Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
PLoS One ; 14(5): e0216320, 2019.
Article in English | MEDLINE | ID: mdl-31048931

ABSTRACT

X-linked juvenile retinoschisis (XLRS) is a hereditary retinal dystrophy, caused by mutations in the RS1 gene which encodes the secreted protein retinoschisin. In recent years, several molecules have been proposed to interact with retinoschisin, including the retinal Na/K-ATPase, L-voltage gated Ca2+ channels, and specific sugars. We recently showed that the retinal Na/K-ATPase consisting of subunits ATP1A3 and ATP1B2 is essential for anchoring retinoschisin to plasma membranes and identified the glycosylated ATP1B2 subunit as the direct interaction partner for retinoschisin. We now aimed to precisely map the retinoschisin binding domain(s) in ATP1B2. In general, retinoschisin binding was not affected after selective elimination of individual glycosylation sites via site-directed mutagenesis as well as after full enzymatic deglycosylation of ATP1B2. Applying the interface prediction tool PresCont, two putative protein-protein interaction patches ("patch I" and "patch II") consisting each of four hydrophobic amino acid stretches on the ATP1B2 surface were identified. These were consecutively altered by site-directed mutagenesis. Functional assays with the ATP1B2 patch mutants identified patch II and, specifically, the associated amino acid at position 240 (harboring a threonine in ATP1B2) as crucial for retinoschisin binding to ATP1B2. These and previous results led us to suggest an induced-fit binding mechanism for the interaction between retinoschisin and the Na/K-ATPase, which is dependent on threonine 240 in ATP1B2 allowing the accommodation of hyperflexible retinoschisin spikes by the associated protein-protein interaction patch on ATP1B2.


Subject(s)
Adenosine Triphosphatases/metabolism , Cation Transport Proteins/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cell Adhesion Molecules/metabolism , Eye Proteins/metabolism , Retina/metabolism , Adenosine Triphosphatases/genetics , Animals , Binding Sites , Cation Transport Proteins/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules, Neuronal/genetics , Eye Proteins/genetics , HEK293 Cells , Humans , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism
2.
Mol Biol Cell ; 28(16): 2178-2189, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28615319

ABSTRACT

Mutations in the RS1 gene cause X-linked juvenile retinoschisis (XLRS), a hereditary retinal dystrophy. We recently showed that retinoschisin, the protein encoded by RS1, regulates ERK signaling and apoptosis in retinal cells. In this study, we explored an influence of retinoschisin on the functionality of the Na/K-ATPase, its interaction partner at retinal plasma membranes. We show that retinoschisin binding requires the ß2-subunit of the Na/K-ATPase, whereas the α-subunit is exchangeable. Our investigations revealed no effect of retinoschisin on Na/K-ATPase-mediated ATP hydrolysis and ion transport. However, we identified an influence of retinoschisin on Na/K-ATPase-regulated signaling cascades and Na/K-ATPase localization. In addition to the known ERK deactivation, retinoschisin treatment of retinoschisin-deficient (Rs1h-/Y ) murine retinal explants decreased activation of Src, an initial transmitter in Na/K-ATPase signal transduction, and of Ca2+ signaling marker Camk2. Immunohistochemistry on murine retinae revealed an overlap of the retinoschisin-Na/K-ATPase complex with proteins involved in Na/K-ATPase signaling, such as caveolin, phospholipase C, Src, and the IP3 receptor. Finally, retinoschisin treatment altered Na/K-ATPase localization in photoreceptors of Rs1h-/Y retinae. Taken together, our results suggest a regulatory effect of retinoschisin on Na/K-ATPase signaling and localization, whereas Na/K-ATPase-dysregulation caused by retinoschisin deficiency could represent an initial step in XLRS pathogenesis.


Subject(s)
Cell Adhesion Molecules/metabolism , Eye Proteins/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Cell Adhesion Molecules/genetics , Cell Membrane/metabolism , Eye Proteins/genetics , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Photoreceptor Cells/metabolism , Retina/metabolism , Retinoschisis/genetics , Retinoschisis/metabolism , Signal Transduction
3.
J Immunol ; 197(10): 4034-4041, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27798163

ABSTRACT

Hypoxia-inducible factor-1α (HIF-1α), which accumulates in mammalian host organisms during infection, supports the defense against microbial pathogens. However, whether and to what extent HIF-1α expressed by myeloid cells contributes to the innate immune response against Leishmania major parasites is unknown. We observed that Leishmania-infected humans and L. major-infected C57BL/6 mice exhibited substantial amounts of HIF-1α in acute cutaneous lesions. In vitro, HIF-1α was required for leishmanicidal activity and high-level NO production by IFN-γ/LPS-activated macrophages. Mice deficient for HIF-1α in their myeloid cell compartment had a more severe clinical course of infection and increased parasite burden in the skin lesions compared with wild-type controls. These findings were paralleled by reduced expression of type 2 NO synthase by lesional CD11b+ cells. Together, these data illustrate that HIF-1α is required for optimal innate leishmanicidal immune responses and, thereby, contributes to the cure of cutaneous leishmaniasis.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Leishmania major/immunology , Leishmaniasis, Cutaneous/immunology , Myeloid Cells/metabolism , Skin/parasitology , Animals , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunity, Innate , Interferon-gamma/pharmacology , Leishmaniasis, Cutaneous/parasitology , Leishmaniasis, Cutaneous/pathology , Lipopolysaccharides/immunology , Macrophages/immunology , Macrophages/parasitology , Mice , Mice, Inbred C57BL , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/genetics , Parasite Load , Skin/pathology
4.
PLoS One ; 7(4): e34723, 2012.
Article in English | MEDLINE | ID: mdl-22509350

ABSTRACT

HIV-1 candidate vaccines expressing an artificial polyprotein comprising Gag, Pol and Nef (GPN) and a secreted envelope protein (Env) were shown in recent Phase I/II clinical trials to induce high levels of polyfunctional T cell responses; however, Env-specific responses clearly exceeded those against Gag. Here, we assess the impact of the GPN immunogen design and variations in the formulation and vaccination regimen of a combined GPN/Env DNA vaccine on the T cell responses against the various HIV proteins. Subtle modifications were introduced into the GPN gene to increase Gag expression, modify the expression ratio of Gag to PolNef and support budding of virus-like particles. I.m. administration of the various DNA constructs into BALB/c mice resulted in an up to 10-fold increase in Gag- and Pol-specific IFNγ(+) CD8(+) T cells compared to GPN. Co-administering Env with Gag or GPN derivatives largely abrogated Gag-specific responses. Alterations in the molar ratio of the DNA vaccines and spatially or temporally separated administration induced more balanced T cell responses. Whereas forced co-expression of Gag and Env from one plasmid induced predominantly Env-specific T cells responses, deletion of the only H-2(d) T cell epitope in Env allowed increased levels of Gag-specific T cells, suggesting competition at an epitope level. Our data demonstrate that the biochemical properties of an artificial polyprotein clearly influence the levels of antigen-specific T cells, and variations in formulation and schedule can overcome competition for the induction of these responses. These results are guiding the design of ongoing pre-clinical and clinical trials.


Subject(s)
Gene Products, gag/immunology , HIV-1/immunology , Vaccines, DNA , env Gene Products, Human Immunodeficiency Virus/immunology , nef Gene Products, Human Immunodeficiency Virus/immunology , pol Gene Products, Human Immunodeficiency Virus/immunology , Animals , Clinical Trials, Phase III as Topic , Female , Gene Products, gag/biosynthesis , Gene Products, gag/genetics , HEK293 Cells , HIV-1/genetics , Humans , Mice , Mice, Inbred BALB C , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Virus-Like Particle/immunology , env Gene Products, Human Immunodeficiency Virus/biosynthesis , env Gene Products, Human Immunodeficiency Virus/genetics , nef Gene Products, Human Immunodeficiency Virus/biosynthesis , nef Gene Products, Human Immunodeficiency Virus/genetics , pol Gene Products, Human Immunodeficiency Virus/biosynthesis , pol Gene Products, Human Immunodeficiency Virus/genetics
5.
PLoS One ; 6(8): e23233, 2011.
Article in English | MEDLINE | ID: mdl-21829720

ABSTRACT

Creation of an effective vaccine for HIV has been an elusive goal of the scientific community for almost 30 years. Neutralizing antibodies are assumed to be pivotal to the success of a prophylactic vaccine but previous attempts to make an immunogen capable of generating neutralizing antibodies to primary "street strain" isolates have resulted in responses of very limited breadth and potency. The objective of the study was to determine the breadth and strength of neutralizing antibodies against autologous and heterologous primary isolates in a cohort of HIV-1 infected Nigerians and to characterize envelopes from subjects with particularly broad or strong immune responses for possible use as vaccine candidates in regions predominated by HIV-1 CRF02_AG and G subtypes. Envelope vectors from a panel of primary Nigerian isolates were constructed and tested with plasma/sera from the same cohort using the PhenoSense HIV neutralizing antibody assay (Monogram Biosciences Inc, USA) to assess the breadth and potency of neutralizing antibodies. The immediate goal of this study was realized by the recognition of three broadly cross-neutralizing sera: (NG2-clade CRF02_AG, NG3-clade CRF02_AG and NG9- clade G). Based on these findings, envelope gp140 sequences from NG2 and NG9, complemented with a gag sequence (Clade G) and consensus tat (CRF02_AG and G) antigens have been codon-optimized, synthesized, cloned and evaluated in BALB/c mice. The intramuscular administration of these plasmid DNA constructs, followed by two booster DNA immunizations, induced substantial specific humoral response against all constructs and strong cellular responses against the gag and tat constructs. These preclinical findings provide a framework for the design of candidate vaccine for use in regions where the HIV-1 epidemic is driven by clades CRF02_AG and G.


Subject(s)
Antibodies, Neutralizing/immunology , Codon , HIV Antibodies/immunology , HIV-1/immunology , Neutralization Tests , AIDS Vaccines , Animals , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , HIV Antibodies/biosynthesis , HIV-1/classification , HIV-1/genetics , Humans , Mice , Mice, Inbred BALB C , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Cytotoxic/immunology
6.
J Virol ; 84(22): 11602-13, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20826695

ABSTRACT

Heterologous gene transfer by viral vector systems is often limited by factors such as preexisting immunity, toxicity, low packaging capacity, or weak immunogenic potential. A novel viral vector system derived from equine herpesvirus type 1 (EHV-1) not only overcomes some of these obstacles but also promotes the robust expression of a delivered transgene and the induction of antigen-specific immune responses. Regarding an enhanced safety profile, we assessed the impact of the gene encoding the sole essential tegument protein, ETIF, on the replication and immunogenicity of recombinant EHVs. The deletion of ETIF severely attenuates replication in permissive RK13 cells and a human lung epithelial cell line but without influencing transgene expression. Whereas the intranasal administration of a recombinant luciferase EHV in BALB/c mice resulted in transgene expression in nasal cavities and lungs for 5 to 6 days, the ETIF deletion limited expression to 2 days and resulted in 30-fold-less luminescence. Attenuated replication was accompanied by a decreased capacity to induce CD8(+) T cells against a delivered HIV Gag transgene in BALB/c mice following repeated intranasal application. However, a single subcutaneous immunization with a gag DNA vaccine primed specific T cells for substantial expansion by two subsequent intranasal booster immunizations with either the gag recombinant ETIF mutant or the parental virus. In addition to inducing Gag-specific serum antibodies, this prime-boost strategy clearly outperformed three sequential immunizations with the parental or EHV-ΔETIF virus or repeated DNA vaccination by inducing substantial specific secretory IgA (sIgA) titers.


