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











Publication year range
1.
Hum Vaccin Immunother ; 9(10): 2228-36, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23896580

ABSTRACT

The efficacy of DNA vaccines is highly dependent on the methods used for their delivery and the choice of delivery sites/targets for gene injection, pointing at the necessity of a strict control over the gene delivery process. Here, we have investigated the effect of the injection site on gene expression and immunogenicity in BALB/c mice, using as a model a weak gene immunogen, DNA encoding firefly luciferase (Luc) delivered by superficial or deep injection with subsequent electroporation (EP). Immunization was assessed by monitoring the in vivo expression of luciferase by 2D- and 3D-bioluminescence imaging (BLI) and by the end-point immunoassays. Anti-Luc antibodies were assessed by ELISA, and T-cell response by IFN-γ and IL-2 FluoroSpot in which mouse splenocytes were stimulated with Luc or a peptide representing its immunodominant CD8+ T-cell epitope GFQSMYTFV. Monitoring of immunization by BLI identified EP parameters supporting the highest Luc gene uptake and expression. Superficial injection of Luc DNA followed by optimal EP led to a low level Luc expression in the mouse skin, and triggered a CD8+ T-cell response characterized by the peptide-specific secretion of IFN-γ and IL-2, but no specific antibodies. Intramuscular gene delivery resulted in a several-fold higher Luc expression and anti-Luc antibody, but induced low IL-2 and virtually no specific IFN-γ. Photon flux from the sites of Luc gene injection was inversely proportional to the immune response against GFQSMYTFV (p<0.05). Thus, BLI permitted to control the accuracy of gene delivery and transfection with respect to the injection site as well as the parameters of electroporation. Further, it confirmed the critical role of the site of DNA administration for the type and magnitude of the vaccine-specific immune response. This argues for the use of luminescent reporters in the preclinical gene vaccine tests to monitor both gene delivery and the immune response development in live animals.


Subject(s)
Immunization/methods , Luminescent Measurements , Optical Imaging , Vaccines, DNA/administration & dosage , Vaccines, DNA/pharmacokinetics , Animals , Antibodies/blood , Enzyme-Linked Immunosorbent Assay , Female , Genes, Reporter , Immunoassay , Insect Proteins/immunology , Interferon-gamma/metabolism , Interleukin-2/metabolism , Luciferases, Firefly/biosynthesis , Luciferases, Firefly/genetics , Mice , Mice, Inbred BALB C , T-Lymphocytes/immunology
2.
Hum Vaccin Immunother ; 9(10): 2111-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23881028

ABSTRACT

HIV-1 infection induces chronic oxidative stress. The resultant neurotoxicity has been associated with Tat protein. Here, we for the first time describe the induction of oxidative stress by another HIV-1 protein, reverse transcriptase (RT). Expression of HIV-1 RT in human embryonic kidney cells generated potent production of the reactive oxygen species (ROS), detected by the fluorescence-based probes. Quantitative RT-PCR demonstrated that expression of RT in HEK293 cells induced a 10- to 15-fold increased transcription of the phase II detoxifying enzymes human NAD(P)H: quinone oxidoreductase (Nqo1) and heme oxygenase 1 (HO-1), indicating the induction of oxidative stress response. The capacity to induce oxidative stress and stress response appeared to be an intrinsic property of a vast variety of RTs: enzymatically active and inactivated, bearing mutations of drug resistance, following different routes of processing and presentation, expressed from viral or synthetic expression-optimized genes. The total ROS production induced by RT genes of the viral origin was found to be lower than that induced by the synthetic/expression-optimized or chimeric RT genes. However, the viral RT genes induced higher levels of ROS production and higher levels of HO-1 mRNA than the synthetic genes per unit of protein in the expressing cell. The capacity of RT genes to induce the oxidative stress and stress response was then correlated with their immunogenic performance. For this, RT genes were administered into BALB/c mice by intradermal injections followed by electroporation. Splenocytes of immunized mice were stimulated with the RT-derived and control antigens and antigen-specific proliferation was assessed by IFN-γ/IL-2 Fluorospot. RT variants generating high total ROS levels induced significantly stronger IFN-γ responses than the variants inducing lower total ROS, while high levels of ROS normalized per unit of protein in expressing cell were associated with a weak IFN-γ response. Poor gene immunogenicity was also associated with a high (per unit of protein) transcription of antioxidant response element (ARE) dependent phase II detoxifying enzyme genes, specifically HO-1. Thus, we have revealed a direct link between the propensity of the microbial proteins to induce oxidative stress and their immunogenicity.


