1
|
Abbasnia S, Hashem Asnaashari AM, Sharebiani H, Soleimanpour S, Mosavat A, Rezaee SA. Mycobacterium tuberculosis and host interactions in the manifestation of tuberculosis. J Clin Tuberc Other Mycobact Dis 2024; 36:100458. [PMID: 38983441 PMCID: PMC11231606 DOI: 10.1016/j.jctube.2024.100458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024] Open
Abstract
The final step of epigenetic processes is changing the gene expression in a new microenvironment in the body, such as neuroendocrine changes, active infections, oncogenes, or chemical agents. The case of tuberculosis (TB) is an outcome of Mycobacterium tuberculosis (M.tb) and host interaction in the manifestation of active and latent TB or clearance. This comprehensive review explains and interprets the epigenetics findings regarding gene expressions on the host-pathogen interactions in the development and progression of tuberculosis. This review introduces novel insights into the complicated host-pathogen interactions, discusses the challengeable results, and shows the gaps in the clear understanding of M.tb behavior. Focusing on the biological phenomena of host-pathogen interactions, the epigenetic changes, and their outcomes provides a promising future for developing effective TB immunotherapies when converting gene expression toward appropriate host immune responses gradually becomes attainable. Overall, this review may shed light on the dark sides of TB pathogenesis as a life-threatening disease. Therefore, it may support effective planning and implementation of epigenetics approaches for introducing proper therapies or effective vaccines.
Collapse
Affiliation(s)
- Shadi Abbasnia
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hiva Sharebiani
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saman Soleimanpour
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arman Mosavat
- Blood Borne Infections Research Center, Academic Center for Education, Culture, and Research (ACECR), Razavi Khorasan, Mashhad, Iran
| | - Seyed Abdolrahim Rezaee
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
2
|
Wang X, Wang Q, Wang J, Wang X, Yin L, Wang C, Fan G, Pan J. A Novel Lipopeptide-Functionalized Metal-Organic Framework for Periodontitis Therapy through the Htra1/FAK/YAP Pathway. Biomater Res 2024; 28:0057. [PMID: 39076893 PMCID: PMC11283871 DOI: 10.34133/bmr.0057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 06/20/2024] [Indexed: 07/31/2024] Open
Abstract
Periodontitis is a chronic inflammatory disease characterized by plaque accumulation, resulting in immune microenvironment disorders and resorption of alveolar bone. To promote bone healing under inflammatory environments, a functional biomaterial based on disease pathophysiology is designed. A novel fatty acid C10-modified polypeptide, C10-KR8, is discovered to have excellent abilities in modulating macrophage repolarization and promoting bone regeneration in periodontitis. To build a multifunctional material localized drug delivery system, C10-KR8@ZIF-8 (C10-KR8-loaded zeolitic imidazolate framework-8) nanoparticles are constructed to sustainedly release the C10-KR8 peptide and Zn elements. By synergistic effects of providing a dynamic immuno-modulatory environment and promoting osteogenesis under pathological conditions, the obtained pH-responsive nanoparticles display excellent bone regeneration capability. Furthermore, coimmunoprecipitation/liquid chromatography-tandem mass spectrometry analysis and proteomics analysis revealed that the C10-KR8 peptide directly interacts with the high-temperature requirement protein A1 (Htra1), and C10-KR8@ZIF-8 nanoparticles promote the osteogenic differentiation of bone mesenchymal stem cells by activating the focal adhesion kinase (FAK)/phosphatidylinositide 3-kinase (PI3K)/AKT pathway and enhancing the nuclear localization of Yes-associated protein (YAP). Taken together, this study demonstrates C10-KR8 peptide regulate osteoimmunology and bone regeneration by Htra1/FAK/YAP pathway and that ZIF-8-based peptide loading platform is a promising strategy for periodontitis.
Collapse
Affiliation(s)
- Xuechun Wang
- Department of Stomatology, Shanghai General Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Qing Wang
- Department of Stomatology, Shanghai General Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Jian Wang
- Department of General Dentistry, Shanghai Ninth People’s Hospital, School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology,
Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Xuan Wang
- Department of Stomatology, Shanghai General Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Linling Yin
- Department of Stomatology, Shanghai General Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Changping Wang
- Department of Orthopaedics, Shanghai General Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Guangjian Fan
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Jinsong Pan
- Department of Stomatology, Shanghai General Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| |
Collapse
|
3
|
Ahmadi Ghezeldasht S, Mosavat A, Rezaee SA. Novel insights into human T-lymphotropic virus type-1 (HTLV-1) pathogenesis-host interactions in the manifestation of HTLV-1-associated myelopathy/tropical spastic paraparesis. Rev Med Virol 2024; 34:e2567. [PMID: 38937135 DOI: 10.1002/rmv.2567] [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: 12/23/2023] [Revised: 04/16/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024]
Abstract
Human T-lymphotropic virus type-1 (HTLV-1) was the first discovered human oncogenic retrovirus, the etiological agent of two serious diseases have been identified as adult T-cell leukaemia/lymphoma malignancy and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP), a debilitating chronic neuro-myelopathy. Despite more than 40 years of molecular, histopathological and immunological studies on HTLV-1-associated diseases, the virulence and pathogenicity of this virus are yet to be clarified. The reason why the majority of HTLV-1-infected individuals (∼95%) remain asymptomatic carriers is still unclear. The deterioration of the immune system towards oncogenicity and autoimmunity makes HTLV-1 a natural probe for the study of malignancy and neuro-inflammatory diseases. Additionally, its slow worldwide spreading has prompted public health authorities and researchers, as urged by the WHO, to focus on eradicating HTLV-1. In contrast, neither an effective therapy nor a protective vaccine has been introduced. This comprehensive review focused on the most relevant studies of the neuro-inflammatory propensity of HTLV-1-induced HAM/TSP. Such an emphasis on the virus-host interactions in the HAM/TSP pathogenesis will be critically discussed epigenetically. The findings may shed light on future research venues in designing and developing proper HTLV-1 therapeutics.
Collapse
Affiliation(s)
- Sanaz Ahmadi Ghezeldasht
- Blood Borne Infections Research Center, Academic Center for Education, Culture and Research (ACECR), Razavi Khorasan, Mashhad, Iran
| | - Arman Mosavat
- Blood Borne Infections Research Center, Academic Center for Education, Culture and Research (ACECR), Razavi Khorasan, Mashhad, Iran
| | - Seyed Abdolrahim Rezaee
- Inflammation and Inflammatory Diseases Division, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Karbalaei M, Mosavat A, Soleimanpour S, Farsiani H, Ghazvini K, Amini AA, Sankian M, Rezaee SA. Production and Evaluation of Ag85B:HspX:hFcγ1 Immunogenicity as an Fc Fusion Recombinant Multi-Stage Vaccine Candidate Against Mycobacterium tuberculosis. Curr Microbiol 2024; 81:127. [PMID: 38575759 DOI: 10.1007/s00284-024-03655-3] [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] [Received: 04/15/2023] [Accepted: 02/29/2024] [Indexed: 04/06/2024]
Abstract
An urgent need is to introduce an effective vaccine against Mycobacterium tuberculosis (M.tb) infection. In the present study, a multi-stage M.tb immunodominant Fcγ1 fusion protein (Ag85B:HspX:hFcγ1) was designed and produced, and the immunogenicity of purified protein was evaluated. This recombinant fusion protein was produced in the Pichia pastoris expression system. The HiTrap-rPA column affinity chromatography purified and confirmed the fusion protein using ELISA and Western blotting methods. The co-localisation assay was used to confirm its proper folding and function. IFN-γ, IL-12, IL-4, and TGF-β expression in C57BL/6 mice then evaluated the immunogenicity of the construct in the presence and absence of BCG. After expression optimisation, medium-scale production and the Western blotting test confirmed suitable production of Ag85B:HspX:hFcγ1. The co-localisation results on antigen-presenting cells (APCs) showed that Ag85B:HspX:hFcγ1 properly folded and bound to hFcγRI. This strong co-localisation with its receptor can confirm inducing proper Th1 responses. The in vivo immunisation assay showed no difference in the expression of IL-4 but a substantial increase in the expression of IFN-γ and IL-12 (P ≤ 0.02) and a moderate increase in TGF-β (P = 0.05). In vivo immunisation assay revealed that Th1-inducing pathways have been stimulated, as IFN-γ and IL-12 strongly, and TGF-β expression moderately increased in Ag85B:HspX:hFcγ1 group and Ag85B:HspX:hFcγ1+BCG. Furthermore, the production of IFN-γ from splenocytes in the Ag85B:HspX:hFcγ1 group was enormously higher than in other treatments. Therefore, this Fc fusion protein can make a selective multi-stage delivery system for inducing appropriate Th1 responses and is used as a subunit vaccine alone or in combination with others.
Collapse
Affiliation(s)
- Mohsen Karbalaei
- Department of Microbiology and Virology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Arman Mosavat
- Blood Borne Infections Research Center, Academic Center for Education, Culture, and Research (ACECR), Razavi Khorasan, Mashhad, Iran
| | - Saman Soleimanpour
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hadi Farsiani
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kiarash Ghazvini
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abbas Ali Amini
- Cancer and Immunology Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mojtaba Sankian
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Abdolrahim Rezaee
- Inflammation and Inflammatory Diseases Division, Faculty of Medicine, Immunology Research Center, Mashhad University of Medical Sciences, Azadi-Square, Medical Campus, Mashhad, 9177948564, Iran.
| |
Collapse
|
5
|
Hashemzaei M, Ghoshoon MB, Jamshidi M, Moradbeygi F, Hashemzehi A. A Review on Romiplostim Mechanism of Action and the Expressive Approach in E. coli. Recent Pat Biotechnol 2024; 18:95-109. [PMID: 38282441 DOI: 10.2174/1872208317666230503094451] [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: 11/01/2022] [Revised: 02/10/2023] [Accepted: 02/24/2023] [Indexed: 01/30/2024]
Abstract
Immune thrombocytopenic purpura (ITP) is an autoimmune disorder determined by immune-mediated platelet demolition and reduction of platelet production. Romiplostim is a new thrombopoiesis motivating peptibody that binds and stimulates the human thrombopoietin receptor the patent of which was registered in 2008. It is used to treat thrombocytopenia in patients with chronic immune thrombocytopenic purpura. Romiplostim is a 60 kDa peptibody designed to inhibit cross-reacting immune responses. It consists of four high-affinity TPO-receptor binding domains for the Mpl receptor and one human IgG1 Fc domain. Escherichia coli is a good host for the fabrication of recombinant proteins such as romiplostim. The expression of a gene intended in E. coli is dependent on many factors such as a protein's inherent ability to fold, mRNA's secondary structure, its solubility, its toxicity preferential codon use, and its need for post-translational modification (PTM). This review focuses on the structure, function, mechanism of action, and expressive approach to romiplostim in E. coli.
Collapse
Affiliation(s)
- Masoud Hashemzaei
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mehrnaz Jamshidi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Moradbeygi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Hashemzehi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Zheng L, Liu H, Chen L, You X, Lv F, Fan H, Hui Q, Liu B, Wang X. Expression and Purification of FGFR1-Fc Fusion Protein and Its Effects on Human Lung Squamous Carcinoma. Appl Biochem Biotechnol 2024; 196:573-587. [PMID: 37160564 DOI: 10.1007/s12010-023-04542-6] [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] [Accepted: 04/11/2023] [Indexed: 05/11/2023]
Abstract
Molecular-targeted therapies for lung squamous cell carcinoma (LSCC) are limited mainly because targetable oncogenic aberrations are absent in LSCC. Recent genomic analyses have revealed that the fibroblast growth factor (FGF) signaling pathway plays a fundamental role in LSCC progression via cancer cell proliferation and angiogenesis. In the present study, we designed, expressed, and purified a fibroblast growth factor receptor fragment (FGFR1-Fc) fusion protein using NS/0 cells. In FGF2-FGFR1 overexpressed NCI-H1703 cells, the FGFR1-Fc fusion protein effectively inhibited proliferation and invasion and arrested the cell cycle at the G0-G1 phase. In NCI-H1703 cells treated with the FGFR1-Fc fusion protein, the phosphorylation levels of FGFR1, FRS2, ERK, and AKT were significantly reduced. Using an siRNA assay, we demonstrated that FGF2-FGFR1 is the major anti-tumor target of FGFR1-Fc fusion the FGFR1-Fc fusion protein, which also significantly inhibited proliferation and invasion by NCI-H1703 cells via the FGF2-FGFR1 signaling pathway. In addition, the FGFR1-Fc fusion protein significantly inhibited angiogenesis in an embryonic chorioallantoic membrane model. The FGFR1-Fc fusion protein may be an effective therapeutic candidate for LSCC.
Collapse
Affiliation(s)
- Lulu Zheng
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Zhejiang, 310000, Hangzhou, China
| | - Huan Liu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Lingfeng Chen
- School of Pharmaceutical Sciences, Hangzhou Medical College, Zhejiang, 310012, Hangzhou, China
| | - Xinyi You
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Fangyi Lv
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Haibing Fan
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Qi Hui
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Baohua Liu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
- Department of Neurological Rehabilitation, The Second Asffiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
| | - Xiaojie Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
7
|
Li S, Wang S, Fordjour E, Liang Y, Wang X, Ye Y, Bai Z, Yang Y, Chen Y. Development and characterization of anti-IL-5 monoclonal antibody Fab fragment for blocking IL-5/IL-5Rα binding. Int Immunopharmacol 2023; 124:111032. [PMID: 37832239 DOI: 10.1016/j.intimp.2023.111032] [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: 05/22/2023] [Revised: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 10/15/2023]
Abstract
Interleukin-5 (IL-5) is a homodimeric cytokine that is a crucial regulator of the proliferation, activation, and maturation of eosinophils. Anti-IL-5 monoclonal antibodies, which block the binding of IL-5 to the IL-5 receptor subunit alpha (IL-5Rα), have been successfully used to treat eosinophilic (EOS) asthma. The currently marketed monoclonal antibody drugs require repeated injections for administration, which seriously affect patient compliance and high systemic exposure for injectable drug delivery. Here we successfully screened and developed the Fab (fragment of antigen binding), which is 1/3rd the molecular weight of IgG, favoring inhalation-mediated delivery to the lungs, making it more effective for asthma treatment. The 20A12-Fab-H12L3 can bind to IL-5 with a binding constant of 1.236E-09 M while significantly inhibiting the IL-5/IL-5Rα complex formation. We found that the light chain amino acids (S46 and F71) significantly affected the antibody expression during humanization. The 20A12-Fab-H12L3 significantly inhibited the proliferation of TF-1 cells and blocked the IL-5 binding to the IL-5Rα-overexpressing human embryonic kidney (HEK)-293 cells in vitro. Therefore, based on the mutant IL-5 binding with Fab, we explained why antibodies blocked IL-5 binding to IL-5Rα. Thus, this study provided a candidate pharmaceutical antibody for inhalation drug delivery.
Collapse
Affiliation(s)
- Shijie Li
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China; National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China; Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi 214122, China
| | - Shijie Wang
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China; National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China; Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi 214122, China
| | - Eric Fordjour
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China; National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China; Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi 214122, China
| | - Yaoji Liang
- Zhuhai Resproly Pharmaceutical Technology Co., Ltd, Zhuhai, 519040, Guangdong, China
| | - Xuelian Wang
- Zhuhai Resproly Pharmaceutical Technology Co., Ltd, Zhuhai, 519040, Guangdong, China
| | - Yonghao Ye
- Zhuhai Resproly Pharmaceutical Technology Co., Ltd, Zhuhai, 519040, Guangdong, China
| | - Zhonghu Bai
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China; National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China; Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi 214122, China.
| | - Yankun Yang
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China; National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 214122, China; Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi 214122, China.
| | - Yongqi Chen
- Zhuhai Resproly Pharmaceutical Technology Co., Ltd, Zhuhai, 519040, Guangdong, China.
| |
Collapse
|
8
|
Chen JP, Gong JS, Su C, Li H, Xu ZH, Shi JS. Improving the soluble expression of difficult-to-express proteins in prokaryotic expression system via protein engineering and synthetic biology strategies. Metab Eng 2023; 78:99-114. [PMID: 37244368 DOI: 10.1016/j.ymben.2023.05.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 05/23/2023] [Indexed: 05/29/2023]
Abstract
Solubility and folding stability are key concerns for difficult-to-express proteins (DEPs) restricted by amino acid sequences and superarchitecture, resolved by the precise distribution of amino acids and molecular interactions as well as the assistance of the expression system. Therefore, an increasing number of tools are available to achieve efficient expression of DEPs, including directed evolution, solubilization partners, chaperones, and affluent expression hosts, among others. Furthermore, genome editing tools, such as transposons and CRISPR Cas9/dCas9, have been developed and expanded to construct engineered expression hosts capable of efficient expression ability of soluble proteins. Accounting for the accumulated knowledge of the pivotal factors in the solubility and folding stability of proteins, this review focuses on advanced technologies and tools of protein engineering, protein quality control systems, and the redesign of expression platforms in prokaryotic expression systems, as well as advances of the cell-free expression technologies for membrane proteins production.
Collapse
Affiliation(s)
- Jin-Ping Chen
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China; Yixing Institute of Food and Biotechnology Co., Ltd, Yixing, 214200, PR China
| | - Jin-Song Gong
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China; Yixing Institute of Food and Biotechnology Co., Ltd, Yixing, 214200, PR China.
| | - Chang Su
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China
| | - Heng Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China
| | - Zheng-Hong Xu
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, School of Biotechnology, Jiangnan University, Wuxi, 214122, PR China; Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi, 214122, PR China; Yixing Institute of Food and Biotechnology Co., Ltd, Yixing, 214200, PR China
| | - Jin-Song Shi
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China; Yixing Institute of Food and Biotechnology Co., Ltd, Yixing, 214200, PR China
| |
Collapse
|
9
|
Kon E, Levy Y, Elia U, Cohen H, Hazan-Halevy I, Aftalion M, Ezra A, Bar-Haim E, Naidu GS, Diesendruck Y, Rotem S, Ad-El N, Goldsmith M, Mamroud E, Peer D, Cohen O. A single-dose F1-based mRNA-LNP vaccine provides protection against the lethal plague bacterium. SCIENCE ADVANCES 2023; 9:eadg1036. [PMID: 36888708 PMCID: PMC9995031 DOI: 10.1126/sciadv.adg1036] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/08/2023] [Indexed: 05/28/2023]
Abstract
Messenger RNA (mRNA) lipid nanoparticle (LNP) vaccines have emerged as an effective vaccination strategy. Although currently applied toward viral pathogens, data concerning the platform's effectiveness against bacterial pathogens are limited. Here, we developed an effective mRNA-LNP vaccine against a lethal bacterial pathogen by optimizing mRNA payload guanine and cytosine content and antigen design. We designed a nucleoside-modified mRNA-LNP vaccine based on the bacterial F1 capsule antigen, a major protective component of Yersinia pestis, the etiological agent of plague. Plague is a rapidly deteriorating contagious disease that has killed millions of people during the history of humankind. Now, the disease is treated effectively with antibiotics; however, in the case of a multiple-antibiotic-resistant strain outbreak, alternative countermeasures are required. Our mRNA-LNP vaccine elicited humoral and cellular immunological responses in C57BL/6 mice and conferred rapid, full protection against lethal Y. pestis infection after a single dose. These data open avenues for urgently needed effective antibacterial vaccines.
Collapse
Affiliation(s)
- Edo Kon
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yinon Levy
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel
| | - Uri Elia
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel
| | - Hila Cohen
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel
| | - Inbal Hazan-Halevy
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Moshe Aftalion
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel
| | - Assaf Ezra
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Erez Bar-Haim
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel
| | - Gonna Somu Naidu
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yael Diesendruck
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Shahar Rotem
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel
| | - Nitay Ad-El
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Meir Goldsmith
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Emanuelle Mamroud
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel
| | - Dan Peer
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ofer Cohen
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel
| |
Collapse
|
10
|
Hu Z, Feng J, Deng J, Zhang Y, He X, Hu J, Wang X, Hu S, Liu X, Liu X. Delivery of Fc-fusion Protein by a Recombinant Newcastle Disease Virus Vector. Appl Biochem Biotechnol 2023; 195:2077-2092. [PMID: 36417109 DOI: 10.1007/s12010-022-04237-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2022] [Indexed: 11/24/2022]
Abstract
Fc-fusion proteins (FCPs), a new generation biological medicine, have revolutionized the practice of medicines that treat diseases. However, complex manufacturing techniques are required for FCP production, casting the affordability and accessibility issues in low- and middle-income economies (LMIEs). Virus-vectored system may serve as a simple and cost-effective platform for FCP delivery. As a proof-of-concept study, Newcastle disease virus (NDV), a widely-used vector for vaccine generation, was used as a vector to express and deliver a model FCP composed of the hemagglutinin (HA) and IgG Fc. A recombinant NDV expressing the HA-Fc fusion protein was generated using reverse genetics, which had comparable replication and virulence to the parental virus. High levels of expression of soluble HA-Fc were detected in cell culture and embryonated chicken eggs inoculated with the recombinant NDV. In addition, the recombinant NDV replicated in the lung of mouse, delivering the HA-Fc protein to this organ. The HA-Fc expressed by NDV specifically bound to murine FcγRI, which was dependent on the presence of the Fc tag. The recombinant NDV induced high vector-specific antibody response, whereas it failed to elicit H7N9-specific antibody immunity in mice. The absence of HA-specific antibodies may be attributed to deficient incorporation of the HA-Fc protein into NDV virion particles. Our results indicated that NDV may be potentially used as a vector for FCP expression and delivery. This strategy may help to enhance the affordability and equal accessibility of FCP biological medicines, especially in LIMEs.
Collapse
Affiliation(s)
- Zenglei Hu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China.,Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Jianing Feng
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China.,College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, China
| | - Jing Deng
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China.,Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yanyan Zhang
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China.,Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Xiaozheng He
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China.,Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Jiao Hu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Xiaoquan Wang
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Shunlin Hu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Xiaowen Liu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, China. .,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China. .,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China.
| |
Collapse
|
11
|
Ahmadi Ghezeldasht S, Momen Heravi M, Valizadeh N, Rafatpanah H, Shamsian SA, Mosavat A, Rezaee SA. Development of a Novel HTLV-1 Protease: Human Fcγ1 Recombinant Fusion Molecule in the CHO Eukaryotic Expression System. Appl Biochem Biotechnol 2023; 195:1862-1876. [PMID: 36399306 PMCID: PMC9673214 DOI: 10.1007/s12010-022-04259-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2022] [Indexed: 11/19/2022]
Abstract
Human T-cell leukaemia virus type 1 (HTLV-1) is the causative agent of two life-threatening diseases, adult T cell leukaemia/lymphoma (ATLL), and HTLV-1-associated myelopathy/tropical spastic (HAM/TSP). HTLV-1 protease (HTLV-1-PR) is an aspartic protease that represents a promising target for therapeutic purposes like human immunodeficiency virus-PR inhibitors (HIV-PR). Therefore, in this study, the human Fc fusion recombinant-PR (HTLV-1-PR:hFcγ1) was designed and expressed for two applications, finding a blocking substrate as a potential therapeutic or a potential subunit peptide vaccine. The PCR amplified DNA sequences encoding the HTLV-1-PR from the MT2-cell line using specific primers with restriction enzyme sites of Not1 and Xba1. The construct was then cloned to pTZ57R/T TA plasmid and, after confirming the PR sequence, subcloned into the pDR2ΔEF1α Fc-expression vector to create pDR2ΔEF1α.HTLV-1-PR:hFcγ1. The integrity of recombinant DNA was confirmed by sequencing to ensure that the engineered construct was in the frame. The recombinant fusion protein was then produced in the Chinese hamster ovary cell (CHO) system and was purified from its supernatant using HiTrap-rPA column affinity chromatography. Then, the immunofluorescence assay (IFA) co-localisation method showed that HTLV-1-PR:hFc recombinant fusion protein has appropriate folding as it binds to the anti-Fcγ antibody; the Fcγ1 tag participates to have HTLV-1-PR:hFcγ1 as a dimeric secretory protein. The development and production of HTLV-1-PR can be used to find a blocking substrate as a potential therapeutic molecule and apply it in an animal model to assess its immunogenicity and potential protection against HTLV-1 infection.
Collapse
Affiliation(s)
- Sanaz Ahmadi Ghezeldasht
- Inflammation and Inflammatory Diseases Division, Medical Campus, Immunology Research Center, Mashhad University of Medical Sciences, Azadi Square, Mashhad, 9177948564 Iran
- Blood Borne Infections Research Center, Academic Center for Education, Culture and Research (ACECR), Azadi Square, Ferdowsi University Campus, Razavi Khorasan, Mashhad, 9177949367 Iran
| | - Mastoureh Momen Heravi
- Inflammation and Inflammatory Diseases Division, Medical Campus, Immunology Research Center, Mashhad University of Medical Sciences, Azadi Square, Mashhad, 9177948564 Iran
| | - Narges Valizadeh
- Inflammation and Inflammatory Diseases Division, Medical Campus, Immunology Research Center, Mashhad University of Medical Sciences, Azadi Square, Mashhad, 9177948564 Iran
| | - Houshang Rafatpanah
- Inflammation and Inflammatory Diseases Division, Medical Campus, Immunology Research Center, Mashhad University of Medical Sciences, Azadi Square, Mashhad, 9177948564 Iran
| | - Seyed Aliakbar Shamsian
- Blood Borne Infections Research Center, Academic Center for Education, Culture and Research (ACECR), Azadi Square, Ferdowsi University Campus, Razavi Khorasan, Mashhad, 9177949367 Iran
- Department of Parasitology and Mycology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arman Mosavat
- Blood Borne Infections Research Center, Academic Center for Education, Culture and Research (ACECR), Azadi Square, Ferdowsi University Campus, Razavi Khorasan, Mashhad, 9177949367 Iran
| | - Seyed Abdolrahim Rezaee
- Inflammation and Inflammatory Diseases Division, Medical Campus, Immunology Research Center, Mashhad University of Medical Sciences, Azadi Square, Mashhad, 9177948564 Iran
| |
Collapse
|
12
|
Hassannia H, Amiri MM, Ghaedi M, Sharifian RA, Golsaz-Shirazi F, Jeddi-Tehrani M, Shokri F. Preclinical Assessment of Immunogenicity and Protectivity of Novel ROR1 Fusion Proteins in a Mouse Tumor Model. Cancers (Basel) 2022; 14:5827. [PMID: 36497309 PMCID: PMC9738141 DOI: 10.3390/cancers14235827] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 10/30/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
The receptor tyrosine kinase-like orphan receptor 1 (ROR1) is a new tumor associated antigen (TAA) which is overexpressed in several hematopoietic and solid malignancies. The present study aimed to produce and evaluate different fusion proteins of mouse ROR1 (mROR1) to enhance immunogenicity and protective efficacy of ROR1. Four ROR1 fusion proteins composed of extracellular region of mROR1, immunogenic fragments of TT as well as Fc region of mouse IgG2a were produced and employed to immunize Balb/C mice. Humoral and cellular immune responses and anti-tumor effects of these fusion proteins were evaluated using two different syngeneic murine ROR1+ tumor models. ROR1-specific antibodies were induced in all groups of mice. The levels of IFN-γ, IL-17 and IL-22 cytokines in culture supernatants of stimulated splenocytes were increased in all groups of immunized mice, particularly mice immunized with TT-mROR1-Fc fusion proteins. The frequency of ROR1-specific CTLs was higher in mice immunized with TT-mROR1-Fc fusion proteins. Finally, results of tumor challenge in immunized mice showed that immunization with TT-mROR1-Fc fusion proteins completely inhibited ROR1+ tumor cells growth in two different syngeneic tumor models until day 120 post tumor challenge. Our preclinical findings, for the first time, showed that our fusion proteins could be considered as a potential candidate vaccine for active immunotherapy of ROR1-expressing malignancies.
Collapse
Affiliation(s)
- Hadi Hassannia
- Immunogenetics Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari P.O. Box 48157-33971, Iran
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran P.O. Box 14155-6559, Iran
| | - Mohammad Mehdi Amiri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran P.O. Box 14155-6559, Iran
| | - Mojgan Ghaedi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran P.O. Box 14155-6559, Iran
| | - Ramezan-Ali Sharifian
- Department of Hematology and Oncology, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran P.O. Box 14197-33141, Iran
| | - Forough Golsaz-Shirazi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran P.O. Box 14155-6559, Iran
| | - Mahmood Jeddi-Tehrani
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran P.O. Box 19839-69412, Iran
| | - Fazel Shokri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran P.O. Box 14155-6559, Iran
| |
Collapse
|
13
|
Selective APC-targeting of a novel Fc-fusion multi-immunodominant recombinant protein ( tTax- tEnv:mFcγ2a) for HTLV-1 vaccine development. Life Sci 2022; 308:120920. [PMID: 36044973 DOI: 10.1016/j.lfs.2022.120920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 11/22/2022]
Abstract
AIMS HTLV-1 causes two life-threatening diseases: adult T-cell leukaemia/lymphoma and HTLV-1-associated myelopathy/tropical spastic paraparesis. Due to the lack of proper treatment, an effective HTLV-1 vaccine is urgently needed. MAIN METHODS DNA sequences of 11-19 and 178-186 amino acids of HTLV-1-Tax and SP2 and P21 were fused to the mouse-Fcγ2a, or His-tag called tTax-tEnv:mFcγ2a and tTax-tEnv:His, respectively. These constructs were produced in Pichia pastoris, and their immunogenicity and protective properties were assessed in a mouse challenging model with an HTLV-1-MT2 cell line. KEY FINDINGS The immunogenicity assessments showed significant increase in IFN-γ production in animals receiving tTax-tEnv:mFcγ2a (1537.2 ± 292.83 pg/mL) compared to tTax-tEnv:His (120.28 ± 23.9, p = 0.02). IL-12 production also increased in group receiving tTax-tEnv:mFcγ2a than tTax-tEnv:His group, (23 ± 2.6 vs 1.5 ± 0.6, p = 0.01), respectively. The IFN-γ and IL-12 levels in the Fc-immunised group were negatively correlated with PVL (R = -0.82, p < 0.04) and (R = -0.87, p = 0.05), respectively. While, IL-4 was increased by tTax-tEnv:His (21.16 ± 1.76 pg/mL) compared to tTax-tEnv:mFcγ2a (13.7 ± 1.49, p = 0.019) with a negative significant correlation to PVL (R = -0.95, p = 0.001). SIGNIFICANCE The mouse challenging assay with tTax-tEnv:mFcγ2a showed 50 % complete protection and a 50 % low level of HTLV-1-PVL compared to the positive control receiving HTLV-1-MT2 (p = 0.001). Challenging experiments for the His-tag protein showed the same outcome (p = 0.002) but by different mechanisms. The Fc-fusion construct induced more robust Th1, and His-tag protein shifted more to Th2 immune responses. Therefore, inducing both T helper responses, but a Th1/Th2 balance in favour of Th1 might be necessary for appropriate protection against HTLV-1 infection, spreading via cell-to-cell contact manner.
Collapse
|
14
|
A century of attempts to develop an effective tuberculosis vaccine: Why they failed? Int Immunopharmacol 2022; 109:108791. [PMID: 35487086 DOI: 10.1016/j.intimp.2022.108791] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/18/2022] [Accepted: 04/18/2022] [Indexed: 11/23/2022]
Abstract
Tuberculosis (TB) remains a major global health problem despite widespread use of the Bacillus BCG vaccine. This situation is worsened by co-infection with HIV, and the development of multidrug-resistant Mycobacterium tuberculosis (Mtb) strains. Thus, novel vaccine candidates and improved vaccination strategies are urgently needed in order to reduce the incidence of TB and even to eradicate TB by 2050. Over the last few decades, 23 novel TB vaccines have entered into clinical trials, more than 13 new vaccines have reached various stages of preclinical development, and more than 50 potential candidates are in the discovery stage as next-generation vaccines. Nevertheless, why has a century of attempts to introduce an effective TB vaccine failed? Who should be blamed -scientists, human response, or Mtb strategies? Literature review reveals that the elimination of latent or active Mtb infections in a given population seems to be an epigenetic process. With a better understanding of the connections between bacterial infections and gene expression conditions in epigenetic events, opportunities arise in designing protective vaccines or therapeutic agents, particularly as epigenetic processes can be reversed. Therefore, this review provides a brief overview of different approaches towards novel vaccination strategies and the mechanisms underlying these approaches.
Collapse
|
15
|
Elia U, Ramishetti S, Rosenfeld R, Dammes N, Bar-Haim E, Naidu GS, Makdasi E, Yahalom-Ronen Y, Tamir H, Paran N, Cohen O, Peer D. Design of SARS-CoV-2 hFc-Conjugated Receptor-Binding Domain mRNA Vaccine Delivered via Lipid Nanoparticles. ACS NANO 2021; 15:9627-9637. [PMID: 33480671 PMCID: PMC7860138 DOI: 10.1021/acsnano.0c10180] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 01/21/2021] [Indexed: 05/20/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been identified as the causal agent of COVID-19 and stands at the center of the current global human pandemic, with death toll exceeding one million. The urgent need for a vaccine has led to the development of various immunization approaches. mRNA vaccines represent a cell-free, simple, and rapid platform for immunization, and therefore have been employed in recent studies toward the development of a SARS-CoV-2 vaccine. Herein, we present the design of an mRNA vaccine, based on lipid nanoparticles (LNPs)-encapsulated SARS-CoV-2 human Fc-conjugated receptor-binding domain (RBD-hFc). Several ionizable lipids have been evaluated in vivo in a luciferase (luc) mRNA reporter assay, and two leading LNPs formulations have been chosen for the subsequent RBD-hFc mRNA vaccine strategy. Intramuscular administration of LNP RBD-hFc mRNA elicited robust humoral response, a high level of neutralizing antibodies and a Th1-biased cellular response in BALB/c mice. The data in the current study demonstrate the potential of these lipids as promising candidates for LNP-based mRNA vaccines in general and for a COVID19 vaccine in particular.
Collapse
Affiliation(s)
- Uri Elia
- Laboratory of Precision NanoMedicine, Shmunis School
for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences,
Tel Aviv University, Tel Aviv 69978,
Israel
- Center for Nanoscience and Nanotechnology,
Tel Aviv University, Tel Aviv 69978,
Israel
- Department of Materials Sciences and Engineering, Iby
and Aladar Fleischman Faculty of Engineering, Tel Aviv
University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv
University, Tel Aviv 69978, Israel
- Department of Biochemistry and Molecular Genetics,
Israel Institute for Biological Research, Ness-Ziona 76100,
Israel
| | - Srinivas Ramishetti
- Laboratory of Precision NanoMedicine, Shmunis School
for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences,
Tel Aviv University, Tel Aviv 69978,
Israel
- Center for Nanoscience and Nanotechnology,
Tel Aviv University, Tel Aviv 69978,
Israel
- Department of Materials Sciences and Engineering, Iby
and Aladar Fleischman Faculty of Engineering, Tel Aviv
University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv
University, Tel Aviv 69978, Israel
| | - Ronit Rosenfeld
- Department of Biochemistry and Molecular Genetics,
Israel Institute for Biological Research, Ness-Ziona 76100,
Israel
| | - Niels Dammes
- Laboratory of Precision NanoMedicine, Shmunis School
for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences,
Tel Aviv University, Tel Aviv 69978,
Israel
- Center for Nanoscience and Nanotechnology,
Tel Aviv University, Tel Aviv 69978,
Israel
- Department of Materials Sciences and Engineering, Iby
and Aladar Fleischman Faculty of Engineering, Tel Aviv
University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv
University, Tel Aviv 69978, Israel
| | - Erez Bar-Haim
- Department of Biochemistry and Molecular Genetics,
Israel Institute for Biological Research, Ness-Ziona 76100,
Israel
| | - Gonna Somu Naidu
- Laboratory of Precision NanoMedicine, Shmunis School
for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences,
Tel Aviv University, Tel Aviv 69978,
Israel
- Center for Nanoscience and Nanotechnology,
Tel Aviv University, Tel Aviv 69978,
Israel
- Department of Materials Sciences and Engineering, Iby
and Aladar Fleischman Faculty of Engineering, Tel Aviv
University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv
University, Tel Aviv 69978, Israel
| | - Efi Makdasi
- Department of Infectious Diseases, Israel
Institute for Biological Research, Ness-Ziona 76100,
Israel
| | - Yfat Yahalom-Ronen
- Department of Infectious Diseases, Israel
Institute for Biological Research, Ness-Ziona 76100,
Israel
| | - Hadas Tamir
- Department of Infectious Diseases, Israel
Institute for Biological Research, Ness-Ziona 76100,
Israel
| | - Nir Paran
- Department of Infectious Diseases, Israel
Institute for Biological Research, Ness-Ziona 76100,
Israel
| | - Ofer Cohen
- Department of Biochemistry and Molecular Genetics,
Israel Institute for Biological Research, Ness-Ziona 76100,
Israel
| | - Dan Peer
- Laboratory of Precision NanoMedicine, Shmunis School
for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences,
Tel Aviv University, Tel Aviv 69978,
Israel
- Center for Nanoscience and Nanotechnology,
Tel Aviv University, Tel Aviv 69978,
Israel
- Department of Materials Sciences and Engineering, Iby
and Aladar Fleischman Faculty of Engineering, Tel Aviv
University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv
University, Tel Aviv 69978, Israel
| |
Collapse
|
16
|
Grindel B, Engel BJ, Hall CG, Kelderhouse LE, Lucci A, Zacharias NM, Takahashi TT, Millward SW. Mammalian Expression and In Situ Biotinylation of Extracellular Protein Targets for Directed Evolution. ACS OMEGA 2020; 5:25440-25455. [PMID: 33043224 PMCID: PMC7542843 DOI: 10.1021/acsomega.0c03990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 09/08/2020] [Indexed: 05/17/2023]
Abstract
Directed evolution is a powerful tool for the selection of functional ligands from molecular libraries. Extracellular domains (ECDs) of cell surface receptors are common selection targets for therapeutic and imaging agent development. Unfortunately, these proteins are often post-translationally modified and are therefore unsuitable for expression in bacterial systems. Directional immobilization of these targets is further hampered by the absence of biorthogonal groups for site-specific chemical conjugation. We have developed a nonadherent mammalian expression system for rapid, high-yield expression of biotinylated ECDs. ECDs from EGFR, HER2, and HER3 were site-specifically biotinylated in situ and recovered from the cell culture supernatant with yields of up to 10 mg/L at >90% purity. Biotinylated ECDs also contained a protease cleavage site for rapid and selective release of the ECD after immobilization on avidin/streptavidin resins and library binding. A model mRNA display selection round was carried out against the HER2 ECD with the HER2 affibody expressed as an mRNA-protein fusion. HER2 affibody-mRNA fusions were selectively released by thrombin and quantitative PCR revealed substantial improvements in the enrichment of functional affibody-mRNA fusions relative to direct PCR amplification of the resin-bound target. This methodology allows rapid purification of high-quality targets for directed evolution and selective elution of functional sequences at the conclusion of each selection round.
Collapse
Affiliation(s)
- Brian
J. Grindel
- Department
of Cancer Systems Imaging, MD Anderson Cancer
Center, Houston, Texas 77030, United States
| | - Brian J. Engel
- Department
of Cancer Systems Imaging, MD Anderson Cancer
Center, Houston, Texas 77030, United States
| | - Carolyn G. Hall
- Department
of Breast Surgical Oncology, MD Anderson
Cancer Center, Houston, Texas 77030, United States
| | - Lindsay E. Kelderhouse
- Department
of Cancer Systems Imaging, MD Anderson Cancer
Center, Houston, Texas 77030, United States
| | - Anthony Lucci
- Department
of Breast Surgical Oncology, MD Anderson
Cancer Center, Houston, Texas 77030, United States
| | - Niki M. Zacharias
- Department
of Urology, MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Terry T. Takahashi
- Department
of Chemistry, University of Southern California, Los Angeles, California 90089, United States
| | - Steven W. Millward
- Department
of Cancer Systems Imaging, MD Anderson Cancer
Center, Houston, Texas 77030, United States
| |
Collapse
|
17
|
Effects of Adjuvants on the Immunogenicity and Efficacy of a Zika Virus Envelope Domain III Subunit Vaccine. Vaccines (Basel) 2019; 7:vaccines7040161. [PMID: 31717890 PMCID: PMC6963592 DOI: 10.3390/vaccines7040161] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/10/2019] [Accepted: 10/24/2019] [Indexed: 02/08/2023] Open
Abstract
Zika virus (ZIKV), a mosquito-borne flavivirus, has attracted global attention due to its close association with congenital Zika syndrome and neurological diseases, and transmission through additional routes, such as sexual contact. Currently there are no vaccines approved for ZIKV, and thus, there is an urgent need to develop an effective and safe ZIKV vaccine. Domain III (DIII) of the ZIKV envelope (E) protein is an important vaccine target, and a vaccine developed using a mutant DIII of E (EDIII) protein protects adult and pregnant mice, and unborn offspring, against ZIKV infection. Here, we have used immunocompetent BALB/c mice treated with anti-interferon-α/β receptor 1 (Ifnar1) antibodies to investigate whether three adjuvants (aluminum (Alum), monophosphoryl lipid A (MPL), and MF59), either alone or in combination, could improve the efficacy of this EDIII subunit vaccine. Our data show that, although vaccine formulated with a single adjuvant induced a specific antibody and cellular immune response, and reduced viral load in mice challenged with ZIKV, the combination of Alum and MPL adjuvants led to a more robust and balanced immune response, stronger neutralizing activity against three recent ZIKV human strains, and greater protection against a high-dose ZIKV challenge. Particularly, the combination of Alum with MPL significantly reduced viral titers and viral RNA copy numbers in sera and tissues, including the male reproductive organs. Overall, this study has identified the combination of Alum and MPL as the most effective adjuvant for ZIKV EDIII subunit vaccines, and it has important implications for subunit vaccines against other enveloped viruses, including non-ZIKV flaviviruses.
