59 results on '"Dna immunization"'
Search Results
2. Mesenchymal Stem Cells Can both Enhance and Inhibit the Cellular Response to DNA Immunization by Genes of Nonstructural Proteins of the Hepatitis C Virus
- Author
-
Klimova Rr, E. I. Lesnova, Alexander V. Ivanov, Alla A. Kushch, and O. V. Masalova
- Subjects
0301 basic medicine ,Genes, Viral ,medicine.medical_treatment ,T-Lymphocytes ,Hepacivirus ,Viral Nonstructural Proteins ,medicine.disease_cause ,HCV vaccine ,immune response ,Mice ,0302 clinical medicine ,Plasmid ,Vaccines, DNA ,Biology (General) ,Spectroscopy ,Immunity, Cellular ,Vaccination ,nonstructural HCV proteins ,General Medicine ,Hepatitis C ,Computer Science Applications ,Chemistry ,medicine.anatomical_structure ,Treatment Outcome ,Mice, Inbred DBA ,030220 oncology & carcinogenesis ,Cytokines ,Female ,Adjuvant ,Plasmids ,DNA immunization ,QH301-705.5 ,Hepatitis C virus ,mesenchymal stem cells (MSC) ,Biology ,Transfection ,Catalysis ,Article ,DNA vaccination ,Proinflammatory cytokine ,Inorganic Chemistry ,03 medical and health sciences ,Immune system ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Physical and Theoretical Chemistry ,Molecular Biology ,QD1-999 ,Organic Chemistry ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Virology ,030104 developmental biology ,Bone marrow ,hepatitis C virus (HCV) - Abstract
Despite extensive research, there is still no vaccine against the hepatitis C virus (HCV). The aim of this study was to investigate whether MSCs can exhibit adjuvant properties during DNA vaccination against hepatitis C. We used the pcNS3-NS5B plasmid encoding five nonstructural HCV proteins and MSCs derived from mice bone marrow. Five groups of DBA mice were immunized with the plasmid and/or MSCs in a different order. Group 1 was injected with the plasmid twice at intervals of 3 weeks, Group 2 with the plasmid, and after 24 h with MSCs, Group 3 with MSCs followed by the plasmid the next day, Group 4 with only MSCs, and Group 5 with saline. When the MSCs were injected prior to DNA immunization, the cell immune response to HCV proteins assessed by the level of IFN-γ synthesis was markedly increased compared to DNA alone. In contrast, MSCs injected after DNA suppressed the immune response. Apparently, the high level of proinflammatory cytokines detected after DNA injection promotes the conversion of MSCs introduced later into the immunosuppressive MSC2. The low level of cytokines in mice before MSC administration promotes the high immunostimulatory activity of MSC1 in response to a DNA vaccine. Thus, when administered before DNA, MSCs are capable of exhibiting promising adjuvant properties.
- Published
- 2021
- Full Text
- View/download PDF
3. Immune protection duration and efficacy stability of DNA vaccine encoding Eimeria tenella TA4 and chicken IL-2 against coccidiosis
- Author
-
Xiaokai Song, Xiaofang Zhao, Xiangrui Li, Ruofeng Yan, and Lixin Xu
- Subjects
Protozoan Vaccines ,0301 basic medicine ,Indirect elisa ,Protozoan Proteins ,Biology ,Eimeria ,DNA vaccination ,03 medical and health sciences ,Vaccines, DNA ,medicine ,Animals ,Poultry Diseases ,General Veterinary ,Coccidiosis ,Immune protection ,Vaccination ,medicine.disease ,biology.organism_classification ,Virology ,030104 developmental biology ,Dna immunization ,Immunization ,Immunology ,biology.protein ,Interleukin-2 ,Antibody ,Chickens ,Eimeria tenella - Abstract
In our previous study, an effective DNA vaccine encoding Eimeria tenella TA4 and chicken IL-2 was constructed. In the present study, the immunization dose of the DNA vaccine pVAX1.0-TA4-IL-2 was further optimized. With the optimized dose, the dynamics of antibodies induced by the DNA vaccine was determined using indirect ELISA. To evaluate the immune protection duration of the DNA vaccine, two-week-old chickens were intramuscularly immunized twice and the induced efficacy was evaluated by challenging with E. tenella at 5, 9, 13, 17 and 21weeks post the last immunization (PLI) separately. To evaluate the efficacy stability of the DNA vaccine, two-week-old chickens were immunized with 3 batches of the DNA vaccine, and the induced efficacy was evaluated by challenging with E. tenella. The results showed that the optimal dose was 25μg. The induced antibody level persisted until 10weeks PPI. For the challenge time of 5 and 9weeks PLI, the immunization resulted in ACIs of 182.28 and 162.23 beyond 160, showing effective protection. However, for the challenge time of 13, 17 and 21weeks PLI, the immunization resulted in ACIs below 160 which means poor protection. Therefore, the immune protection duration of the DNA vaccination was at least 9weeks PLI. DNA immunization with three batches DNA vaccine resulted in ACIs of 187.52, 191.57 and 185.22, which demonstrated that efficacies of the three batches DNA vaccine were effective and stable. Overall, our results indicate that DNA vaccine pVAX1.0-TA4-IL-2 has the potential to be developed as effective vaccine against coccidiosis.
- Published
- 2017
- Full Text
- View/download PDF
4. In vitro simian virus 40 large tumor antigen expression correlates with differential immune responses following DNA immunization
- Author
-
Lowe, Devin B., Shearer, Michael H., Tarbox, James A., Kang, Hyun Seok, Jumper, Cynthia A., Bright, Robert K., and Kennedy, Ronald C.
- Subjects
- *
SIMIAN viruses , *TUMORS , *IMMUNE response , *VIROLOGY - Abstract
Abstract: Simian virus 40 (SV40) contains an essential protein, large tumor antigen (Tag), which assists in viral replication and causes cell transformation and immortalization. Our laboratory has examined plasmid DNA, expressing SV40 Tag under two different promoters, for use in potential cancer vaccination strategies. One plasmid, pSV3-neo, failed to induce SV40 Tag antibody, produced a weak cell-mediated response, and only partial protection in murine experimental tumor challenge systems. The second plasmid, pCMV-Tag, induced antibodies to SV40 Tag, produced a robust cell-mediated response, and invoked complete tumor immunity in vivo. The induction of CD4+ and CD8+ T cell responses following plasmid DNA immunization and tumor cell challenge reflected a type 1 cytokine secretion profile. Our hypothesis for this differential immune response is that pCMV-Tag exhibits a higher level of transgene expression due to a more efficient promoter. We determined that pCMV-Tag levels of SV40 Tag mRNA and protein expression were higher when compared to pSV3-neo. A threshold amount of SV40 Tag may be required to stimulate antibody production and provide complete systemic tumor immunity. [Copyright &y& Elsevier]
- Published
- 2005
- Full Text
- View/download PDF
5. M448R and MGF505-7R: Two African Swine Fever Virus Antigens Commonly Recognized by ASFV-Specific T-Cells and with Protective Potential
- Author
-
Javier Collado, Egbert Mundt, Elisabet López, Laia Bosch-Camós, Veljko Nikolin, María L. Salas, Miguel Blanco-Fuertes, Sonia Pina-Pedrero, Francesc Accensi, Fernando Rodriguez, Maria Jesus Navas, Generalitat de Catalunya, Ministerio de Ciencia e Innovación (España), Red de Investigación en Sanidad Animal (España), Producció Animal, and Sanitat Animal
- Subjects
0301 basic medicine ,DNA immunization ,030106 microbiology ,Immunology ,Priming (immunology) ,Immunopeptidomics ,Biology ,African swine fever virus ,Article ,Virus ,DNA vaccination ,03 medical and health sciences ,Plasmid ,Immune system ,Antigen ,Drug Discovery ,Pharmacology (medical) ,Antigen discovery ,live attenuated virus ,Pharmacology ,Protection ,Attenuated vaccine ,Live attenuated virus ,T-cells ,immunopeptidomics ,protection ,biology.organism_classification ,Virology ,antigen discovery ,030104 developmental biology ,Infectious Diseases ,Medicine ,African swine fever - Abstract
African swine fever (ASF) is today s number one threat for the global swine industry. Neither commercial vaccine nor treatment is available against ASF and, thus far, only live attenuated viruses (LAV) have provided robust protection against lethal ASF virus (ASFV) challenge infections. Identification of ASFV proteins inducing protective immune responses is one of the major challenges to develop safer and efficient subunit vaccines. Immunopeptidomic studies recently performed in our laboratory allowed identifying ASFV antigens recognized by ASFV-specific CD8 T-cells. Here, we used data from the SLAI-peptide repertoire presented by a single set of ASFV-infected porcine alveolar macrophages to generate a complex DNA vaccine composed by 15 plasmids encoding the individual peptide-bearing ORFs. DNA vaccine priming improved the protection afforded by a suboptimal dose of the BA71∆CD2 LAV given as booster vaccination, against Georgia2007/1 lethal challenge. Interestingly, M448R was the only protein promiscuously recognized by the induced ASFV-specific T-cells. Furthermore, priming pigs with DNA plasmids encoding M488R and MGF505-7R, a CD8 T-cell antigen previously described, confirmed these two proteins as T-cell antigens with protective potential. These studies might be useful to pave the road for designing safe and more efficient vaccine formulations in the future., Generalitat de Catalunya (Spain), grant number 2015 DI 037. Studies were co-financed by Boehringer Ingelheim Veterinary Research Center (BIVRC) GmbH & Co. KG, the Ministerio de Ciencia e Innovación of Spain (grant numbers AGL2016-78169-C2-1-R and PID2019-107616RB-I00) and Red de Investigación en Sanidad Animal (RISA)
- Published
- 2021
- Full Text
- View/download PDF
6. DNA immunization as a technology platform for monoclonal antibody induction
- Author
-
Shixia Wang, Shuying Liu, and Shan Lu
- Subjects
0301 basic medicine ,Immunogen ,Epidemiology ,animal diseases ,Review ,immunogenicity ,Communicable Diseases, Emerging ,law.invention ,Mice ,Immunogenicity, Vaccine ,law ,Drug Discovery ,Vaccines, DNA ,Pathogen ,Mice, Inbred BALB C ,Immunogenicity ,Antibodies, Monoclonal ,General Medicine ,Recombinant Proteins ,3. Good health ,Infectious Diseases ,Dna immunization ,Recombinant DNA ,Antibody ,DNA immunization ,medicine.drug_class ,Immunology ,Enzyme-Linked Immunosorbent Assay ,chemical and pharmacologic phenomena ,Biology ,Monoclonal antibody ,Microbiology ,hybridoma ,03 medical and health sciences ,Virology ,medicine ,Animals ,Humans ,Immunization Schedule ,Hybridomas ,antigen delivery ,biochemical phenomena, metabolism, and nutrition ,030104 developmental biology ,Immunization ,monoclonal antibody ,biology.protein ,bacteria ,Parasitology - Abstract
To combat the threat of many emerging infectious diseases, DNA immunization offers a unique and powerful approach to the production of high-quality monoclonal antibodies (mAbs) against various pathogens. Compared with traditional protein-based immunization approaches, DNA immunization is efficient for testing novel immunogen designs, does not require the production or purification of proteins from a pathogen or the use of recombinant protein technology and is effective at generating mAbs against conformation-sensitive targets. Although significant progress in the use of DNA immunization to generate mAbs has been made over the last two decades, the literature does not contain an updated summary of this experience. The current review provides a comprehensive analysis of the literature, including our own work, describing the use of DNA immunization to produce highly functional mAbs, in particular, those against emerging infectious diseases. Critical factors such as immunogen design, delivery approach, immunization schedule, use of immune modulators and the role of final boost immunization are discussed in detail.
