1
|
Akhtar S, Ansari MM, Verma RD, Sharma J, Gupta A, Dhuriya RK, Verma DP, Saroj J, Ali M, Verma NK, Mitra K, Singh BN, Ghosh JK. Generating a Peptide Library Using the Repeats of Amino Acid Scaffolds Created by Sliding the Framework of a 7-mer Human Chemerin Segment and Discovery of Potent Antibacterial and Antimycobacterial Peptides. J Med Chem 2025; 68:566-589. [PMID: 39718360 DOI: 10.1021/acs.jmedchem.4c02351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
The quest for new approaches for generating novel bioactive designer proteins/peptides has continued with their success in various biomedical applications. Previously, we designed a 14-mer α-helical peptide with antimicrobial and antimycobacterial activities by employing a tandem repeat of the 7-mer, "KVLGRLV" human chemerin segment. Herein, we devised a new method of "sliding framework" with this segment to create amino acid scaffolds of varying sizes and sequences and explored the design of a peptide library with antibacterial and antimycobacterial activities. By utilizing 2 to 7 repeats of these 2 to 6-residue scaffolds, we designed and synthesized 30 peptides of 10-16 residue lengths. Thus, we identified novel AMPs with α-helical, β-sheet, and random coil structures, membrane-destabilizing, and intracellular modes of action, and 9 of them showed therapeutic indices between 100 and 750. Three and two of these nine peptides showed in vivo antibacterial and antitubercular efficacies against Escherichia coli ATCC 25922 and Mycobacterium bovis BCG infections, respectively, in a mouse model.
Collapse
Affiliation(s)
- Sariyah Akhtar
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Mohd Mustkim Ansari
- Division of Molecular Microbiology and Immunology, CSIR- Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Rahul Dev Verma
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Juhi Sharma
- Electron Microscopy Unit, SAIF &R Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Arvind Gupta
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rajendra Kumar Dhuriya
- Division of Molecular Microbiology and Immunology, CSIR- Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Devesh Pratap Verma
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Jyotshana Saroj
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mehmood Ali
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Neeraj Kumar Verma
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
- School of Studies in Biotechnology, Shaheed Mahendra Karma Vishwavidyalaya, Bastar, Dharampura-2, Jagdalpur, C.G. 494001, India
| | - Kalyan Mitra
- Electron Microscopy Unit, SAIF &R Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Bhupendra Narain Singh
- Division of Molecular Microbiology and Immunology, CSIR- Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Jimut Kanti Ghosh
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
2
|
Haghighi FH, Farsiani H. Is Lactococcus lactis a Suitable Candidate for Use as a Vaccine Delivery System Against Helicobacter pylori? Curr Microbiol 2024; 82:30. [PMID: 39643816 DOI: 10.1007/s00284-024-03994-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 11/15/2024] [Indexed: 12/09/2024]
Abstract
Helicobacter pylori was described in 1979. This bacterium, which thrives in the harsh conditions of the stomach, is typically acquired during childhood and can remain colonized for life. Approximately, 90% of the global population is affected, and H. pylori is linked to various conditions, including gastritis, peptic ulcers, lymphoproliferative gastric lymphoma, and even gastric cancer. Currently, antibiotics are the primary treatment method, but the associated challenges of antibiotic use have led to the consideration of oral vaccination as a viable preventive measure against this infection. However, the stomach's harsh environment characterized by its acidic conditions and numerous proteolytic enzymes poses significant obstacles to the development and effectiveness of oral vaccines. To address these challenges, researchers have proposed and evaluated several delivery systems. One of the most promising options is the use of probiotics. Among the various probiotics, Lactococcus lactis stands out as a suitable candidate for oral vaccine delivery against H. pylori due to the advancements in genetic engineering that have been applied to it. This review article discusses the limitations of current treatment strategies and rationalizes the shift toward vaccination, particularly oral vaccination for this infection. It also explores the advantages and challenges of using probiotic bacteria, with a focus on L. lactis as a delivery system. Ultimately, despite the existing challenges, L. lactis continues to be recognized as a promising delivery system. Nonetheless, further research is essential to fully assess its effectiveness and address the challenges associated with this approach.
Collapse
Affiliation(s)
- Faria Hasanzadeh Haghighi
- Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hadi Farsiani
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Azadi-Square, Medical Campus, Mashhad, 9177948564, Iran.
| |
Collapse
|
3
|
Huang L, Tang W, He L, Li M, Lin X, Hu A, Huang X, Wu Z, Wu Z, Chen S, Hu Y. Engineered probiotic Escherichia coli elicits immediate and long-term protection against influenza A virus in mice. Nat Commun 2024; 15:6802. [PMID: 39122688 PMCID: PMC11315933 DOI: 10.1038/s41467-024-51182-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Influenza virus infection remains a major global health problem and requires a universal vaccine with broad protection against different subtypes as well as a rapid-response vaccine to provide immediate protection in the event of an epidemic outbreak. Here, we show that intranasal administration of probiotic Escherichia coli Nissle 1917 activates innate immunity in the respiratory tract and provides immediate protection against influenza virus infection within 1 day. Based on this vehicle, a recombinant strain is engineered to express and secret five tandem repeats of the extracellular domain of matrix protein 2 from different influenza virus subtypes. Intranasal vaccination with this strain induces durable humoral and mucosal responses in the respiratory tract, and provides broad protection against the lethal challenge of divergent influenza viruses in female BALB/c mice. Our findings highlight a promising delivery platform for developing mucosal vaccines that provide immediate and sustained protection against respiratory pathogens.
Collapse
Affiliation(s)
- Ling Huang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei Tang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Lina He
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Mengke Li
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xian Lin
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- Hubei JiangXia Laboratory, Wuhan, 430071, China
| | - Ao Hu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xindi Huang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Zhouyu Wu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhiyong Wu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shiyun Chen
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Yangbo Hu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
- Hubei JiangXia Laboratory, Wuhan, 430071, China.
| |
Collapse
|
4
|
Piri-Gharaghie T, Ghajari G, Rezaeizadeh G, Adil M, Mahdi MH. A novel vaccine strategy against Brucellosis using Brucella abortus multi-epitope OMPs vaccine based on Lactococcus lactis live bacterial vectors. Int Immunopharmacol 2024; 134:112204. [PMID: 38703567 DOI: 10.1016/j.intimp.2024.112204] [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: 03/09/2024] [Revised: 04/25/2024] [Accepted: 05/01/2024] [Indexed: 05/06/2024]
Abstract
Brucella infections typically occur in mucosal membranes, emphasizing the need for mucosal vaccinations. This study evaluated the effectiveness of orally administering Lactococcus lactis (L. lactis) for producing the Brucella abortus multi-epitope OMPs peptide. A multi-epitope plasmid was generated through a reverse vaccinology method, and mice were administered the genetically modified L. lactis orally as a vaccine. The plasmid underwent digestion, synthesizing a 39 kDa-sized protein known as OMPs by the target group. The sera of mice that were administered the pNZ8124-OMPs-L. lactis vaccine exhibited a notable presence of IgG1 antibodies specific to outer membrane proteins (OMPs), heightened levels of interferon (IFN-λ) and tumor necrosis factor alpha (TNF-α), and enhanced transcription rates of interleukin 4 (IL-4) and interleukin 10 (IL-10). The spleen sections from the pNZ8124-OMPs-L. lactis and IRIBA group had less morphological damage associated with inflammation, infiltration of lymphocytes, and lesions to the spleen. The findings present a novel approach to utilizing the food-grade, non-pathogenic L. lactis as a protein cell factory to synthesize innovative immunological candidate OMPs. This approach offers a distinctive way to evaluate experimental medicinal items' practicality, safety, affordability, and long-term sustainability.
Collapse
Affiliation(s)
- Tohid Piri-Gharaghie
- Biotechnology Research Center, Faculty of Biological Sciences, East Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Ghazal Ghajari
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Golnoosh Rezaeizadeh
- Department of Microbiology, Falavarjan Branch, Islamic Azad University, Isfahan, Iran
| | - Mohaned Adil
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | |
Collapse
|
5
|
Kunjapur AM. Planting a chemical flag on antigens. Science 2024; 384:41-42. [PMID: 38574130 DOI: 10.1126/science.ado4537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Next-generation live vaccines are created by autonomous production of nitrated antigens.
Collapse
Affiliation(s)
- Aditya M Kunjapur
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA
| |
Collapse
|
6
|
Ashique S, Hussain A, Fatima N, Altamimi MA. HPV pathogenesis, various types of vaccines, safety concern, prophylactic and therapeutic applications to control cervical cancer, and future perspective. Virusdisease 2023:1-19. [PMID: 37363362 PMCID: PMC10208188 DOI: 10.1007/s13337-023-00824-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 05/01/2023] [Indexed: 06/28/2023] Open
Abstract
Over 98% of cervical cancers (CC) are caused by regular infections with "high risk" genotype of the human papilloma virus (HPV). However, this is not always the causative factor. Therefore, production of HPV vaccinations represents a significant chance to minimize the risk of CC. Phase III studies for a number of preventative HPV vaccines based on L1-virus-like particle (VLPs) have just been completed and the preliminary results are very convincing. However, there are a lot of practical concerns that need to be resolved before the use of these vaccinations. These vaccines were challenged with obvious queries such as protection time, subject receiving vaccines, time of vaccination, and how to include them into ongoing screening programs. Although these vaccines were 90% effective at preventing HPV infection as these offered only modest advantages for the removal of pre-existing infections. New advancements in the creation of therapeutic vaccinations have been explored for further improvement and post-vaccination surveillance. Therapeutic vaccines attempted to boost cell-mediated immunities and these are detrimental to the infected cell as opposed to neutralizing antibodies (different from prophylactic vaccines).
Collapse
Affiliation(s)
- Sumel Ashique
- Department of Pharmaceutics, School of Pharmacy, Bharat Institute of Technology (BIT), Meerut, Uttar Pradesh 250103 India
| | - Afzal Hussain
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451 Saudi Arabia
| | - Neda Fatima
- Department of Pharmacology, Sai College of Pharmacy, Mau, Uttar Pradesh 275102 India
| | - Mohammad A. Altamimi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451 Saudi Arabia
| |
Collapse
|
7
|
Li CX, Qi Y, Chen Y, Zhang Y, Li B, Feng J, Zhang XZ. Tuning Bacterial Morphology to Enhance Anticancer Vaccination. ACS NANO 2023; 17:8815-8828. [PMID: 37093563 DOI: 10.1021/acsnano.3c02373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Morphology tuning is a potent strategy to modulate physiological effects of synthetic biomaterials, but it is rarely explored in microbe-based biochemicals due to the lack of artificial adjustability. Inspired by the interesting phenomenon of microbial transformation, Escherichia coli is rationally adjusted into filamentous morphology-adjusted bacteria (MABac) via chemical stimulation to prepare a bacteria-based vaccine adjuvant/carrier. Inactivated MABac display stronger immunogenicity and special delivery patterns (phagosome escape and cytoplasmic retention) that are sharply distinct from the short rod-shaped bacteria parent (Bac). Transcriptomic study further offers solid evidence for deeply understanding the in vivo activity of MABac-based vaccine, which more effectively motivates multiple cytosolic immune pathways (such as NOD-like receptors and STING) and induces pleiotropic immune responses in comparison with Bac. Harnessing the special functions caused by morphology tuning, the MABac-based adjuvant/carrier significantly improves the immunogenicity and delivery profile of cancer antigens in vivo, thus boosting cancer-specific immunity against the melanoma challenge. This study validates the feasibility of tuning bacterial morphology to improve their biological effects, establishing a facile engineering strategy that upgrades bacterial properties and functions without complex procedures like gene editing.
Collapse
Affiliation(s)
- Chu-Xin Li
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Yongdan Qi
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Yingge Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Yu Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Bin Li
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, PR China
| | - Jun Feng
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| |
Collapse
|
8
|
Mukherjee AG, Wanjari UR, Gopalakrishnan AV, Kannampuzha S, Murali R, Namachivayam A, Ganesan R, Renu K, Dey A, Vellingiri B, Prabakaran DS. Exploring the Molecular Pathogenesis, Pathogen Association, and Therapeutic Strategies against HPV Infection. Pathogens 2022; 12:25. [PMID: 36678374 PMCID: PMC9865103 DOI: 10.3390/pathogens12010025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/10/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
The human papillomavirus (HPV), commonly documented as the cause of warts, has gained much interest recently due to its possible links to several types of cancer. HPV infection is discussed in this review from multiple angles, including its virology, epidemiology, etiology, immunology, clinical symptoms, and treatment. Recent breakthroughs in molecular biology have led to the development of new methods for detecting and treating HPV in tissue. There is no cure for HPV, and although vaccines are available to prevent infection with the most common HPV viruses, their utilization is limited. Destruction and excision are the primary treatment modalities. This review sheds light on the epidemiology, molecular pathogenesis, the association of several other pathogens with HPV, the latest treatment strategies available to treat the same, and an overview of the progress made and the obstacles still to be overcome in the fight against HPV infection.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Sandra Kannampuzha
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Reshma Murali
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Arunraj Namachivayam
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Raja Ganesan
- Institute for Liver and Digestive Diseases, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics, Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata 700073, India
| | - Balachandar Vellingiri
- Stem Cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda 151401, India
| | - D. S. Prabakaran
- Department of Radiation Oncology, College of Medicine, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju 28644, Republic of Korea
- Department of Biotechnology, Ayya Nadar Janaki Ammal College (Autonomous), Srivilliputhur Main Road, Sivakasi 626124, India
| |
Collapse
|
9
|
Zhao Z, Wang H, Zhang D, Guan Y, Siddiqui SA, Feng-Shan X, Cong B. Oral vaccination with recombinant Lactobacillus casei expressing Aeromonas hydrophila Aha1 against A. hydrophila infections in common carps. Virulence 2022; 13:794-807. [PMID: 35499101 PMCID: PMC9067532 DOI: 10.1080/21505594.2022.2063484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 11/03/2022] Open
Abstract
The immunogenicity of Aha1, an OMP of Aeromonas hydrophila mediating the adhesion of bacteria onto the mucosal surface of hosts has been established. In this study, recombinant vectors, pPG1 and pPG2, carrying a 1366 bp DNA fragment that was responsible for encoding the 49 kDa Aha1 from A. hydrophila were constructed, respectively, then electroporated into a probiotic strain Lactobacillus casei CC16 separately to generate two types of recombinants, L. casei-pPG1-Aha1 (Lc-pPG1-Aha1) and L. casei-pPG2-Aha1 (Lc-pPG2-Aha1). Subsequently, these were orally administered into common carps to examine their immunogenicity. The expression and localization of the expressed Aha1 protein relative to the carrier L. casei was validated via Western blotting, flow cytometry, and immune fluorescence separately. The recombinant vaccines produced were shown high efficacies, stimulated higher level of antibodies and AKP, ACP, SOD, LZM, C3, C4 in serum in hosts. Immune-related gene expressions of cytokines including IL-10, IL-1β, TNF-α, IFN-γ in the livers, spleens, HK, and intestines were up-regulated significantly. Besides, a more potent phagocytosis response was observed in immunized fish, and higher survival rates were presented in common carps immunized with Lc-pPG1-Aha1 (60%) and Lc-pPG2-Aha1 (50%) after re-infection with virulent strain A. hydrophila. Moreover, the recombinant L. casei were shown a stronger propensity for survivability in the intestine in immunized fish. Taken together, the recombinant L. casei strains might be promising candidates for oral vaccination against A. hydrophila infections in common carps.
Collapse
Affiliation(s)
- Zelin Zhao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Hong Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Dongxing Zhang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Yongchao Guan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Shahrood Ahmad Siddiqui
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Xiao Feng-Shan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Bo Cong
- Institute of special animal and plant sciences of CAAS, Changchun, Jilin, China
| |
Collapse
|
10
|
Kumar V, Kumar S, Sharma PC. Recent advances in the vaccine development for the prophylaxis of SARS Covid-19. Int Immunopharmacol 2022; 111:109175. [PMID: 35994853 PMCID: PMC9381430 DOI: 10.1016/j.intimp.2022.109175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/08/2022] [Accepted: 08/14/2022] [Indexed: 12/14/2022]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-caused Coronavirus Disease 2019 (COVID-19) is currently a global pandemic that has wreaked havoc on public health, lives, and the global economy. The present COVID-19 outbreak has put pressure on the scientific community to develop medications and vaccinations to combat COVID-19. However, according to highly optimistic forecasts, we could not have a COVID-19 vaccine until September 2020. This is due to the fact that a successful COVID-19 vaccine will necessitate a careful validation of effectiveness and adverse reactivity given that the target vaccine population includes high-risk people over 60, particularly those with severe co-morbid conditions, frontline healthcare professionals, and those involved in essential industrial sectors. For passive immunization, which is being considered for Covid-19, there are several platforms for vaccine development, each with its own advantages and disadvantages. The COVID-19 pandemic, which is arguably the deadliest in the last 100 years after the Spanish flu, necessitates a swift assessment of the various approaches for their ability to incite protective immunity and safety to prevent unintended immune potentiation, which is crucial to the pathogenesis of this virus. Considering the pandemic's high fatality rate and rapid spread, an efficient vaccination is critical for its management. As a result, academia, industry, and government are collaborating in unprecedented ways to create and test a wide range of vaccinations. In this review, we summarize the Covid-19 vaccine development initiatives, recent trends, difficulties, comparison between traditional vaccines development and Covid-19 vaccines development also listed the approved/authorized, phase-3 and pre-clinical trials Covid-19 vaccines in different countries.
Collapse
Affiliation(s)
- Vipul Kumar
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India
| | - Sahil Kumar
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India.
| | - Prabodh Chander Sharma
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India
| |
Collapse
|
11
|
Sultana A, Kumar R. Modified bactofection for efficient and functional DNA delivery using invasive E. coli DH10B vector into human epithelial cell line. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
12
|
Soleymani S, Tavassoli A, Housaindokht MR. An overview of progress from empirical to rational design in modern vaccine development, with an emphasis on computational tools and immunoinformatics approaches. Comput Biol Med 2022; 140:105057. [PMID: 34839187 DOI: 10.1016/j.compbiomed.2021.105057] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 11/03/2021] [Accepted: 11/20/2021] [Indexed: 12/15/2022]
Abstract
Vaccination remains the most effective strategy for preventing and controlling infectious diseases. Numerous conventional vaccines, especially live attenuated, inactivated (killed) microorganisms and subunit vaccines, lead to an effective induction of protective immune responses, mainly antibody-mediated responses against pathogens. However, it has become known that a wide range of highly dangerous pathogens are uncontrollable via conventional vaccination strategies. Recent advances in molecular biology, immunology, genetics, biochemistry, and bioinformatics have provided new prospects for vaccine development. As a result of these advances, several new strategies for vaccine design, development, and production have appeared. These strategies show advantages over conventional vaccines. In this review, we discuss some of the major novel approaches, including recombinant protein vaccines, live recombinant viral and bacterial vectors, DNA and RNA vaccines, reverse vaccinology and reverse genetics approaches. Moreover, we have described the recent progresses on computational tools and immunoinformatics approaches for identifying, designing, and developing new candidate vaccines.
Collapse
Affiliation(s)
- Safoura Soleymani
- Research and Technology Center of Biomolecules, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Amin Tavassoli
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Mohammad Reza Housaindokht
- Research and Technology Center of Biomolecules, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Department of Chemistry, Faculty of Sciences, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
13
|
Zahedipour F, Zamani P, Jamialahmadi K, Jaafari MR, Sahebkar A. Vaccines targeting angiogenesis in melanoma. Eur J Pharmacol 2021; 912:174565. [PMID: 34656608 DOI: 10.1016/j.ejphar.2021.174565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/28/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022]
Abstract
Angiogenesis has a significant role in metastasis and progression of melanoma. Even small tumors may be susceptible to metastasis and hence lead to a worse outcome in patients with melanoma. One of the anti-angiogenic treatment approaches that is undergoing comprehensive study is specific immunotherapy. While tumor cells are challenging targets for immunotherapy due to their genetic instability and heterogeneity, endothelial cells (ECs) are genetically stable. Therefore, vaccines targeting angiogenesis in melanoma are appropriate choices that target both tumor cells and ECs while capable of inducing strong, anti-tumor immune responses with limited toxicity. The main targets of angiogenesis are VEGFs and their receptors but other potential targets have also been investigated, especially in preclinical studies. Various types of vaccines that target angiogenesis in melanoma have been studied including DNA, peptide, protein, dendritic cell-based, and endothelial cell vaccines. This review outlines a number of target antigens that are important for potential progress in developing vaccines for targeting angiogenesis in melanoma. We also discuss different types of vaccines that have been investigated, delivery mechanisms and popular adjuvants, and suggest ways to improve future clinical outcomes.
Collapse
Affiliation(s)
- Fatemeh Zahedipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Zamani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khadijeh Jamialahmadi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, The University of Western Australia, Perth, Australia; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
14
|
Salem-Bekhit MM, Youssof AME, Alanazi FK, Aleanizy FS, Abdulaziz A, Taha EI, Amara AAAF. Bacteria from Infectious Particles to Cell Based Anticancer Targeted Drug Delivery Systems. Pharmaceutics 2021; 13:1984. [PMID: 34959266 PMCID: PMC8706210 DOI: 10.3390/pharmaceutics13121984] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 11/17/2022] Open
Abstract
Bacterial ghosts (BGs) are empty cell envelopes of nonliving evacuated bacterial cells. They are free from their cytoplasmic contents; however, they sustain their cellular 3D morphology and antigenic structures, counting on bioadhesive properties. Lately, they have been tested as an advanced drug delivery system (DDS) for different materials like DNA, peptides, or drugs, either single components or combinations. Different studies have revealed that, BG DDS were paid the greatest attention in recent years. The current review explores the impact of BGs on the field of drug delivery and drug targeting. BGs have a varied area of applications, including vaccine and tumor therapy. Moreover, the use of BGs, their synthesis, their uniqueness as a delivery system and application principles in cancer are discussed. Furthermore, the safety issues of BGs and stability aspects of using ghost bacteria as delivery systems are discussed. Future perspective efforts that must be followed for this important system to continue to grow are important and promising.
Collapse
Affiliation(s)
- Mounir M. Salem-Bekhit
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.M.E.Y.); (F.K.A.); (F.S.A.); (A.A.); (E.I.T.)
- Department of Microbiology and Immunology, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt
- Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah M. E. Youssof
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.M.E.Y.); (F.K.A.); (F.S.A.); (A.A.); (E.I.T.)
- Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Fars K. Alanazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.M.E.Y.); (F.K.A.); (F.S.A.); (A.A.); (E.I.T.)
- Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Fadilah Sfouq Aleanizy
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.M.E.Y.); (F.K.A.); (F.S.A.); (A.A.); (E.I.T.)
| | - Alsuwyeh Abdulaziz
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.M.E.Y.); (F.K.A.); (F.S.A.); (A.A.); (E.I.T.)
- Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ehab I. Taha
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.M.E.Y.); (F.K.A.); (F.S.A.); (A.A.); (E.I.T.)
| | - Amro Abd Al Fattah Amara
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications, Alexandria 21934, Egypt;
| |
Collapse
|
15
|
Luzuriaga MA, Herbert FC, Brohlin OR, Gadhvi J, Howlett T, Shahrivarkevishahi A, Wijesundara YH, Venkitapathi S, Veera K, Ehrman R, Benjamin CE, Popal S, Burton MD, Ingersoll MA, De Nisco NJ, Gassensmith JJ. Metal-Organic Framework Encapsulated Whole-Cell Vaccines Enhance Humoral Immunity against Bacterial Infection. ACS NANO 2021; 15:17426-17438. [PMID: 34546723 DOI: 10.1021/acsnano.1c03092] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The increasing rate of resistance of bacterial infection against antibiotics requires next generation approaches to fight potential pandemic spread. The development of vaccines against pathogenic bacteria has been difficult owing, in part, to the genetic diversity of bacteria. Hence, there are many potential target antigens and little a priori knowledge of which antigen/s will elicit protective immunity. The painstaking process of selecting appropriate antigens could be avoided with whole-cell bacteria; however, whole-cell formulations typically fail to produce long-term and durable immune responses. These complications are one reason why no vaccine against any type of pathogenic E. coli has been successfully clinically translated. As a proof of principle, we demonstrate a method to enhance the immunogenicity of a model pathogenic E. coli strain by forming a slow releasing depot. The E. coli strain CFT073 was biomimetically mineralized within a metal-organic framework (MOF). This process encapsulates the bacteria within 30 min in water and at ambient temperatures. Vaccination with this formulation substantially enhances antibody production and results in significantly enhanced survival in a mouse model of bacteremia compared to standard inactivated formulations.
Collapse
|
16
|
Debnath N, Thakur M, Khushboo, Negi NP, Gautam V, Kumar Yadav A, Kumar D. Insight of oral vaccines as an alternative approach to health and disease management: An innovative intuition and challenges. Biotechnol Bioeng 2021; 119:327-346. [PMID: 34755343 DOI: 10.1002/bit.27987] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/06/2021] [Accepted: 11/03/2021] [Indexed: 12/11/2022]
Abstract
Vaccination is the most suitable and persuasive healthcare program for the prohibition of various deadly diseases. However, the higher production cost and purification strategies are out of reach for the developing nations. In this scenario, development of edible vaccine turns out to be the most promising alternative for remodeling the pharmaceutical industry with reduced production and purification costs. Generally, oral route of vaccination is mostly preferred due to its safety, compliance, low manufacturing cost and most importantly the ability to induce immunity in both systemic and mucosal sites. Genetically modified microorganisms and plants could efficiently be used as vehicles for edible vaccines. Edible vaccines are supposed to reduce the risk associated with traditional vaccines. Currently, oral vaccines are available in the market for several viral and bacterial diseases like cholera, hepatitis B, malaria, rabies etc. Herein, the review focuses on the breakthrough events in the area of edible vaccines associated with dietary microbes and plants for better control over diseases.
Collapse
Affiliation(s)
- Nabendu Debnath
- Centre for Molecular Biology, Central University of Jammu, Samba, Jammu & Kashmir (UT), India
| | - Mony Thakur
- Department of Microbiology, Central University of Haryana, Mahendergarh, Haryana, India
| | - Khushboo
- Department of Biotechnology, Central University of Haryana, Mahendergarh, Haryana, India
| | - Neelam P Negi
- Department of Biotechnology, University Institute of Biotechnology, Chandigarh University, Mohali, Punjab, India
| | - Vibhav Gautam
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ashok Kumar Yadav
- Centre for Molecular Biology, Central University of Jammu, Samba, Jammu & Kashmir (UT), India
| | - Deepak Kumar
- Department of Botany, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
17
|
Induction of Immunogenic Response in BALB/c Mice by Live and Killed Form of Recombinant Lactococcus lactis Displaying EG95 of Echinococcus granulosus. IRANIAN BIOMEDICAL JOURNAL 2021; 25:284-96. [PMID: 34217159 PMCID: PMC8334390 DOI: 10.52547/ibj.25.4.284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background: CE is a zoonotic parasitic infection caused by Echinococcus granulosus worldwide and is associated with economic losses among livestock animals. EG95 is an immunogenic antigen from the E. granulosus.Lactococcus lactis has been prested as a safe vehicle for antigen delivery. The goal of this study was to design a novel L. lactis strain displaying EG95 as a vaccine delivery system. Methods: The eg95 encoding gene fragment fused to the M6 anchoring protein was cloned into the pNZ7021 vector, and L. lactis NZ9000 displaying recombinant EG95 was constructed. The expression of an approximately 32-kDa EG95 protein was confirmed by Western blotting and immunofluorescence analysis. The immune responses were evaluated in BALB/c mice immunized orally and subcutaneously with the live and killed recombinant L. lactis, respectively. Results: Total IgG level in mice immunized with heat-killed recombinant L. lactis (pNZ7021-eg95) significantly increased compared to the control group. sIgA was significantly higher in mice received live recombinant L. lactis (pNZ7021-eg95) compared to the control mice. Splenic lymphocytes from immunized mice represented the high levels of IFN-γ and the low-levels of IL-4 and IL-10. Conclusion: Our results indicate that immunization with EG95-expressing L. lactis can induce both specific humoral and cellular immune responses in mice.
Collapse
|
18
|
Afchangi A, Latifi T, Jalilvand S, Marashi SM, Shoja Z. Combined use of lactic-acid-producing bacteria as probiotics and rotavirus vaccine candidates expressing virus-specific proteins. Arch Virol 2021; 166:995-1006. [PMID: 33533975 DOI: 10.1007/s00705-021-04964-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/03/2020] [Indexed: 12/24/2022]
Abstract
Due to the lower efficacy of currently approved live attenuated rotavirus (RV) vaccines in developing countries, a new approach to the development of safe mucosally administered live bacterial vectors is being considered, using probiotic bacteria as an efficient delivery platform for heterologous RV antigens. Lactic acid bacteria (LAB), which are considered food-grade bacteria and normal microbiota, have been utilized throughout history as probiotics and developed since the 1990s as a delivery system for recombinant heterologous proteins. Over the last decade, LAB have frequently been used as a platform for the delivery of various RV antigens to the mucosa. Given the appropriate safety profile for neonates and providing the benefits of probiotics, recombinant LAB-based vaccines could potentially address the need for a subunit RV vaccine. The present review focuses mainly on different recombinant LAB vaccine constructs for RV and their potential as an alternative recombinant vaccine against RV disease.
Collapse
Affiliation(s)
- Atefeh Afchangi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Tayebeh Latifi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Jalilvand
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sayed Mahdi Marashi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Zabihollah Shoja
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
19
|
Abstract
The immune system has evolved over time to protect the host from foreign microorganisms. Activation of the immune system is predicated on a distinction between self and nonself. Unfortunately, cancer is characterized by genetic alterations in the host's cells, leading to uncontrolled cellular proliferation and evasion of immune surveillance. Cancer immunotherapy aims to educate the host's immune system to not only recognize but also attack and kill mutated cancer cells. While immune checkpoint blockers have been proven to be effective against multiple types of advanced cancer, the overall patient response rate still remains below 30%. Therefore, there is an urgent need to improve current cancer immunotherapies. In this Account, we present an overview of our recent progress on nanoparticle-based strategies for improving cancer vaccines and immunotherapies. We also present other complementary strategies to give a well-rounded snapshot of the field of combination cancer immunotherapy. The versatility and tunability of nanoparticles make them promising platforms for addressing individual challenges posed by various cancers. For example, nanoparticles can deliver cargo materials to specific cells, such as vaccines delivered to antigen-presenting cells for strong immune activation. Nanoparticles also allow for stimuli-responsive delivery of various therapeutics to cancer cells, thus forming the basis for combination cancer immunotherapy. Here, we focus on nanoparticle platforms engineered to deliver tumor antigens, whole tumor cells, and chemotherapeutic or phototherapeutic agents in a manner to effectively and safely trigger the host's immune system against tumor cells. For each work, we discuss the nanoparticle platform developed, synthesis chemistry, and in vivo applications. Nanovaccines offer a unique platform for codelivery of personalized tumor neoantigens and adjuvants and elicitation of robust immune responses against aggressive tumors. Nanovaccines either delivering whole tumor cell lysate or formed from tumor cell lysate may increase the repertoire of tumor antigens as immune targets while exploiting immunogenic cell death to prime antitumor immune responses. We also discuss how antigen- and whole tumor cell-based approaches may open the door for personalized cancer vaccination and immunotherapy. On the other hand, chemotherapy, phototherapy, and radiotherapy are more standardized cancer therapies, and nanoparticle-based approaches may promote their ability to initiate T cell activation against tumor cells and improve antitumor efficacy with minimal toxicity. Finally, building on the recent progress in nanoparticle-based cancer immunotherapy, the field should set the ultimate goal to be clinical translation and clinical efficacy. We will discuss regulatory, analytical, and manufacturing hurdles that should be addressed to expedite the clinical translation of nanomedicine-based cancer immunotherapy.
Collapse
Affiliation(s)
- Marisa E Aikins
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan 48109, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Cheng Xu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan 48109, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan 48109, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
20
|
Taghinezhad-S S, Keyvani H, Bermúdez-Humarán LG, Donders GGG, Fu X, Mohseni AH. Twenty years of research on HPV vaccines based on genetically modified lactic acid bacteria: an overview on the gut-vagina axis. Cell Mol Life Sci 2020; 78:1191-1206. [PMID: 32979054 PMCID: PMC7519697 DOI: 10.1007/s00018-020-03652-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 09/03/2020] [Accepted: 09/16/2020] [Indexed: 10/27/2022]
Abstract
Most cervical cancer (CxCa) are related to persistent infection with high-risk human papillomavirus (HR-HPV) in the cervical mucosa, suggesting that an induction of mucosal cell-mediated immunity against HR-HPV oncoproteins can be a promising strategy to fight HPV-associated CxCa. From this perspective, many pre-clinical and clinical trials have proved the potential of lactic acid bacteria (LAB) genetically modified to deliver recombinant antigens to induce mucosal, humoral and cellular immunity in the host. Altogether, the outcomes of these studies suggest that there are several key factors to consider that may offer guidance on improvement protein yield and improving immune response. Overall, these findings showed that oral LAB-based mucosal HPV vaccines expressing inducible surface-anchored antigens display a higher potential to induce particularly specific systemic and mucosal cytotoxic cellular immune responses. In this review, we describe all LAB-based HPV vaccine investigations by reviewing databases from international studies between 2000 and 2020. Our aim is to promote the therapeutic HPV vaccines knowledge and to complete the gaps in this field to empower scientists worldwide to make proper decisions regarding the best strategies for the development of therapeutic HPV vaccines.
Collapse
Affiliation(s)
- Sedigheh Taghinezhad-S
- Department of Microbiology, Faculty of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, 1477893855, Iran
| | - Hossein Keyvani
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | | | - Gilbert G G Donders
- Department of Obstetrics and Gynaecology, Antwerp University Hospital, Antwerp, Belgium.,Femicare Clinical Research for Women, Tienen, Belgium
| | - Xiangsheng Fu
- Department of Gastroenterology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Amir Hossein Mohseni
- Department of Microbiology, Faculty of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, 1477893855, Iran.
| |
Collapse
|
21
|
Kuai R, Singh PB, Sun X, Xu C, Najafabadi AH, Scheetz L, Yuan W, Xu Y, Hong H, Keskin DB, Wu CJ, Jain R, Schwendeman A, Moon JJ. Robust anti-tumor T cell response with efficient intratumoral infiltration by nanodisc cancer immunotherapy. ADVANCED THERAPEUTICS 2020; 3:2000094. [PMID: 38317797 PMCID: PMC10843840 DOI: 10.1002/adtp.202000094] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Indexed: 12/30/2022]
Abstract
Potent anti-tumor T cell response and efficient intratumoral T cell infiltration are the major challenges for therapeutic cancer vaccines. To address these issues, a nano-vaccine system has been designed to promote anti-tumor T cell responses, and intratumoral infiltration was examined in various murine tumor models. Subcutaneous vaccination with nanodiscs carrying human papillomavirus (HPV)-16 E7 antigen elicits as high as ~32% E7-specific CD8 α + T cell responses in circulation, representing a 29-fold improvement over the soluble peptide vaccination. Importantly, nanodisc vaccination also promotes robust intratumoral T cell infiltration and eliminates HPV16 E6/E7-expressing TC-1 tumors at mucosal sites, including lungs, inner lip, and intravaginal tissues. In a benchmark study with a live Listeria vaccine combined with anti-PD-1 IgG, nanodiscs plus anti-PD-1 immune checkpoint blockade elicits comparable levels of T cell responses with anti-tumor efficacy. Furthermore, compared with Complete Freund's Adjuvant combined with tetanus toxoid, nanodisc vaccination in HLA-A02 mice generates >200-fold stronger IFN-γ+ T cell responses against a neoantigen from an HLA-A02 melanoma patient. Overall, these results show that the nanodisc system is a promising cancer vaccine platform for inducing anti-tumor T cell responses.
Collapse
Affiliation(s)
- Rui Kuai
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | | | - Xiaoqi Sun
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Cheng Xu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Alireza Hassani Najafabadi
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Lindsay Scheetz
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Wenmin Yuan
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Yao Xu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Hao Hong
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing University, Nanjing, 210093, China
| | - Derin B. Keskin
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- The Translational Immunogenomics Lab (TIGL), Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Catherine J. Wu
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- The Translational Immunogenomics Lab (TIGL), Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Renu Jain
- Bristol Myers Squibb, Redwood City, CA, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - James J. Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
22
|
Wait LF, Dobson AP, Graham AL. Do parasite infections interfere with immunisation? A review and meta-analysis. Vaccine 2020; 38:5582-5590. [DOI: 10.1016/j.vaccine.2020.06.064] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/12/2020] [Accepted: 06/21/2020] [Indexed: 12/18/2022]
|
23
|
Liu Q, Shen X, Bian X, Kong Q. Effect of deletion of gene cluster involved in synthesis of Enterobacterial common antigen on virulence and immunogenicity of live attenuated Salmonella vaccine when delivering heterologous Streptococcus pneumoniae antigen PspA. BMC Microbiol 2020; 20:150. [PMID: 32513100 PMCID: PMC7278252 DOI: 10.1186/s12866-020-01837-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/01/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Enterobacterial common antigen (ECA) is a family-specific surface antigen shared by all members of the Enterobacteriaceae family. Previous studies showed that the loss of ECA results in Salmonella attenuation, indicating its usefulness as a vaccine candidate for Salmonella infection, but no studies have shown whether the mutation resulting from the deletion of the ECA operon in conjunction with other mutations could be used as an antigen vehicle for heterologous protein antigen delivery. RESULTS In this study, we introduced a nonpolar, defined ECA operon deletion into wild-type S. Typhimurium χ3761 and an attenuated vaccine strain χ9241, obtaining two isogenic ECA operon mutants, namely, χ12357 and χ12358, respectively. A number of in vitro and in vivo properties of the mutants were analyzed. We found that the loss of ECA did not affect the growth, lipopolysaccharide (LPS) production and motility of S. Typhimurium wild type strain χ3761 and its attenuated vaccine strain χ9241 but significantly affected the virulence when administered orally to BALB/c mice. Furthermore, the effects of the ECA mutation on the immunogenicity of a recombinant S. Typhimurium vaccine strain χ9241 when delivering the pneumococcal antigen PspA were determined. The result showed that the total anti-PspA IgG level of χ12358 (pYA4088) was slightly lower than that of χ9241 (pYA4088), but the protection rate was not compromised. CONCLUSIONS ECA affects virulence and benefits the Th2 immunity of Salmonella Typhimurium, therefore, it is feasible to use a reversible ECA mutant mode to design future Salmonella vaccine strains for heterologous protective antigens.
Collapse
Affiliation(s)
- Qing Liu
- College of Animal Science and Technology, Southwest University, Chongqing, China
- Chongqing Engineering Research Center for Herbivores Resource Protection and Utilization, Chongqing, China
| | - Xuegang Shen
- College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Xiaoping Bian
- College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Qingke Kong
- College of Animal Science and Technology, Southwest University, Chongqing, China.
| |
Collapse
|
24
|
Liu Q, Su H, Bian X, Wang S, Kong Q. Live attenuated Salmonella Typhimurium with monophosphoryl lipid A retains ability to induce T-cell and humoral immune responses against heterologous polysaccharide of Shigella flexneri 2a. Int J Med Microbiol 2020; 310:151427. [PMID: 32654768 DOI: 10.1016/j.ijmm.2020.151427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 05/03/2020] [Accepted: 05/10/2020] [Indexed: 12/19/2022] Open
Abstract
Shigella flexneri 2a (Sf2a) is one of the most frequently isolated Shigella strains that causes the endemic shigellosis in developing countries. In this study, we used recombinant attenuated Salmonella vaccine (RASV) strains to deliver Sf2a O-antigen and characterized the immune responses induced by the vectored O-antigen. First, we identified genes sufficient for biosynthesis of Sf2a O-antigen. A plasmid containing the identified genes was then introduced into the RASV strains, which were manipulated to produce only the heterologous O-antigen and modified lipid A. After oral immunization of mice, we demonstrated that RASV strains could induce potent humoral immune responses as well as robust CD4+ T-cell responses against Sf2a Lipopolysaccharide (LPS) and protect mice against virulent Sf2a challenge. The induced serum antibodies mediated high levels of Shigella-specific serum bactericidal activity and C3 deposition. Moreover, the IgG+ B220low/int BM cell and T follicular helper (Tfh) cell responses could also be triggered effectively. The live attenuated Salmonella with the modified lipid A delivering Sf2a O-antigen polysaccharide showed the same ability to induce immune responses against Sf2a LPS as the strain with the original lipid A. These findings underscore the potential of RASV delivered Sf2a O-antigen for induction of robust CD4+ T-cell and IgG responses and warrant further studies toward the development of Shigella vaccine candidates with RASV strains.
Collapse
Affiliation(s)
- Qing Liu
- College of Animal Science and Technology, Southwest University, 400715, Chongqing, China
| | - Huali Su
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, 32611, FL, USA
| | - Xiaoping Bian
- College of Animal Science and Technology, Southwest University, 400715, Chongqing, China
| | - Shifeng Wang
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, 32611, FL, USA
| | - Qingke Kong
- College of Animal Science and Technology, Southwest University, 400715, Chongqing, China; Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, 32611, FL, USA.
| |
Collapse
|
25
|
Wang R, Pan W, Jin L, Huang W, Li Y, Wu D, Gao C, Ma D, Liao S. Human papillomavirus vaccine against cervical cancer: Opportunity and challenge. Cancer Lett 2020; 471:88-102. [DOI: 10.1016/j.canlet.2019.11.039] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/28/2019] [Accepted: 11/30/2019] [Indexed: 12/20/2022]
|
26
|
Bai Y, Wang G, Qi H, Wang Y, Xu C, Yue L, Hou X, Yu L. Immunogenicity of 987P fimbriae of enterotoxigenic Escherichia coli surface-displayed on Lactobacillus casei. Res Vet Sci 2020; 128:308-314. [DOI: 10.1016/j.rvsc.2019.12.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 12/10/2019] [Accepted: 12/22/2019] [Indexed: 12/27/2022]
|
27
|
Gunasekaran B, Gothandam KM. A review on edible vaccines and their prospects. ACTA ACUST UNITED AC 2020; 53:e8749. [PMID: 31994600 PMCID: PMC6984374 DOI: 10.1590/1414-431x20198749] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 11/04/2019] [Indexed: 01/07/2023]
Abstract
For a long time, vaccines have been the main mode of defense and protection against several bacterial, viral, and parasitic diseases. However, the process of production and purification makes them expensive and unaffordable to many developing nations. An edible vaccine is when the antigen is expressed in the edible part of the plant. This reduces the cost of production of the vaccine because of ease of culturing. In this article, various types of edible vaccines that include algal and probiotics in addition to plants are discussed. Various diseases against which research has been carried out are also reviewed. This article focused on the conception of edible vaccines highlighting the various ways by which vaccines can be delivered.
Collapse
Affiliation(s)
- B Gunasekaran
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - K M Gothandam
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
28
|
Qin M, Du G, Sun X. Biomimetic cell-derived nanocarriers for modulating immune responses. Biomater Sci 2020; 8:530-543. [PMID: 31750453 DOI: 10.1039/c9bm01444f] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In this review, we summarize various applications of biomimetic carriers in modulating immune responses and discuss the future perspectives.
Collapse
Affiliation(s)
- Ming Qin
- Key Laboratory of Drug Targeting and Drug Delivery Systems
- Ministry of Education
- West China School of Pharmacy
- Sichuan University
- Chengdu 610041
| | - Guangsheng Du
- Key Laboratory of Drug Targeting and Drug Delivery Systems
- Ministry of Education
- West China School of Pharmacy
- Sichuan University
- Chengdu 610041
| | - Xun Sun
- Key Laboratory of Drug Targeting and Drug Delivery Systems
- Ministry of Education
- West China School of Pharmacy
- Sichuan University
- Chengdu 610041
| |
Collapse
|
29
|
Torres MA, Terraf MCL, Minahk CJ, Delgado MA. Stability of the Salmonella Typhimurium rcsC11 mutant under different stress conditions. MICROBIOLOGY-SGM 2019; 166:157-168. [PMID: 31714197 DOI: 10.1099/mic.0.000873] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The virulence genes of Salmonella are modulated during infection by several regulatory systems, and the RcsCDB system is one of the most important of these. The S. Typhimurium EG14873 (rcsC11) strain harbours the rcsC11 point mutation, displaying a constitutive activation of this system, which is characterized by mucoid colonies and attenuated virulence phenotypes. In this work, the stability of the rcsC11 mutation was analysed under stress conditions. Under acid and anaerobic stresses, we observed the appearance of small and non-mucoid colonies of the rcsC11 strain. The sequencing of the rcsC gene from these colonies showed that the mutation is conserved. Moreover, we found that small colonies were also generated when the wild-type strain grew in acid and anaerobic conditions. It is worth noting that the transition from normal to atypical colonies of both strains only took place after several days of incubation and was not observed during eukaryotic cell infection. Therefore, the appearance of these atypical colonies is a characteristic feature of S. Typhimurium strains under stressful situations and does not involve a reversion of the rcsC11 allele and nor does it imply any risk to mammalian cells. Therefore, we propose that the S. Typhimurium rcsC11 strain is a good candidate for the development of attenuated vaccines.
Collapse
Affiliation(s)
- Mariela A Torres
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT and Instituto de Química Biológica 'Dr. Bernabé Bloj', Facultad de Bioquímica, Química y Farmacia, UNT Chacabuco 461, T4000ILI - San Miguel de Tucumán, Argentina
| | - María C Leccese Terraf
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT and Instituto de Química Biológica 'Dr. Bernabé Bloj', Facultad de Bioquímica, Química y Farmacia, UNT Chacabuco 461, T4000ILI - San Miguel de Tucumán, Argentina
| | - Carlos J Minahk
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT and Instituto de Química Biológica 'Dr. Bernabé Bloj', Facultad de Bioquímica, Química y Farmacia, UNT Chacabuco 461, T4000ILI - San Miguel de Tucumán, Argentina
| | - Mónica A Delgado
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT and Instituto de Química Biológica 'Dr. Bernabé Bloj', Facultad de Bioquímica, Química y Farmacia, UNT Chacabuco 461, T4000ILI - San Miguel de Tucumán, Argentina
| |
Collapse
|
30
|
Mustafa AD, Kalyanasundram J, Sabidi S, Song AAL, Abdullah M, Abdul Rahim R, Yusoff K. Recovery of recombinant Mycobacterium tuberculosis antigens fused with cell wall-anchoring motif (LysM) from inclusion bodies using non-denaturing reagent (N-laurylsarcosine). BMC Biotechnol 2019; 19:27. [PMID: 31088425 PMCID: PMC6518676 DOI: 10.1186/s12896-019-0522-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 04/30/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The current limitations of conventional BCG vaccines highlights the importance in developing novel and effective vaccines against tuberculosis (TB). The utilization of probiotics such as Lactobacillus plantarum for the delivery of TB antigens through in-trans surface display provides an effective and safe vaccine approach against TB. Such non-recombinant probiotic surface display strategy involves the fusion of candidate proteins with cell wall binding domain such as LysM, which enables the fusion protein to anchor the L. plantarum cell wall externally, without the need for vector genetic modification. This approach requires sufficient production of these recombinant fusion proteins in cell factory such as Escherichia coli which has been shown to be effective in heterologous protein production for decades. However, overexpression in E. coli expression system resulted in limited amount of soluble heterologous TB-LysM fusion protein, since most of it are accumulated as insoluble aggregates in inclusion bodies (IBs). Conventional methods of denaturation and renaturation for solubilizing IBs are costly, time-consuming and tedious. Thus, in this study, an alternative method for TB antigen-LysM protein solubilization from IBs based on the use of non-denaturating reagent N-lauroylsarcosine (NLS) was investigated. RESULTS Expression of TB antigen-LysM fusion genes was conducted in Escherichia coli, but this resulted in IBs deposition in contrast to the expression of TB antigens only. This suggested that LysM fusion significantly altered solubility of the TB antigens produced in E. coli. The non-denaturing NLS technique was used and optimized to successfully solubilize and purify ~ 55% of the recombinant cell wall-anchoring TB antigen from the IBs. Functionality of the recovered protein was analyzed via immunofluorescence microscopy and whole cell ELISA which showed successful and stable cell wall binding to L. plantarum (up to 5 days). CONCLUSION The presented NLS purification strategy enables an efficient and rapid method for obtaining higher yields of soluble cell wall-anchoring Mycobacterium tuberculosis antigens-LysM fusion proteins from IBs in E. coli.
Collapse
Affiliation(s)
- Anhar Danial Mustafa
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Jeevanathan Kalyanasundram
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Sarah Sabidi
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Adelene Ai-Lian Song
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.,Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Maha Abdullah
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Raha Abdul Rahim
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.,Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Khatijah Yusoff
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia. .,Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia. .,Malaysia Genome Institute, 43000, Kajang, Selangor, Malaysia.
| |
Collapse
|
31
|
Kowalski PS, Rudra A, Miao L, Anderson DG. Delivering the Messenger: Advances in Technologies for Therapeutic mRNA Delivery. Mol Ther 2019; 27:710-728. [PMID: 30846391 PMCID: PMC6453548 DOI: 10.1016/j.ymthe.2019.02.012] [Citation(s) in RCA: 712] [Impact Index Per Article: 118.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/11/2019] [Accepted: 02/12/2019] [Indexed: 12/18/2022] Open
Abstract
mRNA has broad potential as a therapeutic. Current clinical efforts are focused on vaccination, protein replacement therapies, and treatment of genetic diseases. The clinical translation of mRNA therapeutics has been made possible through advances in the design of mRNA manufacturing and intracellular delivery methods. However, broad application of mRNA is still limited by the need for improved delivery systems. In this review, we discuss the challenges for clinical translation of mRNA-based therapeutics, with an emphasis on recent advances in biomaterials and delivery strategies, and we present an overview of the applications of mRNA-based delivery for protein therapy, gene editing, and vaccination.
Collapse
Affiliation(s)
- Piotr S Kowalski
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Arnab Rudra
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Lei Miao
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
32
|
Jiang B, Li Z, Ou B, Duan Q, Zhu G. Targeting ideal oral vaccine vectors based on probiotics: a systematical view. Appl Microbiol Biotechnol 2019; 103:3941-3953. [PMID: 30915504 DOI: 10.1007/s00253-019-09770-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/11/2019] [Accepted: 03/12/2019] [Indexed: 12/29/2022]
Abstract
Probiotics have great potential to be engineered into oral vaccine delivery systems, which can facilitate elicitation of mucosal immunity without latent risks of pathogenicity. Combined with the progressive understanding of probiotics and the mucosal immune system as well as the advanced biotechniques of genetic engineering, the development of promising oral vaccine vectors based on probiotics is available while complicated and demanding. Therefore, a systematical view on the design of practical probiotic vectors is necessary, which will help to logically analyze and resolve the problems that might be neglected during our exploration. Here, we attempt to systematically summarize several fundamental issues vital to the effectiveness of the vector of probiotics, including the stability of the engineered vectors, the optimization of antigen expression, the improvement of colonization, and the enhancement of immunoreactivity. We also compared the existent strategies and some developing ones, attempting to figure out an optimal strategy that might deserve to be referred in the future development of oral vaccine vectors based on probiotics.
Collapse
Affiliation(s)
- Boyu Jiang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.,Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou, 225009, China
| | - Zhendong Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.,Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou, 225009, China
| | - Bingming Ou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.,Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou, 225009, China.,College of Life Science, Zhaoqing University, Zhaoqing, 526061, China
| | - Qiangde Duan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China. .,Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou, 225009, China.
| | - Guoqiang Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China. .,Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou, 225009, China.
| |
Collapse
|
33
|
Immunotherapy of gynecological cancers. Best Pract Res Clin Obstet Gynaecol 2019; 60:97-110. [PMID: 31003902 DOI: 10.1016/j.bpobgyn.2019.03.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 03/01/2019] [Indexed: 12/29/2022]
Abstract
Oncology treatments have evolved from intuitive, via empiric, to the present precision medicine, with the integration of molecular targeted therapies in our treatment arsenal. The use of the patients' powerful immune system has long been contemplated and recently led to the integration of immunotherapy to overturn the well-documented inhibitory effects of the tumor on the immune system and restore it to a state of activity against the cancer. Recent favorable results have shown the value and effectiveness of immunotherapy against gynecological cancers. In particular, the checkpoint inhibitors, targeting the programmed death-1 (PD-1) pathway, have shown durable clinical responses with manageable toxicity. Several phase II and III clinical trials testing the association of different regimen of chemotherapy and immunotherapy are ongoing in gynecological cancers, and important results are expected. In this chapter, we outline the main principles of immunotherapy for gynecological cancers and summarize the current strategies used in clinical trials.
Collapse
|
34
|
Yang H, Yan Z, Zhang Z, Realivazquez A, Ma B, Liu Y. Anti-caries vaccine based on clinical cold-adapted influenza vaccine: A promising alternative for scientific and public-health protection against dental caries. Med Hypotheses 2019; 126:42-45. [PMID: 31010498 DOI: 10.1016/j.mehy.2019.03.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/21/2019] [Accepted: 03/20/2019] [Indexed: 01/20/2023]
Abstract
Dental caries remains one of the most pervasive infectious disease around the world. Protection against dental caries can be achieved experimentally by eliciting salivary IgA targeting surficial antigens of S. mutans, however, no such a vaccine has been launched for human use yet. Live vectored vaccines hold the greatest feasibility to induce potent and long-lasting immunity in the host. The FDA approved intranasal cold-adapted influenza vaccine has been used in clinical settings for many years. The vaccine can not only induce broad adaptive immune responses especially mucosal immunity, but the member strains can also circumvent existing immunity, presenting promising candidates for live vectored anti-caries vaccine. Moreover, the genetic techniques for modification of cold-adapted influenza viruses are well developed and widely applicable. Thus, we hypothesize that effective anti-caries vaccine can be developed with the backbone of cold-adapted influenza viruses by inserting specific antigenic identifier sequences of S. mutans into the viral genome, which is anticipated to protect against dental caries in humans with easy inoculation. The immune efficacies of recombinant cold-adapted influenza viruses expressing exogenous antigens have been verified by in vivo experiments for multiple infectious diseases, giving us great confidence to validate the safety properties and protection effect with this chimeric vaccine in animals or even humans. Existing data suggests that the live anti-caries vaccine may help improve public oral health by controlling the caries disease itself.
Collapse
Affiliation(s)
- Huixiao Yang
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatological Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510140, PR China
| | - Zhonghai Yan
- Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Zijian Zhang
- Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Adilene Realivazquez
- Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Binger Ma
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatological Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510140, PR China
| | - Yi Liu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610065, PR China.
| |
Collapse
|
35
|
del Rio B, Redruello B, Fernandez M, Martin MC, Ladero V, Alvarez MA. Lactic Acid Bacteria as a Live Delivery System for the in situ Production of Nanobodies in the Human Gastrointestinal Tract. Front Microbiol 2019. [PMCID: PMC6346216 DOI: 10.3389/fmicb.2018.03179] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
36
|
Thinbanmai T, Lulitanond V, Mayo B, Lulitanond A, Panya M. Cloning and expression of enterovirus 71 capsid protein 1 in a probiotic Bifidobacterium pseudocatenulatum. Lett Appl Microbiol 2018; 68:9-16. [PMID: 30357884 DOI: 10.1111/lam.13089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 10/03/2018] [Accepted: 10/03/2018] [Indexed: 12/19/2022]
Abstract
This study investigated cloning and expression of enterovirus 71 viral capsid protein 1 (EV71-VP1) in Bifidobacterium pseudocatenulatum (B. pseudocatenulatum) M115. To achieve this, a codon-optimized gene coding for EV71-VP1 was analysed, designed, synthesized and cloned into a plasmid vector flanked by a transcriptional promoter and terminator sequences. The promoter was based on that of P919, a constitutive promoter of the gene encoding the large ribosomal protein of B. bifidum BGN4, while the terminator was based on that of the peptidase N gene of Lactococcus lactis. The construct was amplified in Escherichia coli XL1-blue and then transferred into B. pseudocatenulatum M115 by electrotransformation. Western blot analysis revealed that the EV71-VP1 was intracellularly expressed in B. pseudocatenulatum M115 under the control of the selected heterologous promoter. In addition, plasmid stability analysis showed the construct was maintained stably for more than 160 generations, enough for most future applications. The results derived from this study open the possibility to utilize the bacterium carrying a specific expression plasmid as cell factory for the production of proteins with high commercial and health-promoting value. SIGNIFICANCE AND IMPACT OF THE STUDY: This study demonstrated the first successful expression of a codon-optimized gene coding for enterovirus 71 viral capsid protein 1 (EV71-VP1) in Bifidobacterium pseudocatenulatum M115, a novel probiotic strain isolated from human intestines. The EV71-VP1 was constitutively expressed under the control of P919 promoter derived from B. bifidum BGN4 in the cytoplasm of bacterial cells supporting the use of heterologous promoter and terminator sequences for viral gene expression in Bifidobacterium species.
Collapse
Affiliation(s)
- T Thinbanmai
- Department of Microbiology and Research and Diagnostic Center for Emerging Infectious Diseases, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - V Lulitanond
- Department of Microbiology and Research and Diagnostic Center for Emerging Infectious Diseases, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - B Mayo
- Departamento de Microbiología y Bioquímica, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Spain
| | - A Lulitanond
- Department of Clinical Microbiology, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - M Panya
- College of Medicine and Public Health, Ubon Ratchathani University, Ubon Ratchathani, Thailand
| |
Collapse
|
37
|
Maqsood I, Shi W, Wang L, Wang X, Han B, Zhao H, Nadeem A, Moshin B, Saima K, Jamal S, Din M, Xu Y, Tang L, Li Y. Immunogenicity and protective efficacy of orally administered recombinant Lactobacillus plantarum expressing VP2 protein against IBDV in chicken. J Appl Microbiol 2018; 125:1670-1681. [PMID: 30118165 PMCID: PMC7166448 DOI: 10.1111/jam.14073] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 07/27/2018] [Accepted: 08/13/2018] [Indexed: 11/29/2022]
Abstract
AIM To develop an effective oral vaccine against the very virulent infectious bursal disease virus (vvIBDV), we generated two recombinant Lactobacillus plantarum strains (pPG612-VP2/LP and pPG612-T7g10-VP2/LP, which carried the T7g10 translational enhancer) that displayed the VP2 protein on the surface, and compared the humoral and cellular immune responses against vvIBDV in chickens. METHODS AND RESULTS We genetically engineered the L. plantarum strains pPG612-VP2/LP and pPG612-T7g10-VP2/LP constitutively expressing the VP2 protein of vvIBDV. We found that the T7g10 enhancer efficiently upregulates VP2 expression in pPG612-T7g10-VP2/LP. Orally administered, pPG612-T7g10-VP2/LP exhibited significant levels of protection (87·5%) against vvIBDV in chickens, indicating improved immunogenicity. Chickens in the pPG612-T7g10-VP2/LP group produced higher levels of interferons (IFN-γ) and interleukins (IL-2 and IL-4) than those in the pPG612-VP2/LP group. CD8+ and CD4+ lymphocyte counts indicated greater stimulation in the pPG612-T7g10-VP2/LP group (13·3 and 21·0% respectively) than in the pPG612-VP2/LP group (10·4 and 14·0% respectively). Thus, pPG612-T7g10-VP2/LP could induce strong humoral and cellular immune responses against vvIBDV. CONCLUSIONS The recombinant L. plantarum that expresses pPG612-T7g10-VP2 is a promising candidate for oral vaccine development against vvIBDV. SIGNIFICANCE AND IMPACT OF THE STUDY The recombinant Lactobacillus delivery system provides a promising strategy for vaccine development against vvIBDV in chickens.
Collapse
Affiliation(s)
- I. Maqsood
- College of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
| | - W. Shi
- College of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
| | - L. Wang
- College of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
| | - X. Wang
- College of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
| | - B. Han
- College of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
| | - H. Zhao
- College of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
| | - A.M. Nadeem
- College of Life SciencesAnhui Agricultural UniversityHefeiChina
| | - B.S. Moshin
- College of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
| | - K. Saima
- College of Life SciencesAnhui Agricultural UniversityHefeiChina
| | - S.S. Jamal
- Department of ManagementHarbin Institute of TechnologyHarbinChina
| | - M.F. Din
- Department of Molecular GeneticsChinese Academy of Science (CAS)University of Science and Technology (USTC)HefeiChina
| | - Y. Xu
- College of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
| | - L. Tang
- College of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
| | - Y. Li
- College of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
| |
Collapse
|
38
|
Han Y, Liu Q, Yi J, Liang K, Wei Y, Kong Q. A biologically conjugated polysaccharide vaccine delivered by attenuated Salmonella Typhimurium provides protection against challenge of avian pathogenic Escherichia coli O1 infection. Pathog Dis 2018; 75:4085839. [PMID: 28911037 DOI: 10.1093/femspd/ftx102] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 08/17/2017] [Indexed: 01/12/2023] Open
Abstract
Avian pathogenic Escherichia coli (APEC) causes avian airsacculitis and colibacillosis, resulting in significant economic loss to the poultry industry. O1, O2 and O78 are the three predominant serotypes. O-antigen of lipopolysaccharide is serotype determinant and highly immunogenic, and O-antigen polysaccharide-based vaccines have great potential for preventing bacterial infections. In this study, we utilized a novel yeast/bacterial shuttle vector pSS26 to clone the 10.8 kb operon synthesizing APEC O1 O-antigen polysaccharide. The resulting plasmid was introduced into attenuated Salmonella vaccines to deliver this O-antigen polysaccharide. O1 O-antigen was stably synthesized in attenuated Salmonella Typhimurium, demonstrated by slide agglutination, silver staining and western blot. Our results also showed that APEC O1 O-antigen produced in the Salmonella vaccines was attached to bacterial cell surfaces, and the presence of heterologous O-antigen did not alter the resistance to surface-acting agents. Furthermore, birds immunized orally or intramuscularly provided protection against the virulent O1 APEC challenge. Salmonella vaccines carrying APEC O1 O-antigen gene cluster also induced high IgG and IgA immune responses against lipopolysaccharide from the APEC O1 strain. The use of our novel shuttle vector facilitates cloning of large DNA fragments, and this strategy could pave the way for production of Salmonella-vectored vaccines against prevalent APEC serotypes.
Collapse
Affiliation(s)
- Yue Han
- Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Qing Liu
- Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Jie Yi
- Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Kang Liang
- Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yunan Wei
- Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Qingke Kong
- Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.,Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ 85287-5401, USA.,Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL 32608, USA
| |
Collapse
|
39
|
Chabeda A, Yanez RJR, Lamprecht R, Meyers AE, Rybicki EP, Hitzeroth II. Therapeutic vaccines for high-risk HPV-associated diseases. PAPILLOMAVIRUS RESEARCH (AMSTERDAM, NETHERLANDS) 2018; 5:46-58. [PMID: 29277575 PMCID: PMC5887015 DOI: 10.1016/j.pvr.2017.12.006] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 12/13/2017] [Accepted: 12/17/2017] [Indexed: 12/16/2022]
Abstract
Cancer is the second leading cause of death worldwide, and it is estimated that Human papillomavirus (HPV) related cancers account for 5% of all human cancers. Current HPV vaccines are extremely effective at preventing infection and neoplastic disease; however, they are prophylactic and do not clear established infections. Therapeutic vaccines which trigger cell-mediated immune responses for the treatment of established infections and malignancies are therefore required. The E6 and E7 early genes are ideal targets for vaccine therapy due to their role in disruption of the cell cycle and their constitutive expression in premalignant and malignant tissues. Several strategies have been investigated for the development of therapeutic vaccines, including live-vector, nucleic acid, peptide, protein-based and cell-based vaccines as well as combinatorial approaches, with several vaccine candidates progressing to clinical trials. With the current understanding of the HPV life cycle, molecular mechanisms of infection, carcinogenesis, tumour biology, the tumour microenvironment and immune response mechanisms, an approved HPV therapeutic vaccine seems to be a goal not far from being achieved. In this article, the status of therapeutic HPV vaccines in clinical trials are reviewed, and the potential for plant-based vaccine production platforms described.
Collapse
Affiliation(s)
- Aleyo Chabeda
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, South Africa
| | - Romana J R Yanez
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, South Africa
| | - Renate Lamprecht
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, South Africa
| | - Ann E Meyers
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, South Africa
| | - Edward P Rybicki
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, South Africa; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Inga I Hitzeroth
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch 7701, South Africa.
| |
Collapse
|
40
|
Lalsiamthara J, Kim JH, Lee JH. Engineering of a rough auxotrophic mutant Salmonella Typhimurium for effective delivery. Oncotarget 2018; 9:25441-25457. [PMID: 29876000 PMCID: PMC5986645 DOI: 10.18632/oncotarget.25192] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 04/05/2018] [Indexed: 11/25/2022] Open
Abstract
Live Salmonella vaccine vectors offer a remarkable platform for delivering immunogens and therapeutic molecules by mimicking natural intracellular infections; however, pre-existing anti-vector immunity can impede effective deployment. Measures to alleviate pre-existing immunity include the use of heterologous vectors, development of highly attenuated strain enabling greater payload, removal of major immunoreactive components from the vector, and/or augmentation of delivered antigens via increased presentation in antigen presenting cells. Here we report a Salmonella Typhimurium (ST) vector-JOL1800 that embodies these requisite properties. JOL1800 is a highly attenuated, auxotrophic, and O-antigen deficient rough-mutant strain. Heterologous bacterial and viral antigens were expressed and delivered using JOL1800 in mice, irrespective of the inoculation route successful inductions of the mucosal and systemic humoral responses were observed. Compared to smooth LPS vector delivery, we observed an increased fraction of delivered-antigen presenting dendritic cells and a higher frequency of delivered-antigen displayed per macrophage. Upon post-priming with JOL1800 delivery, efficacy of the delivery was minimally affected as indicated by insignificant decrease in colonization, humoral and cellular responses. Our results show that the generated vector is capable of remote antigen delivery, manifests higher antigen presentation, is Differentiating Infected from Vaccinated Animals (DIVA) capable, evades normal pre-existing immunity, and can be deployed for effective delivery.
Collapse
Affiliation(s)
- Jonathan Lalsiamthara
- College of Veterinary Medicine, Chonbuk National University, Iksan Campus, Iksan 54596, Republic of Korea
| | - Je Hyoung Kim
- College of Veterinary Medicine, Chonbuk National University, Iksan Campus, Iksan 54596, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Chonbuk National University, Iksan Campus, Iksan 54596, Republic of Korea
| |
Collapse
|
41
|
Yurina V. Live Bacterial Vectors-A Promising DNA Vaccine Delivery System. Med Sci (Basel) 2018; 6:E27. [PMID: 29570602 PMCID: PMC6024733 DOI: 10.3390/medsci6020027] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/19/2018] [Accepted: 03/19/2018] [Indexed: 12/14/2022] Open
Abstract
Vaccination is one of the most successful immunology applications that has considerably improved human health. The DNA vaccine is a new vaccine being developed since the early 1990s. Although the DNA vaccine is promising, no human DNA vaccine has been approved to date. The main problem facing DNA vaccine efficacy is the lack of a DNA vaccine delivery system. Several studies explored this limitation. One of the best DNA vaccine delivery systems uses a live bacterial vector as the carrier. The live bacterial vector induces a robust immune response due to its natural characteristics that are recognized by the immune system. Moreover, the route of administration used by the live bacterial vector is through the mucosal route that beneficially induces both mucosal and systemic immune responses. The mucosal route is not invasive, making the vaccine easy to administer, increasing the patient's acceptance. Lactic acid bacterium is one of the most promising bacteria used as a live bacterial vector. However, some other attenuated pathogenic bacteria, such as Salmonella spp. and Shigella spp., have been used as DNA vaccine carriers. Numerous studies showed that live bacterial vectors are a promising candidate to deliver DNA vaccines.
Collapse
Affiliation(s)
- Valentina Yurina
- Department of Pharmacy, Medical Faculty, Universitas Brawijaya, East Java 65145, Malang, Indonesia.
| |
Collapse
|
42
|
|
43
|
Sinha S, Kuo CY, Ho JK, White PJ, Jazayeri JA, Pouton CW. A suicidal strain of Listeria monocytogenes is effective as a DNA vaccine delivery system for oral administration. Vaccine 2017; 35:5115-5122. [PMID: 28822642 DOI: 10.1016/j.vaccine.2017.08.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 07/10/2017] [Accepted: 08/07/2017] [Indexed: 01/24/2023]
Abstract
In this study we determined the in vivo activity of model ovalbumin vaccines delivered by direct intramuscular delivery of plasmid DNA or oral delivery using a recombinant suicidal Listeria monocytogenes strain (rsΔ2). In a previous report we described how rsΔ2 is capable of delivering luciferase, as protein or DNA, in vitro, into non-dividing intestinal epithelial cells (Kuo et al., 2009). This is achieved by engineering a dual expression shuttle vector, pDuLX-Luc, that replicates in E. coli and rsΔ2 and drives gene expression from the Listeria promoter (Phly) as well as the eukaryotic cytomegalovirus promoter (CMV), thereby delivering both protein and plasmid DNA to the cell cytoplasm. For the current in vivo study rsΔ2 containing pDuLX-OVA was used to deliver both ovalbumin protein and the mammalian expression plasmid by the oral route. Controls were used to investigate the activity of this system versus positive and negative controls, as well as quantifying activity against direct intramuscular injection of expression plasmids. Oral administration of rsΔ2(pDuLX-OVA) produced significant titres of antibody and was effective at inducing targeted T-cell lysis (approximately 30% lysis relative to an experimental positive control, intravenous OVA-coated splenocytes+lipopolysaccharide). Intramuscular injection of plasmids pDuLX-OVA or p3L-OVA (which lacks the prokaryotic promoter) also produced significant CTL-mediated cell lysis. The delivery of the negative control rsΔ2 (pDuLX-Luc) confirmed that the observed activity was induced specifically by the ovalbumin vaccination. The data suggest that the oral activity of rsΔ2(pDuLX-OVA) is explained by delivery of OVA protein, expressed in rsΔ2 from the prokaryotic promoter present in pDuLX-OVA, but transfection of mammalian cells in vivo may also play a role. Antibody titres were also produced by oral delivery (in rsΔ2) of the p3L-OVA plasmid in which does not include a prokaryotic promoter.
Collapse
Affiliation(s)
- Shubhra Sinha
- Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Melbourne, Victoria, Australia
| | - Cheng-Yi Kuo
- Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Melbourne, Victoria, Australia
| | - Joan K Ho
- Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Melbourne, Victoria, Australia
| | - Paul J White
- Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Melbourne, Victoria, Australia
| | - Jalal A Jazayeri
- Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Melbourne, Victoria, Australia
| | - Colin W Pouton
- Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Melbourne, Victoria, Australia.
| |
Collapse
|
44
|
Kamble NM, Hyoung KJ, Lee JH. Intracellular delivery of HA1 subunit antigen through attenuated Salmonella Gallinarum act as a bivalent vaccine against fowl typhoid and low pathogenic H5N3 virus. Vet Res 2017; 48:40. [PMID: 28784184 PMCID: PMC5545839 DOI: 10.1186/s13567-017-0446-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 08/01/2017] [Indexed: 11/22/2022] Open
Abstract
Introduction of novel inactivated oil-emulsion vaccines against different strains of prevailing and emerging low pathogenic avian influenza (LPAI) viruses is not an economically viable option for poultry. Engineering attenuated Salmonella Gallinarum (S. Gallinarum) vaccine delivering H5 LPAI antigens can be employed as a bivalent vaccine against fowl typhoid and LPAI viruses, while still offering economic viability and sero-surveillance capacity. In this study, we developed a JOL1814 bivalent vaccine candidate against LPAI virus infection and fowl typhoid by engineering the attenuated S. Gallinarum to deliver the globular head (HA1) domain of hemagglutinin protein from H5 LPAI virus through pMMP65 constitutive expression plasmid. The important feature of the developed JOL1814 was the delivery of the HA1 antigen to cytosol of peritoneal macrophages. Immunization of chickens with JOL1814 produced significant level of humoral, mucosal, cellular and IL-2, IL-4, IL-17 and IFN-γ cytokine immune response against H5 HA1 and S. Gallinarum antigens in the immunized chickens. Post-challenge, only the JOL1814 immunized chicken showed significantly faster clearance of H5N3 virus in oropharyngeal and cloacal swabs, and 90% survival rate against lethal challenge with a wild type S. Gallinarum. Furthermore, the JOL1814 immunized were differentiated from the H5N3 LPAI virus infected chickens by matrix (M2) gene-specific real-time PCR. In conclusion, the data from the present showed that the JOL1814 can be an effective bivalent vaccine candidate against H5N3 LPAI and fowl typhoid infection in poultry while still offering sero-surveillance property against H5 avian influenza virus.
Collapse
MESH Headings
- Animals
- Antigens, Viral/administration & dosage
- Antigens, Viral/immunology
- Chickens/immunology
- Chickens/microbiology
- Chickens/virology
- Genetic Engineering/methods
- Genetic Engineering/veterinary
- Influenza A Virus, H5N8 Subtype/immunology
- Influenza Vaccines/immunology
- Influenza Vaccines/therapeutic use
- Influenza in Birds/immunology
- Influenza in Birds/prevention & control
- Influenza in Birds/virology
- Poultry Diseases/immunology
- Poultry Diseases/microbiology
- Poultry Diseases/prevention & control
- Poultry Diseases/virology
- Salmonella/immunology
- Salmonella Infections, Animal/immunology
- Salmonella Infections, Animal/microbiology
- Salmonella Infections, Animal/prevention & control
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/therapeutic use
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/therapeutic use
Collapse
Affiliation(s)
- Nitin Machindra Kamble
- College of Veterinary Medicine, Chonbuk National University, Iksan Campus, Jeonju, 570-752 Republic of Korea
| | - Kim Je Hyoung
- College of Veterinary Medicine, Chonbuk National University, Iksan Campus, Jeonju, 570-752 Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Chonbuk National University, Iksan Campus, Jeonju, 570-752 Republic of Korea
| |
Collapse
|
45
|
Kamble NM, Hyoung KJ, Lee JH. Live-attenuated auxotrophic mutant of Salmonella Typhimurium expressing immunogenic HA1 protein enhances immunity and protective efficacy against H1N1 influenza virus infection. Future Microbiol 2017; 12:739-752. [PMID: 28594235 DOI: 10.2217/fmb-2016-0190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To evaluate the efficacy of attenuated Salmonella Typhimurium (JOL912) as a live bacterial vaccine vector. MATERIALS & METHODS The JOL912 engineered to deliver HA1 protein from influenza A/Puerto Rico/8/1934 (H1N1; PR8) virus was coined as JOL1635 and further evaluated for immunogenicity and protective efficacy. RESULTS The JOL1635 stably harbored the HA1 gene within pMMP65 plasmid with periplasmic expression and effective delivery of HA1 protein to RAW264.7 cells. The JOL1635 immunized chickens showed the significant increase in HA1-specific IgG, sIgA antibody, IFN-γ, IL-6 cytokine and cellular immune responses. The postoral challenge, the JOL1635-immunized chickens showed a faster clearance of PR8 virus cloacal shedding than the control group. CONCLUSION Generated JOL1635 can establish specific immunogenicity and protection against the PR8 virus in chickens.
Collapse
Affiliation(s)
- Nitin Machindra Kamble
- Veterinary Public Health, College of Veterinary Medicine, Chonbuk National University, Iksan Campus, Jeonju 570-752, Republic of Korea
| | - Kim Je Hyoung
- Veterinary Public Health, College of Veterinary Medicine, Chonbuk National University, Iksan Campus, Jeonju 570-752, Republic of Korea
| | - John Hwa Lee
- Veterinary Public Health, College of Veterinary Medicine, Chonbuk National University, Iksan Campus, Jeonju 570-752, Republic of Korea
| |
Collapse
|
46
|
Bi X, Yin J, Nguyen GKT, Rao C, Halim NBA, Hemu X, Tam JP, Liu CF. Enzymatic Engineering of Live Bacterial Cell Surfaces Using Butelase 1. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201703317] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Xiaobao Bi
- School of Biological Sciences; Nanyang Technological University; 60 Nanyang Drive Singapore 637551 Singapore
| | - Juan Yin
- Lee Kong Chian School of Medicine; Nanyang Technological University; 59 Nanyang Drive Singapore 636921 Singapore
| | - Giang K. T. Nguyen
- School of Biological Sciences; Nanyang Technological University; 60 Nanyang Drive Singapore 637551 Singapore
| | - Chang Rao
- School of Biological Sciences; Nanyang Technological University; 60 Nanyang Drive Singapore 637551 Singapore
| | - Nurashikin Bte Abdul Halim
- School of Biological Sciences; Nanyang Technological University; 60 Nanyang Drive Singapore 637551 Singapore
| | - Xinya Hemu
- School of Biological Sciences; Nanyang Technological University; 60 Nanyang Drive Singapore 637551 Singapore
| | - James P. Tam
- School of Biological Sciences; Nanyang Technological University; 60 Nanyang Drive Singapore 637551 Singapore
| | - Chuan-Fa Liu
- School of Biological Sciences; Nanyang Technological University; 60 Nanyang Drive Singapore 637551 Singapore
| |
Collapse
|
47
|
Bi X, Yin J, Nguyen GKT, Rao C, Halim NBA, Hemu X, Tam JP, Liu CF. Enzymatic Engineering of Live Bacterial Cell Surfaces Using Butelase 1. Angew Chem Int Ed Engl 2017; 56:7822-7825. [DOI: 10.1002/anie.201703317] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Indexed: 11/08/2022]
Affiliation(s)
- Xiaobao Bi
- School of Biological Sciences; Nanyang Technological University; 60 Nanyang Drive Singapore 637551 Singapore
| | - Juan Yin
- Lee Kong Chian School of Medicine; Nanyang Technological University; 59 Nanyang Drive Singapore 636921 Singapore
| | - Giang K. T. Nguyen
- School of Biological Sciences; Nanyang Technological University; 60 Nanyang Drive Singapore 637551 Singapore
| | - Chang Rao
- School of Biological Sciences; Nanyang Technological University; 60 Nanyang Drive Singapore 637551 Singapore
| | - Nurashikin Bte Abdul Halim
- School of Biological Sciences; Nanyang Technological University; 60 Nanyang Drive Singapore 637551 Singapore
| | - Xinya Hemu
- School of Biological Sciences; Nanyang Technological University; 60 Nanyang Drive Singapore 637551 Singapore
| | - James P. Tam
- School of Biological Sciences; Nanyang Technological University; 60 Nanyang Drive Singapore 637551 Singapore
| | - Chuan-Fa Liu
- School of Biological Sciences; Nanyang Technological University; 60 Nanyang Drive Singapore 637551 Singapore
| |
Collapse
|
48
|
Lactobacillus plantarum producing a Chlamydia trachomatis antigen induces a specific IgA response after mucosal booster immunization. PLoS One 2017; 12:e0176401. [PMID: 28467432 PMCID: PMC5415134 DOI: 10.1371/journal.pone.0176401] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 04/09/2017] [Indexed: 01/23/2023] Open
Abstract
Mucosal immunity is important for the protection against a wide variety of pathogens. Traditional vaccines administered via parenteral routes induce strong systemic immunity, but they often fail to generate mucosal IgA. In contrast, bacteria-based vaccines comprise an appealing strategy for antigen delivery to mucosal sites. Vaginal infection with Chlamydia trachomatis can develop into upper genital tract infections that can lead to infertility. Therefore, the development of an effective vaccine against Chlamydia is a high priority. In the present study, we have explored the use of a common lactic acid bacterium, Lactobacillus plantarum, as a vector for delivery of a C. trachomatis antigen to mucosal sites. The antigen, referred as Hirep2 (H2), was anchored to the surface of L. plantarum cells using an N-terminal lipoprotein anchor. After characterization, the constructed strain was used as an immunogenic agent in mice. We explored a heterologous prime-boost strategy, consisting of subcutaneous priming with soluble H2 antigen co-administered with CAF01 adjuvant, followed by an intranasal boost with H2-displaying L. plantarum. The results show that, when used as a booster, the recombinant L. plantarum strain was able to evoke cellular responses. Most importantly, booster immunization with the Lactobacillus-based vaccine induced generation of antigen-specific IgA in the vaginal cavity.
Collapse
|
49
|
Immunogenic Properties of Lactobacillus plantarum Producing Surface-Displayed Mycobacterium tuberculosis Antigens. Appl Environ Microbiol 2016; 83:AEM.02782-16. [PMID: 27815271 DOI: 10.1128/aem.02782-16] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 10/27/2016] [Indexed: 12/18/2022] Open
Abstract
Tuberculosis (TB) remains among the most deadly diseases in the world. The only available vaccine against tuberculosis is the bacille Calmette-Guérin (BCG) vaccine, which does not ensure full protection in adults. There is a global urgency for the development of an effective vaccine for preventing disease transmission, and it requires novel approaches. We are exploring the use of lactic acid bacteria (LAB) as a vector for antigen delivery to mucosal sites. Here, we demonstrate the successful expression and surface display of a Mycobacterium tuberculosis fusion antigen (comprising Ag85B and ESAT-6, referred to as AgE6) on Lactobacillus plantarum The AgE6 fusion antigen was targeted to the bacterial surface using two different anchors, a lipoprotein anchor directing the protein to the cell membrane and a covalent cell wall anchor. AgE6-producing L. plantarum strains using each of the two anchors induced antigen-specific proliferative responses in lymphocytes purified from TB-positive donors. Similarly, both strains induced immune responses in mice after nasal or oral immunization. The impact of the anchoring strategies was reflected in dissimilarities in the immune responses generated by the two L. plantarum strains in vivo The present study comprises an initial step toward the development of L. plantarum as a vector for M. tuberculosis antigen delivery. IMPORTANCE This work presents the development of Lactobacillus plantarum as a candidate mucosal vaccine against tuberculosis. Tuberculosis remains one of the top infectious diseases worldwide, and the only available vaccine, bacille Calmette-Guérin (BCG), fails to protect adults and adolescents. Direct antigen delivery to mucosal sites is a promising strategy in tuberculosis vaccine development, and lactic acid bacteria potentially provide easy, safe, and low-cost delivery vehicles for mucosal immunization. We have engineered L. plantarum strains to produce a Mycobacterium tuberculosis fusion antigen and to anchor this antigen to the bacterial cell wall or to the cell membrane. The recombinant strains elicited proliferative antigen-specific T-cell responses in white blood cells from tuberculosis-positive humans and induced specific immune responses after nasal and oral administrations in mice.
Collapse
|
50
|
Oldiges DP, Laughery JM, Tagliari NJ, Leite Filho RV, Davis WC, da Silva Vaz I, Termignoni C, Knowles DP, Suarez CE. Transfected Babesia bovis Expressing a Tick GST as a Live Vector Vaccine. PLoS Negl Trop Dis 2016; 10:e0005152. [PMID: 27911903 PMCID: PMC5135042 DOI: 10.1371/journal.pntd.0005152] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 11/01/2016] [Indexed: 11/18/2022] Open
Abstract
The Rhipicephalus microplus tick is a notorious blood-feeding ectoparasite of livestock, especially cattle, responsible for massive losses in animal production. It is the main vector for transmission of pathogenic bacteria and parasites, including Babesia bovis, an intraerythrocytic apicomplexan protozoan parasite responsible for bovine Babesiosis. This study describes the development and testing of a live B. bovis vaccine expressing the protective tick antigen glutathione-S-transferase from Haemaphysalis longicornis (HlGST). The B. bovis S74-T3B parasites were electroporated with a plasmid containing the bidirectional Ef-1α (elongation factor 1 alpha) promoter of B. bovis controlling expression of two independent genes, the selectable marker GFP-BSD (green fluorescent protein–blasticidin deaminase), and HlGST fused to the MSA-1 (merozoite surface antigen 1) signal peptide from B. bovis. Electroporation followed by blasticidin selection resulted in the emergence of a mixed B. bovis transfected line (termed HlGST) in in vitro cultures, containing parasites with distinct patterns of insertion of both exogenous genes, either in or outside the Ef-1α locus. A B. bovis clonal line termed HlGST-Cln expressing intracellular GFP and HlGST in the surface of merozoites was then derived from the mixed parasite line HlGST using a fluorescent activated cell sorter. Two independent calf immunization trials were performed via intravenous inoculation of the HlGST-Cln and a previously described control consisting of an irrelevant transfected clonal line of B. bovis designated GFP-Cln. The control GFP-Cln line contains a copy of the GFP-BSD gene inserted into the Ef-1α locus of B. bovis in an identical fashion as the HIGST-Cln parasites. All animals inoculated with the HlGST-Cln and GFP-Cln transfected parasites developed mild babesiosis. Tick egg fertility and fully engorged female tick weight was reduced significantly in R. microplus feeding on HlGST-Cln-immunized calves. Collectively, these data show the efficacy of a transfected HlGST-Cln B. bovis parasite to induce detectable anti-glutathione-S-transferase antibodies and a reduction in tick size and fecundity of R. microplus feeding in experimentally inoculated animals. The cattle tick Rhipicephalus microplus is a hematophagous ectoparasite, responsible for the transmission of lethal parasites such as Babesia sp, limiting cattle production in tropical and subtropical regions of the world. There is an urgent emerging need for improved methods of control for these currently neglected tick and tick borne diseases. It is hypothesized that a dual attenuated-live vector vaccine containing a stably transfected tick antigen elicits protective immune responses against the parasite and the tick vector in vaccinated cattle. Live Babesia vaccines based on attenuated parasites are the only effective method available for preventing acute babesiosis. On the other hand, glutathione-S-transferase from Haemaphysalis longicornis (HlGST) is a known effective antigen against Rhipicephalus microplus, the most common vector for B. bovis. This study describes the development and testing of a transfected, B. bovis vaccine expressing HlGST against the tick R. microplus. A B. bovis clonal line designated HlGST-Cln expressing HlGST and GFP/BSD, and separately a control transfected B. bovis clonal line expressing only GFP/BSD was used to vaccinate calves in two independent experiments. All immunized calves developed mild babesiosis, and only calves immunized with the HlGST-Cln parasite line generated anti-HlGST antibodies. Tick egg fertility and fully engorged female tick weight were reduced significantly in R. microplus feeding on HlGST-Cln-vaccinated calves. Taken together, these data demonstrates the ability of transfected B. bovis to elicit antibodies against a heterologous tick antigen in cattle and to induce partial protection in the vaccinated animals against the cattle tick for the first time, representing a step toward the goal to produce a live vector anti-tick vaccine.
Collapse
Affiliation(s)
- Daiane P. Oldiges
- Centro de Biotecnologia Universidade Federal do Rio Grande do Sul, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Jacob M. Laughery
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, United States of America
| | - Nelson Junior Tagliari
- Faculdade de Veterinária Universidade Federal do Rio Grande do Sul; Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Ronaldo Viana Leite Filho
- Faculdade de Veterinária Universidade Federal do Rio Grande do Sul; Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - William C. Davis
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, United States of America
| | - Itabajara da Silva Vaz
- Centro de Biotecnologia Universidade Federal do Rio Grande do Sul, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Faculdade de Veterinária Universidade Federal do Rio Grande do Sul; Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Carlos Termignoni
- Centro de Biotecnologia Universidade Federal do Rio Grande do Sul, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Departamento de Bioquímica Universidade Federal do Rio Grande do Sul; Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Donald P. Knowles
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, United States of America
- Animal Disease Research Unit, Agricultural Research Service, United States Department of Agriculture, Pullman, Washington, United States of America
| | - Carlos E. Suarez
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, United States of America
- Animal Disease Research Unit, Agricultural Research Service, United States Department of Agriculture, Pullman, Washington, United States of America
- * E-mail: ,
| |
Collapse
|