Subject(s)
AIDS Vaccines/immunology , HIV Reverse Transcriptase/immunology , Immunization/methods , Oxidative Stress , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Animals , Cell Line , Gene Expression Profiling , HIV Reverse Transcriptase/genetics , Humans , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred BALB C , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
3.
Mol Imaging ; 11(6): 471-86, 2012.
Article in English | MEDLINE | ID: mdl-23084248

ABSTRACT

The efficient cell-mediated immune response clears cells expressing deoxyribonucleic acid (DNA) immunogens, but there are no methods to monitor this in vivo. We hypothesized that immune-mediated clearance can be monitored in vivo if DNA immunogens are coexpressed with reporter(s). To test this, we designed genes encoding human immunodeficiency virus 1 (HIV-1) reverse transcriptase (RT) fused via its N- or C-terminus to 30-amino acid-long Gly-Ala-repeat of Epstein-Barr virus nuclear antigen 1 or via the N-terminus to the transport signal of invariant chain/Ii or inserted between the cytoplasmic and luminal domains of lysosome-associated membrane protein I (LAMP). DNA immunogens mixed with luciferase gene were injected into BALB/c mice with subsequent electroporation. Reporter expression seen as luminescence was monitored by in vivo imaging. When luminescence faded, mice were sacrificed, and their splenocytes were stimulated with RT-derived antigens. Fading of luminescence correlated with the RT-specific secretion of interferon-γ and interleukin-2. Both immune and in vivo imaging techniques concordantly demonstrated an enhanced immunogenicity of RT-LAMP and of the N-terminal Gly-Ala-RT fusion genes. In vivo imaging performed as an animal-sparing method to estimate the overall performance of DNA immunogens, predicting it early in the experiment. So far, in vivo imaging cannot be a substitute for conventional immune assays, but it is supplementary to them. Further experiments are needed to identify which arms of cellular immune response in vivo imaging monitors best.


Subject(s)
Optical Imaging , Vaccines, Synthetic/immunology , Amino Acid Sequence , Animals , Base Sequence , CD4-Positive T-Lymphocytes/immunology , Epitopes/immunology , Female , HEK293 Cells , HIV Reverse Transcriptase/immunology , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , T-Lymphocytes/immunology , Vaccines, DNA/immunology , Vaccines, Synthetic/genetics
4.
Proteomics ; 8(11): 2211-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18528842

ABSTRACT

The driving force behind oncoproteomics is to identify protein signatures that are associated with a particular malignancy. Here, we have used a recombinant scFv antibody microarray in an attempt to classify sera derived from pancreatic adenocarcinoma patients versus healthy subjects. Based on analysis of nonfractionated, directly labeled, whole human serum proteomes we have identified a protein signature based on 19 nonredundant analytes, that discriminates between cancer patients and healthy subjects. Furthermore, a potential protein signature, consisting of 21 protein analytes, could be defined that was shown to be associated with cancer patients having a life expectancy of <12 months. Taken together, the data suggest that antibody microarray analysis of complex proteomes will be a useful tool to define disease associated protein signatures.


Subject(s)
Blood Proteins/chemistry , Gene Expression Regulation, Neoplastic , Immunoglobulin Fragments/chemistry , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/diagnosis , Protein Array Analysis/methods , Adult , Aged , Aged, 80 and over , Antibodies, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Female , Humans , Male , Middle Aged , Neoplasm Proteins/chemistry , Pancreatic Neoplasms/metabolism , Proteomics/methods
5.
Infect Agent Cancer ; 2: 14, 2007 Jul 10.
Article in English | MEDLINE | ID: mdl-17623060

ABSTRACT

BACKGROUND: The human immunodeficiency virus type 1 (HIV-1) regulatory protein, Nef, is an attractive vaccine target because it is involved in viral pathogenesis, is expressed early in the viral life cycle and harbors many T and B cell epitopes. Several clinical trials include gene-based vaccines encoding this protein. However, Nef has been shown to transform certain cell types in vitro. Based on these findings we performed a long-term toxicity and immunogenicity study of Nef, encoded either by Modified Vaccinia virus Ankara or by plasmid DNA. BALB/c mice were primed twice with either DNA or MVA encoding Nef and received a homologous or heterologous boost ten months later. In the meantime, the Nef-specific immune responses were monitored and at the time of sacrifice an extensive toxicological evaluation was performed, where presence of tumors and other pathological changes were assessed. RESULTS: The toxicological evaluation showed that immunization with MVAnef is safe and does not cause cellular transformation or other toxicity in somatic organs.Both DNAnef and MVAnef immunized animals developed potent Nef-specific cellular responses that declined to undetectable levels over time, and could readily be boosted after almost one year. This is of particular interest since it shows that plasmid DNA vaccine can also be used as a potent late booster of primed immune responses. We observed qualitative differences between the T cell responses induced by the two different vectors: DNA-encoded nef induced long-lasting CD8+ T cell memory responses, whereas MVA-encoded nef induced CD4+ T cell memory responses. In terms of the humoral immune responses, we show that two injections of MVAnef induce significant anti-Nef titers, while repeated injections of DNAnef do not. A single boost with MVAnef could enhance the antibody response following DNAnef prime to the same level as that observed in animals immunized repeatedly with MVAnef. We also demonstrate the possibility to boost HIV-1 Nef-specific immune responses using the MVAnef construct despite the presence of potent anti-vector immunity. CONCLUSION: This study shows that the nef gene vectored by MVA does not induce malignancies or other adverse effects in mice. Further, we show that when the nef gene is delivered by plasmid or by a viral vector, it elicits potent and long-lasting immune responses and that these responses can be directed towards a CD4+ or a CD8+ T cell response depending on the choice of vector.

