1
|
Demissie EA, Park SY, Moon JH, Lee DY. Comparative Analysis of Codon Optimization Tools: Advancing toward a Multi-Criteria Framework for Synthetic Gene Design. J Microbiol Biotechnol 2025; 35:e2411066. [PMID: 40223268 PMCID: PMC12010093 DOI: 10.4014/jmb.2411.11066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 04/15/2025]
Abstract
Codon optimization is an essential technique in synthetic biology and biopharmaceutical production, enhancing recombinant protein expression by fine-tuning genetic sequences to match the translational machinery and codon usage preferences of specific host organisms. This study presents a comprehensive comparative analysis of widely used codon optimization tools, focusing on their capacity to reflect host-specific codon biases, design principles, and parameters. Industrially relevant target proteins were evaluated in Escherichia coli, Saccharomyces cerevisiae, and CHO cells, uncovering significant variability in sequence design and clustering patterns across tools. Tools such as JCat, OPTIMIZER, ATGme, and GeneOptimizer demonstrated strong alignment with genome-wide and highly expressed gene-level codon usage, achieving high codon adaptation index (CAI) values and efficient codon-pair utilization. Conversely, tools like TISIGNER and IDT employed different optimization strategies that frequently produced divergent results. Other key parameters, including GC content, mRNA secondary structure stability (ΔG), and codon-pair bias (CPB), were analyzed to elucidate their influence on translational efficiency. While increased GC content enhanced mRNA stability in E. coli, A/T-rich codons in S. cerevisiae minimized secondary structure formation, and moderate GC content in CHO cells balanced mRNA stability and translation efficiency. Our findings highlight the limitations of single-metric approaches and advocate for a multi-criteria framework that integrates CAI, GC content, mRNA folding energy, and codon-pair considerations. This integrative strategy enables the design of tailored genetic sequences that meet host-specific requirements, advancing synthetic gene design for biotechnological innovation and precision biopharmaceutical applications.
Collapse
Affiliation(s)
- Eden A. Demissie
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Seo-Young Park
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
- School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Je Hun Moon
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Dong-Yup Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
2
|
Sen A, Singh V, Ashique S, Jagriti, Biswas S, Islam A, Ehsan I, Mojgani N. DNA and mRNA vaccines: Significant therapeutic approach against cancer management. Adv Immunol 2024; 165:1-36. [PMID: 40449971 DOI: 10.1016/bs.ai.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2025]
Abstract
Cancer's complex nature and personal variety make it among the toughest cancers to conquer. Innovative treatment strategies can be achieved through new biotechnology developments. DNA and mRNA vaccinations deliver an opportunity to take a new path for cancer. The ways in which DNA and mRNA vaccinations generate immune reactions that specifically focus on cancer cells are discussed in this section. This chapter focuses on the development and creation of these vaccines. We will focus on the latest research that proves the effectiveness of these vaccines and their safety over different types of cancer. Also, we discuss the technological and biological barriers in the process of vaccine development that hinder the development of these vaccines, such as the stability of delivery methods and a patient-specific design for vaccines. DNA and mRNA vaccinations are an important therapeutic approach against cancer with genetic information. They offer an opportunity for the future to develop tailored as well as more efficient treatment options.
Collapse
Affiliation(s)
- Aniruddha Sen
- Department of Biochemistry, All India Institute of Medical Sciences, Gorakhpur, Uttar Pradesh, India.
| | - Vijay Singh
- Department of Biochemistry, All India Institute of Medical Sciences, Gorakhpur, Uttar Pradesh, India.
| | - Sumel Ashique
- Department of Pharmaceutical Technology, Bharat Technology, Uluberia, West Bengal, India.
| | - Jagriti
- Department of Biochemistry, All India Institute of Medical Sciences, Gorakhpur, Uttar Pradesh, India.
| | - Sombuddha Biswas
- Department of Preventive and Social Medicine (Public Health), JIPMER Puducherry, India.
| | - Anas Islam
- Faculty of Pharmacy, Integral University, Lucknow, Uttar Pradesh, India.
| | - Iman Ehsan
- Research Associate, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India.
| | - Naheed Mojgani
- Biotechnology Department, Razi vaccine and serum Research Institute, Agricultural Research, Education and Extension Organization, Karaj, Iran.
| |
Collapse
|
3
|
Yang M, Liu J, Yang W, Li Z, Hai Y, Duan B, Zhang H, Yang X, Xia C. Analysis of codon usage patterns in 48 Aconitum species. BMC Genomics 2023; 24:703. [PMID: 37993787 PMCID: PMC10664653 DOI: 10.1186/s12864-023-09650-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 09/05/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND The Aconitum genus is a crucial member of the Ranunculaceae family. There are 350 Aconitum species worldwide, with about 170 species found in China. These species are known for their various pharmacological effects and are commonly used to treat joint pain, cold abdominal pain, and other ailments. Codon usage bias (CUB) analysis contributes to evolutionary relationships and phylogeny. Based on protein-coding sequences (PCGs), we selected 48 species of Aconitum for CUB analysis. RESULTS The results revealed that Aconitum species had less than 50% GC content. Furthermore, the distribution of GC content was irregular and followed a trend of GC1 > GC2 > GC3, indicating a bias towards A/T bases. The relative synonymous codon usage (RSCU) heat map revealed the presence of conservative codons with slight variations within the genus. The effective number of codons (ENC)-Plot and the parity rule 2 (PR2)-bias plot analysis indicate that natural selection is the primary factor influencing the variation in codon usage. As a result, we screened various optimal codons and found that A/T bases were preferred as the last codon. Furthermore, our Maximum Likelihood (ML) analysis based on PCGs among 48 Aconitum species yielded results consistent with those obtained from complete chloroplast (cp.) genome data. This suggests that analyzing mutation in PCGs is an efficient method for demonstrating the phylogeny of species at the genus level. CONCLUSIONS The CUB analysis of 48 species of Aconitum was mainly influenced by natural selection. This study reveals the CUB pattern of Aconitum and lays the foundation for future genetic modification and phylogenetic analyses.
Collapse
Affiliation(s)
- Meihua Yang
- College of Pharmaceutical Science, Dali University, Dali, Yunnan, 671000, China
- Key Laboratory of Yunnan Provincial Higher Education Institutions for Development of Yunnan Daodi Medicinal Materials Resources, Dali, Yunnan, 671000, China
| | - Jiahao Liu
- College of Pharmaceutical Science, Dali University, Dali, Yunnan, 671000, China
- Key Laboratory of Yunnan Provincial Higher Education Institutions for Development of Yunnan Daodi Medicinal Materials Resources, Dali, Yunnan, 671000, China
| | - Wanqing Yang
- College of Pharmaceutical Science, Dali University, Dali, Yunnan, 671000, China
- Key Laboratory of Yunnan Provincial Higher Education Institutions for Development of Yunnan Daodi Medicinal Materials Resources, Dali, Yunnan, 671000, China
| | - Zhen Li
- College of Pharmaceutical Science, Dali University, Dali, Yunnan, 671000, China
- Key Laboratory of Yunnan Provincial Higher Education Institutions for Development of Yunnan Daodi Medicinal Materials Resources, Dali, Yunnan, 671000, China
| | - Yonglin Hai
- College of Pharmaceutical Science, Dali University, Dali, Yunnan, 671000, China
- Key Laboratory of Yunnan Provincial Higher Education Institutions for Development of Yunnan Daodi Medicinal Materials Resources, Dali, Yunnan, 671000, China
| | - Baozhong Duan
- College of Pharmaceutical Science, Dali University, Dali, Yunnan, 671000, China
- Key Laboratory of Yunnan Provincial Higher Education Institutions for Development of Yunnan Daodi Medicinal Materials Resources, Dali, Yunnan, 671000, China
| | - Haizhu Zhang
- College of Pharmaceutical Science, Dali University, Dali, Yunnan, 671000, China
- Western Yunnan Traditional Chinese Medicine and Ethnic Drug Engineering Center, Dali, Yunnan, 671000, China
| | - Xiaoli Yang
- College of Pharmaceutical Science, Dali University, Dali, Yunnan, 671000, China.
- Key Laboratory of Yunnan Provincial Higher Education Institutions for Development of Yunnan Daodi Medicinal Materials Resources, Dali, Yunnan, 671000, China.
| | - Conglong Xia
- College of Pharmaceutical Science, Dali University, Dali, Yunnan, 671000, China.
- Key Laboratory of Yunnan Provincial Higher Education Institutions for Development of Yunnan Daodi Medicinal Materials Resources, Dali, Yunnan, 671000, China.
| |
Collapse
|
4
|
Li Y, Hu X, Xiao M, Huang J, Lou Y, Hu F, Fu X, Li Y, He H, Cheng J. An analysis of codon utilization patterns in the chloroplast genomes of three species of Coffea. BMC Genom Data 2023; 24:42. [PMID: 37558997 PMCID: PMC10413492 DOI: 10.1186/s12863-023-01143-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 07/28/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND The chloroplast genome of plants is known for its small size and low mutation and recombination rates, making it a valuable tool in plant phylogeny, molecular evolution, and population genetics studies. Codon usage bias, an important evolutionary feature, provides insights into species evolution, gene function, and the expression of exogenous genes. Coffee, a key crop in the global tropical agricultural economy, trade, and daily life, warrants investigation into its codon usage bias to guide future research, including the selection of efficient heterologous expression systems for coffee genetic transformation. RESULTS Analysis of the codon utilization patterns in the chloroplast genomes of three Coffea species revealed a high degree of similarity among them. All three species exhibited similar base compositions, with high A/T content and low G/C content and a preference for A/T-ending codons. Among the 30 high-frequency codons identified, 96.67% had A/T endings. Fourteen codons were identified as ideal. Multiple mechanisms, including natural selection, were found to influence the codon usage patterns in the three coffee species, as indicated by ENc-GC3s mapping, PR2 analysis, and neutral analysis. Nicotiana tabacum and Saccharomyces cerevisiae have potential value as the heterologous expression host for three species of coffee genes. CONCLUSION This study highlights the remarkable similarity in codon usage patterns among the three coffee genomes, primarily driven by natural selection. Understanding the gene expression characteristics of coffee and elucidating the laws governing its genetic evolution are facilitated by investigating the codon preferences in these species. The findings can enhance the efficacy of exogenous gene expression and serve as a basis for future studies on coffee evolution.
Collapse
Affiliation(s)
- Yaqi Li
- Institute of Tropical and Subtropical Cash Crops, Yunnan Academy of Agricultural Sciences, Baoshan, Yunnan, China
| | - Xiang Hu
- Institute of Tropical Eco-Agricultural, Yunnan Academy of Agricultural Sciences, Yuanmou, Yunnan, China
| | - Mingkun Xiao
- Institute of Tropical and Subtropical Cash Crops, Yunnan Academy of Agricultural Sciences, Baoshan, Yunnan, China
| | - Jiaxiong Huang
- Institute of Tropical and Subtropical Cash Crops, Yunnan Academy of Agricultural Sciences, Baoshan, Yunnan, China
| | - Yuqiang Lou
- Institute of Tropical and Subtropical Cash Crops, Yunnan Academy of Agricultural Sciences, Baoshan, Yunnan, China
| | - Faguang Hu
- Institute of Tropical and Subtropical Cash Crops, Yunnan Academy of Agricultural Sciences, Baoshan, Yunnan, China
| | - Xingfei Fu
- Institute of Tropical and Subtropical Cash Crops, Yunnan Academy of Agricultural Sciences, Baoshan, Yunnan, China
| | - Yanan Li
- Institute of Tropical and Subtropical Cash Crops, Yunnan Academy of Agricultural Sciences, Baoshan, Yunnan, China
| | - Hongyan He
- Institute of Tropical and Subtropical Cash Crops, Yunnan Academy of Agricultural Sciences, Baoshan, Yunnan, China
- Yunnan Academy of Agricultural Engineering, Kunming, Yunnan, China
| | - Jinhuan Cheng
- Institute of Tropical and Subtropical Cash Crops, Yunnan Academy of Agricultural Sciences, Baoshan, Yunnan, China.
- Yunnan Academy of Agricultural Engineering, Kunming, Yunnan, China.
| |
Collapse
|
5
|
Gao Y, Lu Y, Song Y, Jing L. Analysis of codon usage bias of WRKY transcription factors in Helianthus annuus. BMC Genom Data 2022; 23:46. [PMID: 35725374 PMCID: PMC9210703 DOI: 10.1186/s12863-022-01064-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 06/13/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The phenomenon of codon usage bias is known to exist in many genomes and is mainly determined by mutation and selection. Codon usage bias analysis is a suitable strategy for identifying the principal evolutionary driving forces in different organisms. Sunflower (Helianthus annuus L.) is an annual crop that is cultivated worldwide as ornamentals, food plants and for their valuable oil. The WRKY family genes in plants play a central role in diverse regulation and multiple stress responses. Evolutionary analysis of WRKY family genes of H. annuus can provide rich genetic information for developing hybridization resources of the genus Helianthus.
Results
Bases composition analysis showed the average GC content of WRKY genes of H. annuus was 43.42%, and the average GC3 content was 39.60%, suggesting that WRKY gene family prefers A/T(U) ending codons. There were 29 codons with relative synonymous codon usage (RSCU) greater than 1 and 22 codons ending with A and U base. The effective number of codons (ENC) and codon adaptation index (CAI) in WRKY genes ranged from 43.47–61.00 and 0.14–0.26, suggesting that the codon bias was weak and WRKY genes expression level was low. Neutrality analysis found a significant correlation between GC12 and GC3. ENC-plot showed most genes on or close to the expected curve, suggesting that mutational bias played a major role in shaping codon usage. The Parity Rule 2 plot (PR2) analysis showed that the usage of AT and GC was disproportionate. A total of three codons were identified as the optimal codons.
Conclusion
Apart from natural selection effects, most of the genetic evolution in the H. annuus WRKY genome might be driven by mutation pressure. Our results provide a theoretical foundation for elaborating the genetic architecture and mechanisms of H. annuus and contributing to enrich H. annuus genetic resources.
Collapse
|
6
|
Wang Y, Li J, Zhang L, Sun HX, Zhang Z, Xu J, Xu Y, Lin Y, Zhu A, Luo Y, Zhou H, Wu Y, Lin S, Sun Y, Xiao F, Chen R, Wen L, Chen W, Li F, Ou R, Zhang Y, Kuo T, Li Y, Li L, Sun J, Sun K, Zhuang Z, Lu H, Chen Z, Mai G, Zhuo J, Qian P, Chen J, Yang H, Wang J, Xu X, Zhong N, Zhao J, Li J, Zhao J, Jin X. Plasma cell-free RNA characteristics in COVID-19 patients. Genome Res 2022; 32:228-241. [PMID: 35064006 PMCID: PMC8805721 DOI: 10.1101/gr.276175.121] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/21/2021] [Indexed: 12/15/2022]
Abstract
The pathogenesis of COVID-19 is still elusive, which impedes disease progression prediction, differential diagnosis, and targeted therapy. Plasma cell-free RNAs (cfRNAs) carry unique information from human tissue and thus could point to resourceful solutions for pathogenesis and host-pathogen interactions. Here, we performed a comparative analysis of cfRNA profiles between COVID-19 patients and healthy donors using serial plasma. Analyses of the cfRNA landscape, potential gene regulatory mechanisms, dynamic changes in tRNA pools upon infection, and microbial communities were performed. A total of 380 cfRNA molecules were up-regulated in all COVID-19 patients, of which seven could serve as potential biomarkers (AUC > 0.85) with great sensitivity and specificity. Antiviral (NFKB1A, IFITM3, and IFI27) and neutrophil activation (S100A8, CD68, and CD63)–related genes exhibited decreased expression levels during treatment in COVID-19 patients, which is in accordance with the dynamically enhanced inflammatory response in COVID-19 patients. Noncoding RNAs, including some microRNAs (let 7 family) and long noncoding RNAs (GJA9-MYCBP) targeting interleukin (IL6/IL6R), were differentially expressed between COVID-19 patients and healthy donors, which accounts for the potential core mechanism of cytokine storm syndromes; the tRNA pools change significantly between the COVID-19 and healthy group, leading to the accumulation of SARS-CoV-2 biased codons, which facilitate SARS-CoV-2 replication. Finally, several pneumonia-related microorganisms were detected in the plasma of COVID-19 patients, raising the possibility of simultaneously monitoring immune response regulation and microbial communities using cfRNA analysis. This study fills the knowledge gap in the plasma cfRNA landscape of COVID-19 patients and offers insight into the potential mechanisms of cfRNAs to explain COVID-19 pathogenesis.
Collapse
|
7
|
Analysis of Codon Usage Patterns in Giardia duodenalis Based on Transcriptome Data from GiardiaDB. Genes (Basel) 2021; 12:genes12081169. [PMID: 34440343 PMCID: PMC8393687 DOI: 10.3390/genes12081169] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/24/2021] [Accepted: 07/27/2021] [Indexed: 12/03/2022] Open
Abstract
Giardia duodenalis, a flagellated parasitic protozoan, the most common cause of parasite-induced diarrheal diseases worldwide. Codon usage bias (CUB) is an important evolutionary character in most species. However, G. duodenalis CUB remains unclear. Thus, this study analyzes codon usage patterns to assess the restriction factors and obtain useful information in shaping G. duodenalis CUB. The neutrality analysis result indicates that G. duodenalis has a wide GC3 distribution, which significantly correlates with GC12. ENC-plot result—suggesting that most genes were close to the expected curve with only a few strayed away points. This indicates that mutational pressure and natural selection played an important role in the development of CUB. The Parity Rule 2 plot (PR2) result demonstrates that the usage of GC and AT was out of proportion. Interestingly, we identified 26 optimal codons in the G. duodenalis genome, ending with G or C. In addition, GC content, gene expression, and protein size also influence G. duodenalis CUB formation. This study systematically analyzes G. duodenalis codon usage pattern and clarifies the mechanisms of G. duodenalis CUB. These results will be very useful to identify new genes, molecular genetic manipulation, and study of G. duodenalis evolution.
Collapse
|
8
|
Sefidi-Heris Y, Jahangiri A, Mokhtarzadeh A, Shahbazi MA, Khalili S, Baradaran B, Mosafer J, Baghbanzadeh A, Hejazi M, Hashemzaei M, Hamblin MR, Santos HA. Recent progress in the design of DNA vaccines against tuberculosis. Drug Discov Today 2020; 25:S1359-6446(20)30345-7. [PMID: 32927065 DOI: 10.1016/j.drudis.2020.09.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/31/2020] [Accepted: 09/04/2020] [Indexed: 12/12/2022]
Abstract
Current tuberculosis (TB) vaccines have some disadvantages and many efforts have been undertaken to produce effective TB vaccines. As a result of their advantages, DNA vaccines are promising future vaccine candidates. This review focuses on the design and delivery of novel DNA-based vaccines against TB.
Collapse
Affiliation(s)
- Youssof Sefidi-Heris
- Department of Biology, College of Sciences, Shiraz University, 7146713565, Shiraz, Iran
| | - Abolfazl Jahangiri
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, 193955487, Tehran, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, 5166614731, Tabriz, Iran.
| | - Mohammad-Ali Shahbazi
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland; Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, 45139-56184 Zanjan, Iran.
| | - Saeed Khalili
- Department of Biology Sciences, Faculty of Sciences, Shahid Rajaee Teacher Training University, 1678815811, Tehran, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, 5166614731, Tabriz, Iran
| | - Jafar Mosafer
- Research Center of Advanced Technologies in Medicine, Torbat Heydariyeh University of Medical Sciences, 9516915169, Torbat Heydariyeh, Iran; Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, 9196773117, Mashhad, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, 5166614731, Tabriz, Iran
| | - Maryam Hejazi
- Immunology Research Center, Tabriz University of Medical Sciences, 5166614731, Tabriz, Iran
| | - Mahmoud Hashemzaei
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Zabol University of Medical Sciences, 9861615881, Zabol, Iran
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA; Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa.
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland; Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki FI-00014, Finland.
| |
Collapse
|
9
|
Chapman R, Rybicki EP. Use of a Novel Enhanced DNA Vaccine Vector for Preclinical Virus Vaccine Investigation. Vaccines (Basel) 2019; 7:vaccines7020050. [PMID: 31200559 PMCID: PMC6632145 DOI: 10.3390/vaccines7020050] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/07/2019] [Accepted: 06/11/2019] [Indexed: 12/14/2022] Open
Abstract
DNA vaccines are stable, safe, and cost effective to produce and relatively quick and easy to manufacture. However, to date, DNA vaccines have shown relatively poor immunogenicity in humans despite promising preclinical results. Consequently, a number of different approaches have been investigated to improve the immunogenicity of DNA vaccines. These include the use of improved delivery methods, adjuvants, stronger promoters and enhancer elements to increase antigen expression, and codon optimization of the gene of interest. This review describes the creation and use of a DNA vaccine vector containing a porcine circovirus (PCV-1) enhancer element that significantly increases recombinant antigen expression and immunogenicity and allows for dose sparing. A 172 bp region containing the PCV-1 capsid protein promoter (Pcap) and a smaller element (PC; 70 bp) within this were found to be equally effective. DNA vaccines containing the Pcap region expressing various HIV-1 antigens were found to be highly immunogenic in mice, rabbits, and macaques at 4-10-fold lower doses than normally used and to be highly effective in heterologous prime-boost regimens. By lowering the amount of DNA used for immunization, safety concerns over injecting large amounts of DNA into humans can be overcome.
Collapse
Affiliation(s)
- Rosamund Chapman
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town 7925, South Africa.
| | - Edward P Rybicki
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town 7925, South Africa.
- Biopharming Research Unit, Department of Molecular & Cell Biology, University of Cape Town, PB X3 Rondebosch, Cape Town 7701, South Africa.
| |
Collapse
|
10
|
Immunization of mice by the co-administration of codon-optimized HPV16 E7 and lL12 genes against HPV16-associated cervical cancer. Microb Pathog 2019; 132:20-25. [PMID: 31004722 DOI: 10.1016/j.micpath.2019.04.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 04/11/2019] [Accepted: 04/16/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND Various promising procedures have been used to improve the potency of DNA vaccines for the treatment of human papillomavirus type 16 (HPV16) infections. Interleukin-12 (IL12) is a powerful adjuvant that can contribute to T cell-mediated protection against many pathogens, specifically viruses. Considering the important role of T cell-mediated immunity in tumor clearance, the induction of these responses can help control the progression of tumors in animal models. We have demonstrated that the co-administration of codon-optimized E7 (uE7) gene of HPV16 with interleukin-12 is effective in the development of antitumor responses. OBJECTIVES The present study examined the co-administration of codon-optimized HPV16 E7 gene with murine interleukin-12 gene (mIL-12) as a vaccine adjuvant in tumor mice model. MATERIALS AND METHODS C57BL/6 mice were studied for tumor progression after injection of recombinant DNA vaccines. Lactate dehydrogenase (LDH) and IFN-γ were measured to evaluate the activity of cytotoxic T lymphocytes (CTLs). Measurements of tumor volume and MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay were used for assessment of therapeutic antitumor effects of the vaccines. RESULTS Results showed that DNA vaccines, specifically codon-optimized E7/murine interleukin-12 (mIL-12), elicited significant differences in levels of IFN-γ and cytotoxic T lymphocyte (CTLs) responses compared to control groups. Furthermore, higher antitumor response and lower tumor size in the vaccine group was significantly evident compared to control group. CONCLUSION The co-administration of codon-optimized HPV16 E7 gene with IL12 significantly enhances the DNA vaccine potency against HPV16-associated cervical cancer.
Collapse
|
11
|
Tang J, Cai Y, Liang J, Tan Z, Tang X, Zhang C, Cheng L, Zhou J, Wang H, Yam WC, Chen X, Wang H, Chen Z. In vivo electroporation of a codon-optimized BER opt DNA vaccine protects mice from pathogenic Mycobacterium tuberculosis aerosol challenge. Tuberculosis (Edinb) 2018; 113:65-75. [PMID: 30514515 DOI: 10.1016/j.tube.2018.07.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 07/06/2018] [Accepted: 07/08/2018] [Indexed: 12/17/2022]
Abstract
DNA vaccines have been extensively studied as preventative and therapeutic interventions for various infectious diseases such as tuberculosis, HIV/AIDS and influenza. Despite promising progresses made, improving the immunogenicity of DNA vaccine remains a technical challenge for clinical development. In this study, we investigated a tuberculosis DNA vaccine BERopt, which contained a codon-optimized fusion immunogen Ag85B-ESAT-6-Rv2660c for enhanced mammalian cell expression and immunogenicity. BERopt immunization through in vivo electroporation in BALB/c mice induced surprisingly high frequencies of Ag85B tetramer+ CD8+ T cells in peripheral blood and IFN-γ-secreting CD8+ T cells in splenocytes. Meanwhile, the BERopt vaccine-induced long-lasting T cell immunity protected BALB/c mice from high dose viral challenge using a modified vaccinia virus Tiantan strain expressing mature Ag85B protein (MVTT-m85B) and the virulent M. tb H37Rv aerosol challenge. Since the BERopt DNA vaccine does not induce anti-vector immunity, the strong immunogenicity and protective efficacy of this novel DNA vaccine warrant its future development for M. tb prevention and immunotherapy to alleviate the global TB burden.
Collapse
Affiliation(s)
- Jiansong Tang
- AIDS Institute and Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong SAR, PR China; HKU AIDS Institute Shenzhen Research Laboratory and Guangdong Key Laboratory for Emerging Infectious Disease, Shenzhen Third People's Hospital, Guangdong Medical College, Shenzhen, PR China
| | - Yi Cai
- HKU AIDS Institute Shenzhen Research Laboratory and Guangdong Key Laboratory for Emerging Infectious Disease, Shenzhen Third People's Hospital, Guangdong Medical College, Shenzhen, PR China
| | - Jianguo Liang
- AIDS Institute and Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong SAR, PR China
| | - Zhiwu Tan
- AIDS Institute and Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong SAR, PR China
| | - Xian Tang
- HKU AIDS Institute Shenzhen Research Laboratory and Guangdong Key Laboratory for Emerging Infectious Disease, Shenzhen Third People's Hospital, Guangdong Medical College, Shenzhen, PR China
| | - Chi Zhang
- HKU AIDS Institute Shenzhen Research Laboratory and Guangdong Key Laboratory for Emerging Infectious Disease, Shenzhen Third People's Hospital, Guangdong Medical College, Shenzhen, PR China
| | - Lin Cheng
- AIDS Institute and Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong SAR, PR China
| | - Jingying Zhou
- AIDS Institute and Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong SAR, PR China
| | - Haibo Wang
- AIDS Institute and Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong SAR, PR China
| | - Wing-Cheong Yam
- Department of Microbiology, Queen Mary Hospital, The University of Hong Kong, Hong Kong SAR, PR China
| | - Xinchun Chen
- HKU AIDS Institute Shenzhen Research Laboratory and Guangdong Key Laboratory for Emerging Infectious Disease, Shenzhen Third People's Hospital, Guangdong Medical College, Shenzhen, PR China
| | - Hui Wang
- HKU AIDS Institute Shenzhen Research Laboratory and Guangdong Key Laboratory for Emerging Infectious Disease, Shenzhen Third People's Hospital, Guangdong Medical College, Shenzhen, PR China.
| | - Zhiwei Chen
- AIDS Institute and Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong SAR, PR China; HKU AIDS Institute Shenzhen Research Laboratory and Guangdong Key Laboratory for Emerging Infectious Disease, Shenzhen Third People's Hospital, Guangdong Medical College, Shenzhen, PR China.
| |
Collapse
|
12
|
Codon optimization and improved delivery/immunization regimen enhance the immune response against wild-type and drug-resistant HIV-1 reverse transcriptase, preserving its Th2-polarity. Sci Rep 2018; 8:8078. [PMID: 29799015 PMCID: PMC5967322 DOI: 10.1038/s41598-018-26281-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 05/01/2018] [Indexed: 02/06/2023] Open
Abstract
DNA vaccines require a considerable enhancement of immunogenicity. Here, we optimized a prototype DNA vaccine against drug-resistant HIV-1 based on a weak Th2-immunogen, HIV-1 reverse transcriptase (RT). We designed expression-optimized genes encoding inactivated wild-type and drug-resistant RTs (RT-DNAs) and introduced them into mice by intradermal injections followed by electroporation. RT-DNAs were administered as single or double primes with or without cyclic-di-GMP, or as a prime followed by boost with RT-DNA mixed with a luciferase-encoding plasmid (“surrogate challenge”). Repeated primes improved cellular responses and broadened epitope specificity. Addition of cyclic-di-GMP induced a transient increase in IFN-γ production. The strongest anti-RT immune response was achieved in a prime-boost protocol with electroporation by short 100V pulses done using penetrating electrodes. The RT-specific response, dominated by CD4+ T-cells, targeted epitopes at aa 199–220 and aa 528–543. Drug-resistance mutations disrupted the epitope at aa 205–220, while the CTL epitope at aa 202–210 was not affected. Overall, multiparametric optimization of RT strengthened its Th2- performance. A rapid loss of RT/luciferase-expressing cells in the surrogate challenge experiment revealed a lytic potential of anti-RT response. Such lytic CD4+ response would be beneficial for an HIV vaccine due to its comparative insensitivity to immune escape.
Collapse
|
13
|
Hacobian A, Hercher D. Pushing the Right Buttons: Improving Efficacy of Therapeutic DNA Vectors. TISSUE ENGINEERING PART B-REVIEWS 2017; 24:226-239. [PMID: 29264951 DOI: 10.1089/ten.teb.2017.0353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Gene therapy represents a potent therapeutical application for regenerative medicine. So far, viral and nonviral approaches suffer from major drawbacks hindering efficient gene therapeutic applicability: the immunogenicity of viral systems on the one hand, and the low gene transfer efficiency of nonviral systems on the other hand. Therefore, there is a high demand for improvements of therapeutical systems at several levels. This review summarizes different DNA vector modifications to enhance biological efficacy and efficiency of therapeutical vectors, aiming for low toxicity, high specificity, and biological efficacy-the cornerstones for successful translation of gene therapy into the clinic. We aim to provide a step-by-step instruction to optimize their vectors to achieve the desired outcome of gene therapy. Our review provides the means to either construct a potent gene therapeutic vector de novo or to specifically address a bottleneck in the chain of events mandatory for therapeutic success. Although most of the introduced techniques can be translated into different areas, this review primarily addresses improvements for applications in transient gene therapy in the field of tissue engineering.
Collapse
Affiliation(s)
- Ara Hacobian
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Department of Molecular Biology, AUVA Research Center, The Austrian Cluster for Tissue Regeneration , Vienna, Austria
| | - David Hercher
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Department of Molecular Biology, AUVA Research Center, The Austrian Cluster for Tissue Regeneration , Vienna, Austria
| |
Collapse
|
14
|
Lopes A, Vanvarenberg K, Préat V, Vandermeulen G. Codon-Optimized P1A-Encoding DNA Vaccine: Toward a Therapeutic Vaccination against P815 Mastocytoma. MOLECULAR THERAPY-NUCLEIC ACIDS 2017; 8:404-415. [PMID: 28918040 PMCID: PMC5537203 DOI: 10.1016/j.omtn.2017.07.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 07/11/2017] [Accepted: 07/11/2017] [Indexed: 12/20/2022]
Abstract
DNA vaccine can be modified to increase protein production and modulate immune response. To enhance the efficiency of a P815 mastocytoma DNA vaccine, the P1A gene sequence was optimized by substituting specific codons with synonymous ones while modulating the number of CpG motifs. The P815A murine antigen production was increased with codon-optimized plasmids. The number of CpG motifs within the P1A gene sequence modulated the immunogenicity by inducing a local increase in the cytokines involved in innate immunity. After prophylactic immunization with the optimized vaccines, tumor growth was significantly delayed and mice survival was improved. Consistently, a more pronounced intratumoral recruitment of CD8+ T cells and a memory response were observed. Therapeutic vaccination was able to delay tumor growth when the codon-optimized DNA vaccine containing the highest number of CpG motifs was used. Our data demonstrate the therapeutic potential of optimized P1A vaccine against P815 mastocytoma, and they show the dual role played by codon optimization on both protein production and innate immune activation.
Collapse
Affiliation(s)
- Alessandra Lopes
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue E. Mounier 73, B1.73.12, 1200 Brussels, Belgium
| | - Kevin Vanvarenberg
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue E. Mounier 73, B1.73.12, 1200 Brussels, Belgium
| | - Véronique Préat
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue E. Mounier 73, B1.73.12, 1200 Brussels, Belgium.
| | - Gaëlle Vandermeulen
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue E. Mounier 73, B1.73.12, 1200 Brussels, Belgium
| |
Collapse
|
15
|
Martínez O, Bravo Cruz A, Santos S, Ramírez M, Miranda E, Shisler J, Otero M. Vaccination with a codon-optimized A27L-containing plasmid decreases virus replication and dissemination after vaccinia virus challenge. Vaccine 2017. [PMID: 28629922 DOI: 10.1016/j.vaccine.2017.05.091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Smallpox is a disease caused by Variola virus (VARV). Although eradicated by WHO in 1980, the threat of using VARV on a bioterror attack has increased. The current smallpox vaccine ACAM2000, which consists of live vaccinia virus (VACV), causes complications in individuals with a compromised immune system or with previously reported skin diseases. Thus, a safer and efficacious vaccine needs to be developed. Previously, we reported that our virus-free DNA vaccine formulation, a pVAX1 plasmid encoding codon-optimized VACV A27L gene (pA27LOPT) with and without Imiquimod adjuvant, stimulates A27L-specific production of IFN-γ and increases humoral immunity 7days post-vaccination. Here, we investigated the immune response of our novel vaccine by measuring the frequency of splenocytes producing IFN-γ by ELISPOT, the TH1 and TH2 cytokine profiles, and humoral immune responses two weeks post-vaccination, when animals were challenged with VACV. In all assays, the A27-based DNA vaccine conferred protective immune responses. Specifically, two weeks after vaccination, mice were challenged intranasally with vaccinia virus, and viral titers in mouse lungs and ovaries were significantly lower in groups immunized with pA27LOPT and pA27LOPT+Imiquimod. These results demonstrate that our vaccine formulation decreases viral replication and dissemination in a virus-free DNA vaccine platform, and provides an alternative towards a safer an efficacious vaccine.
Collapse
Affiliation(s)
- Osmarie Martínez
- Department of Microbiology and Medical Zoology, University of Puerto Rico, Medical Sciences Campus, School of Medicine, San Juan, PR 00936, United States
| | - Ariana Bravo Cruz
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, IL 61801, United States
| | - Saritza Santos
- Department of Microbiology and Medical Zoology, University of Puerto Rico, Medical Sciences Campus, School of Medicine, San Juan, PR 00936, United States
| | - Maite Ramírez
- Department of Microbiology and Medical Zoology, University of Puerto Rico, Medical Sciences Campus, School of Medicine, San Juan, PR 00936, United States
| | - Eric Miranda
- Department of Microbiology and Medical Zoology, University of Puerto Rico, Medical Sciences Campus, School of Medicine, San Juan, PR 00936, United States
| | - Joanna Shisler
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, IL 61801, United States
| | - Miguel Otero
- Department of Microbiology and Medical Zoology, University of Puerto Rico, Medical Sciences Campus, School of Medicine, San Juan, PR 00936, United States.
| |
Collapse
|
16
|
Huang X, Xu J, Chen L, Wang Y, Gu X, Peng X, Yang G. Analysis of transcriptome data reveals multifactor constraint on codon usage in Taenia multiceps. BMC Genomics 2017; 18:308. [PMID: 28427327 PMCID: PMC5397707 DOI: 10.1186/s12864-017-3704-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 04/12/2017] [Indexed: 12/04/2022] Open
Abstract
Background Codon usage bias (CUB) is an important evolutionary feature in genomes that has been widely observed in many organisms. However, the synonymous codon usage pattern in the genome of T. multiceps remains to be clarified. In this study, we analyzed the codon usage of T. multiceps based on the transcriptome data to reveal the constraint factors and to gain an improved understanding of the mechanisms that shape synonymous CUB. Results Analysis of a total of 8,620 annotated mRNA sequences from T. multiceps indicated only a weak codon bias, with mean GC and GC3 content values of 49.29% and 51.43%, respectively. Our analysis indicated that nucleotide composition, mutational pressure, natural selection, gene expression level, amino acids with grand average of hydropathicity (GRAVY) and aromaticity (Aromo) and the effective selection of amino-acids all contributed to the codon usage in T. multiceps. Among these factors, natural selection was implicated as the major factor affecting the codon usage variation in T. multiceps. The codon usage of ribosome genes was affected mainly by mutations, while the essential genes were affected mainly by selection. In addition, 21codons were identified as “optimal codons”. Overall, the optimal codons were GC-rich (GC:AU, 41:22), and ended with G or C (except CGU). Furthermore, different degrees of variation in codon usage were found between T. multiceps and Escherichia coli, yeast, Homo sapiens. However, little difference was found between T. multiceps and Taenia pisiformis. Conclusions In this study, the codon usage pattern of T. multiceps was analyzed systematically and factors affected CUB were also identified. This is the first study of codon biology in T. multiceps. Understanding the codon usage pattern in T. multiceps can be helpful for the discovery of new genes, molecular genetic engineering and evolutionary studies. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3704-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xing Huang
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.,Chengdu Agricultural College, Chengdu, 611130, China
| | - Jing Xu
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Lin Chen
- Meat-processing Application Key Laboratory of Sichuan Province, College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Yu Wang
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xiaobin Gu
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xuerong Peng
- College of Science, Sichuan Agricultural University, Ya'an, 625014, China
| | - Guangyou Yang
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
17
|
Prados-Rosales R, Carreño L, Cheng T, Blanc C, Weinrick B, Malek A, Lowary TL, Baena A, Joe M, Bai Y, Kalscheuer R, Batista-Gonzalez A, Saavedra NA, Sampedro L, Tomás J, Anguita J, Hung SC, Tripathi A, Xu J, Glatman-Freedman A, Jacobs WR, Chan J, Porcelli SA, Achkar JM, Casadevall A. Enhanced control of Mycobacterium tuberculosis extrapulmonary dissemination in mice by an arabinomannan-protein conjugate vaccine. PLoS Pathog 2017; 13:e1006250. [PMID: 28278283 PMCID: PMC5360349 DOI: 10.1371/journal.ppat.1006250] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/21/2017] [Accepted: 02/17/2017] [Indexed: 12/22/2022] Open
Abstract
Currently there are a dozen or so of new vaccine candidates in clinical trials for prevention of tuberculosis (TB) and each formulation attempts to elicit protection by enhancement of cell-mediated immunity (CMI). In contrast, most approved vaccines against other bacterial pathogens are believed to mediate protection by eliciting antibody responses. However, it has been difficult to apply this formula to TB because of the difficulty in reliably eliciting protective antibodies. Here, we developed capsular polysaccharide conjugates by linking mycobacterial capsular arabinomannan (AM) to either Mtb Ag85b or B. anthracis protective antigen (PA). Further, we studied their immunogenicity by ELISA and AM glycan microarrays and protection efficacy in mice. Immunization with either Abg85b-AM or PA-AM conjugates elicited an AM-specific antibody response in mice. AM binding antibodies stimulated transcriptional changes in Mtb. Sera from AM conjugate immunized mice reacted against a broad spectrum of AM structural variants and specifically recognized arabinan fragments. Conjugate vaccine immunized mice infected with Mtb had lower bacterial numbers in lungs and spleen, and lived longer than control mice. These findings provide additional evidence that humoral immunity can contribute to protection against Mtb. Vaccine design in the TB field has been driven by the imperative of attempting to elicit strong cell-mediated responses. However, in recent decades evidence has accumulated that humoral immunity can protect against many intracellular pathogens through numerous mechanisms. In this work, we demonstrate that immunization with mycobacterial capsular arabinomannan (AM) conjugates elicited responses that contributed to protection against Mtb infection. We developed two different conjugates including capsular AM linked to the Mtb related protein Ag85b or the Mtb unrelated PA from B. anthracis and found that immunization with AM conjugates elicited antibody populations with different specificities. These surface-specific antibodies could directly modify the transcriptional profile and metabolism of mycobacteria. In addition, we observed a prolonged survival and a reduction in bacterial numbers in lungs and spleen in mice immunized with Ag85b-AM conjugates after infection with Mtb and that the presence of AM-binding antibodies was associated with modest prolongation in survival and a marked reduction in mycobacterial dissemination. Finally, we show that AM is antigenically variable and could potentially form the basis for a serological characterization of mycobacteria based on serotypes.
Collapse
Affiliation(s)
- Rafael Prados-Rosales
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx NY, United States of America
- CIC bioGUNE, Bizkaia Technology Park, Derio, Bizkaia, Spain
- * E-mail:
| | - Leandro Carreño
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx NY, United States of America
- Millennium Institute on Immunology and Immunotherapy, Programa Disciplinario de Inmunologia, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Tingting Cheng
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx NY, United States of America
- Department of Medicine, Albert Einstein College of Medicine, Bronx NY, United States of America
| | - Caroline Blanc
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx NY, United States of America
- Department of Medicine, Albert Einstein College of Medicine, Bronx NY, United States of America
| | - Brian Weinrick
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx NY, United States of America
- Howard Hughes Medical Institute, Albert Einstein College of Medicine, Bronx NY, United States of America
| | - Adel Malek
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx NY, United States of America
- Howard Hughes Medical Institute, Albert Einstein College of Medicine, Bronx NY, United States of America
| | - Todd L. Lowary
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Gunning-Lemieux Chemistry Center, Edmonton, Alberta, Canada
| | - Andres Baena
- Grupo de Inmunologia Celular e inmunogenetica, Universidad de Antioquia, Medellin, Colombia
| | - Maju Joe
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Gunning-Lemieux Chemistry Center, Edmonton, Alberta, Canada
| | - Yu Bai
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Gunning-Lemieux Chemistry Center, Edmonton, Alberta, Canada
| | - Rainer Kalscheuer
- Institute for Medical Microbiology and Hospital Hygiene, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - Ana Batista-Gonzalez
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx NY, United States of America
| | - Noemi A. Saavedra
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx NY, United States of America
| | | | - Julen Tomás
- CIC bioGUNE, Bizkaia Technology Park, Derio, Bizkaia, Spain
| | - Juan Anguita
- CIC bioGUNE, Bizkaia Technology Park, Derio, Bizkaia, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Bizkaia, Spain
| | - Shang-Cheng Hung
- Genomics Research Center, Academia Sinica, Section 2, Nankang, Taipei, Taiwan
| | - Ashish Tripathi
- Genomics Research Center, Academia Sinica, Section 2, Nankang, Taipei, Taiwan
| | - Jiayong Xu
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx NY, United States of America
- Department of Medicine, Albert Einstein College of Medicine, Bronx NY, United States of America
| | - Aharona Glatman-Freedman
- Infectious Diseases Unit, Israel Center for Disease Control, Israel Ministry of Health, Tel Hashomer, Israel
- Department of Pediatrics, and Department of Family and Community Medicine, New York Medical College, Valhalla, NY, United States of America
| | - Williams R. Jacobs
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx NY, United States of America
- Howard Hughes Medical Institute, Albert Einstein College of Medicine, Bronx NY, United States of America
| | - John Chan
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx NY, United States of America
- Department of Medicine, Albert Einstein College of Medicine, Bronx NY, United States of America
| | - Steven A. Porcelli
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx NY, United States of America
| | - Jacqueline M. Achkar
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx NY, United States of America
- Department of Medicine, Albert Einstein College of Medicine, Bronx NY, United States of America
| | - Arturo Casadevall
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx NY, United States of America
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
| |
Collapse
|
18
|
Extracellular expression and antiviral activity of a bovine interferon-alpha through codon optimization in Pichia pastoris. Microbiol Res 2016; 191:12-8. [PMID: 27524649 DOI: 10.1016/j.micres.2016.05.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/29/2016] [Accepted: 05/18/2016] [Indexed: 11/23/2022]
Abstract
Interferons (IFNs) are the primary line of defense against infectious agents. In particular, IFN-α is an important antiviral cytokine and has a wide range of immune-modulating functions. Porcine and human IFN-α have been successfully prepared and play important roles in the prevention and therapy of viral diseases. To date, there has been limited applied research on bovine IFN-α. To achieve high-level expression of recombinant bovine IFN-α (bIFN-α) in Pichia pastoris for large-scale application, the bIFN-α gene was optimized and synthesized on the basis of codon bias of P. pastoris. Optimized bIFN-α (opti-bIFN-α) was successfully expressed in P. pastoris and directly secreted into the culture supernatant. The amount of extracellular soluble opti-bIFN-α was observed to be 200μg/mL in a shake flask. Expression efficiency of opti-bIFN-α was found to be about three times that of wild-type bIFN-α when the expression yield was compared at the same copies of the targeted gene. In addition, both the original cultural supernatant and purified opti-bIFN-α showed strong antiviral activity in MDBK cells (2×10(6)AU/mL and 1×10(7)AU/mg, respectively) and IBRS-2 cells (3×10(5)AU/mL and 1.5×10(6)AU/mg, respectively) against a recombinant vesicular stomatitis virus expressing the green fluorescence protein. In this study, we demonstrated high-level extracellular expression of opti-bIFN-α by P. pastoris. To the best of our knowledge, the opti-bIFN-α yield observed in this study is the highest to be reported to date. Our results demonstrated that the extracellular opti-bIFN-α with strong antiviral activity could be easily prepared and purified at a low cost and that it may be a potential biological therapeutic drug against bovine viral infections.
Collapse
|
19
|
Zhang Y, Feng L, Li L, Wang D, Li C, Sun C, Li P, Zheng X, Liu Y, Yang W, Niu X, Zhong N, Chen L. Effects of the fusion design and immunization route on the immunogenicity of Ag85A-Mtb32 in adenoviral vectored tuberculosis vaccine. Hum Vaccin Immunother 2016; 11:1803-13. [PMID: 26076321 DOI: 10.1080/21645515.2015.1042193] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Vaccines containing multiple antigens may induce broader immune responses and provide better protection against Mycobacterium tuberculosis (Mtb) infection as compared to a single antigen. However, strategies for incorporating multiple antigens into a single vector and the immunization routes may affect their immunogenicity. In this study, we utilized recombinant adenovirus type 5 (rAd5) as a model vaccine vector, and Ag85A (Rv3804c) and Mtb32 (Rv0125) as model antigens, to comparatively evaluate the influence of codon usage optimization, signal sequence, fusion linkers, and immunization routes on the immunogenicity of tuberculosis (TB) vaccine containing multiple antigens in C57BL/6 mice. We showed that codon-optimized Ag85A and Mtb32 fused with a GSG linker induced the strongest systemic and pulmonary cell-mediated immune (CMI) responses. Strong CMI responses were characterized by the generation of a robust IFN-γ ELISPOT response as well as antigen-specific CD4(+) T and CD8(+) T cells, which secreted mono-, dual-, or multiple cytokines. We also found that subcutaneous (SC) and intranasal (IN)/oral immunization with this candidate vaccine exhibited the strongest boosting effects for Mycobacterium bovis bacille Calmette-Guérin (BCG)-primed systemic and pulmonary CMI responses, respectively. Our results supported that codon optimized Ag85A and Mtb32 fused with a proper linker and immunized through SC and IN/oral routes can generate the strongest systemic and pulmonary CMI responses in BCG-primed mice, which may be particularly important for the design of TB vaccines containing multiple antigens.
Collapse
Key Words
- APC, Allophycocyanin
- BCG, Mycobacterium bovis bacille Calmette-Guérin
- BSA, bovine serum album
- CMI, cell-mediated immune responses
- DAPI, 4′,6-diamidino-2-phenylindole
- DMSO, Dimethyl sulfoxide
- ELISPOT, Enzyme-linked immune-sorbent spot
- FACS, Fluorescence Activated Cell Sorter
- FBS, fetal bovine serum
- FITC, fluorescein isothiocyanate
- HA tag, hemagglutinin tag
- HEK, human embryo kidney
- ICS, Intracellular cytokine staining
- IFN-γ, interferon gamma
- IL-2, Interleukin 2
- IM, intramuscular
- IN, intranasal
- Mtb, Mycobacterium tuberculosis
- NBT/BCIP, Nitro blue tetrazolium/ 5-Bromo-4-chloro-3-indolyl phosphate
- PBS, Phosphate Buffered Saline
- PCR, polymerase chain reaction
- PE, Phycoerythrin
- PerCP, Peridinin-ChlorophylL-Protein Complex
- RPMI, Roswell Park Memorial Institute
- SC, subcutaneous
- SDS-PAGE, sodium dodecyl sulfate polyacrylamide gel electrophoresis
- SFC, spot-forming cells
- TB, tuberculosis
- TNF-α, tumor necrosis factor α
- fusion strategies
- immunization routes
- immunogenicity
- multiple antigens
- mycobacterium tuberculosis
- rAd5, recombinant adenovirus type 5
- tPA, tissue plasminogen activator
- vp, viral particles
Collapse
Affiliation(s)
- Yiling Zhang
- a State Key Laboratory of Respiratory Diseases; The First Affiliated Hospital of Guangzhou Medical University ; Guangzhou , China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Jia X, Liu S, Zheng H, Li B, Qi Q, Wei L, Zhao T, He J, Sun J. Non-uniqueness of factors constraint on the codon usage in Bombyx mori. BMC Genomics 2015; 16:356. [PMID: 25943559 PMCID: PMC4422305 DOI: 10.1186/s12864-015-1596-z] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 04/28/2015] [Indexed: 11/10/2022] Open
Abstract
Background The analysis of codon usage is a good way to understand the genetic and evolutionary characteristics of an organism. However, there are only a few reports related with the codon usage of the domesticated silkworm, Bombyx mori (B. mori). Hence, the codon usage of B. mori was analyzed here to reveal the constraint factors and it could be helpful to improve the bioreactor based on B. mori. Results A total of 1,097 annotated mRNA sequences from B. mori were analyzed, revealing there is only a weak codon bias. It also shows that the gene expression level is related to the GC content, and the amino acids with higher general average hydropathicity (GRAVY) and aromaticity (Aromo). And the genes on the primary axis are strongly positively correlated with the GC content, and GC3s. Meanwhile, the effective number of codons (ENc) is strongly correlated with codon adaptation index (CAI), gene length, and Aromo values. However, the ENc values are correlated with the second axis, which indicates that the codon usage in B. mori is affected by not only mutation pressure and natural selection, but also nucleotide composition and the gene expression level. It is also associated with Aromo values, and gene length. Additionally, B. mori has a greater relative discrepancy in codon preferences with Drosophila melanogaster (D. melanogaster) or Saccharomyces cerevisiae (S. cerevisiae) than with Arabidopsis thaliana (A. thaliana), Escherichia coli (E. coli), or Caenorhabditis elegans (C. elegans). Conclusions The codon usage bias in B. mori is relatively weak, and many influence factors are found here, such as nucleotide composition, mutation pressure, natural selection, and expression level. Additionally, it is also associated with Aromo values, and gene length. Among them, natural selection might play a major role. Moreover, the “optimal codons” of B. mori are all encoded by G and C, which provides useful information for enhancing the gene expression in B. mori through codon optimization. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1596-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xian Jia
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, and Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Centre, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.
| | - Shuyu Liu
- Guangzhou East Campus Lab Centre, Sun Yat-sen University, Guangzhou, 510006, People's Republic of China.
| | - Hao Zheng
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, and Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Centre, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.
| | - Bo Li
- Innovative Drug Research Centre, Chongqing University, Chongqing, 401331, People's Republic of China.
| | - Qi Qi
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, and Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Centre, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.
| | - Lei Wei
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, and Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Centre, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.
| | - Taiyi Zhao
- Wageningen University, Wageningen, 6708 PG, Netherlands.
| | - Jian He
- Guangzhou East Campus Lab Centre, Sun Yat-sen University, Guangzhou, 510006, People's Republic of China.
| | - Jingchen Sun
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, and Subtropical Sericulture and Mulberry Resources Protection and Safety Engineering Research Centre, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.
| |
Collapse
|
21
|
Martínez O, Miranda E, Ramírez M, Santos S, Rivera C, Vázquez L, Sánchez T, Tremblay RL, Ríos-Olivares E, Otero M. Immunomodulator-based enhancement of anti smallpox immune responses. PLoS One 2015; 10:e0123113. [PMID: 25875833 PMCID: PMC4395221 DOI: 10.1371/journal.pone.0123113] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 02/27/2015] [Indexed: 12/23/2022] Open
Abstract
Background The current live vaccinia virus vaccine used in the prevention of smallpox is contraindicated for millions of immune-compromised individuals. Although vaccination with the current smallpox vaccine produces protective immunity, it might result in mild to serious health complications for some vaccinees. Thus, there is a critical need for the production of a safe virus-free vaccine against smallpox that is available to everyone. For that reason, we investigated the impact of imiquimod and resiquimod (Toll-like receptors agonists), and the codon-usage optimization of the vaccinia virus A27L gene in the enhancement of the immune response, with intent of producing a safe, virus-free DNA vaccine coding for the A27 vaccinia virus protein. Methods We analyzed the cellular-immune response by measuring the IFN-γ production of splenocytes by ELISPOT, the humoral-immune responses measuring total IgG and IgG2a/IgG1 ratios by ELISA, and the TH1 and TH2 cytokine profiles by ELISA, in mice immunized with our vaccine formulation. Results The proposed vaccine formulation enhanced the A27L vaccine-mediated production of IFN-γ on mouse spleens, and increased the humoral immunity with a TH1-biased response. Also, our vaccine induced a TH1 cytokine milieu, which is important against viral infections. Conclusion These results support the efforts to find a new mechanism to enhance an immune response against smallpox, through the implementation of a safe, virus-free DNA vaccination platform.
Collapse
Affiliation(s)
- Osmarie Martínez
- Department of Microbiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| | - Eric Miranda
- Department of Microbiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
- Department of Microbiology Universidad Central del Caribe School of Medicine, Bayamón, Puerto Rico
| | - Maite Ramírez
- Department of Microbiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| | - Saritza Santos
- Department of Microbiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| | - Carlos Rivera
- Department Biology, University of Puerto Rico, Rio Piedras Campus, San Juan, Puerto Rico
| | - Luis Vázquez
- Department Biology, University of Puerto Rico, Rio Piedras Campus, San Juan, Puerto Rico
| | - Tomás Sánchez
- Department Biology, University of Puerto Rico, Rio Piedras Campus, San Juan, Puerto Rico
| | - Raymond L. Tremblay
- Department of Biology, University of Puerto Rico, Humacao, Puerto Rico
- Department of Biology, University of Puerto Rico, Rio Piedras Campus, San Juan, Puerto Rico
- Center for Applied Tropical Ecology and Conservation, University of Puerto Rico, Rio Piedras campus, San Juan, Puerto Rico
| | - Eddy Ríos-Olivares
- Department of Microbiology Universidad Central del Caribe School of Medicine, Bayamón, Puerto Rico
| | - Miguel Otero
- Department of Microbiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
- * E-mail:
| |
Collapse
|
22
|
Kato G, Yamashita K, Kondo H, Hirono I. Protective efficacy and immune responses induced by a DNA vaccine encoding codon-optimized PPA1 against Photobacterium damselae subsp. piscicida in Japanese flounder. Vaccine 2015; 33:1040-5. [DOI: 10.1016/j.vaccine.2015.01.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/02/2015] [Accepted: 01/03/2015] [Indexed: 01/23/2023]
|
23
|
Jones CH, Hakansson AP, Pfeifer BA. Biomaterials at the interface of nano- and micro-scale vector-cellular interactions in genetic vaccine design. J Mater Chem B 2014; 46:8053-8068. [PMID: 29887986 PMCID: PMC5990286 DOI: 10.1039/c4tb01058b] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The development of safe and effective vaccines for the prevention of elusive infectious diseases remains a public health priority. Immunization, characterized by adaptive immune responses to specific antigens, can be raised by an array of delivery vectors. However, current commercial vaccination strategies are predicated on the retooling of archaic technology. This review will discuss current and emerging strategies designed to elicit immune responses in the context of genetic vaccination. Selected strategies at the biomaterial-biological interface will be emphasized to illustrate the potential of coupling both fields towards a common goal.
Collapse
Affiliation(s)
- Charles H Jones
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260-4200, USA
| | - Anders P Hakansson
- Department of Microbiology and Immunology, University at Buffalo, The State University of New York, Buffalo, NY 14260-4200, USA
- The Witebsky Center for Microbial Pathogenesis and Immunology, University at Buffalo, The State University of New York, Buffalo, NY 14260-4200, USA
| | - Blaine A Pfeifer
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260-4200, USA
| |
Collapse
|
24
|
Abstract
INTRODUCTION Tuberculosis (TB) remains a major health problem and novel vaccination regimens are urgently needed. AREAS COVERED DNA vaccines against TB have been tested in various preclinical models and strategies have been developed to increase their immunogenicity in large animal species. DNA vaccines are able to induce a wide variety of immune responses, including CD8(+) T-cell-mediated cytolytic and IFN-γ responses. DNA vaccination may be valuable in heterologous prime-boost strategies with the currently used bacillus Calmette-Guérin (BCG) vaccine. This approach could broaden the antigenic repertoire of BCG and enhance its weak induction of MHC class I-restricted immune responses. EXPERT OPINION DNA vaccines offer a number of advantages over certain other types of vaccines, such as the induction of robust MHC class I-restricted cytotoxic T lymphocyte (CTL), their generic manufacturing platform and their relatively low manufacturing costs. Because of their strong potential for inducing memory responses, DNA vaccines are particularly suited for priming immune responses. Furthermore, DNA vaccine technology may help antigen discovery by facilitating screening of candidate vaccines. Co-administration of BCG with plasmid DNA coding for immunodominant, subdominant and phase-specific antigens, poorly expressed by BCG, may lead to the development of improved TB vaccines.
Collapse
Affiliation(s)
- Nicolas Bruffaerts
- Scientific Institute of Public Health, O.D. CID-Immunology , Engelandstraat 642, Brussels, B1180 , Belgium
| | | | | |
Collapse
|
25
|
Imani Fooladi AA, Bagherpour G, Khoramabadi N, Fallah Mehrabadi J, Mahdavi M, Halabian R, Amin M, Izadi Mobarakeh J, Einollahi B. Cellular immunity survey against urinary tract infection using pVAX/fimH cassette with mammalian and wild type codon usage as a DNA vaccine. Clin Exp Vaccine Res 2014; 3:185-93. [PMID: 25003092 PMCID: PMC4083071 DOI: 10.7774/cevr.2014.3.2.185] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 02/04/2014] [Accepted: 02/15/2014] [Indexed: 12/01/2022] Open
Abstract
Purpose FimH (the adhesion fragment of type 1 fimbriae) is implicated in uropathogenic Escherichia coli (UPEC) attachment to epithelial cells through interaction with mannose. Recently, some studies have found that UPEC can thrive intracellularly causing recurrent urinary tract infection (UTI). Almost all vaccines have been designed to induce antibodies against UPEC. Yet, the humoral immune response is not potent enough to overcome neither the primary UTI nor recurrent infections. However, DNA vaccines offer the possibility of inducing cell mediated immune responses and may be a promising preventive tool. Materials and Methods In this study, we employed two different open reading frames within mammalian (mam) and wild type (wt) codons of fimH gene. Optimized fragments were cloned in pVAX-1. Expression of the protein in COS-7 was confirmed by western blot analysis after assessing pVAX/fimH(mam) and pVAX/fimH(wt). The constructs were injected to BALB/c mice at plantar surface of feet followed by electroporation. Results The mice immunized with both constructs following booster injection with recombinant FimH showed increased interferon-γ and interleukin-12 responses significantly higher than non-immunized ones (p<0.05). The immunized mice were challenged with UPEC and then the number of bacteria recovered from the immunized mice was compared with the non-immunized ones. Decreased colony count in immunized mice along with cytokine responses confirmed the promising immune response by the DNA vaccines developed in this study. Conclusion In conclusion, DNA vaccines of UPEC proteins may confer some levels of protection which can be improved by multiple constructs or boosters.
Collapse
Affiliation(s)
- Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ghasem Bagherpour
- Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Nima Khoramabadi
- Bacteriology Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Jalil Fallah Mehrabadi
- Department of Genetic Engineering, Faculty of Biosciences and Biotechnology, Malekashtar University of Technology, Tehran, Iran
| | - Mehdi Mahdavi
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Raheleh Halabian
- Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohsen Amin
- Department of Drug and Food Control, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Behzad Einollahi
- Nephrology and Urology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
|
27
|
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.
Collapse
Affiliation(s)
- Shixia Wang
- University of Massachusetts Medical School, Worcester, Massachusetts
| | - Shan Lu
- University of Massachusetts Medical School, Worcester, Massachusetts
| |
Collapse
|
28
|
Chen J, Lin M, Li N, Lin L, She F. Therapeutic vaccination with Salmonella-delivered codon-optimized outer inflammatory protein DNA vaccine enhances protection in Helicobacter pylori infected mice. Vaccine 2012; 30:5310-5. [PMID: 22749593 DOI: 10.1016/j.vaccine.2012.06.052] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 05/31/2012] [Accepted: 06/16/2012] [Indexed: 02/07/2023]
Abstract
Vaccination had demonstrated as an alternative way to combat Helicobacter pylori challenge. In the present study, codon-optimized outer inflammatory protein gene (oipA) for Mus species codon usage, the inclusion of optimal Kozak sequence, and modified of GC content was applied to construct a novel DNA construct. The Salmonella-delivered wild type oipA construct (SL7207/poipA) and the Salmonella-delivered codon-optimized oipA construct (SL7207/poipA-opt) were prepared and their therapeutic efficacy was evaluated in H. pylori-infected mice. The codon-optimized oipA construct (poipA-opt) expressed almost six-fold higher protein than that of wild type construct (poipA) as normalized to the β-actin expression in AGS cells. Oral therapeutic immunization with SL7207/poipA-opt significantly eliminated H. pylori colonization in the stomach; and protection was related to a robust Th1/Th2 immune response. Therefore, our results suggested that fine therapeutic efficacy was related to sufficient expression of the antigen. It is supposed that codon-optimized oipA gene improves protein expression and consequently enhances the immunogenicity of DNA vaccine, which resulted in a significant reduction of bacterial loads in H. pylori infected mice. The Salmonella-delivered codon-optimized DNA construct could be a candidate vaccine against H. pylori for the clinical application.
Collapse
Affiliation(s)
- Jiansen Chen
- Department of Hospital Infection Control, Union Hospital, Fujian Medical University, Fuzhou 350001, Fujian, China
| | | | | | | | | |
Collapse
|
29
|
Bagherpour G, Fooladi A, Mehrabadi J, Nourani M, Einollahi B. Evaluation of mammalian codon usage of fimH in DNA vaccine design. Acta Microbiol Immunol Hung 2011; 58:259-71. [PMID: 22207284 DOI: 10.1556/amicr.58.2011.4.2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Uropathogenic Escherichia coli (UPEC) bacteria are the principal cause of urinary tract infections (UTI). Because these bacteria propagate intracellularly, the cellular immune response is an important factor in UTIs. Therefore, we designed a genetic construct to induce a cellular immune response. In order to develop a genetic construct that induces strong cellular immunity against this pathogen, we used the fimH synthetic gene according to mammalian codon usage, and the gene expression was compared with wild type codon usage. Initially, we designed two constructs, pVAX/fimH mam and pVAX/fimH wt, which contain mammalian and wild type codon usage, respectively. The Cos-7 cell line was transfected separately with a complex of pVAX/fimH mam-ExGene 500 poly cationic polymer and pVAX/fimH wt-ExGene 500 poly cationic polymer. Expression of the fimH gene in both constructs in COS7 cells was confirmed by RT-PCR, SDS-PAGE, and Western blotting. Both of the pVAX/fimH cassettes expressed inserted fimH genes (mam and wt) in Cos-7 cells. Our results suggest that codon optimization successfully expressed the fimH gene because the fimH gene with mammalian codon usage is compatible with the eukaryotic expression system. Therefore, mammalian codon usage could be appropriate in a pVAX/fimH construct as a DNA vaccine.
Collapse
Affiliation(s)
- Ghasem Bagherpour
- 1 Baqiyatallah University of Medical Sciences Molecular Biology, Research Center Tehran Iran
| | - Abbas Fooladi
- 2 Baqiyatallah University of Medical Sciences Applied Microbiology Research Center Tehran Iran
| | - Jalil Mehrabadi
- 3 Malekashtar University of Technology Department of Genetic Engineering, Faculty of Biosciences and Biotechnology Tehran Iran
| | - Mohammad Nourani
- 4 Baqiyatallah University of Medical Sciences Chemical Injury Research Center (CIRC) Tehran Iran
| | - Behzad Einollahi
- 5 Baqiyatallah University of Medical Sciences Nephrology and Urology Research Center Tehran Iran
| |
Collapse
|
30
|
Jørgensen LVG, Buchmann K. Cysteine proteases as potential antigens in antiparasitic DNA vaccines. Vaccine 2011; 29:5575-83. [PMID: 21664399 DOI: 10.1016/j.vaccine.2011.05.091] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 05/12/2011] [Accepted: 05/25/2011] [Indexed: 12/17/2022]
Abstract
Cysteine proteases in parasites are potent inducers of vertebrate host immune responses and may under certain circumstances take part in the pathogen's immune evasion strategies. These capacities place these parasite molecules as interesting candidate antigens in antiparasitic vaccines for use in vertebrates. Parasite cysteine proteases are able to skew the Th1/Th2 profile in mammals towards a response which allows sustainable parasite burdens in the host. DNA vaccines are also able to skew the Th1/Th2 profile by different administration techniques and the use of cysteine proteases in these genetic immunizations open perspectives for manipulation of the host immune response towards higher protection.
Collapse
Affiliation(s)
- Louise von Gersdorff Jørgensen
- Laboratory of Aquatic Pathobiology, Department of Veterinary Disease Biology, Faculty of Life Sciences, University of Copenhagen, Denmark.
| | | |
Collapse
|
31
|
Shan S, Jiang Y, Bu Z, Ellis T, Zeng X, Edwards J, Tian G, Li Y, Ge J, Chen H, Fenwick S. Strategies for improving the efficacy of a H6 subtype avian influenza DNA vaccine in chickens. J Virol Methods 2011; 173:220-6. [PMID: 21333689 DOI: 10.1016/j.jviromet.2011.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Revised: 02/10/2011] [Accepted: 02/10/2011] [Indexed: 10/18/2022]
Abstract
A low-pathogenicity avian influenza H6N2 virus was used to investigate approaches to improve DNA vaccine efficacy. The viral hemagglutinin (HA) gene or its chicken biased HA gene, incorporating a Kozak sequence, was cloned into a pCAGGS vector to produce the pCAG-HAk and pCAG-optiHAk constructs. Following two intramuscular injections, the seroconversion rate in vaccinated chickens with 10, 100 or 300 μg pCAG-HAk were 87.5%, 75% and 75%, respectively. The profile of H6 hemagglutination inhibition (HI) antibodies induced by different doses of pCAG-HAk during the 8-week study period was similar. The HI titer rose significantly in the three different dose groups following the booster and reached a plateau 2-3 weeks post-booster. In a single dose vaccination group with 100 μg pCAG-HAk, a maximum seroconversion rate reached 53.3% at 5 weeks post-vaccination. The earliest time of seroconversion appeared two weeks after DNA immunization. Following two electroporation (EP) vaccinations with 100 μg pCAG-HAk, all birds seroconverted and the HI antibody titers were significantly higher than those using intramuscular immunization, suggesting that EP was more efficient than intramuscular delivery of the DNA vaccines. In comparison, chickens immunized with 10 or 100 μg pCAG-optiHAk showed 37.5% and 87.5% seroconversion rates, respectively, at 3 weeks following the booster. The pCAG-HAk was not significantly different from the pCAG-optiHAk in either the seroconversion rate or H6 HI titer, suggesting that the codon-optimized HA DNA vaccine did not achieve significantly better immunogenicity than the pCAG-HAk vaccine.
Collapse
Affiliation(s)
- Songhua Shan
- School of Veterinary and Biomedical Sciences, Murdoch University, Perth, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Hallengärd D, Haller BK, Petersson S, Boberg A, Maltais AK, Isaguliants M, Wahren B, Bråve A. Increased expression and immunogenicity of HIV-1 protease following inactivation of the enzymatic activity. Vaccine 2010; 29:839-48. [PMID: 21109032 DOI: 10.1016/j.vaccine.2010.10.083] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Revised: 10/21/2010] [Accepted: 10/25/2010] [Indexed: 10/18/2022]
Abstract
HIV-1 protease is an important target for anti-HIV therapy but has not received much attention as a vaccine antigen. To investigate the immunogenic properties of HIV-1 protease, we designed DNA plasmids encoding variants of the protease gene. Mutations resulting in enzymatic inactivation (D25N) and resistance to standard antiretroviral drugs (V82F/I84V) were introduced in order to examine the impact of the enzymatic activity on immunogenicity and the possibility to induce immune responses against drug resistant protease, respectively. The enzymatic inactivation of protease resulted in significantly increased in vitro expression as well as in vivo immunogenicity. The inactivated protease was highly immunogenic in both BALB/c and HLA-A0201 transgenic C57Bl/6 mice, and the immunogenicity was retained when the gene was delivered as a part of a multigene HIV-1 DNA vaccine. The drug resistance mutations hampered both the cellular and humoral immune responses, as the mutations also affect both CD4 and CD8 T cell epitopes. Taken together, our data demonstrates the possibility to drastically increase the immunogenicity of HIV-1 protease.