Collapse
|
18
|
Parker CL, McSweeney MD, Lucas AT, Jacobs TM, Wadsworth D, Zamboni WC, Lai SK. Pretargeted delivery of PEG-coated drug carriers to breast tumors using multivalent, bispecific antibody against polyethylene glycol and HER2. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2019; 21:102076. [PMID: 31394261 PMCID: PMC7224238 DOI: 10.1016/j.nano.2019.102076] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/18/2019] [Accepted: 07/29/2019] [Indexed: 12/19/2022]
Abstract
Pretargeting is an increasingly explored strategy to improve nanoparticle targeting, in which pretargeting molecules that bind both selected epitopes on target cells and nanocarriers are first administered, followed by the drug-loaded nanocarriers. Bispecific antibodies (bsAb) represent a promising class of pretargeting molecules, but how different bsAb formats may impact the efficiency of pretargeting remains poorly understood, in particular Fab valency and Fc receptor (FcR)-binding of bsAb. We found the tetravalent bsAb markedly enhanced PEGylated nanoparticle binding to target HER2+ cells relative to the bivalent bsAb in vitro. Pretargeting with tetravalent bsAb with abrogated FcR binding increased tumor accumulation of PEGylated liposomal doxorubicin (PLD) 3-fold compared to passively targeted PLD alone, and 5-fold vs pretargeting with tetravalent bsAb with normal FcR binding in vivo. Our work demonstrates that multivalency and elimination of FcRn recycling are both important features of pretargeting molecules, and further supports pretargeting as a promising nanoparticle delivery strategy.
Collapse
MESH Headings
- Animals
- Antibodies, Bispecific/chemistry
- Antibodies, Bispecific/pharmacology
- Antineoplastic Agents, Immunological/chemistry
- Antineoplastic Agents, Immunological/pharmacology
- Cell Line, Tumor
- Drug Carriers/chemistry
- Drug Carriers/pharmacology
- Female
- Humans
- Mice, Nude
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Polyethylene Glycols/chemistry
- Polyethylene Glycols/pharmacology
- Receptor, ErbB-2/antagonists & inhibitors
- Xenograft Model Antitumor Assays
- omega-Chloroacetophenone
Collapse
Affiliation(s)
- Christina L Parker
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, United States
| | - Morgan D McSweeney
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, United States
| | - Andrew T Lucas
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, United States; UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, United States; Carolina Center for Nanotechnology Excellence, University of North Carolina at Chapel Hill, United States
| | - Timothy M Jacobs
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, United States
| | - Daniel Wadsworth
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, United States
| | - William C Zamboni
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, United States; UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, United States; Carolina Center for Nanotechnology Excellence, University of North Carolina at Chapel Hill, United States
| | - Samuel K Lai
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, United States; Department of Biomedical Engineering, University of North Carolina at Chapel Hill, United States; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, United States.
| |
Collapse
|
19
|
Karbalaei Zadeh Babaki M, Taghiabadi M, Soleimanpour S, Saleh Moghadam M, Mosavat A, Amini AA, Mohammadi A, Rezaee SA. Mycobacterium tuberculosis Ag85b:hfcγ1 recombinant fusion protein as a selective receptor-dependent delivery system for antigen presentation. Microb Pathog 2019; 129:68-73. [DOI: 10.1016/j.micpath.2019.01.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 01/29/2019] [Accepted: 01/30/2019] [Indexed: 02/03/2023]
|
20
|
A fully automated three-step protein purification procedure for up to five samples using the NGC chromatography system. Protein Expr Purif 2019; 153:1-6. [DOI: 10.1016/j.pep.2018.08.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/31/2018] [Accepted: 08/09/2018] [Indexed: 01/15/2023]
|
21
|
Siegemund M, Beha N, Müller D. Production, Purification, and Characterization of Antibody-TNF Superfamily Ligand Fusion Proteins. Methods Mol Biol 2018; 1827:351-364. [PMID: 30196506 DOI: 10.1007/978-1-4939-8648-4_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Antibody-fusion proteins with ligands, e.g., of the TNF superfamily (TNFSF) can be adequately produced in mammalian expression systems. Here, we describe the transient production in adherent and suspension human embryonic kidney cells at laboratory scale, followed by purification procedures applying protein A and immobilized metal affinity chromatography for proteins with Fc domain and 6 × histidine-tag, respectively. In addition, characterization of the purified proteins by size exclusion chromatography is described.
Collapse
Affiliation(s)
- Martin Siegemund
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Nadine Beha
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Dafne Müller
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.
| |
Collapse
|
22
|
Khademi F, Derakhshan M, Yousefi-Avarvand A, Tafaghodi M, Soleimanpour S. Multi-stage subunit vaccines against Mycobacterium tuberculosis: an alternative to the BCG vaccine or a BCG-prime boost? Expert Rev Vaccines 2017; 17:31-44. [DOI: 10.1080/14760584.2018.1406309] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Farzad Khademi
- Department of Microbiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Derakhshan
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arshid Yousefi-Avarvand
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Tafaghodi
- Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saman Soleimanpour
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Reference Tuberculosis Laboratory, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
23
|
Construction and immunogenicity of a new Fc-based subunit vaccine candidate against Mycobacterium tuberculosis. Mol Biol Rep 2016; 43:911-22. [PMID: 27251218 DOI: 10.1007/s11033-016-4024-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 05/27/2016] [Indexed: 12/17/2022]
Abstract
As an ancient disease, tuberculosis (TB) is a major global health threat. Therefore, there is an urgent need for an effective and safe anti-TB vaccine. In the current study, a delivery system of Fc domain of mouse IgG2a and early secreted antigenic target protein 6 (ESAT-6) was evaluated for the selective uptake of antigens by antigen-presenting cells (APCs). Thus, it was based on the immunogenicity of a fusion protein. The study was initiated by the transfer of recombinant expression vectors of pPICZαA-ESAT-6:Fcγ2a and pPICZαA-ESAT-6: His into Pichia pastoris (P. pastoris). Recombinant proteins were assessed for immunogenicity following the immunoblotting analysis. High levels of IFN-γ and IL-12 were produced to induce Th1-type cellular responses through vaccination with both recombinant proteins [ESAT-6:Fcγ2a (EF) and ESAT-6:His (EH)]. The Fc-tagged recombinant protein induced more effective Th1-type cellular responses with a low increment in IL-4 compared to PBS, BCG, and EH groups. Although in all the immunized groups, the ratio of IFN-γ/IL-4 was in favor of Th1 responses, the highest Th1/Th2 balance was observed in EF immunized group. Fc fragment of mouse IgG2a may induce a selective uptake of APCs towards the cross-presentation and formation of Th1 responses in favor of an appropriate protective anti-tuberculosis reaction. Thus, further research on Fc-fusion proteins is required to develop Fc-based TB vaccines.
Collapse
|