- Published
- 2016
- Full Text
- View/download PDF
7. Removal of the N-glycosylation sequon at position N116 located in p27 of the respiratory syncytial virus fusion protein elicits enhanced antibody responses after DNA immunization
- Author
-
Peter Delputte, Xavier Saelens, Paul Cos, Annelies Leemans, Dalan Bailey, Isabel Pintelon, Louis Maes, Kenny Roose, Wim Martinet, Winke Van der Gucht, Guy Caljon, Bert Schepens, and Marlies Boeren
- Subjects
0301 basic medicine ,Models, Molecular ,class I fusion protein ,Glycosylation ,MONOCLONAL-ANTIBODY ,lcsh:QR1-502 ,Antibodies, Viral ,Protein Engineering ,lcsh:Microbiology ,Mice ,vaccine ,Medicine and Health Sciences ,Vaccines, DNA ,REVERSE GENETICS ,Neutralizing antibody ,CELL-FUSION ,Mice, Inbred BALB C ,Immunogenicity ,Hydrolysis ,ENVELOPE PROTEINS ,Viral Load ,LINKED GLYCANS ,Infectious Diseases ,Female ,Antibody ,virus glycosylation ,Plasmids ,DNA immunization ,medicine.drug_class ,Protein subunit ,030106 microbiology ,Respiratory Syncytial Virus Infections ,MEMBRANE-FUSION ,Biology ,Monoclonal antibody ,Virus ,Article ,03 medical and health sciences ,NEUTRALIZING ANTIBODY ,Virology ,medicine ,Respiratory Syncytial Virus Vaccines ,Animals ,Humans ,pneumovirus ,humoral responses ,Biology and Life Sciences ,IN-VITRO ,Sequon ,F-PROTEIN ,Fusion protein ,Antibodies, Neutralizing ,WEST NILE VIRUS ,Protein Subunits ,030104 developmental biology ,Respiratory Syncytial Virus, Human ,Mutation ,biology.protein ,Immunization ,Human medicine ,Viral Fusion Proteins - Abstract
Prevention of severe lower respiratory tract infections in infants caused by the human respiratory syncytial virus (hRSV) remains a major public health priority. Currently, the major focus of vaccine development relies on the RSV fusion (F) protein since it is the main target protein for neutralizing antibodies induced by natural infection. The protein conserves 5 N-glycosylation sites, two of which are located in the F2 subunit (N27 and N70), one in the F1 subunit (N500) and two in the p27 peptide (N116 and N126). To study the influence of the loss of one or more N-glycosylation sites on RSV F immunogenicity, BALB/c mice were immunized with plasmids encoding RSV F glycomutants. In comparison with F WT DNA immunized mice, higher neutralizing titres were observed following immunization with F N116Q. Moreover, RSV A2-K-line19F challenge of mice that had been immunized with mutant F N116Q DNA was associated with lower RSV RNA levels compared with those in challenged WT F DNA immunized animals. Since p27 is assumed to be post-translationally released after cleavage and thus not present on the mature RSV F protein, it remains to be elucidated how deletion of this glycan can contribute to enhanced antibody responses and protection upon challenge. These findings provide new insights to improve the immunogenicity of RSV F in potential vaccine candidates.
- Published
- 2018
8. Efficacy Testing of H56 cDNA Tattoo Immunization against Tuberculosis in a Mouse Model
- Author
-
Platteel, Anouk C M, Nieuwenhuizen, Natalie E, Domaszewska, Teresa, Schürer, Stefanie, Zedler, Ulrike, Brinkmann, Volker, Sijts, Alice, Kaufmann, Stefan H E, dI&I RA-I&I I&I, LS Immunologie, dI&I RA-I&I I&I, and LS Immunologie
- Subjects
0301 basic medicine ,lcsh:Immunologic diseases. Allergy ,DNA immunization ,T cell ,Immunology ,complex mixtures ,Epitope ,03 medical and health sciences ,Immune system ,Antigen ,vaccine ,medicine ,Immunology and Allergy ,Original Research ,biology ,business.industry ,H56 ,Toxoid ,vaccination ,Virology ,Mycobacterium bovis bacillus Calmette–Guérin ,3. Good health ,Vaccination ,030104 developmental biology ,medicine.anatomical_structure ,Immunization ,tuberculosis ,biology.protein ,bacteria ,Antibody ,business ,lcsh:RC581-607 - Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains a global threat. The only approved vaccine against TB, Mycobacterium bovis bacillus Calmette-Guérin (BCG), provides insufficient protection and, being a live vaccine, can cause disseminated disease in immunocompromised individuals. Previously, we found that intradermal cDNA tattoo immunization with cDNA of tetanus toxoid fragment C domain 1 fused to cDNA of the fusion protein H56, comprising the Mtb antigens Ag85B, ESAT-6, and Rv2660c, induced antigen-specific CD8+ T cell responses in vivo. As cDNA tattoo immunization would be safer than a live vaccine in immunocompromised patients, we tested the protective efficacy of intradermal tattoo immunization against TB with H56 cDNA, as well as with H56_E, a construct optimized for epitope processing in a mouse model. As Mtb antigens can be used in combination with BCG to boost immune responses, we also tested the protective efficacy of heterologous prime-boost, using dermal tattoo immunization with H56_E cDNA to boost BCG immunization in mice. Dermal H56 and H56_E cDNA immunization induced H56-specific CD4+ and CD8+ T cell responses and Ag85B-specific IgG antibodies, but did not reduce bacterial loads, although immunization with H56_E ameliorated lung pathology. Both subcutaneous and intradermal immunization with BCG resulted in broad cellular immune responses, with increased frequencies of CD4+ T effector memory cells, T follicular helper cells, and germinal center B cells, and resulted in reduced bacterial loads and lung pathology. Heterologous vaccination with BCG/H56_E cDNA induced increased H56-specific CD4+ and CD8+ T cell cytokine responses compared to vaccination with BCG alone, and lung pathology was significantly decreased in BCG/H56_E cDNA immunized mice compared to unvaccinated controls. However, bacterial loads were not decreased after heterologous vaccination compared to BCG alone. CD4+ T cells responding to Ag85B- and ESAT-6-derived epitopes were predominantly IFN-γ+TNF-α+ and TNF-α+IL-2+, respectively. In conclusion, despite inducing appreciable immune responses to Ag85B and ESAT-6, intradermal H56 cDNA tattoo immunization did not substantially enhance the protective effect of BCG under the conditions tested.
- Published
- 2017
- Full Text
- View/download PDF
9. Efficacy of a DNA Vaccine Carrying Eimeria maxima Gam56 Antigen Gene against Coccidiosis in Chickens
- Author
-
Jinjun Xu, Yan Zhang, and Jianping Tao
- Subjects
Protozoan Vaccines ,DNA vaccine ,DNA immunization ,Antibodies, Protozoan ,Antigens, Protozoan ,Lymphocyte proliferation ,Injections, Intramuscular ,Eimeria ,DNA vaccination ,Antigen ,Vaccines, DNA ,medicine ,Animals ,Lymphocytes ,Cell Proliferation ,biology ,Coccidiosis ,Immunogenicity ,Vaccination ,biology.organism_classification ,medicine.disease ,Gam56 antigen ,Virology ,Disease Models, Animal ,Infectious Diseases ,Eimeria maxima ,Peripheral blood lymphocyte ,Original Article ,Parasitology ,Chickens ,control - Abstract
To control coccidiosis without using prophylactic medications, a DNA vaccine targeting the gametophyte antigen Gam56 from Eimeria maxima in chickens was constructed, and the immunogenicity and protective effects were evaluated. The ORF of Gam56 gene was cloned into an eukaryotic expression vector pcDNA3.1(zeo)+. Expression of Gam56 protein in COS-7 cells transfected with recombinant plasmid pcDNA-Gam56 was confirmed by indirect immunofluorescence assay. The DNA vaccine was injected intramuscularly to yellow feathered broilers of 1-week old at 3 dosages (25, 50, and 100 µg/chick). Injection was repeated once 1 week later. One week after the second injection, birds were challenged orally with 5×10(4) sporulated oocysts of E. maxima, then weighed and killed at day 8 post challenge. Blood samples were collected and examined for specific peripheral blood lymphocyte proliferation activity and serum antibody levels. Compared with control groups, the administration of pcDNA-Gam56 vaccine markedly increased the lymphocyte proliferation activity (P
- Published
- 2013
- Full Text
- View/download PDF
10. Immune responses induced by DNA vaccines bearing Spike gene of PEDV combined with porcine IL-18
- Author
-
Yudong Ren, Dingding Su, Pengchong Li, Guangxing Li, Xunliang Li, Dante S. Zarlenga, Ri-e Bu, Fandan Meng, Ren Xiaofeng, Siqingaowa Suo, and Chao Wang
- Subjects
Cancer Research ,DNA immunization ,T-Lymphocytes ,Spike protein ,Immunofluorescence ,medicine.disease_cause ,Porcine interleukin-18 gene ,Antibodies, Viral ,Article ,DNA vaccination ,Interferon-gamma ,Mice ,Immune system ,Antigen ,Adjuvants, Immunologic ,Viral Envelope Proteins ,Virology ,medicine ,Vaccines, DNA ,Cytotoxic T cell ,Animals ,Adjuvant ,Coronavirus ,Cell Proliferation ,Membrane Glycoproteins ,medicine.diagnostic_test ,biology ,Porcine epidemic diarrhea virus ,Interleukin-18 ,biology.organism_classification ,Molecular biology ,Infectious Diseases ,Blood ,Spike Glycoprotein, Coronavirus ,biology.protein ,Interleukin-4 ,Antibody ,Spleen ,Plasmids ,T-Lymphocytes, Cytotoxic - Abstract
Highlights ► Generation of three distinct DNA constructs encoding S protein of PEDV, the N-terminal fragment (S1) of PEDV and porcine interleukin-18 (pIL-18). ► Immune responses of above-mentioned DNA constructs were evaluated systematically using mouse model. ► PEDV-S and the combination of PEDV-S1 with pIL-18 induced the strongest responses but that pIL-18 had no adjuvant effects when given in combination with PEDV-S1., Porcine epidemic diarrhea virus (PEDV) is the causative agent of porcine epidemic diarrhea, a highly contagious enteric disease of swine. The Spike (S) protein is one of the main structural proteins of PEDV capable of inducing neutralizing antibodies in vivo. Herein, we generated three distinct DNA constructs in the eukaryotic expression plasmid pVAX1; one encoding the S protein [pVAX1-(PEDV-S)], the second encoding the N-terminal fragment (S1) [pVAX1-(PEDV-S1)] containing potent antigenic sites, and the third expressing the porcine interleukin-18 (pIL-18) [pVAX1-(IL-18)]. Immunofluorescence assays in BHK-21 cells demonstrated successful protein expression from all 3 constructs. Kunming mice were injected separately with each of these constructs or with a pVAX1-(PEDV-S1)/pVAX1-(IL-18) combination, an attenuated PEDV vaccine, or vector only control. Animals were examined for T lymphocyte proliferation, anti-PEDV antibodies, IFN-γ and IL-4 protein levels, and cytotoxic T cell function in mouse peripheral blood and spleen. In all cases, results showed that pVAX1-(PEDV-S) and the combination of pVAX1-(PEDV-S1) with pVAX1-(IL-18) induced the strongest responses; however, pIL-18 had no adjuvant effects when given in combination with pVAX1-(PEDV-S1).