6.
Vaccine ; 25(11): 2145-54, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17254672

ABSTRACT

A DNA plasmid encoding human immunodeficiency virus type 1 (HIV-1) env, nef and tat genes was used in mice in a prime-boost immunization regimen with the corresponding recombinant proteins. The genetic immunogen was delivered with a gene gun and the proteins were injected intramuscularly together with the adjuvant AS02A. Immunizations were followed by experimental challenge with pseudotyped HIV-1 subtype A or B virus. In an initial experiment in which animals were challenged four weeks after the final immunization, all single modality and prime-boost vaccinations resulted in a significant level of protection as compared to control animals. There was a trend for DNA-alone immunization yielding the highest protection. In a subsequent study, a late challenge was performed 19 weeks after the final immunization. All groups having received the DNA vaccine, either alone or in combination with adjuvanted protein, exhibited strong protection against HIV replication. The subtype-specific protection against the experimental HIV challenge was significantly stronger than the cross-protection. Cellular and humoral immune responses were assessed during immunization and after challenge, but without clear correlation to protection against HIV replication. The data suggest that either DNA or protein antigens alone provide partial protection against an HIV-1/MuLV challenge and that DNA immunization is essential for achieving very high levels of efficacy in this murine HIV-1 challenge model. While prime-boost combinations were more immunogenic than DNA alone, they did not appear to provide any further enhancement over DNA vaccine mediated efficacy. The DNA immunogen might prime low levels of CD8+ T cells responsible for virus clearance or possibly a yet unidentified mechanism of protection.


Subject(s)
AIDS Vaccines/immunology , Adjuvants, Immunologic , HIV Infections/prevention & control , Lipid A/analogs & derivatives , Retroviridae Infections/prevention & control , Saponins/immunology , Vaccines, DNA/immunology , Animals , Biolistics , Disease Models, Animal , Drug Combinations , HIV Antibodies/blood , HIV Antigens/genetics , HIV Antigens/immunology , HIV-1/immunology , Humans , Immunization, Secondary , Injections, Intramuscular , Leukemia Virus, Murine , Lipid A/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Recombinant Proteins/genetics , Recombinant Proteins/immunology , T-Lymphocytes/immunology , Tumor Virus Infections/prevention & control , Vaccination , Vaccines, Subunit/immunology
7.
Vaccine ; 24(21): 4524-6, 2006 May 22.
Article in English | MEDLINE | ID: mdl-16174543

ABSTRACT

We investigated the effects of immunizing with several genes and subtypes of HIV-1. The genes used as immunogens were: gp160 envelope (env subtypes A, B and C), p37gag (gag subtypes A and B), rev (subtype B) and reverse transcriptase (RT subtype B). The different genes are all carried by separate plasmids. C57BL/6 and BALB/c mice were immunized with different combinations of the genes together with recombinant cytokine granulocyte macrophage-colony stimulating factor. The env genes injected alone induced significantly stronger cellular responses to envelope in both strains of mice than when env genes were injected together with gag and RT genes. In the C57BL/6 mice, the envelope specific responses were significantly increased after spatial separation of env genes from gag and RT genes as compared to when all vaccine genes were injected as a mixture. The gag responses were strong in gag-immunized animals and were not significantly affected by the spatial separation of gag and RT genes from the env genes. Our results illustrate the importance of being cautious when formulating multivalent genetic vaccines and that it might be possible to overcome lost immune responses through spatial separation of vaccine antigens.