Subject(s)
Gene Deletion , Genetic Vectors/adverse effects , Genetic Vectors/immunology , Herpesvirus 1, Equid/genetics , Vaccines/immunology , Viral Structural Proteins/genetics , Animals , Cell Line , Drug-Related Side Effects and Adverse Reactions , Female , Genetic Vectors/genetics , Herpesvirus 1, Equid/immunology , Herpesvirus 1, Equid/physiology , Humans , Immunization , Mice , Mice, Inbred BALB C , Vaccines/genetics , Viral Structural Proteins/immunology , Virus Replication
7.
Viral Immunol ; 22(5): 309-19, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19811088

ABSTRACT

As part of a European initiative (EuroVacc), we report the design, construction, and immunogenicity of two HIV-1 vaccine candidates based on a clade C virus strain (CN54) representing the current major epidemic in Asia and parts of Africa. Open reading frames encoding an artificial 160-kDa GagPolNef (GPN) polyprotein and the external glycoprotein gp120 were fully RNA and codon optimized. A DNA vaccine (DNA-GPN and DNA-gp120, referred to as DNA-C), and a replication-deficient vaccinia virus encoding both reading frames (NYVAC-C), were assessed regarding immunogenicity in Balb/C mice. The intramuscular administration of both plasmid DNA constructs, followed by two booster DNA immunizations, induced substantial T-cell responses against both antigens as well as Env-specific antibodies. Whereas low doses of NYVAC-C failed to induce specific CTL or antibodies, high doses generated cellular as well as humoral immune responses, but these did not reach the levels seen following DNA vaccination. The most potent immune responses were detectable using prime:boost protocols, regardless of whether DNA-C or NYVAC-C was used as the priming or boosting agent. These preclinical findings revealed the immunogenic response triggered by DNA-C and its enhancement by combining it with NYVAC-C, thus complementing the macaque preclinical and human phase I clinical studies of EuroVacc.


Subject(s)
HIV Infections/prevention & control , HIV-1/immunology , Immunity, Humoral/drug effects , Vaccines, Attenuated/immunology , Vaccines, DNA/immunology , Animals , Cell Line, Tumor , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/immunology , HIV Infections/immunology , HIV Infections/virology , HIV-1/genetics , Humans , Mice , Mice, Inbred BALB C , Retroviridae Proteins/genetics , Retroviridae Proteins/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccinia virus/immunology
8.
Virology ; 382(1): 46-58, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-18945465

ABSTRACT

Human immunodeficiency virus type-1 (HIV-1) Pr55(Gag) virus-like particles (VLP) represent an interesting HIV vaccine component since they stimulate strong humoral and cellular immune responses. We demonstrated that VLP expressed by recombinant baculoviruses activate human PBMC to release pro-inflammatory (lL-6, TNF-alpha), anti-inflammatory (IL-10) and Th1-polarizing (IFN-gamma) cytokines as well as GM-CSF and MIP-1alpha in a dose-and time-dependent manner. Herein, residual baculoviruses within the VLP preparations showed no or minor effects. Monocytes could be identified as a main target for VLP to induce cytokine production. Furthermore, VLP-induced monocyte activation was shown by upregulation of molecules involved in antigen presentation (MHC II, CD80, CD86) and cell adhesion (CD54). Exposure of VLP to serum inactivates its capacity to stimulate cytokine production. In summary, these investigations establish VLP as strong activators of PBMC and monocytes therein, potently enhancing their functionality and potency to promote an efficient immune response. This capacity makes VLP an interesting component of combination vaccines.


Subject(s)
Cytokines/biosynthesis , HIV-1/immunology , Monocytes/immunology , Protein Precursors/immunology , Virosomes/immunology , B7-2 Antigen/biosynthesis , Cell Adhesion Molecules/biosynthesis , Cells, Cultured , Histocompatibility Antigens Class II/biosynthesis , Humans , Oligopeptides/biosynthesis , Receptors, Immunologic/biosynthesis
9.
Hum Gene Ther ; 19(6): 622-34, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18533892

ABSTRACT

Trans-dominant human immunodeficiency virus type 1 (HIV-1) Gag derivatives have been shown to efficiently inhibit late steps of HIV-1 replication in vitro by interfering with Gag precursor assembly, thus ranking among the interesting candidates for gene therapy approaches. However, efficient antiviral activities of corresponding transgenes are likely to be counteracted in particular by cell-mediated host immune responses toward the transgene-expressing cells. To decrease this potential immunogenicity, a 24-amino acid Gly-Ala (GA) stretch derived from Epstein-Barr virus nuclear antigen-1 (EBNA1) and known to overcome proteasomal degradation was fused to a trans-dominant Gag variant (sgD1). To determine the capacity of this fusion polypeptide to repress viral replication, PM-1 cells were transduced with sgD1 and GAsgD1 transgenes, using retroviral gene transfer. Challenge of stably transfected permissive cell lines with various viral strains indicated that N-terminal GA fusion even enhanced the inhibitory properties of sgD1. Further studies revealed that the GA stretch increased protein stability by blocking proteasomal degradation of Gag proteins. Immunization of BALB/c mice with a DNA vaccine vector expressing sgD1 induced substantial Gag-specific immune responses that were, however, clearly diminished in the presence of GA. Furthermore, recognition of cells expressing the GA-fused transgene by CD8(+) T cells was drastically reduced, both in vitro and in vivo, resulting in prolonged survival of the transduced cells in recipient mice.


Subject(s)
Epstein-Barr Virus Nuclear Antigens/genetics , Genetic Therapy , HIV Infections/therapy , HIV-1 , Transduction, Genetic , gag Gene Products, Human Immunodeficiency Virus/genetics , AIDS Vaccines/immunology , Alanine/genetics , Animals , Cell Line , Cell Survival , Epstein-Barr Virus Nuclear Antigens/metabolism , Glycine/genetics , Humans , Mice , Peptides/genetics , Peptides/metabolism , Proteasome Endopeptidase Complex/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repetitive Sequences, Amino Acid/genetics , Transgenes , Vaccines, DNA/immunology , Virus Replication , gag Gene Products, Human Immunodeficiency Virus/metabolism
10.
Vaccine ; 25(11): 1969-92, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17224219

ABSTRACT

Recombinants based on the attenuated vaccinia virus strains MVA and NYVAC are considered candidate vectors against different human diseases. In this study we have generated and characterized in BALB/c and in transgenic HHD mice the immunogenicity of two attenuated poxvirus vectors expressing in a single locus (TK) the codon optimized HIV-1 genes encoding gp120 and Gag-Pol-Nef (GPN) polyprotein of clade C (referred as MVA-C and NYVAC-C). In HHD mice primed with either MVA-C or NYVAC-C, or primed with DNA-C and boosted with the poxvirus vectors, the splenic T cell responses against clade C peptides spanning gp120/GPN was broad and mainly directed against Gag-1, Env-1 and Env-2 peptide pools. In BALB/c mice immunized with the homologous or the heterologous combination of poxvirus vectors or with Semliki forest virus (SFV) vectors expressing gp120/GPN, the immune response was also broad but the most immunogenic peptides were Env-1, GPN-1 and GPN-2. Differences in the magnitude of the cellular immune responses were observed between the poxvirus vectors depending on the protocol used. The specific cellular immune response triggered by the poxvirus vectors was Th1 type. The cellular response against the vectors was higher for NYVAC than for MVA in both HHD and BALB/c mice, but differences in viral antigen recognition between the vectors was observed in sera from the poxvirus-immunized animals. These results demonstrate the immunogenic potential of MVA-C and NYVAC-C as novel vaccine candidates against clade C of HIV-1.


Subject(s)
AIDS Vaccines/immunology , HIV Antigens/immunology , HIV-1/immunology , AIDS Vaccines/genetics , Animals , Base Sequence , Codon/genetics , Gene Products, gag/genetics , Gene Products, gag/immunology , Gene Products, nef/genetics , Gene Products, nef/immunology , Gene Products, pol/genetics , Gene Products, pol/immunology , Genetic Vectors , HIV Antigens/genetics , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/immunology , HIV-1/genetics , Humans , Immunization, Secondary , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Transgenic , Models, Animal , Molecular Sequence Data , Semliki forest virus , Spleen/immunology , T-Lymphocytes/immunology , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccinia virus , Viral Vaccines , nef Gene Products, Human Immunodeficiency Virus
11.
J Virol ; 79(9): 5445-54, 2005 May.
Article in English | MEDLINE | ID: mdl-15827159

ABSTRACT

Key problems using viral vectors for vaccination and gene therapy are antivector immunity, low transduction efficiencies, acute toxicity, and limited capacity to package foreign genetic information. It could be demonstrated that animal and human cells were efficiently transduced with equine herpesvirus 1 (EHV-1) reconstituted from viral DNA maintained and manipulated in Escherichia coli. Between 13 and 23% of primary human CD3+, CD4+, CD8+, CD11b+, and CD19+ cells and more than 70% of CD4+ MT4 cells or various human tumor cell lines (MeWo, Huh7, HeLa, 293T, or H1299) could be transduced with one infectious unit of EHV-1 per cell. After intranasal instillation of EHV-1 into mice, efficient transgene expression in lungs was detectable. Successful immunization using EHV-1 was shown after delivery of the human immunodeficiency virus type 1 Pr55gag precursor by the induction of a Gag-specific CD8+ immune response in mice. Because EHV-1 was not neutralized by human sera containing high titers of antibodies directed against human herpesviruses 1 to 5, it is concluded that this animal herpesvirus has enormous potential as a vaccine vector, because it is able to efficiently transduce a variety of animal and human cells, has high DNA packaging capacity, and can conveniently be maintained and manipulated in prokaryotic cells.


Subject(s)
Genetic Vectors/immunology , Herpesvirus 1, Equid/immunology , Immunization , Administration, Intranasal , Animals , Antibodies, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cattle , Cell Line , Cross Reactions , Gene Products, gag/biosynthesis , Gene Products, gag/genetics , Genetic Vectors/genetics , HIV Infections/immunology , HIV Infections/prevention & control , Herpesvirus 1, Equid/genetics , Horses , Humans , Immune Sera , Immunity, Cellular , Leukocytes, Mononuclear/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Precursors/biosynthesis , Protein Precursors/genetics , Spleen/immunology , Transduction, Genetic , Vaccines, Synthetic/biosynthesis , Vaccines, Synthetic/genetics
12.
Vaccine ; 22(13-14): 1732-43, 2004 Apr 16.
Article in English | MEDLINE | ID: mdl-15068857

ABSTRACT

We have analysed the influence of size, intracellular localisation, and sorting of various human immunodeficiency virus type 1 (HIV-1)-derived Gag and Env polypeptides containing well defined H2(d)-restricted cytotoxic T lymphocyte (CTL) epitopes on the induction of a humoral and cellular immune response after DNA vaccination. Thus, expression vectors were generated based on RNA- and codon-optimised genes encoding (i). budding competent full-length Gag, (ii). a myristylation defect mutant GagMyr(-), (iii). the isolated p24 capsid moiety of Gag as well as variants of these proteins, which were C-terminally fused HIV gp120-derived V3 epitope (R10I), respectively. These constructs were compared to different minitopes each encoding one of the H2(d)-restricted Gag epitopes A9I and E10F or the V3 epitope R10I that were directly linked to the C-terminus of an Ad2-E3 protein-derived ER signal peptide. Immunological evaluation of these constructs in BALB/c mice revealed that both, the budding competent as well as the intracellular Gag proteins were-irrespective of their molecular weights-equally efficient in the priming of Gag-specific humoral and cellular immune responses. In addition, the capacity of these constructs to stimulate Gag-specific humoral as well as H2-K(d) and H2-L(d) restricted cellular immune responses was not influenced by C-terminal fusion of the immunodominant H2-D(d) restricted V3 epitope. Chimeric GagV3 polyproteins encoding all three major CTL epitopes within a continuous polyprotein were more efficient to stimulate epitope-specific cellular immune responses than the selected minitopes. In addition, the minitopes failed to induce epitope-specific antibody responses. These results clearly show the advantages of complex polypeptides over minitopes regarding the induction of strong humoral and cellular immune responses.


Subject(s)
AIDS Vaccines/immunology , Epitopes/immunology , HIV-1/immunology , Immunity, Cellular/immunology , Peptides/immunology , T-Lymphocytes, Cytotoxic/immunology , AIDS Vaccines/genetics , Animals , Antibody Formation , Codon/genetics , Cytokines/analysis , Cytokines/biosynthesis , DNA Primers , Epitopes/genetics , Female , Gene Products, env/immunology , Gene Products, gag/immunology , Genes, MHC Class I/immunology , HIV-1/genetics , Immunity, Cellular/genetics , Mice , Mice, Inbred BALB C , Peptides/genetics , Plasmids/genetics , Plasmids/immunology , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction , Vaccines, DNA/genetics , Vaccines, DNA/immunology
13.
Methods Mol Med ; 94: 133-57, 2004.
Article in English | MEDLINE | ID: mdl-14959827

ABSTRACT

T-cells play a crucial role in the control of various viral infections such as HIV and herpes viruses. Thus, the development of advanced techniques for the stimulation and measurement of both antigen-specific T-helper and CTL responses is one of most meaningful objectives in vaccinology. Herein, we present HIV-1 Pr55gag lipoprotein particles (VLPs) to be a potent antigen for introducing epitopes into the MHC-class-I and -II processing and presentation pathway. These VLPs can easily be produced in insect cells by using the baculovirus expression system. Immunization studies in mice revealed the strong capacity of these VLPs to stimulate Gag-specific T-helper-1 cell-biased humoral and cellular immune responses. In addition, these VLPs can be used as a stimulator antigen for the detection of Gag-specific T-helper and CTL responses, as determined by conventional ELISA, ELISpot, FACS, and 51Cr-release assays. These results strongly underline the value of VLPs as a stimulator of MHC-class-I and -II mediated epitope presentation for preventive, therapeutic, and diagnostic purposes.


Subject(s)
Antigens, Viral , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antigen Presentation , Antigens, Viral/genetics , Baculoviridae/genetics , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression , Gene Products, gag/genetics , Gene Products, gag/immunology , HIV Infections/immunology , HIV-1/genetics , HIV-1/immunology , Humans , Immunization , In Vitro Techniques , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Precursors/genetics , Protein Precursors/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology
14.
Vaccine ; 20(15): 1980-4, 2002 May 06.
Article in English | MEDLINE | ID: mdl-11983258

ABSTRACT

Most studies on DNA-based immunization have used viral promoters to drive antigen expression. Recently, the use of tissue-specific DNA vaccines has been favored regarding safety issues. In this study, we determined the impact of antigen localization and tissue-specific expression on the induction of humoral as well as cellular immune responses in a BALB/c mouse model. Thereby, we show that using the muscle-specific muscle creatine kinase (MCK) promoter/enhancer the efficiency of immune stimulation is strictly dependent on the ability of HIV-1 Pr55(gag) to be released from cells. By contrast, localization of Pr55(gag) and derivatives thereof plays only a minor role when antigen is constitutively expressed using the ubiquitous viral CMV promoter.


Subject(s)
AIDS Vaccines/immunology , Creatine Kinase/genetics , Enhancer Elements, Genetic , Gene Products, gag/immunology , HIV Core Protein p24/immunology , HIV-1/immunology , Isoenzymes/genetics , Muscle, Skeletal/metabolism , Promoter Regions, Genetic , Protein Precursors/immunology , Vaccines, DNA/immunology , Animals , Antibody Specificity , Creatine Kinase, MM Form , Gene Products, gag/analysis , Gene Products, gag/genetics , Genes, Synthetic , Genes, gag , HIV Antibodies/biosynthesis , HIV Core Protein p24/analysis , HIV Core Protein p24/genetics , HIV-1/genetics , Humans , Immunity, Cellular , Mice , Mice, Inbred BALB C , Organ Specificity , Protein Precursors/analysis , Protein Precursors/genetics , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
15.
J Antimicrob Chemother ; 49(4): 625-30, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11909836

ABSTRACT

The mechanisms of fluoroquinolone resistance in two isolates of Enterobacter cloacae, Ecl#1 and Ecl#2, from the same patient and with identical pulsed-field gel electrophoresis patterns, have been analysed. MICs of ciprofloxacin were 0.25 and 1 mg/L for Ecl#1 and Ecl#2, respectively. Ecl#2 was also more resistant to chloramphenicol and organic solvents. The quinolone resistance determining regions of gyrA/B and parC/E, and the marORA and acrB genes, were sequenced. Expression of marR, acrB, soxS, robA, ramA and fis was analysed by northern blotting. The activity of a 90 bp E. cloacae mar promoter fragment was examined with the reporter plasmid pIGJ-1mar. Sequencing the gyrAB and parCE genes revealed a single amino acid substitution in GyrA (corresponding to position 83 in GyrA of Escherichia coli) in Ecl#1 and Ecl#2 (Phe83) compared with reference strain E. cloacae DSMZ 3264 (Thr83). Ecl#2 accumulated significantly less norfloxacin and displayed higher levels of expression of marR and acrB than Exl#1, indicative of greater fluoroquinolone efflux activity. Sequencing gyrB, parC/E and marORA, and northern blotting of robA, ramA and fis, did not reveal any further differences between the two strains. No homologue of soxRS was detected in E. cloacae. Expression of GFP from pIGJ1-mar in Ecl#2 was higher than in Ecl#1. In these two closely related clinical isolates of E. cloacae, a target mutation in GyrA (Ecl#1 and Ecl#2) and increased fluoroquinolone efflux by AcrAB (Ecl#2) contribute to the resistance phenotypes, corroborating findings in vitro and in vivo about the sequential development of fluoroquinolone resistance.


Subject(s)
Anti-Infective Agents/pharmacology , Ciprofloxacin/pharmacology , Enterobacter cloacae/drug effects , Drug Resistance, Bacterial/genetics , Humans , Microbial Sensitivity Tests
16.
Intervirology ; 45(4-6): 275-86, 2002.
Article in English | MEDLINE | ID: mdl-12566710

ABSTRACT

In this study, we analyzed the in vitro expression, potency and longevity of immune responses induced in a Balb/c mouse model by a synthetic HIV-1 GAG gene exhibiting a codon usage that was adapted to that of highly expressed mammalian genes (syngag). In contrast to a vector containing the wild-type (wt) GAG gene, the syngag construct enabled highly efficient Gag expression in both human and rodent cell lines in complete absence of Rev and Rev-responsive element. Immunization of Balb/c mice with the wt gag plasmid DNA induced only weak and inconsistent humoral immune responses. Mice vaccinated by syngag but not wt gag developed substantial and highly consistent Gag-specific antibody titers showing a clear T helper 1 polarization even with low doses of DNA. Moreover, vaccinated mice developed a strong Gag-specific cellular immune response, including cytotoxic T cells, which was not observed in wt gag-immunized animals. Both humoral and cellular immunity were efficient and lasted for more than 20 weeks. Furthermore, the induction of the humoral as well as the cellular immune response was independent of the immunization route (intramuscular or subcutaneous). These results clearly show the advantages of codon-optimized genes with respect to the expression and immunogenicity of plasmid DNA constructs, making them promising vaccine candidates for further studies.


Subject(s)
AIDS Vaccines/immunology , Codon , Gene Products, gag/immunology , HIV-1/immunology , T-Lymphocytes/immunology , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , HIV-1/genetics , Humans , Injections, Intramuscular , Injections, Subcutaneous , Tumor Cells, Cultured , Vaccines, DNA/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...