- Published
- 2012
11. A DNA Vaccine Encoding for TcSSP4 Induces Protection against Acute and Chronic Infection in Experimental Chagas Disease
- Author
-
Patricia Talamás-Rohana, Minerva Arce-Fonseca, Aracely López-Monteon, Angel Ramos-Ligonio, Berenice Salgado-Jiménez, and José Luis Rosales-Encina
- Subjects
Chagas disease ,DNA immunization ,Trypanosoma cruzi ,Protozoan Proteins ,Applied Microbiology and Biotechnology ,DNA vaccination ,law.invention ,Interferon-gamma ,Mice ,Plasmid ,law ,TcSSP4 ,Complementary DNA ,medicine ,Vaccines, DNA ,Animals ,Chagas Disease ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Mice, Inbred BALB C ,biology ,Cell Biology ,Th1 Cells ,biology.organism_classification ,medicine.disease ,Virology ,Chronic infection ,Myocarditis ,Immunization ,Recombinant DNA ,Cytokines ,Female ,Developmental Biology ,Research Paper ,Plasmids - Abstract
Immunization of mice with plasmids containing genes of Trypanosoma cruzi induces protective immunity in the murine model of Chagas disease. A cDNA clone that codes for an amastigote-specific surface protein (TcSSP4) was used as a candidate to develop a DNA vaccine. Mice were immunized with the recombinant protein rTcSSP4 and with cDNA for TcSSP4, and challenged with bloodstream trypomastigotes. Immunization with rTcSSP4 protein makes mice more susceptible to trypomastigote infection, with high mortality rates, whereas mice immunized with a eukaryotic expression plasmid containing the TcSSP4 cDNA were able to control the acute phase of infection. Heart tissue of gene-vaccinated animals did not show myocarditis and tissue damage at 365 days following infection, as compared with control animals. INF-γ was detected in sera of DNA vaccinated mice shortly after immunization, suggesting the development of a Th1 response. The TcSSP4 gene is a promising candidate for the development of an anti-T. cruzi DNA vaccine.
- Published
- 2011
12. Immunogenicity difference between the SARS coronavirus and the bat SARS-like coronavirus spike (S) proteins
- Author
-
Zhenggang Han, Zhengli Shi, Peng Zhou, and Lin-Fa Wang
- Subjects
DNA immunization ,Immunodominant region ,viruses ,Biophysics ,Spike protein ,Biology ,Severe Acute Respiratory Syndrome ,medicine.disease_cause ,Biochemistry ,Article ,Epitope ,Neutralization ,Cell Line ,Mice ,Viral Envelope Proteins ,Chiroptera ,medicine ,Animals ,Humans ,Molecular Biology ,Gene ,Coronavirus ,Genomic organization ,Membrane Glycoproteins ,SL-CoV ,Immunogenicity ,virus diseases ,Viral Vaccines ,Cell Biology ,biochemical phenomena, metabolism, and nutrition ,Virology ,respiratory tract diseases ,Titer ,Severe acute respiratory syndrome-related coronavirus ,Spike Glycoprotein, Coronavirus ,biology.protein ,Antibody - Abstract
SARS-like coronavirus (SL-CoV) in bats have a similar genomic organization to the human SARS-CoV. Their cognate gene products are highly conserved with the exception of the N-terminal region of the S proteins, which have only 63-64% sequence identity. The N-terminal region of coronavirus S protein is responsible for virus-receptor interaction. In this study, the immunogenicity of the SL-CoV S protein (S(SL)) was studied and compared with that of SARS-CoV (S(SARS)). DNA immunization in mice with S(SL) elicited a high titer of antibodies against HIV-pseudotyped S(SL). The sera had low cross-reactivity, but no neutralization activity, for the HIV-pseudotyped S(SARS). Studies using wild bat sera revealed that it is highly likely that the immunodominant epitopes overlap with the major neutralizing sites of the SL-CoV S protein. These results demonstrated that SL-CoV and SARS-CoV shared only a limited number of immunogenic epitopes in their S proteins and the major neutralization epitopes are substantially different. This work provides useful information for future development of differential serologic diagnosis and vaccines for coronaviruses with different S protein sequences.
- Published
- 2009
- Full Text
- View/download PDF
13. Advances in DNA immunization against hepatitis C virus infection: Opportunities and challenges
- Author
-
Santiago Dueñas-Carrera and Liz Alvarez-Lajonchere
- Subjects
Cirrhosis ,Hepatitis C virus ,Immunogenicity ,Immunology ,Biology ,medicine.disease ,medicine.disease_cause ,Virology ,digestive system diseases ,Virus ,DNA vaccination ,Chronic infection ,Dna immunization ,Hepatocellular carcinoma ,medicine ,General Pharmacology, Toxicology and Pharmaceutics - Abstract
Hepatitis C virus (HCV) causes chronic infection in approximately two thirds of cases, leading to chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma in a substantial proportion of the 170 million HCV-infected individuals worldwide. As there is neither prophylactic nor therapeutic vaccine for this virus, the research in this area is of special importance. Several vaccine candidates have been evaluated in pre-clinic, but just a few has reached the clinical evaluation. DNA immunization is one of the most evaluated approaches to obtain an effective vaccine against HCV infection. In the last few years a group of technical refinements in DNA vaccines has allowed to increase their immunogenicity. Two DNA vaccine candidates against HCV have already reached clinical evaluation, being well tolerated and immunogenic in HCV-chronically infected individuals. The main results, opportunities and challenges of DNA immunization against HCV are discussed further in the present commentary. DNA vaccination is a...
- Published
- 2009
- Full Text
- View/download PDF
14. Search for potential target site of nucleocapsid gene for the design of an epitope-based SARS DNA vaccine
- Author
-
Le Quynh Mai, Noton K. Dutta, Dong Jae Kim, Hyun Kyoung Lee, Byoung-Hee Lee, Min Won Baek, Hiroaki Kariwa, Sung-Hoon Park, Kaushiki Mazumdar, Jae-Hak Park, and Yi Rang Na
- Subjects
Cellular immunity ,DNA immunization ,T-Lymphocytes ,Immunology ,Epitope ,Article ,Antibodies ,law.invention ,DNA vaccination ,Cell Line ,Epitopes ,Mice ,Immune system ,Antigen ,law ,Immunity ,Escherichia coli ,Vaccines, DNA ,Immunology and Allergy ,Animals ,Humans ,Nucleocapsid ,Cell Proliferation ,Mice, Inbred BALB C ,biology ,SARS-CoV ,Viral Vaccines ,Virology ,Molecular biology ,Severe acute respiratory syndrome-related coronavirus ,Recombinant DNA ,biology.protein ,Female ,Antibody - Abstract
It is believed today that nucleocapsid protein (N) of severe acute respiratory syndrome (SARS)-CoV is one of the most promising antigen candidates for vaccine design. In this study, three fragments [N1 (residues: 1-422); N2 (residues: 1-109); N3 (residues: 110-422)] of N protein of SARS-CoV were expressed in Escherichia coli and analyzed by pooled sera of convalescence phase of SARS patients. Three gene fragments [N1 (1-1269 nt), N2 (1-327 nt) and N3 (328-1269 nt)-expressing the same proteins of N1, N2 and N3, respectively] of SARS-N were cloned into pVAX-1 and used to immunize BALB/c mice by electroporation. Humoral (by enzyme-linked immunosorbent assay, ELISA) and cellular (by cell proliferation and CD4(+):CD8(+) assay) immunity was detected by using recombinant N1 and N3 specific antigen. Results showed that N1 and N3 fragments of N protein expressed by E. coli were able to react with sera of SARS patients but N2 could not. Specific humoral and cellular immunity in mice could be induced significantly by inoculating SARS-CoV N1 and N3 DNA vaccine. In addition, the immune response levels in N3 were significantly higher for antibody responses (IgG and IgG1 but not IgG2a) and cell proliferation but not in CD4(+):CD8(+) assay compared to N1 vaccine. The identification of antigenic N protein fragments has implications to provide basic information for the design of DNA vaccine against SARS-CoV. The present results not only suggest that DNA immunization with pVax-N3 could be used as potential DNA vaccination approaches to induce antibody in BALB/c mice, but also illustrates that gene immunization with these SARS DNA vaccines can generate different immune responses.
- Published
- 2008
15. Potential der DNA-Vakzinierung zur Prävention und Therapie von allergischen Krankheiten
- Author
-
A. B. Reske-Kunz, M. Bros, R. Ross, and S. Sudowe
- Subjects
Type i allergy ,Biology ,medicine.disease_cause ,Virology ,respiratory tract diseases ,Immune therapy ,law.invention ,DNA vaccination ,Vaccination ,Allergen ,Dna immunization ,immune system diseases ,law ,Immunology ,Recombinant DNA ,medicine ,Immunology and Allergy ,Gene - Abstract
Preclinical studies in animal models of type I allergy support the notion that the transfer of allergen genes by means ofDNA vaccination represents a promising alternative to immune therapy with allergen extracts or recombinant allergens. We outline the basic mechanisms of DNA immunization and report on further developments of this vaccination strategy to enhance efficiency and optimize safety of DNA vaccines.
- Published
- 2007
- Full Text
- View/download PDF
16. Co-Expression of Interleukin-2 by a Bicistronic Plasmid Increases the Efficacy of DNA Immunization to Prevent Influenza Virus Infections
- Author
-
Roland Zell, Peter Wutzler, Andreas Henke, and Nadine Rohland
- Subjects
Interleukin 2 ,Biology ,medicine.disease_cause ,Virus ,Cell Line ,Microbiology ,DNA vaccination ,Mice ,Viral Proteins ,Dogs ,Influenza A Virus, H1N1 Subtype ,Plasmid ,Adjuvants, Immunologic ,Orthomyxoviridae Infections ,Virology ,Vaccines, DNA ,medicine ,Influenza A virus ,Animals ,Gene ,Mice, Inbred BALB C ,Infectious Diseases ,Dna immunization ,Immunization ,Influenza Vaccines ,Interleukin-2 ,Female ,Plasmids ,medicine.drug - Abstract
A promising approach to protect susceptible individuals against severe diseases is the inoculation of plasmids. Such DNA vaccines against influenza virus infections were quite efficient in different animal models; but still this procedure is not in clinical use until today. The present study reports the generation and characterization of bicistronic plasmids which enables the expression of influenza A virus gene sequences together with immunostimulatory cytokines demonstrating that among these cytokines especially interleukin-2 (IL-2) was efficient to prevent a lethal influenza virus infection in mice.
- Published
- 2006
- Full Text
- View/download PDF
17. Comparative study of macrophage response in mice after DNA immunization and infection with herpes simplex virus type 1
- Author
-
L. G. Zaitseva, O. V. Nekrasova, L. N. Shingarova, T. M. Andronova, Kobets Nv, E. F. Boldyreva, S. V. Guryanova, A. Yu. Kozlov, I. V. Kireeva, V. A. Bekhalo, E. V. Nagurskaya, and Klimova Rr
- Subjects
Male ,viruses ,medicine.medical_treatment ,Inflammation ,Herpesvirus 1, Human ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,law.invention ,DNA vaccination ,Mice ,Cell Movement ,law ,Vaccines, DNA ,medicine ,Animals ,Macrophage ,Mice, Inbred BALB C ,business.industry ,Macrophages ,Granulocyte-Macrophage Colony-Stimulating Factor ,Herpes Simplex ,General Medicine ,Virology ,Vaccination ,Herpes simplex virus ,Dna immunization ,Immunology ,Recombinant DNA ,medicine.symptom ,business ,Acetylmuramyl-Alanyl-Isoglutamine ,Adjuvant - Abstract
Functional activity of macrophages and intensity of T cell immune response in mice were studied after intravaginal and intraperitoneal infection with herpes simplex virus type 1 and DNA vaccination in combination with adjuvant treatment (recombinant granulocyte-macrophage colony-stimulating factor and glucosaminylmuramyl dipeptide). DNA vaccination induced a virus-specific T cell immune response with no macrophagic inflammatory reaction. Infection with herpes simplex virus type 1 was accompanied by sustained inflammation, but not by the T cell immune response.
- Published
- 2005
- Full Text
- View/download PDF
18. Priming B cell-mediated anti-HIV envelope responses by vaccination allows for the long-term control of infection in macaques exposed to a R5-tropic SHIV
- Author
-
Jeffrey T. Safrit, Leonidas Stamatatos, Cheryl J. Saunders, Leoned G Gines, James Blanchard, Indresh K. Srivastava, Susan W. Barnett, Lucia Vojtech, Agegnehu Gettie, Clarisa Buckner, and Rudolph Bohm
- Subjects
CD4-Positive T-Lymphocytes ,DNA immunization ,Time Factors ,Receptors, CCR5 ,Simian Acquired Immunodeficiency Syndrome ,Priming (immunology) ,HIV Antibodies ,Macaques ,Antibodies ,ΔV2gp140 ,Viral Envelope Proteins ,Immunity ,Virology ,medicine ,Animals ,Neutralizing antibody ,B cell ,Glycoproteins ,AIDS Vaccines ,Recombination, Genetic ,B-Lymphocytes ,SHIVSF162P4 ,biology ,CD8+ depletion ,Vaccination ,virus diseases ,HIV envelope protein ,Macaca mulatta ,HIV neutralization ,medicine.anatomical_structure ,Immunity, Active ,Immunology ,Vaccines, Subunit ,CTL ,biology.protein ,HIV-1 ,Simian Immunodeficiency Virus ,Antibody ,CD8 - Abstract
The potential of vaccine-elicited anti-HIV envelope antibodies to control HIV-infection was evaluated by immunizing macaques with the HIV envelope protein and transiently depleting them of their CD8+ cells before intravenous challenge with the pathogenic CCR5-tropic SIV/HIV chimeric virus, SHIVSF162P4. Although sterilizing immunity was not achieved, all vaccinated animals effectively controlled infection and remained free of disease for the duration of observation (over 3 years). In contrast, during the same period, the control animals progressed to disease. Both the vaccinees and the controls developed robust cell-mediated antiviral and neutralizing antibody responses following infection. A comparative analysis of these responses suggests that the more effective long-term control of infection by the vaccinated animals is due to the more rapid development of anti-HIV envelope antibodies. These studies suggest that priming by vaccination of B cell anti-HIV envelope responses maybe crucial for the long-term control of HIV infection.
- Published
- 2004
- Full Text
- View/download PDF
19. Protection evaluation against Chlamydophila abortus challenge by DNA vaccination with a dnaK-encoding plasmid in pregnant and non-pregnant mice
- Author
-
Olivier Grépinet, Céline Héchard, Annie Rodolakis, and Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)
- Subjects
Biology ,Injections, Intramuscular ,DNA vaccination ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Plasmid ,Animal model ,Pregnancy ,Vaccines, DNA ,Animals ,HSP70 Heat-Shock Proteins ,Chlamydophila Infections ,0303 health sciences ,General Veterinary ,Genetic vaccine ,030306 microbiology ,Chlamydophila ,Escherichia coli Proteins ,biology.organism_classification ,[SDV.MP.BAC]Life Sciences [q-bio]/Microbiology and Parasitology/Bacteriology ,Antibodies, Bacterial ,Virology ,Molecular biology ,Non pregnant ,Recombinant Proteins ,3. Good health ,Chlamydophila abortus ,Disease Models, Animal ,Dna immunization ,Immunoglobulin G ,Female ,Spleen ,Plasmids ,030215 immunology - Abstract
Des souris ont ete immunisees par l'injection intramusculaire d'un plasmide codant pour la proteine DnaK. L'effet protecteur de la vaccination ADN contre une infection par Chlamydophila abortus a ete evalue dans des modeles de souris gestantes ou non-gestantes. La vaccination ADN avec le gene dnaK induit une reponse humorale specifique chez les souris non-gestantes avec une predominance d'IgG2a qui n'ont pas presente d'activite neutralisante in vitro. Aucune reaction d'hypersensibilite retardee n'a ete observee et les rates des souris vaccinees avec le vaccin ADN-dnaK n'ont pas ete protegees contre l'infection par C. abortus. Chez les souris gestantes, le vaccin ADN-dnaKreduit partiellement et de facon non specifique les avortements. Les ilots CpG de l'ADN bacterien, connus pour leur proprietes immunogeniques, pourraient etre responsables de cet effet protecteur non specifique. Cependant, aucune elimination des bacteries n'a ete observee dans les rates des souris gestantes.
- Published
- 2002
- Full Text
- View/download PDF
20. Local and systemic inoculation of DNA or protein gB1s-based vaccines induce a protective immunity against rabbit ocular HSV-1 infection
- Author
-
P. G. Balboni, Elisabetta Caselli, Francesco Parmeggiani, Rafaela Argnani, Carlo Incorvaia, Roberto Manservigi, and Enzo Cassai
- Subjects
Male ,DNA immunization ,Ocular infection ,HSV-1 vaccine ,Enzyme-Linked Immunosorbent Assay ,Herpesvirus 1, Human ,Herpesvirus Vaccines ,Antibodies, Viral ,medicine.disease_cause ,Herpesviridae ,DNA vaccination ,Immune system ,Viral Envelope Proteins ,Vaccines, DNA ,medicine ,Animals ,Neutralizing antibody ,General Veterinary ,General Immunology and Microbiology ,biology ,Immunogenicity ,Public Health, Environmental and Occupational Health ,Vaccine efficacy ,medicine.disease ,Virology ,Infectious Diseases ,Humoral immunity ,Immunology ,Keratitis, Herpetic ,biology.protein ,Molecular Medicine ,Rabbits ,Encephalitis - Abstract
A secreted form of gB1 (gB1s), previously shown to protect rabbits against HSV-1 ocular infection when inoculated systemically, was delivered to rabbit periocular area to evaluate its vaccine efficacy upon local administration. The efficacy of local or systemic inoculation of a gB1s-DNA-based vaccine in the rabbit model of ocular HSV-1 infection was assessed in parallel flow. Rabbits received four inoculations of the different immunogens, then immune responses and clinical symptoms were evaluated. Both the local protein and the systemic DNA administration elicited a neutralizing antibody response, reduced ocular symptoms with respect to controls (P
- Published
- 2000
- Full Text
- View/download PDF
21. Recent advances in the use of DNA vaccines for the treatment of diseases of farmed animals
- Author
-
Lorne A. Babiuk, Reno Pontarollo, Robert Rankin, Sylvia van Drunen Littel-van den Hurk, B. I. Loehr, Richard R. E. Uwiera, and Volker Gerdts
- Subjects
Veterinary Medicine ,IMMUNE STIMULANTS ,Pharmaceutical Science ,Biology ,Virology ,Animal Diseases ,DNA vaccination ,Vaccination ,Disease Models, Animal ,Immune system ,Plasmid ,Dna immunization ,Pharmaceutical technology ,Immunization ,Animals, Domestic ,Immunology ,Vaccines, DNA ,Animals ,Biotechnology - Abstract
DNA-based vaccination constitutes one of the most recent approaches to vaccine development. This technology is in principle one of the most simple and yet versatile methods of inducing both humoral and cellular immune responses, as well as protection against a variety of infectious agents. However, although immune responses have been induced in a number of larger species, most information on the efficacy of DNA immunization has been generated in mice. In this review the information available to date about the use of DNA vaccines in farmed animals, including cattle, pigs and poultry, is presented. The areas that need specific attention in the future to bring this technology to the market are discussed, including the issues concerning delivery, safety, compatibility of plasmids in multivalent vaccines and the potential of using immune stimulants as part of a DNA vaccine.
- Published
- 2000
- Full Text
- View/download PDF
22. Eukaryotic expression of enzymatically active human immunodeficiency virus type 1 reverse transcriptase
- Author
-
Sergey N. Kochetkov, Jorma Hinkula, Katja Pokrovskaya, Britta Wahren, Vladimir I. Kashuba, Dmitrii Pokholok, and Maria G. Isaguliants
- Subjects
DNA immunization ,Protein Conformation ,Protein subunit ,Genetic Vectors ,DNA, Recombinant ,Biophysics ,Human immunodeficiency virus type 1 ,Cytomegalovirus ,Gene Expression ,HIV Antibodies ,Transfection ,Biochemistry ,Cell Line ,Mice ,Plasmid ,Immune system ,Structural Biology ,Chlorocebus aethiops ,Reverse transcriptase ,Genetics ,Animals ,Humans ,Molecular Biology ,AIDS Vaccines ,Chemistry ,3T3 Cells ,Cell Biology ,Virology ,Molecular biology ,HIV Reverse Transcriptase ,In vitro ,Mice, Inbred C57BL ,Molecular Weight ,Eukaryotic expression ,Cell culture ,COS Cells ,HIV-1 ,Immunization ,Immunostaining - Abstract
Reverse transcriptase of human immunodeficiency virus type 1 is a vitalenzyme in the HIV-1 replication cycle and an attractive target of attempts to arrest a primary viral infection. We designed a vector for eukaryotic expression of the 66 kDa subunit of reverse transcriptase under the control of the immediate early cytomegalovirus promoter. Efficient transient expression of the 66 kDa subunit of reverse transcriptase was achieved in a variety of cells. Immunostaining of the transfected cells revealed the cytoplasmatic localization of reverse transcriptase. Reverse transcriptase activity was detected in all transfected cell lines. Injection of this plasmid encoding the 66 kDa subunit of reverse transcriptase into mice resulted in strong reverse transcriptase-specific immune responses indicating that the 66 kDa subunit of reverse transcriptase is expressed in vivo. Sera from DNA-immunized mice inhibited reverse transcription in vitro.
- Published
- 1999
- Full Text
- View/download PDF
23. Protection of Mice against Lethal Coxsackievirus B3 Infection by Using DNA Immunization
- Author
-
Andreas Henke, J. Lindsay Whitton, Elke Wagner, Roland Zell, and Axel Stelzner
- Subjects
viruses ,Immunology ,Coxsackievirus Infections ,Viral Pathogenesis and Immunity ,Enzyme-Linked Immunosorbent Assay ,Vaccinia virus ,Biology ,Antibodies, Viral ,Microbiology ,DNA vaccination ,law.invention ,Mice ,chemistry.chemical_compound ,Plasmid ,law ,Virology ,Vaccines, DNA ,Animals ,Viral Structural Proteins ,Mice, Inbred BALB C ,virus diseases ,biochemical phenomena, metabolism, and nutrition ,Enterovirus B, Human ,Vaccination ,Dna immunization ,chemistry ,Coxsackievirus b3 ,Insect Science ,Recombinant DNA ,biology.protein ,Antibody ,DNA ,Plasmids - Abstract
Vaccination with DNA and recombinant vaccinia viruses (rec.VV) has been studied with the coxsackievirus B3 (CVB3) model system. Plasmids encoding all structural proteins of CVB3, when injected intramuscularly, induced only low levels of virus-specific antibodies. However, DNA vaccination with the major structural protein VP1 protected 72.2% of mice from lethal challenge, whereas VP1 expressed by rec.VV was much less efficient.
- Published
- 1998
- Full Text
- View/download PDF
24. Protective Cellular Immunity Against Influenza VirusInduced by Plasmid Inoculation of Newborn Mice
- Author
-
Constantin A. Bona, Adolfo García-Sastre, Adrian Bot, and Simona Bot
- Subjects
lcsh:Immunologic diseases. Allergy ,Cellular immunity ,DNA immunization ,newborns ,T-Lymphocytes ,Immunology ,Biology ,Cross Reactions ,medicine.disease_cause ,Virus ,influenza virus ,Interferon-gamma ,Mice ,Immune system ,Antigen ,Orthomyxoviridae Infections ,Immunity ,Influenza A virus ,medicine ,Vaccines, DNA ,Animals ,Lung ,Mice, Inbred BALB C ,Vaccination ,Virology ,Tolerance induction ,Animals, Newborn ,Influenza Vaccines ,cytotoxicity ,lcsh:RC581-607 ,Spleen ,Developmental Biology ,Research Article ,Plasmids ,T-Lymphocytes, Cytotoxic - Abstract
Neonate organisms display an intrinsic disability to mount effective immune responses to infectious agents or conventional vaccines. Whereas low. doses of antigens trigger a suboptimal response, higher doses are frequently associated with tolerance induction. We investigated the ability of a plasmid-expressing nucleoprotein of influenza virus to prime a specific cellular immune response when administered to newborn mice. We found that persistent exposure to antigen following plasmid inoculation of neonates leads to a vigorous priming of specific CTLs rather than tolerance induction. The CTLs were cross-reactive against multiple strains of type A influenza viruses and produced IFNγbut no IL-4. The immunity triggered by plasmid inoculation of neonates was protective in terms of pulmonary virus clearance as well as survival rate following lethal challenge with influenza virus. Whereas the persistence of the plasmid at the site of injection was readily demonstrable in adult mice at 3 months after inoculation, mice immunized as newborns displayed no plasmid at 3 months and very little at 1 month after injection. Thus, DNA-based immunization of neonates may prove an effective and safe vaccination strategy for induction of cellular immunity against microbes that cause serious infectious diseases in the early period of life.
- Published
- 1998
25. Tetravalent DNA Vaccine Product as a Vaccine Candidate Against Dengue
- Author
-
Nimfa Teneza-Mora, Kanakatte Raviprakash, and Kevin R. Porter
- Subjects
Serotype ,Dna immunization ,Immunity ,business.industry ,medicine ,Disease ,medicine.disease ,business ,Virology ,Arbovirus ,Virus ,DNA vaccination ,Dengue fever - Abstract
Dengue is the most important arbovirus worldwide and is the virus that causes dengue fever and the more severe dengue hemorrhagic fever. There are four serotypes of dengue with each possessing the ability to cause disease. Developing a preventive vaccine is the most efficient and effective way to prevent these diseases, and because immunity to one serotype does not protect against the other serotypes, a vaccine must provide tetravalent protection. We used DNA immunization as a platform to develop a tetravalent vaccine. In this chapter, we describe the laboratory, regulatory, and clinical methodology for evaluating a candidate tetravalent vaccine in a Phase 1 clinical trial.
- Published
- 2014
- Full Text
- View/download PDF
26. DNA Immunization
- Author
-
Shixia Wang and Shan Lu
- Subjects
Research areas ,medicine.drug_class ,030231 tropical medicine ,Monoclonal antibody ,Microbiology ,Article ,Injections ,Gene gun ,DNA vaccination ,03 medical and health sciences ,0302 clinical medicine ,Virology ,Vaccines, DNA ,medicine ,Animals ,Humans ,030212 general & internal medicine ,biology ,Electroporation ,General Medicine ,biochemical phenomena, metabolism, and nutrition ,Biolistics ,Dna immunization ,Immunization ,Polyclonal antibodies ,biology.protein ,bacteria ,Parasitology - Abstract
DNA immunization was discovered in early 1990s, and its use has been expanded from vaccine studies to a broader range of biomedical research areas, such as the generation of high-quality polyclonal and monoclonal antibodies as research reagents. In this unit, three common DNA immunization methods are described: needle injection, electroporation, and gene gun. In addition, several common considerations related to DNA immunization are discussed. Curr. Protoc. Microbiol. 31:18.3.1-18.3.24. ©2013 by John Wiley & Sons, Inc. Keywords: DNA vaccine; immunization; electroporation; gene gun
- Published
- 2013
- Full Text
- View/download PDF
27. Co-expression of Ubiquitin gene and capsid protein gene enhances the potency of DNA immunization of PCV2 in mice
- Author
-
Yuekun Lang, Guoxin Li, Fang Fu, Xi Li, Xuesong Li, Guangzhi Tong, Yuju Yang, and Yan-Jun Zhou
- Subjects
Circovirus ,DNA immunization ,animal diseases ,medicine.medical_treatment ,Immunization, Secondary ,Gene Expression ,Lymphocyte proliferation ,Biology ,Antibodies, Viral ,Virus ,lcsh:Infectious and parasitic diseases ,Cell Line ,Mice ,Plasmid ,Immune system ,Virology ,Vaccines, DNA ,medicine ,Animals ,Humans ,lcsh:RC109-216 ,Viremia ,Cell Proliferation ,Mice, Inbred BALB C ,Microscopy, Confocal ,Ubiquitin ,Research ,Vaccination ,Antibody titer ,Flow Cytometry ,biology.organism_classification ,Cap ,Molecular biology ,Lymphocyte Subsets ,PCV2 ,Porcine circovirus ,Infectious Diseases ,Cytokine ,Immunoglobulin G ,biology.protein ,Cytokines ,Capsid Proteins ,Female ,Antibody ,Plasmids - Abstract
A recombinant plasmid that co-expressed ubiquitin and porcine circovirus type 2 (PCV2) virus capsid protein (Cap), denoted as pc-Ub-Cap, and a plasmid encoding PCV2 virus Cap alone, denoted as pc-Cap, were transfected into 293T cells. Indirect immunofluorescence (IIF) and confocal microscopy were performed to measure the cellular expression of Cap. Three groups of mice were then vaccinated once every three weeks for a total of three doses with pc-Ub-Cap, pc-Cap or the empty vector pCAGGS, followed by challenging all mice intraperitoneally with 0.5 mL 106.5 TCID50/mL PCV2. To characterize the protective immune response against PCV2 infection in mice, assays of antibody titer (including different IgG isotypes), flow cytometric analysis (FCM), lymphocyte proliferation, cytokine production and viremia were evaluated. The results showed that pc-Ub-Cap and pc-Cap were efficiently expressed in 293T cells. However, pc-Ub-Cap-vaccinated animals had a significantly higher level of Cap-specific antibody and induced a stronger Th1 type cellular immune response than did pc-Cap-vaccinated animals, suggesting that ubiquitin conjugation improved both the cellular and humoral immune responses. Additionally, viral replication in blood was lower in the pc-Ub-Cap-vaccinated group than in the pc-Cap and empty vector groups, suggesting that the protective immunity induced by pc-Ub-Cap is superior to that induced by pc-Cap.
- Published
- 2011
- Full Text
- View/download PDF
28. Induction of specific cytotoxic activity for bovine herpesvirus-1 by DNA immunization with different adjuvants
- Author
-
Cecilia Langellotti, C. Mongini, Valeria Quattrocchi, Patricia Ines Zamorano, and Juan Sebastian Pappalardo
- Subjects
Cytotoxicity, Immunologic ,Herpesvirus Vaccines ,Biology ,DNA vaccination ,Mice ,Viral Proteins ,Immune system ,Adjuvants, Immunologic ,Virology ,Vaccines, DNA ,Cytotoxic T cell ,Glycoprotein D ,Animals ,Vector (molecular biology) ,Infectious Bovine Rhinotracheitis ,Herpesvirus 1, Bovine ,Pharmacology ,Immunity, Cellular ,Mice, Inbred BALB C ,Viral Vaccine ,biology.organism_classification ,Bovine herpesvirus 1 ,Immunity, Humoral ,Dna immunization ,Immunology ,Cattle ,Female ,Immunization - Abstract
It is well documented that adjuvants improve the immune response generated by traditional viral vaccines; however, less is known about their effects on the immune response elicited by DNA vaccines. In this study, we have investigated the use of adjuvants, and have analyzed the humoral and cellular specific immune responses elicited by DNA vaccines based on the BoHV-1 glycoprotein D (secreted version) in pCIneo vector with and without Montanide ISA25 (O/W), ISA206 VG (SEPPIC) and Cliptox™ (natural microparticles of clinoptilolite). The comparison of the immune response induced in mice by pCIgD formulated with or without adjuvants showed that the immunomodulators affect the total specific humoral and cellular response. The isotypes induced by these adjuvants were of the type Th1/Th2. A significant increase in the mac-3+ and F4/80+ populations of the groups receiving pCIneo with ISA25, ISA206; and an increase in CD4+ populations of the group receiving pCIneo ISA25, in comparison with the pCIneo group was observed. On the other hand, mice vaccinated with pCIgD/ISA25, pCIgD/ISA206, or pCIgD/Cliptox developed a significantly higher specific cytotoxic activity against BoHV-1 than the pCIgD and pCIneo groups. In this report we propose the use of ISA25, ISA206 or Cliptox as adjuvants in a DNA vaccine since they are able to induce not only a specific humoral immune response but also a specific cellular immune response.
- Published
- 2011
29. Characterization of murine T-cell epitopes on mycobacterial DNA-binding protein 1 (MDP1) using DNA vaccination
- Author
-
Ghada Eweda, Kunio Tsujimura, Daisuke Suzuki, Makoto Matsumoto, Yukio Koide, Sohkichi Matsumoto, and Toshi Nagata
- Subjects
CD4-Positive T-Lymphocytes ,DNA immunization ,Molecular Sequence Data ,Epitopes, T-Lymphocyte ,CD8-Positive T-Lymphocytes ,Epitope ,DNA vaccination ,Mycobacterium tuberculosis ,Interferon-gamma ,Mice ,Immune system ,Antigen ,Bacterial Proteins ,Interferon ,medicine ,Vaccines, DNA ,Animals ,Amino Acid Sequence ,Tuberculosis Vaccines ,Cells, Cultured ,Antigens, Bacterial ,Mice, Inbred BALB C ,Mice, Inbred C3H ,General Veterinary ,General Immunology and Microbiology ,biology ,Public Health, Environmental and Occupational Health ,biology.organism_classification ,Virology ,Vaccination ,DNA-Binding Proteins ,Mice, Inbred C57BL ,T-cell epitope ,Infectious Diseases ,Molecular Medicine ,Female ,CD8 ,Algorithms ,Spleen ,medicine.drug ,Plasmids - Abstract
Mycobacterial DNA-binding protein 1 (MDP1) is a major protein antigen in mycobacteria and induces protective immunity against Mycobacterium tuberculosis infection in mice. In this study we determined murine T-cell epitopes on MDP1 with MDP1 DNA immunization in mice. We analyzed interferon-gamma production from the MDP1 DNA-immune splenocytes in response to 20-mer overlapping peptides covering MDP1 protein. We identified several CD4+ T-cell epitopes in three inbred mouse strains and one CD8+ T-cell epitope in C57BL/6 mice. These T-cell epitopes would be feasible for analysis of the role of MDP1-specific T-cells in protective immunity and for future vaccine design against M. tuberculosis infection.
- Published
- 2010
30. Vaccine Development: Past, Present and Future
- Author
-
Coenraad F.M. Hendriksen
- Subjects
Dna immunization ,business.industry ,Immunology ,Medicine ,business ,Virology - Published
- 2008
- Full Text
- View/download PDF
31. Genetic vaccine for tuberculosis (pVAXhsp65) primes neonate mice for a strong immune response at the adult stage
- Author
-
Sofia Fernanda Gonçalves Zorzella, Robson Francisco Carvalho, A. C. Pelizon, Julio C. C. Lorenzi, Alexandrina Sartori, Célio Lopes Silva, Arlete Aparecida Martins Coelho-Castelo, Izaíra T. Brandão, Ana Paula Favaro Trombone, Douglas Rodrigues Martins, Universidade Estadual Paulista (Unesp), and Universidade de São Paulo (USP)
- Subjects
drug safety ,heat shock protein 65 ,animal cell ,immunogenicity ,immunomodulation ,bacterial protein ,immune response ,newborn ,Immunology and Allergy ,antibody production ,biology ,Southern blotting ,Th1 cell ,genetic immunization ,Immunogenicity ,Mus ,BCG vaccination ,gene induction ,Mycobacterium leprae ,Vaccination ,Tolerance induction ,glutamine ,cytokine production ,Molecular Medicine ,Antibody ,tuberculosis control ,granulocyte macrophage colony stimulating factor ,Biotechnology ,DNA immunization ,animal experiment ,Immunology ,interleukin 5 ,gentamicin ,interleukin 4 ,DNA vaccination ,reverse transcription polymerase chain reaction ,Immune system ,Antigen ,Animalia ,controlled study ,BCG vaccine ,mouse ,nonhuman ,Research ,Virology ,enzyme linked immunosorbent assay ,Th2 cell ,monoclonal antibody ,biology.protein ,interleukin 12 - Abstract
Submitted by Vitor Silverio Rodrigues (vitorsrodrigues@reitoria.unesp.br) on 2014-05-27T11:22:39Z No. of bitstreams: 0Bitstream added on 2014-05-27T14:29:55Z : No. of bitstreams: 1 2-s2.0-38949193197.pdf: 345078 bytes, checksum: 55ba86a1ab3942619638fa0903a67433 (MD5) Made available in DSpace on 2014-05-27T11:22:39Z (GMT). No. of bitstreams: 0 Previous issue date: 2007-11-29 Background: Vaccination of neonates is generally difficult due to the immaturity of the immune system and consequent higher susceptibility to tolerance induction. Genetic immunization has been described as an alternative to trigger a stronger immune response in neonates, including significant Th1 polarization. In this investigation we analysed the potential use of a genetic vaccine containing the heat shock protein (hsp65) from Mycobacterium leprae (pVAXhsp65) against tuberculosis (TB) in neonate mice. Aspects as antigen production, genomic integration and immunogenicity were evaluated. Methods: Hsp65 message and genomic integration were evaluated by RT-PCR and Southern blot, respectively. Immunogenicity of pVAXhsp65 alone or combined with BCG was analysed by specific induction of antibodies and cytokines, both quantified by ELISA. Results: This DNA vaccine was transcribed by muscular cells of neonate mice without integration into the cellular genome. Even though this vaccine was not strongly immunogenic when entirely administered (three doses) during early animal's life, it was not tolerogenic. In addition, pVAXhsp65 and BCG were equally able to prime newborn mice for a strong and mixed immune response (Th1 + Th2) to pVAXhsp65 boosters administered later, at the adult life. Conclusion: These results suggest that pVAXhsp65 can be safely used as a priming stimulus in neonate animals in prime-boost similar strategies to control TB. However, priming with BCG or pVAXhsp65, directed the ensuing immune response triggered by an heterologous or homologous booster, to a mixed Th1/Th2 pattern of response. Measures as introduction of IL-12 or GM-CSF genes in the vaccine construct or even IL-4 neutralization, are probably required to increase the priming towards Th1 polarization to ensure control of tuberculosis infection. © 2007 Pelizon et al; licensee BioMed Central Ltd. Department of Microbiology and Immunology Biosciences Institute São Paulo State University (UNESP), Botucatu, São Paulo 18618-000 Department of Morphology Biosciences Institute São Paulo State University (UNESP), Botucatu, São Paulo 18618-000 Department of Biochemistry and Immunology University of São Paulo (USP), Ribeirão Preto, São Paulo 14049-900 Department of Microbiology and Immunology Biosciences Institute São Paulo State University (UNESP), Botucatu, São Paulo 18618-000 Department of Morphology Biosciences Institute São Paulo State University (UNESP), Botucatu, São Paulo 18618-000
- Published
- 2007
32. Efficacy of DNA immunization with F and G protein genes of Nipah virus
- Author
-
Hualan Chen, Zhiyun Wen, Xijun Wang, Jin-Ying Ge, Zhigao Bu, Qinghua Wang, and Sen Hu
- Subjects
G protein ,viruses ,Nipah virus ,Biology ,Antibodies, Viral ,General Biochemistry, Genetics and Molecular Biology ,Neutralization ,Mice ,Viral Proteins ,History and Philosophy of Science ,Viral Envelope Proteins ,Vaccines, DNA ,Animals ,Humans ,Gene ,chemistry.chemical_classification ,Henipavirus Infections ,General Neuroscience ,Nipah Virus ,Viral Vaccines ,biochemical phenomena, metabolism, and nutrition ,Virology ,Dna immunization ,chemistry ,Immunization ,Immunoglobulin G ,biology.protein ,Antibody ,Glycoprotein ,Viral Fusion Proteins - Abstract
We investigated the antibody response of DNA immunization with two mammalian codon optimized envelope glycoprotein genes, F and G, of Nipah virus in a mouse model. The results indicated that G gene immunization elicited more significant specific serum IgG response and neutralization antibody response than F gene did, suggesting that the G gene DNA immunization is a potential vaccine strategy against Nipah virus.
- Published
- 2006
33. Efficiency of HPV 16 L1/E7 DNA immunization:Influence of cellular localization and capsid assembly
- Author
-
Martin Müller, Lutz Gissmann, Christoph Leder, Andrea Kern, Jürgen A. Kleinschmidt, Konrad Piuko, and Dirk Kuck
- Subjects
DNA immunization ,Papillomavirus E7 Proteins ,Nuclear Localization Signals ,Enzyme-Linked Immunosorbent Assay ,Biology ,Antibodies, Viral ,Virus ,DNA vaccination ,Mice ,Immune system ,Capsid ,Vaccines, DNA ,Animals ,Humans ,Cellular localization ,Sequence Deletion ,Cell Nucleus ,Human papillomavirus 16 ,Vaccines, Synthetic ,General Veterinary ,General Immunology and Microbiology ,Immunogenicity ,Capsomere ,Public Health, Environmental and Occupational Health ,Viral Vaccines ,Oncogene Proteins, Viral ,Papillomavirus ,Virology ,Protein Structure, Tertiary ,Mice, Inbred C57BL ,Infectious Diseases ,biology.protein ,Molecular Medicine ,Capsid Proteins ,Female ,Antibody ,HeLa Cells ,T-Lymphocytes, Cytotoxic - Abstract
Infections by human papillomaviruses (HPV) are the major cause of uterine cancer in women worldwide. Aiming to develop a combined prophylactic and therapeutic vaccine we have previously demonstrated immunogenicity of chimeric virus-like particles consisting of a C-terminally truncated HPV 16 L1 capsid protein fused to an E7 portion. Here we show that genetic vaccination with a corresponding DNA was inefficient in the induction of a L1-specific prophylactic immune response. DNA immunization with C-terminally truncated HPV 16 L1 genes of different lengths revealed that only short deletions (L1 1–498 ) were tolerated for eliciting a humoral immune response against viral capsids. This correlates with the observation that the C-terminal sequences are critical for nuclear localization, capsomere and capsid assembly. However, only the ability of L1 protein to form capsomeres or capsids showed a direct influence on the outcome of the immune response. C-terminal insertion of 60 amino acids of E7 was tolerated in fusion constructs, whereas insertion of full-length E7 1–98 or shuffled E7 (149 aa) completely abolished the humoral immune response. The L1 1–498 /E7 1–60 fusion construct not only induced L1-specific antibodies but also L1- and E7-specific CTL responses after DNA vaccination.
- Published
- 2006
- Full Text
- View/download PDF
34. Genetic immunization of ducks for production of antibodies specific to Helicobacter pylori UreB in egg yolks
- Author
-
Agnieszka Sirko, Urszula Szczyrk, Aneta Targosz, Bénédicte Ndeboko, Lucyna Cova, Kacper Kazimierczuk, and Tomasz Brzozowski
- Subjects
DNA immunization ,Immunoglobulins ,Enzyme-Linked Immunosorbent Assay ,Biology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Microbiology ,law.invention ,Plasmid ,antigen ,Antigen ,law ,antibody ,IgY ,medicine ,Animals ,Escherichia coli ,urease ,Helicobacter pylori ,biology.organism_classification ,Virology ,Antibodies, Bacterial ,Egg Yolk ,Urease ,Recombinant Proteins ,Ducks ,Immunization ,Polyclonal antibodies ,Recombinant DNA ,biology.protein ,Antibody - Abstract
Following genetic immunization of laying ducks with a plasmid expressing Helicobacter pylori UreB (large subunit of urease), IgY against UreB were obtained from egg yolks. These polyclonal and monospecific IgY antibodies are of higher-titer and specifically recognize recombinant H. pylori urease purified from Escherichia coli. To our knowledge this is the first report describing generation of IgY antibodies directed against antigens of H. pylori by DNA-based immunization.
- Published
- 2005
35. A DNA vaccine expressing the E2 protein of classical swine fever virus elicits T cell responses that can prime for rapid antibody production and confer total protection upon viral challenge
- Author
-
Fernando Rodriguez, Llilianne Ganges, Maritza Barrera, Isabel Blanco, Maria T. Frías, José I. Núñez, and Francisco Sobrino
- Subjects
DNA immunization ,Swine ,T cell ,T-Lymphocytes ,Molecular Sequence Data ,Marker vaccine ,Antibodies, Viral ,Kidney ,Virus ,DNA vaccination ,Classical Swine Fever ,Viral Envelope Proteins ,medicine ,Vaccines, DNA ,Animals ,Amino Acid Sequence ,Viremia ,Neutralizing antibody ,Cell Proliferation ,General Veterinary ,General Immunology and Microbiology ,biology ,Vaccines, Marker ,Public Health, Environmental and Occupational Health ,Viral Vaccines ,biology.organism_classification ,T cell response ,Virology ,Infectious Diseases ,medicine.anatomical_structure ,Immunization ,Classical swine fever ,Classical Swine Fever Virus ,Immunology ,biology.protein ,Molecular Medicine ,CSFV ,Lymph Nodes ,Antibody ,Sequence Alignment ,Spleen - Abstract
Immunization of domestic pigs with a DNA vaccine expressing the complete E2 protein of classical swine fever virus (CSFV) conferred total protection against a severe viral challenge. Immunization with three doses of plasmid pcDNA3.1/E2 elicited a consistent and specific, MHC class II restricted T cell response in the three domestic pigs analyzed, in the absence of detectable anti-CSFV antibodies in serum. Upon challenge specific T cell responses were boosted in the three vaccinated pigs, and a rapid rise in the titers of CSFV neutralizing antibodies was noticed in two of them, which correlated with a total protection. In these two pigs, neither disease symptoms were observed nor was virus detected at any time after CSFV infection. Neutralizing antibody titers were lower in the third vaccine, which developed a mild and transient peak of pyrexia. As expected, similar analyses in three control pigs (injected with the empty vector or PBS) did not reveal the induction of specific T cells or viral antibodies and, upon challenge, animals developed severe symptoms of the disease, including high titers of viremia, hyperthermia and virus spread to different organs. Control pigs developed, also, a marked leucopenia, resulting in SWC3+ (myelomonocytic cells) being the major PBMC population, and a drastic decrease CD3+ T cells. This T cell depletion was prevented in animals immunized with pcDNA3.1/E2. The total protection achieved, in the absence of CSFV antibodies before challenge, supports the relevance in the antiviral response observed of specific T cell responses primed by pcDNA3.1/E2 vaccine, which, upon challenge, led to a rapid induction of neutralizing antibodies. The observation that CSFV antibodies could only be detected in protected animals after viral challenge opens the possibility of exploring the potential of the DNA vaccine approach used to develop marker vaccines against CSF. © 2005 Elsevier Ltd. All rights reserved.
- Published
- 2005
36. A combined bovine herpesvirus 1 gB-gD DNA vaccine induces immune response in mice
- Author
-
Elisabetta Caselli, Dario Di Luca, Alberto Vita, D Salvatori, Michela Boni, and Enzo Cassai
- Subjects
Cellular immunity ,DNA immunization ,Glycoprotein genes ,Immunology ,Cattle Diseases ,Enzyme-Linked Immunosorbent Assay ,Herpesvirus Vaccines ,Biology ,Antibodies, Viral ,Microbiology ,Polymerase Chain Reaction ,DNA vaccination ,Mice ,Viral Proteins ,Plasmid ,Immune system ,Secreted antigens ,Antigen ,Viral Envelope Proteins ,Neutralization Tests ,Vaccines, DNA ,Immunology and Allergy ,Animals ,Vaccines, Combined ,BHV-1 ,Administration, Intranasal ,Herpesvirus 1, Bovine ,Mice, Inbred BALB C ,General Veterinary ,General Medicine ,Herpesviridae Infections ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Virology ,Bovine herpesvirus 1 ,Vaccination ,Infectious Diseases ,DNA, Viral ,biology.protein ,Cattle ,Female ,Immunization ,Antibody ,Plasmids - Abstract
Although DNA vaccines have several advantages over conventional vaccines, antibody production and protection are often not adequate, particularly in single plasmid vaccine formulations. Here we assessed the potential for a combined vaccine based on plasmids encoding the membrane-anchored or secreted forms of bovine herpesvirus type 1 (BHV-1) glycoprotein B and D (gB and gD) to induce neutralizing and cell mediated immune responses in mice. Animals were injected by intramuscular, subcutaneous and intranasal routes. Mice immunized with the combined vaccine containing the secreted forms of BHV-1 glycoproteins developed higher titers of anti-BHV-1 neutralizing antibodies, compared to wild type gB/gD combined plasmids and to single plasmid injected groups. Cellular immunity was also developed in mice immunized with combined vaccines, whereas low or no response were observed in single plasmid injected animals. The data suggest the potential use of this combined vaccine in in vivo trials of calves, in order to evaluate its protective efficacy.
- Published
- 2005
37. Immunization with plasmid DNA encoding the hemagglutinin and the nucleoprotein confers robust protection against a lethal canine distemper virus challenge
- Author
-
Peter Karlskov-Mortensen, Mads Klindt Andersen, Trine Hammer Jensen, Lotte Dahl, Line Hagner Nielsen, T. Fabian Wild, Elisabeth Gottschalck, Robin Buckland, Tove Dannemann Jensen, and Merete Blixenkrone-Møller
- Subjects
DNA immunization ,Paramyxoviridae ,Genes, Viral ,Injections, Intradermal ,viruses ,animal diseases ,CDV ,Antibodies, Viral ,Injections, Intramuscular ,Virus ,Plasmid ,Dogs ,Morbillivirus ,Neutralization Tests ,biology.animal ,medicine ,Vaccines, DNA ,Animals ,Mink ,Distemper ,Mononegavirales ,Antigens, Viral ,Distemper Virus, Canine ,General Veterinary ,General Immunology and Microbiology ,biology ,Canine distemper ,Reverse Transcriptase Polymerase Chain Reaction ,Public Health, Environmental and Occupational Health ,Viral Vaccines ,biology.organism_classification ,medicine.disease ,Virology ,Infectious Diseases ,Hemagglutinins ,Nucleoproteins ,Immunization ,Molecular Medicine ,Female - Abstract
We have investigated the protective effect of immunization of a highly susceptible natural host of canine distemper virus (CDV) with DNA plasmids encoding the viral nucleoprotein (N) and hemagglutinin (H). The combined intradermal and intramuscular routes of immunization elicited high virus-neutralizing serum antibody titres in mink (Mustela vison). To mimic natural exposure, we also conducted challenge infection by horizontal transmission from infected contact animals. Other groups received a lethal challenge infection by administration to the mucosae of the respiratory tract and into the muscle. One of the mink vaccinated with N plasmid alone developed severe disease after challenge. In contrast, vaccination with the H plasmid together with the N plasmid conferred solid protection against disease and we were unable to detect CDV infection in PBMCs or in different tissues after challenge. Our findings show that DNA immunization by the combined intradermal and intramuscular routes can confer solid protective immunity against naturally transmitted morbillivirus infection and disease.
- Published
- 2004
- Full Text
- View/download PDF
38. Induction of Anti-Hepatitis B Virus Immune Responses Through DNA Immunization
- Author
-
Michael Geissler, C.F. Grimm, Robert Weth, Dörte Ortmann, and Hubert E. Blum
- Subjects
Hepatitis B virus ,Dna immunization ,Immune system ,business.industry ,Medicine ,business ,medicine.disease_cause ,Virology - Published
- 2004
- Full Text
- View/download PDF
39. Induction of Humoral and Cellular Immune Responses to Hepatitis Delta Virus Through DNA Immunization in BALB/c Mice
- Author
-
Shih-Jer Hsu, Hui-Lin Wu, Ren-Shiang Lee, Pei-Jer Chen, Shiou-Lin Lin, Li-Rung Huang, and Ding-Shinn Chen
- Subjects
Dna immunization ,Immune system ,biology ,HEPATITIS DELTA ,Immunology ,biology.organism_classification ,Virology ,Virus ,BALB/c - Published
- 2004
- Full Text
- View/download PDF
40. Immune Responses to Hepatitis B Virus Surface and Core Antigens in Mice, Monkeys, and Pigs after Accell® Particle-Mediated DNA Immunization
- Author
-
Deborah H. Fuller, Dennis E. McCabe, James T. Fuller, Joel R. Haynes, and Georg Widera
- Subjects
Hepatitis B virus ,Swine ,Receptors, Antigen, T-Cell, alpha-beta ,Recombinant Fusion Proteins ,Genetic Vectors ,DNA, Recombinant ,Immunization, Secondary ,Hepatitis B virus surface ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Immune system ,Species Specificity ,History and Philosophy of Science ,Antigen ,Animals ,Hepatitis B Vaccines ,Hepatitis B Antibodies ,Mice, Inbred BALB C ,Vaccines, Synthetic ,Hepatitis B Surface Antigens ,General Neuroscience ,Vaccination ,H-2 Antigens ,HIV ,Haplorhini ,Hepatitis B Core Antigens ,Virology ,Core (optical fiber) ,Dna immunization ,Injections, Jet - Published
- 1995
- Full Text
- View/download PDF
41. DNA Immunization as a Means to Generate Antibodies to Proteins
- Author
-
Partha S. Chowdhury
- Subjects
Dna immunization ,biology ,Chemistry ,biology.protein ,Antibody ,Virology - Published
- 2003
- Full Text
- View/download PDF
42. Induction of cytotoxic T lymphocyte activity by immunization with recombinant Semliki Forest virus: indications for cross-priming
- Author
-
Jan Wilschut, Toos Daemen, Anke Huckriede, Jacqueline de Vries, Barry-Lee Waarts, Laura Bungener, Marijke Holtrop, Targeted Gynaecologic Oncology (TARGON), and Translational Immunology Groningen (TRIGR)
- Subjects
viruses ,Genes, MHC Class I ,Mice ,Cricetinae ,Cytotoxic T cell ,Cloning, Molecular ,GENE-EXPRESSION ,DNA IMMUNIZATION ,Vaccines, Synthetic ,Antigen processing ,EXPRESSION SYSTEM ,Flow Cytometry ,Infectious Diseases ,medicine.anatomical_structure ,Molecular Medicine ,alphavirus vector ,Plasmids ,dendritic cell ,T cell ,Antigen presentation ,Bone Marrow Cells ,Biology ,Cross Reactions ,Semliki Forest virus ,Transfection ,Cell Line ,IN-VIVO TRANSFECTION ,Antigen ,medicine ,antigen processing ,ANTIGEN PRESENTATION ,APOPTOTIC CELLS ,Animals ,Humans ,Antigen-presenting cell ,HUMORAL IMMUNE-RESPONSES ,General Veterinary ,General Immunology and Microbiology ,Ubiquitin ,Public Health, Environmental and Occupational Health ,ANTIVIRAL PROTECTION ,EQUINE ENCEPHALITIS-VIRUS ,Viral Vaccines ,Dendritic cell ,Dendritic Cells ,biology.organism_classification ,Virology ,Semliki forest virus ,Chromium Radioisotopes ,Nucleoproteins ,Spleen ,T-Lymphocytes, Cytotoxic - Abstract
For the rational design of vaccines capable of inducing CD8+ T cell responses knowledge of the identity of the antigen-presenting cell (APC) and the mechanism of antigen presentation is very important. Here, we address these issues for alphavirus-based immunization, in particular immunization with recombinant Semliki Forest virus (rSFV). Studies with dendritic cells (DCs) from various origins revealed that rSFV has a very limited capacity to transfect this cell type in vitro. To further investigate in vivo whether rSFV transfects professional antigen-presenting cells directly or whether the antigens reach APCs via a mechanism of cross-priming we compared the immunological effects of three different SFV-constructs encoding the influenza nucleoprotein (NP). These constructs differ in the amount of NP produced per cell or in the stability of the NP, respectively. Induction of cytotoxic T lymphocytes (CTLs) appeared to benefit from a large amount of stable antigen. In contrast, rapid antigen degradation, and thus availability of antigenic peptides in the transfected cell, was found to be disadvantageous. Based on these in vitro and in vivo results, we hypothesize that antigen presentation after SFV-based immunization proceeds via a mechanism in which APCs are not transfected directly but acquire antigen from other transfected cells and present it to CTLs in a process of cross-priming. (C) 2003 Elsevier Ltd. All rights reserved.
- Published
- 2003
43. Enhanced cellular immune response against SIV Gag induced by immunization with DNA vaccines expressing assembly and release-defective SIV Gag proteins
- Author
-
Richard W. Compans, Chinglai Yang, Ling Ye, and Zhigao Bu
- Subjects
CD4-Positive T-Lymphocytes ,DNA immunization ,viruses ,Recombinant Fusion Proteins ,Mutant ,Assembly ,Molecular Sequence Data ,Simian Acquired Immunodeficiency Syndrome ,Gene Products, gag ,Biology ,Protein degradation ,CD8-Positive T-Lymphocytes ,DNA vaccination ,chemistry.chemical_compound ,Mice ,Viral Proteins ,Virology ,Simian immunodeficiency virus (SIV) ,Vaccines, DNA ,Animals ,Amino Acid Sequence ,Gene ,Gag ,Mice, Inbred BALB C ,Immunogenicity ,Virus Assembly ,3C Viral Proteases ,SAIDS Vaccines ,Cellular immune response ,Group-specific antigen ,Cysteine Endopeptidases ,chemistry ,Mutation ,DNA construct ,Female ,Immunization ,Simian Immunodeficiency Virus ,DNA - Abstract
Codon-optimized genes were synthesized for the SIVmac239 Gag, a mutant Gag with mutations in the major homology region, and a chimeric Gag containing a protein destruction signal at the N-terminus of Gag. The mutant and chimeric Gag were expressed at levels comparable to that observed for the wild-type Gag protein but their stability and release into the medium were found to be significantly reduced. Immunization of mice with DNA vectors encoding the mutant or chimeric Gag induced fourfold higher levels of anti-SIV Gag CD4 T cell responses than the DNA vector encoding the wild-type SIV Gag. Moreover, anti-SIV Gag CD8 T cell responses induced by DNA vectors encoding the mutant or chimeric Gag were found to be 5- to 10-fold higher than those induced by the DNA construct for the wild-type Gag. These results indicate that mutations disrupting assembly and/or stability of the SIV Gag protein effectively enhance its immunogenicity when expressed from DNA vaccines.
- Published
- 2003
44. Prospects for an HIV vaccine: conventional approaches and DNA immunization
- Author
-
Mark A. Baumeister, David B. Weiner, and Michael A. Chattergoon
- Subjects
CD4-Positive T-Lymphocytes ,Cytotoxicity, Immunologic ,Cellular immunity ,T-Lymphocytes ,Human immunodeficiency virus (HIV) ,Bioengineering ,HIV Infections ,medicine.disease_cause ,Lymphocyte Activation ,Virus ,Adjuvants, Immunologic ,medicine ,Vaccines, DNA ,Humans ,HIV vaccine ,Molecular Biology ,AIDS Vaccines ,B-Lymphocytes ,biology ,Genetic vaccine ,HIV ,biology.organism_classification ,Virology ,Dna immunization ,Immunization ,Drug Design ,Lentivirus ,Immunology ,Biotechnology - Published
- 2003
45. Immunogenicity of plasmids encoding T and B cell epitopes of foot-and-mouth disease virus (FMDV) in swine
- Author
-
Mildred Foster-Cuevas, Leticia Cedillo-Barrón, Benito Gutiérrez-Castañeda, R. Michael E. Parkhouse, François Lefèvre, Simon Kollnberger, Audra Cook, Institute for Animal Health, University of Oxford [Oxford], Centro de Investigacion y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Unité de recherche Virologie et Immunologie Moléculaires (VIM (UR 0892)), Institut National de la Recherche Agronomique (INRA), and Instituto Gulbenkian de Ciência
- Subjects
Swine ,T-Lymphocytes ,animal diseases ,viruses ,[SDV]Life Sciences [q-bio] ,Enzyme-Linked Immunosorbent Assay ,Antibodies, Viral ,Virus ,Epitope ,Epitopes ,03 medical and health sciences ,Plasmid ,VACCINES ,Animals ,Hypersensitivity, Delayed ,Neutralizing antibody ,Peptide sequence ,030304 developmental biology ,DNA IMMUNIZATION ,B-Lymphocytes ,0303 health sciences ,Aphthovirus ,General Veterinary ,General Immunology and Microbiology ,biology ,030306 microbiology ,Immunogenicity ,Public Health, Environmental and Occupational Health ,Granulocyte-Macrophage Colony-Stimulating Factor ,virus diseases ,DNA-Directed RNA Polymerases ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Virology ,3. Good health ,Infectious Diseases ,FOOT-AND-MOUTH DISEASE ,Foot-and-Mouth Disease Virus ,biology.protein ,Molecular Medicine ,Foot-and-mouth disease virus ,Cell Division ,Plasmids - Abstract
International audience; In this work, we have investigated the immune response in pigs to two recombinant plasmids containing immunodominant neutralizing antibody epitopes of foot-and-mouth disease virus structural protein (VP1) coexpressed with viral non-structural proteins as a source of T cell epitopes. The plasmid pcDNA3.1/3D15 contained a sequence coding for the 3D polymerase upstream of a sequence coding for peptide FMDV15, a peptide derived from VP1, previously shown to stimulate protective immunity to foot-and-mouth disease virus (FMDV), that consisted of the carboxy terminal peptide [VP1200-213] linked by ProProSer to the “loop” peptide [VP1143-160] and terminating in CysGly. The plasmid, pcDNA3.1/2B15 contained a sequence coding for the non-structural protein 2B, and the same FMDV15 peptide sequence. Pigs injected with both constructs showed antibody and T cell responses to 3D and 2B, but not to the FMDV15 peptide. Additionally, delayed type hypersensitivity responses were observed in some cases to both 3D or 2B and to FMDV virus. Finally, no protection was seen against FMDV infection in animals immunized with either of the two FMDV DNA constructs. The additional co-immunization of plasmids encoding for GMCSF did not result in any significant change in the immune responses to the plasmids encoding for FMDV. This work gives some optimism for the construction of a DNA vaccine for FMDV in the future.
- Published
- 2003
- Full Text
- View/download PDF
46. Proteic boost enhances humoral response induced by DNA vaccination with the dnaK gene of Chlamydophila abortus but fails to protect pregnant mice against a virulence challenge
- Author
-
Annie Rodolakis, Olivier Grépinet, Céline Héchard, and Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)
- Subjects
Chlamydophila abortus ,law.invention ,DnaK ,Hsp70 ,0403 veterinary science ,Mice ,Pregnancy ,law ,[SDV.BC.IC]Life Sciences [q-bio]/Cellular Biology/Cell Behavior [q-bio.CB] ,Vaccines, DNA ,Chlamydophila Infections ,0303 health sciences ,Virulence ,biology ,Escherichia coli Proteins ,[SDV.BA]Life Sciences [q-bio]/Animal biology ,04 agricultural and veterinary sciences ,Antibodies, Bacterial ,Isotype ,3. Good health ,[SDV.MP]Life Sciences [q-bio]/Microbiology and Parasitology ,Bacterial Vaccines ,Recombinant DNA ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Female ,[SDV.NEU]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC] ,Antibody ,DNA immunization ,040301 veterinary sciences ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Microbiology ,DNA vaccination ,03 medical and health sciences ,Immune system ,Heat shock protein ,Animals ,HSP70 Heat-Shock Proteins ,General Veterinary ,030306 microbiology ,proteic boost ,Chlamydophila ,[SDV.BBM.BM]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,biology.organism_classification ,[SDV.MP.BAC]Life Sciences [q-bio]/Microbiology and Parasitology/Bacteriology ,Virology ,[SDV.GEN.GA]Life Sciences [q-bio]/Genetics/Animal genetics ,Immunoglobulin G ,Humoral immunity ,biology.protein ,bacteria ,[SDV.SPEE]Life Sciences [q-bio]/Santé publique et épidémiologie - Abstract
International audience; In order to enhance the quantity and the protective properties of the antibodies induced by DNA vaccination with the heat shock protein dnaK gene of Chlamydophila abortus AB7 as well as to elicit an efficient cellular immune response, we vaccinated mice with a DNA prime followed by a boost with the recombinant DnaK protein. In non-pregnant mice, this strategy induced the same predominance of the IgG2a isotype as DNA immunization alone with a substantial increased antibody level. The induced antibodies had no in vitro neutralizing properties on C. abortus infectivity. Moreover, the proteic boost probably failed to elicit an efficient cellular immune response since the pregnant or non-pregnant mice were not protected against the bacterial challenge.
- Published
- 2003
- Full Text
- View/download PDF
47. In vitro simian virus 40 large tumor antigen expression correlates with differential immune responses following DNA immunization
- Author
-
Ronald C. Kennedy, Robert K. Bright, Michael H. Shearer, Devin B. Lowe, James Tarbox, Hyun Seok Kang, and Cynthia A. Jumper
- Subjects
CD4-Positive T-Lymphocytes ,DNA immunization ,Antibodies, Neoplasm ,Antigens, Polyomavirus Transforming ,Simian virus 40 ,CD8-Positive T-Lymphocytes ,Virus ,Mice ,Immune system ,Plasmid ,Antigen ,Virology ,Vaccines, DNA ,Animals ,Antigens, Viral ,Cancer immunology ,Cell Line, Transformed ,Mice, Inbred BALB C ,Simian virus 40 large tumor antigen ,biology ,Molecular biology ,Viral replication ,biology.protein ,Cytokine secretion ,Immunization ,Sarcoma, Experimental ,Antibody - Abstract
Simian virus 40 (SV40) contains an essential protein, large tumor antigen (Tag), which assists in viral replication and causes cell transformation and immortalization. Our laboratory has examined plasmid DNA, expressing SV40 Tag under two different promoters, for use in potential cancer vaccination strategies. One plasmid, pSV3-neo, failed to induce SV40 Tag antibody, produced a weak cell-mediated response, and only partial protection in murine experimental tumor challenge systems. The second plasmid, pCMV-Tag, induced antibodies to SV40 Tag, produced a robust cell-mediated response, and invoked complete tumor immunity in vivo. The induction of CD4+ and CD8+ T cell responses following plasmid DNA immunization and tumor cell challenge reflected a type 1 cytokine secretion profile. Our hypothesis for this differential immune response is that pCMV-Tag exhibits a higher level of transgene expression due to a more efficient promoter. We determined that pCMV-Tag levels of SV40 Tag mRNA and protein expression were higher when compared to pSV3-neo. A threshold amount of SV40 Tag may be required to stimulate antibody production and provide complete systemic tumor immunity.
- Published
- 2002
48. DNA immunization--a new chance in vaccine research?
- Author
-
Andreas Henke
- Subjects
Microbiology (medical) ,medicine.medical_specialty ,Immunology ,Antibodies, Viral ,DNA vaccination ,chemistry.chemical_compound ,Mice ,Medical microbiology ,Immune system ,Immunity ,medicine ,Vaccines, DNA ,Immunology and Allergy ,Animals ,Humans ,Immunity, Cellular ,biology ,Models, Genetic ,Research ,Viral Vaccines ,General Medicine ,Virology ,Dna immunization ,Immunization ,chemistry ,Virus Diseases ,Liposomes ,biology.protein ,Antibody ,DNA ,Plasmids - Abstract
A novel class of vaccines, based on the immunization with "naked" DNA, may hold the promise of protecting against human disease without the disadvantages associated with vaccines presently used, and may help to prevent infections which are not curable today. Direct intramuscular or intradermal inoculation of plasmid DNA encoding sequences of viral proteins results in the synthesis of these proteins, causing humoral and/or cellular immune responses in the recipient. Several advantages are associated with DNA immunization, e.g., cheap to produce, heat stability, amenable to genetic manipulation, mimic viral infection, and no risk of reversion to pathogenicity. Nevertheless, some concerns remain regarding their safety, e.g., the possible integration of plasmid DNA into host chromosomes. In summary, the results concerning the efficiency of DNA vaccination demonstrate clearly that these new vaccines may have a promising future in human immunization.
- Published
- 2002
49. Additives and Protein-DNA Combinations Modulate the Humoral Immune Response Elicited by a Hepatitis C Virus Core-encoding Plasmid in Mice
- Author
-
Thelvia Ramos, Liz Alvarez-Lajonchere, Santiago Dueñas-Carrera, Juan Morales, Ariel Viña, and Dagmara Pichardo
- Subjects
Microbiology (medical) ,Viral Hepatitis Vaccines ,hepatitis C virus ,DNA immunization ,lcsh:Arctic medicine. Tropical medicine ,lcsh:RC955-962 ,medicine.medical_treatment ,lcsh:QR1-502 ,Enzyme-Linked Immunosorbent Assay ,Hepacivirus ,lcsh:Microbiology ,Immunoglobulin G ,law.invention ,Mice ,Plasmid ,Immune system ,law ,PEG ratio ,medicine ,Vaccines, DNA ,Animals ,Mice, Inbred BALB C ,Expression vector ,biology ,Viral Core Proteins ,core ,Hepatitis C Antibodies ,Virology ,Molecular biology ,Recombinant Proteins ,Antibody Formation ,Recombinant DNA ,biology.protein ,Female ,Antibody ,Adjuvant ,Plasmids - Abstract
Humoral and cellular immune responses are currently induced against hepatitis C virus (HCV) core following vaccination with core-encoding plasmids. However, the anti-core antibody response is frequently weak or transient. In this paper, we evaluated the effect of different additives and DNA-protein combinations on the anti-core antibody response. BALB/c mice were intramuscularly injected with an expression plasmid (pIDKCo), encoding a C-terminal truncated variant of the HCV core protein, alone or combined with CaCl2, PEG 6000, Freund's adjuvant, sonicated calf thymus DNA and a recombinant core protein (Co. 120). Mixture of pIDKCo with PEG 6000 and Freund's adjuvant accelerated the development of the anti-core Ab response. Combination with PEG 6000 also induced a bias to IgG2a subclass predominance among anti-core antibodies. The kinetics, IgG2a/IgG1 ratio and epitope specificity of the anti-core antibody response elicited by Co. 120 alone or combined with pIDKCo was different regarding that induced by the pIDKCo alone. Our data indicate that the antibody response induced following DNA immunization can be modified by formulation strategies.
- Published
- 2002
50. Immunotherapeutic Approach to Cancer with Cutaneous DNA Vaccination
- Author
-
William G. Hawkins, Wilbur B. Bowne, and Jonathan J. Lewis
- Subjects
chemistry.chemical_compound ,Molecular Immunology ,Dna immunization ,Plasmid dna ,Chemistry ,Gold particles ,medicine ,Cancer ,medicine.disease ,Virology ,DNA ,Gene gun ,DNA vaccination - Abstract
Innovations for the development of cancer vaccines are emerging from advances in molecular immunology and cancer biology (1). Of these, DNA-based vaccination has become a powerful and potentially versatile method for eliciting an immune response against cancer. One method for DNA immunization involves the delivery of plasmid DNA by particle bombardment. Originally developed for plant hybridization, this approach has proven to be readily transferable to mammalian applications (2-3). Otherwise known as the gene gun, this method allows for the introduction of exogenous "naked" DNA into skin (4) (Fig. 1). Fig. 1. Photograph of the helium-driven gene gun. This is a hand-held device attached to a high pressure helium line and electrical source which operates the trigger. As shown, plastic bullets containing gold particles coated with plasmid DNA are measured and cut to fit within a cartridge. The cartridge, filled with 12 bullets, is easily placed within the barrel of the gun.
- Published
- 2001
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.