromolar concentrations of nucleotide and is readily reversible, whereas NAD-dependent stimulation begins at low micromolar concentrations and is more stable. Under conditions of cell stress or inflammation, ATP and NAD are released into the extracellular space from intracellular stores by lytic and non-lytic mechanisms, and may serve as "danger signals" to alert the immune response to tissue damage. Since ART expression is limited to naïve/resting T cells, P2X7-mediated NAD-induced cell death (NICD) specifically targets this cell population. In inflamed tissue, NICD may inhibit bystander activation of unprimed T cells, reducing the risk of autoimmunity. In draining lymph nodes, NICD may eliminate regulatory T cells or provide space for the preferential expansion of primed cells, and thus help to augment an immune response.

SELECTION OF CITATIONS
SEARCH DETAIL
...