Collapse
Affiliation(s)
- David Hallengärd
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute and Swedish Institute for Infectious Disease Control, Nobels väg 18, 171 82 Solna, Stockholm, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Zhu Y, Lu F, Dai Y, Wang X, Tang J, Zhao S, Zhang C, Zhang H, Lu S, Wang S. Synergistic enhancement of immunogenicity and protection in mice against Schistosoma japonicum with codon optimization and electroporation delivery of SjTPI DNA vaccines. Vaccine 2010; 28:5347-55. [PMID: 20483191 DOI: 10.1016/j.vaccine.2010.05.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Revised: 04/09/2010] [Accepted: 05/04/2010] [Indexed: 11/19/2022]
Abstract
Schistosomiasis is an endemic, zoonotic parasitic disease caused by Schistosoma japonicum that remains a public health concern and an effective vaccine is needed. Triose-phosphate isomerase from S. japonicum is a promising schistosome vaccine antigen shown to be immunogenic when delivered as a DNA vaccine, however, the previous S. japonicum triose-phosphate isomerase (SjTPI) DNA vaccine needs to be further optimized to achieve higher protection. In the current study, codon optimization of SjTPI DNA insert, combined with electroporation but not with the addition of a tPA leader or heat-shock protein in-frame with the SjTPI gene insert, enhanced Th1-type antibody and cytokine production and most significantly, achieved great than 50% reduction of infection against challenge with S. japonicum cercariae, a major milestone in S. japonicum vaccine development. Our results suggest that the combination of a codon optimized vaccine design and an efficient vaccine delivery system can greatly improve the potential of a SjTPI DNA vaccine as a viable schistosome vaccine candidate.
Collapse
Affiliation(s)
- Yinchang Zhu
- Jiangsu Institute of Parasitic Diseases, Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health, 117 Yangxiang Meiyuan, Wuxi 214064, Jiangsu, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Bioinformatics in new generation flavivirus vaccines. J Biomed Biotechnol 2010; 2010:864029. [PMID: 20467477 PMCID: PMC2867002 DOI: 10.1155/2010/864029] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Revised: 12/21/2009] [Accepted: 03/02/2010] [Indexed: 12/22/2022] Open
Abstract
Flavivirus infections are the most prevalent arthropod-borne infections world wide, often causing severe disease especially among children, the elderly, and the immunocompromised. In the absence of effective antiviral treatment, prevention through vaccination would greatly reduce morbidity and mortality associated with flavivirus infections. Despite the success of the empirically developed vaccines against yellow fever virus, Japanese encephalitis virus and tick-borne encephalitis virus, there is an increasing need for a more rational design and development of safe and effective vaccines. Several bioinformatic tools are available to support such rational vaccine design. In doing so, several parameters have to be taken into account, such as safety for the target population, overall immunogenicity of the candidate vaccine, and efficacy and longevity of the immune responses triggered. Examples of how bio-informatics is applied to assist in the rational design and improvements of vaccines, particularly flavivirus vaccines, are presented and discussed.
Collapse
|
35
|
Improved efficacy of a gene optimised adenovirus-based vaccine for venezuelan equine encephalitis virus. Virol J 2009; 6:118. [PMID: 19646224 PMCID: PMC2732613 DOI: 10.1186/1743-422x-6-118] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2009] [Accepted: 07/31/2009] [Indexed: 01/03/2023] Open
Abstract
Background Optimisation of genes has been shown to be beneficial for expression of proteins in a range of applications. Optimisation has increased protein expression levels through improved codon usage of the genes and an increase in levels of messenger RNA. We have applied this to an adenovirus (ad)-based vaccine encoding structural proteins (E3-E2-6K) of Venezuelan equine encephalitis virus (VEEV). Results Following administration of this vaccine to Balb/c mice, an approximately ten-fold increase in antibody response was elicited and increased protective efficacy compared to an ad-based vaccine containing non-optimised genes was observed after challenge. Conclusion This study, in which the utility of optimising genes encoding the structural proteins of VEEV is demonstrated for the first time, informs us that including optimised genes in gene-based vaccines for VEEV is essential to obtain maximum immunogenicity and protective efficacy.
Collapse
|
36
|
Dobaño C, Sedegah M, Rogers WO, Kumar S, Zheng H, Hoffman SL, Doolan DL. Plasmodium: Mammalian codon optimization of malaria plasmid DNA vaccines enhances antibody responses but not T cell responses nor protective immunity. Exp Parasitol 2009; 122:112-23. [DOI: 10.1016/j.exppara.2009.02.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2008] [Revised: 12/27/2008] [Accepted: 02/18/2009] [Indexed: 11/24/2022]
|
37
|
Yao W, Liu S, Qu X, Xiao S, Liu Y, Liu J. Enhanced immune response and protection efficacy of a DNA vaccine constructed by linkage of the Mycobacterium tuberculosis Ag85B-encoding gene with the BVP22-encoding gene. J Med Microbiol 2009; 58:462-468. [DOI: 10.1099/jmm.0.004267-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Plasmid DNA vaccines have been widely explored for use in tuberculosis immunization but their immunogenicity needs improvement. In the present study, we incorporated the bovine herpesvirus 1 VP22 (BVP22)-encoding gene, which encodes a protein that demonstrates a capability for disseminating the expressed antigen to neighbouring cells, into a DNA vector in which it was fused to the Ag85B-encoding gene of Mycobacterium tuberculosis (Mtb), and investigated whether this linkage could enhance immune response and protective efficacy in C57BL/6 mice compared to plasmid DNA encoding Ag85B alone. After immunization in mice, Ag85B-specific ELISA antibodies and spleen lymphocyte proliferative responses induced by DNA co-expressing BVP22 and Ag85B were significantly higher than those obtained in mice immunized with Ag85B-encoding DNA alone, except for the number of gamma interferon secreting cells. In addition, based on histopathological examination and bacterial-load determination in lung and spleen, protection against intravenous Mtb H37Rv challenge evoked by the BVP22–Ag85B DNA immunization exceeded the response elicited by Ag85B DNA alone, which was not significantly different from that provided by Bacillus Calmette–Guérin (BCG). These results suggested that DNA vaccine consisting of BVP22 and Ag85B-encoding DNA enhanced immune response and protection against intravenous Mtb H37Rv challenge in mice, indicating that BVP22-encoding DNA might be a promising tool to enhance TB DNA vaccine efficacy.
Collapse
Affiliation(s)
- Wanhong Yao
- Department of Microbiology and Parasitology, School of Medicine, Wuhan University, Wuhan, PR China
| | - Shengwu Liu
- Department of Immunology, School of Medicine, Wuhan University, Wuhan, PR China
| | - Xueju Qu
- Department of Immunology, School of Medicine, Wuhan University, Wuhan, PR China
| | - Shaobo Xiao
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430071, Hubei, PR China
- Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430071, Hubei, PR China
| | - Yan Liu
- Department of Immunology, School of Medicine, Wuhan University, Wuhan, PR China
| | - Junyan Liu
- Animal Center of Wuhan University, Wuhan University, Wuhan, PR China
- Department of Immunology, School of Medicine, Wuhan University, Wuhan, PR China
| |
Collapse
|
38
|
Liu H, He R, Zhang H, Huang Y, Tian M, Zhang J. Analysis of synonymous codon usage in Zea mays. Mol Biol Rep 2009; 37:677-84. [DOI: 10.1007/s11033-009-9521-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2008] [Accepted: 03/17/2009] [Indexed: 11/29/2022]
|
39
|
Abstract
We have focused our research on understanding the basic biology of and developing novel therapeutic and prophylactic DNA vaccines. We have among others three distinct primary areas of interest which include: 1. Enhancing in vivo delivery and transfection of DNA vaccine vectors 2. Improving DNA vaccine construct immunogenicity 3. Using molecular adjuvants to modulate and skew immune responses. Key to the immunogenicity of DNA vaccines is the presentation of expressed antigen to antigen-presenting cells. To improve expression and presentation of antigen, we have investigated various immunization methods with current focus on a combination of intramuscular injection and electroporation. To improve our vaccine constructs, we also employed methods such as RNA/codon optimization and antigen consensus to enhance expression and cellular/humoral cross-reactivity, respectively. Our lab also researches the potential of various molecular adjuvants to skew Th1/Th2 responses, enhance cellular/humoral responses, and improve protection in various animal models. Through improving our understanding of basic immunology as it is related to DNA vaccine technology, our goal is to develop the technology to the point of utility for human and animal health.
Collapse
Affiliation(s)
- Shaheed A Abdulhaqq
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 422 Curie Blvd, 505 Stellar-Chance Laboratories, Philadelphia, PA, 19104, USA
| | | |
Collapse
|
40
|
Ko HJ, Kim YJ, Kim YS, Chang WS, Ko SY, Chang SY, Sakaguchi S, Kang CY. A combination of chemoimmunotherapies can efficiently break self-tolerance and induce antitumor immunity in a tolerogenic murine tumor model. Cancer Res 2007; 67:7477-86. [PMID: 17671218 DOI: 10.1158/0008-5472.can-06-4639] [Citation(s) in RCA: 159] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Her-2/neu is a well-characterized tumor-associated antigen overexpressed in human carcinomas such as breast cancer. Because Her-2/neu is a self-antigen with poor immunogenicity due to immunologic tolerance, active immunotherapy targeting Her-2/neu should incorporate methods to overcome immunologic tolerance to self-proteins. In this study, we developed a tolerogenic tumor model in mice using mouse Her-2/neu as self-antigen and investigated whether genetic vaccination with DNA plasmid and/or adenoviral vector expressing the extracellular and transmembrane domain of syngeneic mouse Her-2/neu or xenogenic human Her-2/neu could induce mouse Her-2/neu-specific CTL responses. Interestingly, adenoviral vectors expressing xenogenic human Her-2/neu (AdhHM) proved capable of breaking immune tolerance and of thereby inducing self-reactive CTL and antibodies, but not to the degree required to induce therapeutic antitumor immunity. In attempting to generate therapeutic antitumor immunity against established tumors, we adopted several approaches. Treatment with agonistic anti-glucocorticoid-induced TNFR family-related receptor (GITR) antibody plus AdhHM immunization significantly increased self-reactive CTL responses, and alpha-galactosylceramide (alphaGalCer)-loaded dendritic cells (DC) transduced with AdhHM were shown to break self-tolerance in a tolerogenic murine tumor model. Furthermore, gemcitabine treatment together with either AdhHM plus agonistic anti-GITR antibody administration or alphaGalCer-loaded DC transduced with AdhHM showed potent therapeutic antitumor immunity and perfect protection against preexisting tumors. Gemcitabine treatment attenuated the tumor-suppressive environment by eliminating CD11b(+)/Gr-1(+) myeloid-derived suppressor cells. When combined with immunotherapies, gemcitabine offers a promising strategy for the Ag-specific treatment of human cancer.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Antigens, Differentiation/genetics
- Antigens, Differentiation/immunology
- Antigens, Differentiation/metabolism
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Breast Neoplasms/genetics
- Breast Neoplasms/immunology
- Breast Neoplasms/prevention & control
- CD8-Positive T-Lymphocytes/immunology
- CTLA-4 Antigen
- Cell Line, Tumor
- Cell Proliferation
- Colonic Neoplasms/genetics
- Colonic Neoplasms/immunology
- Colonic Neoplasms/prevention & control
- Combined Modality Therapy
- Cyclophosphamide/administration & dosage
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Deoxycytidine/administration & dosage
- Deoxycytidine/analogs & derivatives
- Docetaxel
- Doxorubicin/administration & dosage
- Female
- Flow Cytometry
- Galactosylceramides/immunology
- Genetic Vectors/administration & dosage
- Glucocorticoid-Induced TNFR-Related Protein
- Humans
- Immunization
- Immunotherapy
- Mice
- Myeloid Cells/cytology
- Myeloid Cells/immunology
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- Peptide Fragments/pharmacology
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Receptors, Nerve Growth Factor/genetics
- Receptors, Nerve Growth Factor/metabolism
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Regulatory
- Taxoids/administration & dosage
- Transfection
- Vaccines, DNA/administration & dosage
- Gemcitabine
Collapse
Affiliation(s)
- Hyun-Jeong Ko
- Laboratory of Immunology, Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Kamrud KI, Custer M, Dudek JM, Owens G, Alterson KD, Lee JS, Groebner JL, Smith JF. Alphavirus replicon approach to promoterless analysis of IRES elements. Virology 2007; 360:376-87. [PMID: 17156813 PMCID: PMC1885372 DOI: 10.1016/j.virol.2006.10.049] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2006] [Revised: 08/30/2006] [Accepted: 10/30/2006] [Indexed: 02/05/2023]
Abstract
Here we describe a system for promoterless analysis of putative internal ribosome entry site (IRES) elements using an alphavirus (family Togaviridae) replicon vector. The system uses the alphavirus subgenomic promoter to produce transcripts that, when modified to contain a spacer region upstream of an IRES element, allow analysis of cap-independent translation of genes of interest (GOI). If the IRES element is removed, translation of the subgenomic transcript can be reduced >95% compared to the same transcript containing a functional IRES element. Alphavirus replicons, used in this manner, offer an alternative to standard dicistronic DNA vectors or in vitro translation systems currently used to analyze putative IRES elements. In addition, protein expression levels varied depending on the spacer element located upstream of each IRES. The ability to modulate the level of expression from alphavirus vectors should extend the utility of these vectors in vaccine development.
Collapse
Affiliation(s)
- K I Kamrud
- AlphaVax, Inc., 2 Triangle Drive, Research Triangle Park, NC 27709-0307, USA.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Vaccination with DNA is one of the most promising novel immunization techniques against a variety of pathogens and tumors, for which conventional vaccination regimens have failed. DNA vaccines are able to stimulate both arms of the immune system simultaneously, without carrying the safety risks associated with live vaccines, therefore representing not only an alternative to conventional vaccines but also significant progress in the prevention and treatment of fatal diseases and infections. However, translation of the excellent results achieved in small animals to similar success in primates or large animals has so far proved to be a major hurdle. Moreover, biosafety issues, such as the removal of antibiotic resistance genes present in plasmid DNA used for vaccination, remain to be addressed adequately. This review describes strategies to improve the design and production of conventional plasmid DNA, including an overview of safety and regulatory issues. It further focuses on novel systems for the optimization of plasmid DNA and the development of diverse plasmid DNA delivery systems for vaccination purposes.
Collapse
Affiliation(s)
- Wolfgang Jechlinger
- Institute of Bacteriology, Mycology and Hygiene, Department of Pathobiology, University of Veterinary Medicine, Veterinärplatz 1, A- 1210, Vienna, Austria.
| |
Collapse
|
43
|
Aurisicchio L, Mennuni C, Giannetti P, Calvaruso F, Nuzzo M, Cipriani B, Palombo F, Monaci P, Ciliberto G, La Monica N. Immunogenicity and safety of a DNA prime/adenovirus boost vaccine against rhesus CEA in nonhuman primates. Int J Cancer 2007; 120:2290-300. [PMID: 17304509 DOI: 10.1002/ijc.22555] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Scaling up experimental protocols from rodents to humans is often not a straightforward procedure, and this particularly applies to cancer vaccines, where vaccination technology must be especially effective to overcome a variety of immune suppressive mechanisms. DNA electroporation (DNA-EP) and adenoviral vectors (Ad) have shown high potency and therapeutic efficacy for different antigens in several pre-clinical models. To evaluate the ability of DNA-EP and Ad to break tolerance to a self-antigen in large animals, we have cloned the CEA homologue (rhCEA) from rhesus monkeys (Macaca mulatta) colon tissue samples. rhCEA is a 705 aa protein and shares 78.9% homology to human CEA protein. Immunogenicity of rhCEA expressing vectors was tested in mice and subsequently in rhesus monkeys. To further increase the immunogenic potency of these vectors, a synthetic codon optimized rhCEA cDNA (rhCEAopt) was constructed. Genetic vaccination of rhesus monkeys was effective in breaking immune tolerance to rhCEA in all immunized animals, maintaining over time the elicited immune response, and most importantly, neither autoimmunity nor other side-effects were observed upon treatment. Our data confirm the efficacy of genetic cancer vaccines in large animals such as nonhuman primates and show that development of modified expression cassettes that result in increased potency of plasmid DNA and adenovirus may have a significant impact on vaccine development against malignancies expressing tumor associated antigens in patients.
Collapse
|
44
|
Wang S, Taaffe J, Parker C, Solórzano A, Cao H, García-Sastre A, Lu S. Hemagglutinin (HA) proteins from H1 and H3 serotypes of influenza A viruses require different antigen designs for the induction of optimal protective antibody responses as studied by codon-optimized HA DNA vaccines. J Virol 2006; 80:11628-37. [PMID: 16987975 PMCID: PMC1642598 DOI: 10.1128/jvi.01065-06] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Effective antibody responses provide crucial immunity against influenza virus infection. The hemagglutinin (HA) protein is the major target of protective antibody responses induced by viral infection and by vaccination with both inactivated and live-attenuated flu vaccines, but knowledge about the optimal designs of protective HA antigens from different flu serotypes is still limited. In this study, we have significantly improved the immunogenicity of HA-expressing DNA vaccines by using codon-optimized HA sequences for either an H1 serotype (A/NewCal/20/99) or an H3 serotype (A/Panama/2007/99) human influenza A virus and then used these constructs as model antigens to identify the optimal HA antigen designs to elicit high-level protective antibody responses. Two forms of HA antigen, a wild-type, full-length HA and a secreted form with transmembrane (TM) domain-truncated HA, were produced. Both forms of HA DNA vaccines, from either H1 or H3 serotypes, were able to elicit high levels of HA-specific immunoglobulin G responses in immunized rabbits as measured by enzyme-linked immunosorbent assay. Interestingly, the abilities of H1 HA and H3 HA antigens to elicit hemagglutination inhibition (HI) and neutralizing antibody (NAb) responses differ. For the H1 HA antigens, the full-length HA induced significantly higher HI and NAb responses than did the TM-truncated HA. For the H3 HA antigen, both the full-length HA and TM-truncated HA induced high levels of HI and NAb responses. These data indicate that H1 and H3 antigens have different expression requirements for the induction of an optimal protective antibody response and that the structure integrity of HA antigens is critical for eliciting type-specific protective antibody responses. Our findings will have an important impact on future subunit-based flu vaccine development.
Collapse
Affiliation(s)
- Shixia Wang
- Department of Medicine, University of Massachusetts Medical School, 364 Plantation Street, Lazare Research Building, Worcester, MA 01605-2397, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Du L, He Y, Wang Y, Zhang H, Ma S, Wong CK, Wu SH, Ng F, Huang JD, Yuen KY, Jiang S, Zhou Y, Zheng BJ. Recombinant adeno-associated virus expressing the receptor-binding domain of severe acute respiratory syndrome coronavirus S protein elicits neutralizing antibodies: Implication for developing SARS vaccines. Virology 2006; 353:6-16. [PMID: 16793110 PMCID: PMC7111904 DOI: 10.1016/j.virol.2006.03.049] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2005] [Revised: 01/27/2006] [Accepted: 03/31/2006] [Indexed: 12/31/2022]
Abstract
Development of an effective vaccine for severe acute respiratory syndrome (SARS) remains to be a priority to prevent possible re-emergence of SARS coronavirus (SARS-CoV). We previously demonstrated that the receptor-binding domain (RBD) of SARS-CoV S protein is a major target of neutralizing antibodies. This suggests that the RBD may serve as an ideal vaccine candidate. Recombinant adeno-associated virus (rAAV) has been proven to be an effective system for gene delivery and vaccine development. In this study, a novel vaccine against SARS-CoV was developed based on the rAAV delivery system. The gene encoding RBD was cloned into a pAAV-IRES-hrGFP plasmid. The immunogenicity induced by the resulting recombinant RBD-rAAV was evaluated in BALB/c mice. The results demonstrated that (1) a single dose of RBD-rAAV vaccination could induce sufficient neutralizing antibody against SARS-CoV infection; (2) two more repeated doses of the vaccination boosted the neutralizing antibody to about 5 times of the level achieved by a single dose of the immunization and (3) the level of the antibody continued to increase for the entire duration of the experiment of 5.5 months. These results suggested that RBD-rAAV is a promising SARS candidate vaccine.
Collapse
Affiliation(s)
- Lanying Du
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yuxian He
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
- Lindsley F. Kimball Research Institute, The New York Blood Center, New York, NY10021, USA
| | - Yijia Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Haojie Zhang
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Selene Ma
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Charlotte K.L. Wong
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Sharon H.W. Wu
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Fai Ng
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Jian-Dong Huang
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kwok-Yung Yuen
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Shibo Jiang
- Lindsley F. Kimball Research Institute, The New York Blood Center, New York, NY10021, USA
| | - Yusen Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
- Corresponding authors. Y. Zhou is to be contacted at fax: +86 10 6381 5259. B.-J. Zheng, fax: +8 52 2855 1241.
| | - Bo-Jian Zheng
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Corresponding authors. Y. Zhou is to be contacted at fax: +86 10 6381 5259. B.-J. Zheng, fax: +8 52 2855 1241.
| |
Collapse
|
46
|
Abstract
Plasmid DNA vaccination is a very powerful and easy method for the induction of strong humoral and cell-mediated immune responses in mice. The technique has also been successfully applied for the definition of immunodominant, human T-cell epitopes using HLA-transgenic mice. By virtue of its strong capacity to induce CD4+-mediated Th1 and CD8+-mediated cytotoxic T-lymphocyte responses, this vaccine approach is particularly attractive for the prophylaxis of intracellular pathogens, such as Mycobacterium tuberculosis (TB) and other pathogenic mycobacteria. In small rodents, the potential of mycobacterial DNA vaccines is well established. In humans, DNA vaccines are clearly less immunogenic and, so far, TB-specific DNA vaccines have not been assessed in humans. However, a number of studies in cattle and sheep have demonstrated the potential of mycobacterial DNA vaccines in larger animals. Also, immunization protocols combining the potent priming capacity of plasmid DNA with subsequent boosting with recombinant protein, recombinant pox-viruses or with Mycobacterium bovis bacille Calmette-Guerin (BCG) vaccine are particularly promising for future applications. The potential of mycobacterial DNA vaccines for immunotherapy and post-exposure prophylaxis is still not clear.
Collapse
Affiliation(s)
- Kris Huygen
- Mycobacterial Immunology, Pasteur Institute Brussels, Wetenschappelijk Instituut voor Volksgezondheid, Brussels, Belgium.
| |
Collapse
|