Subject(s)
AIDS Vaccines/immunology , Genes, Viral , HIV-1/genetics , HIV-1/immunology , Immunity, Cellular , AIDS Vaccines/administration & dosage , Animals , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plasmids
8.
Vaccine ; 24(21): 4572-5, 2006 May 22.
Article in English | MEDLINE | ID: mdl-16159686

ABSTRACT

C57BL/6 and BALB/c mice were immunized with two different plasmids (p91023B and pKCEA66) encoding different forms of the tumor-associated antigen carcinoembryonic antigen (CEA). The wild type form of CEA (p91023B), which is expressed at the cell surface, induces stronger anti-CEA IgG response after DNA-plasmid immunizations than the modified intracellular form of CEA (pKCEA66), which was designed to mount strong cellular responses. Boosting with recombinant CEA (rCEA) increased the anti-CEA IgG response significantly. In the tumor protection model used, where SCID mice are challenged with human tumor cells mixed with splenocytes from immunized mice, both innate and specific immune responses are responsible for the protective effect.


Subject(s)
Carcinoembryonic Antigen/immunology , Vaccines, DNA/immunology , Animals , Epitopes/immunology , Killer Cells, Natural/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , T-Lymphocytes/immunology , Tetanus Toxoid/immunology , Vaccines, DNA/administration & dosage
9.
Vaccine ; 22(13-14): 1791-8, 2004 Apr 16.
Article in English | MEDLINE | ID: mdl-15068863

ABSTRACT

We evaluated the compound imiquimod as a possible adjuvant for DNA immunization against human immunodeficiency virus (HIV). We found that gene-gun epidermal delivery of the DNA in combination with imiquimod resulted in the strongest HIV specific immune responses. The effect of imiquimod was further compared to that of recombinant granulocyte macrophage-colony stimulating factor (GM-CSF), a known DNA vaccine adjuvant. Both adjuvants were able to enhance the immune responses induced by the HIV-1 genes alone. The delivery of an adjuvant as a topical cream rather than through injections has a clear clinical benefit. We show for the first time that imiquimod can act as an adjuvant for DNA vaccination.


Subject(s)
AIDS Vaccines/immunology , Adjuvants, Immunologic/pharmacology , Aminoquinolines/pharmacology , Adjuvants, Immunologic/administration & dosage , Administration, Topical , Aminoquinolines/administration & dosage , Animals , Antibody Formation/immunology , Biolistics , Cytokines/biosynthesis , Cytokines/blood , Enzyme-Linked Immunosorbent Assay , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , HIV-1/genetics , HIV-1/immunology , Imiquimod , Immunity, Cellular/immunology , Immunization , Injections, Intradermal , Interferon-gamma/analysis , Interferon-gamma/biosynthesis , Leukemia Virus, Murine/immunology , Mice , Spleen/cytology , Spleen/immunology , Vaccines, DNA/immunology
10.
Cancer Gene Ther ; 10(5): 365-76, 2003 May.
Article in English | MEDLINE | ID: mdl-12719706

ABSTRACT

Carcinoembryonic antigen (CEA, CEACAM5) is expressed on several human carcinomas including colon cancer. CEA contains signal peptides that target the protein through the endoplasmic reticulum and to the cell membrane. We constructed a plasmid DNA vaccine encoding a truncated CEA (deltaCEA), devoid of its signal peptides, and demonstrated that it was retained inside the cell, while full-length CEA (wtCEA) was expressed on the membrane. We hypothesized that intracellular retention of deltaCEA would enhance MHC class I presentation of CEA peptides, thus favoring cellular immune responses. In addition, a promiscuous T-helper epitope (Q830-L844 of tetanus toxoid) was fused to the N-terminal of the truncated CEA gene (tetdeltaCEA). C57BL/6 mice immunized with DNA encoding wtCEA or tetdeltaCEA developed both humoral and cellular immune responses to CEA. SCID mice transplanted with spleen cells from tetdeltaCEA but not wtCEA-immunized C57BL/6 mice showed strong suppression of tumor growth after inoculation of human CEA-expressing colon carcinoma cells. Immune spleen cell populations depleted for either B, T or both B and T cells were active, indicating that effector cells might also reside in other populations. The present approach to manipulating antigen presentation may open new possibilities for immunotherapy against colon and other CEA-secreting carcinomas.


Subject(s)
Carcinoembryonic Antigen/immunology , Colonic Neoplasms/prevention & control , Protein Sorting Signals , Tetanus Toxoid/genetics , Vaccines, DNA/administration & dosage , Animals , Artificial Gene Fusion , B-Lymphocytes/immunology , Cell Division , Colonic Neoplasms/immunology , Gene Transfer Techniques , Genetic Vectors , Humans , Immunization , Mice , Mice, Inbred C57BL , Mice, SCID , Peptide Fragments , Plasmids/genetics , Sequence Deletion , Spleen/immunology , Survival Rate , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL