1
|
Vinayak S, Cecil DL, Disis ML. Vaccines for breast cancer prevention: Are we there yet? Mol Aspects Med 2024; 98:101292. [PMID: 38991631 DOI: 10.1016/j.mam.2024.101292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 06/10/2024] [Accepted: 06/30/2024] [Indexed: 07/13/2024]
Affiliation(s)
- Shaveta Vinayak
- University of Washington, Division of Oncology, Seattle, WA, USA; Fred Hutchinson Cancer Center, Seattle, WA, USA.
| | - Denise L Cecil
- University of Washington, Division of Oncology, Seattle, WA, USA
| | - Mary L Disis
- University of Washington, Division of Oncology, Seattle, WA, USA; Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|
2
|
Nugraha MF, Changestu DA, Ramadhan R, Salsabila T, Nurizati A, Pratiwi SE, Ysrafil Y. Novel prophylactic and therapeutic multi-epitope vaccine based on Ag85A, Ag85B, ESAT-6, and CFP-10 of Mycobacterium tuberculosis using an immunoinformatics approach. Osong Public Health Res Perspect 2024; 15:286-306. [PMID: 39091165 PMCID: PMC11391370 DOI: 10.24171/j.phrp.2024.0026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/26/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Current tuberculosis (TB) control strategies face limitations, such as low antibiotic treatment compliance and a rise in multidrug resistance. Furthermore, the lack of a safe and effective vaccine compounds these challenges. The limited efficacy of existing vaccines against TB underscores the urgency for innovative strategies, such as immunoinformatics. Consequently, this study aimed to design a targeted multi-epitope vaccine against TB infection utilizing an immunoinformatics approach. METHODS The multi-epitope vaccine targeted Ag85A, Ag85B, ESAT-6, and CFP-10 proteins. The design adopted various immunoinformatics tools for cytotoxic T lymphocyte (CTL), helper T lymphocyte (HTL), and linear B lymphocyte (LBL) epitope prediction, the assessment of vaccine characteristics, structure modeling, population coverage analysis, disulfide engineering, solubility prediction, molecular docking/dynamics with toll-like receptors (TLRs), codon optimization/cloning, and immune simulation. RESULTS The multi-epitope vaccine, which was assembled using 12 CTL, 25 HTL, and 21 LBL epitopes associated with CpG adjuvants, showed promising characteristics. The immunoinformatics analysis confirmed the antigenicity, immunogenicity, and lack of allergenicity. Physicochemical evaluations indicated that the proteins were stable, thermostable, hydrophilic, and highly soluble. Docking simulations suggested high-affinity binding to TLRs, including TLR2, TLR4, and TLR9. In silico immune simulation predicted strong T cell (cytokine release) and B cell (immunoglobulin release) responses. CONCLUSION This immunoinformatics-designed multi-epitope vaccine targeting Ag85A, Ag85B, ESAT-6, and CFP-10 proteins showed promising characteristics in terms of stability, immunogenicity, antigenicity, solubility, and predicted induction of humoral and adaptive immune responses. This suggests its potential as a prophylactic and therapeutic vaccine against TB.
Collapse
Affiliation(s)
| | | | - Rizky Ramadhan
- Medical Program, Faculty of Medicine, Universitas Tanjungpura, Pontianak, Indonesia
| | - Tasya Salsabila
- Medical Program, Faculty of Medicine, Universitas Tanjungpura, Pontianak, Indonesia
| | - Arsila Nurizati
- Medical Program, Faculty of Medicine, Universitas Tanjungpura, Pontianak, Indonesia
| | - Sari Eka Pratiwi
- Department of Biology and Pathobiology, Faculty of Medicine, Universitas Tanjungpura, Pontianak, Indonesia
| | - Ysrafil Ysrafil
- Department of Pharmacotherapy, Faculty of Medicine, Universitas Palangka Raya, Palangka Raya, Indonesia
| |
Collapse
|
3
|
Samman N, Mohabatkar H, Behbahani M, Ganjlikhani Hakemi M. Bioinformatics design of a peptide vaccine containing sarcoma antigen NY-SAR-35 epitopes against breast cancer and evaluation of its immunological function in BALB/c mouse model. PLoS One 2024; 19:e0306117. [PMID: 38923980 PMCID: PMC11207152 DOI: 10.1371/journal.pone.0306117] [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: 01/21/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
The development of a cancer vaccine has become an essential focus in the field of medical biotechnology and immunology. In our study, the NY-SAR-35 cancer/testis antigen was targeted to design a novel peptide vaccine using bioinformatics tools, and BALB/c mice were used to evaluate the vaccine's immunological function. This evaluation involved assessing peptide-specific IgG levels in the serum via ELISA and measuring the levels of IFN-γ, IL-4, and granzyme B in the supernatant of cultured splenocytes. The final vaccine construct consisted of two T lymphocyte epitopes linked by the AAY linker. This construct displayed high antigenicity, non-allergenicity, non-toxicity, stability, and ability to induce IFN-γ and IL-4. It showed stable dynamics with both human MHC-I and II molecules, as well as mouse MHC-II molecules, and revealed strong Van der Waals and electrostatic energies. Emulsifying our peptide vaccine in incomplete Freund's adjuvant resulted in a remarkable increase in the levels of IgG. The splenocytes of mice that received the combination of peptide and adjuvant displayed a noteworthy increase in IFN-γ, IL-4, and granzyme B secretion. Additionally, their lymphocytes exhibited higher proliferation rates compared to the control group. Our data demonstrated that our vaccine could stimulate a robust immune response, making it a promising candidate for cancer prevention. However, clinical trials are necessary to assess its efficacy in humans.
Collapse
Affiliation(s)
- Nour Samman
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Hassan Mohabatkar
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mandana Behbahani
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mazdak Ganjlikhani Hakemi
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
4
|
Mugunthan SP, Venkatesan D, Govindasamy C, Selvaraj D, Harish MC. Systems approach to design multi-epitopic peptide vaccine candidate against fowl adenovirus structural proteins for Gallus gallus domesticus. Front Cell Infect Microbiol 2024; 14:1351303. [PMID: 38881736 PMCID: PMC11177691 DOI: 10.3389/fcimb.2024.1351303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 05/06/2024] [Indexed: 06/18/2024] Open
Abstract
Introduction Fowl adenovirus (FAdV) is a significant pathogen in poultry, causing various diseases such as hepatitis-hydropericardium, inclusion body hepatitis, and gizzard erosion. Different serotypes of FAdV are associated with specific conditions, highlighting the need for targeted prevention strategies. Given the rising prevalence of FAdV-related diseases globally, effective vaccination and biosecurity measures are crucial. In this study, we explore the potential of structural proteins to design a multi-epitope vaccine targeting FAdV. Methods We employed an in silico approach to design the multi-epitope vaccine. Essential viral structural proteins, including hexon, penton, and fiber protein, were selected as vaccine targets. T-cell and B-cell epitopes binding to MHC-I and MHC-II molecules were predicted using computational methods. Molecular docking studies were conducted to validate the interaction of the multi-epitope vaccine candidate with chicken Toll-like receptors 2 and 5. Results Our in silico methodology successfully identified potential T-cell and B-cell epitopes within the selected viral structural proteins. Molecular docking studies revealed strong interactions between the multi-epitope vaccine candidate and chicken Toll-like receptors 2 and 5, indicating the structural integrity and immunogenic potential of the designed vaccine. Discussion The designed multi-epitope vaccine presents a promising approach for combating FAdV infections in chickens. By targeting essential viral structural proteins, the vaccine is expected to induce a robust immunological response. The in silico methodology utilized in this study provides a rapid and cost-effective means of vaccine design, offering insights into potential vaccine candidates before experimental validation. Future studies should focus on in vitro and in vivo evaluations to further assess the efficacy and safety of the proposed vaccine.
Collapse
Affiliation(s)
| | | | - Chandramohan Govindasamy
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Dhivya Selvaraj
- Artificial Intelligence Laboratory, School of Computer Information and Communication Engineering, Kunsan National University, Gunsan, Republic of Korea
| | - Mani Chandra Harish
- Department of Biotechnology, Thiruvalluvar University, Vellore, Tamil Nadu, India
| |
Collapse
|
5
|
Saleh RO, Ibrahim FM, Pallathadka H, Kaur I, Ahmad I, Ali SHJ, Redhee AH, Ghildiyal P, Jawad MA, Alsaadi SB. Nucleic acid vaccines-based therapy for triple-negative breast cancer: A new paradigm in tumor immunotherapy arena. Cell Biochem Funct 2024; 42:e3992. [PMID: 38551221 DOI: 10.1002/cbf.3992] [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: 01/21/2024] [Revised: 03/04/2024] [Accepted: 03/10/2024] [Indexed: 04/02/2024]
Abstract
Nucleic acid vaccines (NAVs) have the potential to be economical, safe, and efficacious. Furthermore, just the chosen antigen in the pathogen is the target of the immune responses brought on by NAVs. Triple-negative breast cancer (TNBC) treatment shows great promise for nucleic acid-based vaccines, such as DNA (as plasmids) and RNA (as messenger RNA [mRNA]). Moreover, cancer vaccines offer a compelling approach that can elicit targeted and long-lasting immune responses against tumor antigens. Bacterial plasmids that encode antigens and immunostimulatory molecules serve as the foundation for DNA vaccines. In the 1990s, plasmid DNA encoding the influenza A nucleoprotein triggered a protective and targeted cytotoxic T lymphocyte (CTL) response, marking the first instance of DNA vaccine-mediated immunity. Similarly, in vitro transcribed mRNA was first successfully used in animals in 1990. At that point, mice were given an injection of the gene encoding the mRNA sequence, and the researchers saw the production of a protein. We begin this review by summarizing our existing knowledge of NAVs. Next, we addressed NAV delivery, emphasizing the need to increase efficacy in TNBC.
Collapse
Affiliation(s)
- Raed Obaid Saleh
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq
| | - Fatma M Ibrahim
- Community Health Nursing, RAK Medical and Health Sciences University, Ras Al Khaimah, UAE
- Geriatric Nursing, Mansoura University, Mansoura, Egypt
| | | | - Irwanjot Kaur
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, India
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Saad Hayif Jasim Ali
- Department of Medical Laboratory, College of Health and Medical Technololgy, Al-Ayen University, Thi-Qar, Iraq
| | - Ahmed Huseen Redhee
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| | - Pallavi Ghildiyal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | | | - Salim B Alsaadi
- Department of Pharmaceutics, Al-Hadi University College, Baghdad, Iraq
| |
Collapse
|
6
|
Shahab M, Aiman S, Alshammari A, Alasmari AF, Alharbi M, Khan A, Wei DQ, Zheng G. Immunoinformatics-based potential multi-peptide vaccine designing against Jamestown Canyon Virus (JCV) capable of eliciting cellular and humoral immune responses. Int J Biol Macromol 2023; 253:126678. [PMID: 37666399 DOI: 10.1016/j.ijbiomac.2023.126678] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/21/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023]
Abstract
Jamestown Canyon virus (JCV) is a deadly viral infection transmitted by various mosquito species. This mosquito-borne virus belongs to Bunyaviridae family, posing a high public health threat in the in tropical regions of the United States causing encephalitis in humans. Common symptoms of JCV include fever, headache, stiff neck, photophobia, nausea, vomiting, and seizures. Despite the availability of resources, there is currently no vaccine or drug available to combat JCV. The purpose of this study was to develop an epitope-based vaccine using immunoinformatics approaches. The vaccine aimed to be secure, efficient, bio-compatible, and capable of stimulating both innate and adaptive immune responses. In this study, the protein sequence of JCV was obtained from the NCBI database. Various bioinformatics methods, including toxicity evaluation, antigenicity testing, conservancy analysis, and allergenicity assessment were utilized to identify the most promising epitopes. Suitable linkers and adjuvant sequences were used in the design of vaccine construct. 50s ribosomal protein sequence was used as an adjuvant at the N-terminus of the construct. A total of 5 CTL, 5 HTL, and 5 linear B cell epitopes were selected based on non-allergenicity, immunological potential, and antigenicity scores to design a highly immunogenic multi-peptide vaccine construct. Strong interactions between the proposed vaccine and human immune receptors, i.e., TLR-2 and TLR-4, were revealed in a docking study using ClusPro software, suggesting their possible relevance in the immunological response to the vaccine. Immunological and physicochemical properties assessment ensured that the proposed vaccine demonstrated high immunogenicity, solubility and thermostability. Molecular dynamics simulations confirmed the strong binding affinities, as well as dynamic and structural stability of the proposed vaccine. Immune simulation suggest that the vaccine has the potential to effectively stimulate cellular and humoral immune responses to combat JCV infection. Experimental and clinical assays are required to validate the results of this study.
Collapse
Affiliation(s)
- Muhammad Shahab
- State key laboratories of chemical Resources Engineering Beijing University of chemical technology, Beijing 100029, China
| | - Sara Aiman
- Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing 100124, China
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh 11451, Saudi Arabia
| | - Abdullah F Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh 11451, Saudi Arabia
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh 11451, Saudi Arabia
| | - Abbas Khan
- Deparment of Biostatistics and Bioinformatics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China; School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia.
| | - Dong-Qing Wei
- Deparment of Biostatistics and Bioinformatics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
| | - Guojun Zheng
- State key laboratories of chemical Resources Engineering Beijing University of chemical technology, Beijing 100029, China.
| |
Collapse
|
7
|
Zeng X, Nong WX, Zou XQ, Li F, Ge YY, Zhang QM, Luo B, Huang W, Zou JX, Fan R, Xie XX. Prediction and identification of HLA-A*0201-restricted epitopes from cancer testis antigen CT23. Hum Vaccin Immunother 2023; 19:2293299. [PMID: 38100550 PMCID: PMC10730135 DOI: 10.1080/21645515.2023.2293299] [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: 09/03/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023] Open
Abstract
Cancer-testis antigen CT23 is a class of tumor-associated antigens (TAA) characterized by restricted expression in male germ cells and a variety of tumor tissues. Numerous studies have shown that CT23 is closely related to tumor cell viability, proliferation, metastasis and invasion. CT23 is immunogenic and can cause specific immune response in tumor patients. Therefore, it is considered to be one of the best target antigens for designing therapeutic tumor vaccines and T-cell-mediated tumor immunotherapy. In this study, we initially obtained seven HLA-A*0201-restricted CT23 epitope candidate peptides through the T cell epitope prediction program. Subsequently, a T2 cell binding assay revealed the potential binding of all candidate peptides with HLA-A2 molecules. Notably, peptide P7 (ALLVLCYSI) exhibited the highest affinity, as evidenced by a fluorescence index (FI) of 2.19. Dendritic cells (DCs) loaded with CT23 candidate peptide can stimulate CD8+T cell activation and proliferation, and compared with other candidate peptides, candidate peptide P7 is superior. The cytotoxic T lymphocytes (CTLs) stimulated by the peptide P7 had killing effect on tumor cells (HLA-A*0201+, CT23+), but no killing effect on tumor cells (HLA-A*0201-, CT23+). The CTLs induced by the peptide P7 also had a specific killing effect on T2 cells bearing the peptide P7. In summary, our findings suggest that the CT23 peptide P7 (ALLVLCYSI) can induce immune responses and holds potential for tumor-specific CTL therapy.
Collapse
Affiliation(s)
- Xia Zeng
- Department of Immunology, School of Basic Medicine Science, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Wei-Xia Nong
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Xiao-Qiong Zou
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Feng Li
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Ying-Ying Ge
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Qing-Mei Zhang
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Nanning, P. R. China
| | - Bin Luo
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Nanning, P. R. China
| | - Wei Huang
- Department of Gynecology, First Affiliated Hospital, Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Jian-Xia Zou
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Rong Fan
- Department of Histology and Embryology, School of Pre-Clinical Medicine, Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Xiao-Xun Xie
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Nanning, P. R. China
- Ministry of Education, Key Laboratory of Early Prevention and Treatment of Regional High Frequency Tumor (Guangxi Medical University), Nanning, P. R. China
| |
Collapse
|
8
|
Zhang C, Li M, Yu T. Bioinformatics analysis of Muscovy duck parvovirus REP and VP1 proteins. J Biomol Struct Dyn 2023; 41:7174-7189. [PMID: 36065642 DOI: 10.1080/07391102.2022.2118170] [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: 05/09/2022] [Accepted: 08/20/2022] [Indexed: 10/14/2022]
Abstract
This article was aimed at analyzing the sequence, structure, and function of the two Muscovy duck parvovirus proteins, including REP and VP1. The antigenicity, physical and chemical properties, transmembrane regions, phosphorylation sites, glycosylation sites, three-dimensional structure, and linear epitope of VP1 and REP were predicted and analyzed through bioinformatics methods. A multi-epitope vaccine was also constructed based on the screened epitopes, and the vaccine was characterized, modeled, molecularly docked and molecularly cloned. The epitopes were screened according to the criteria of antigenicity, non-allergenicity and non-toxicity, and 12 epitope fragments were obtained. The B cell epitopes were analyzed according to four scales: β-turn, hydrophilicity, surface accessibility and antigenicity. Combined with the epitope prediction results based on structure, the final epitope prediction results were obtained. The multi-epitope vaccine used an EAAAK-linked adjuvant, a GPGPG-linked T-cell epitope, and a KK-linked B-cell epitope. The analysis showed that the vaccine was stable hydrophilic, antigenic, conserved and non-allergenic. Based on molecular docking it was shown that good interactions between the vaccine and the immune receptor were generated and were essential to generate an immune response. The final vaccine was reverse translated into cDNA and the DNA vaccine was designed by codon optimization and molecular cloning. Further trials are still needed to demonstrate the immunogenicity and other aspects of vaccine efficacy.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Chi Zhang
- College of Computer and Control Engineering, Qiqihar University, Qiqihar, China
| | - Ming Li
- College of Computer and Control Engineering, Qiqihar University, Qiqihar, China
| | - Tianfei Yu
- College of Computer and Control Engineering, Qiqihar University, Qiqihar, China
| |
Collapse
|
9
|
Mohammadzadeh Hosseini Moghri SAH, Ranjbar M, Hassannia H, Khakdan F. In silico analysis of the conserved surface-exposed epitopes to design novel multiepitope peptide vaccine for all variants of the SARS-CoV-2. J Biomol Struct Dyn 2023; 41:7603-7615. [PMID: 36124826 DOI: 10.1080/07391102.2022.2123395] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/04/2022] [Indexed: 10/14/2022]
Abstract
Recently the prevalence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a pervasive threat to generic health. The SARS-CoV-2 spike (S) glycoprotein plays a fundamental role in binds and fusion to the angiotensin-converting enzyme 2 (ACE2). The multi-epitope peptide vaccines would be able to elicit both long-lasting humoral and cellular immune responses, resulting the eliminating SARS-CoV-2 infections as asymptomatic patients are in large numbers. Recently, the omicron variant of the SARS-CoV-2 became a variant of concern that contained just 15-point mutations in the receptor-binding domain of the spike protein. In order to eliminate new evidence on coronavirus variants of concern detected through epidemic intelligence, the conserved epitopes of the receptor-binding domain (RBD) and spike cleavage site is the most probable target for vaccine development to inducing binds and fusion inhibitors neutralizing antibodies respectively. In this study, we utilized bioinformatics tools for identifying and analyzing the spike (S) glycoprotein sequence, e.g. the prediction of the potential linear B-cell epitopes, B-cell multi‑epitope design, secondary and tertiary structures, physicochemical properties, solubility, antigenicity, allergenicity, the molecular docking and molecular dynamics simulation for the promising vaccine candidate against all variant of concern of SARS-CoV-2. Among the epitopes of the RBD region are surface-exposed epitopes SVYAWNRKRISNCV and ATRFASVYAWNRKR as the conserved sequences in all variants of concern can be a good candidate to induce an immune response.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - Mojtaba Ranjbar
- Microbial Biotechnology Department, Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Islamic Republic of Iran
| | - Hadi Hassannia
- Immunogenetic Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | | |
Collapse
|
10
|
Shi D, Chen Y, Chen M, Zhou T, Xu F, Zhang C, Wang C, Li Z. Bioinformatics analysis of Omp19 and Omp25 proteins for designing multi-epitope vaccines against Brucella. Medicine (Baltimore) 2023; 102:e33182. [PMID: 36930131 PMCID: PMC10019172 DOI: 10.1097/md.0000000000033182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/14/2023] [Indexed: 03/18/2023] Open
Abstract
Brucellosis is a zoonotic disease caused by Brucella. There is no effective vaccine against human brucellosis. Omp19 and Omp25 are the outer membrane proteins of Brucella. They are widely expressed and highly conserved in Brucella and have high immunogenicity. Herein, we aim to identify multi-epitope vaccine candidates based on Omp19 and Omp25. We analyzed the physicochemical properties and protein structure of Omp19 and Omp25, and predicted the corresponding B cell and T cell epitopes using bioinformatics analysis. Omp19 and Omp25 were composed of 177 amino acids and 213 amino acids, respectively. They were both stable hydrophilic proteins. The instability indices were 44.8 and 23, respectively. The hydrophilicity was -0.1 and -0.317, respectively. In the secondary structure of Omp19 and Omp25 proteins, the α-helix accounted for 12.43% and 23.94%, the β-sheet was 18.64% and 23.47%, the β-turn was 6.78% and 4.23%, and the random coil was 62.15% and 48.36%. Finally, 5 B cell epitopes, 3 Th-cell epitopes and 5 CTL cell epitopes of Omp19 protein, and 4 B cell epitopes, 3 Th-cell epitopes, and 5 CTL cell epitopes of Omp25 protein were selected as vaccine candidates. In conclusion, we obtained potential B cell and T cell epitopes of the Brucella outer membrane Omp19 and Omp25 proteins. This lays the foundation for the further design of multi-epitope vaccine of Brucella.
Collapse
Affiliation(s)
- Donghao Shi
- Clinical Laboratory Center, Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, Urumqi, P.R. China
| | - Yuan Chen
- Clinical Laboratory Center, People`s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, P.R. China
| | - Muzhi Chen
- Department of Rheumatology, Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, Urumqi, P.R. China
| | - Tingting Zhou
- Department of Public Health, People`s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, P.R. China
| | - Feili Xu
- Clinical Laboratory Center, Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, Urumqi, P.R. China
| | - Chao Zhang
- Urumqi OE Biotech Co., Ltd., Urumqi, Xinjiang, P.R. China
| | - Changmin Wang
- Clinical Laboratory Center, People`s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, P.R. China
| | - Zhiwei Li
- Clinical Laboratory Center, People`s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, P.R. China
| |
Collapse
|
11
|
Rahimnahal S, Yousefizadeh S, Mohammadi Y. Novel multi-epitope vaccine against bovine brucellosis: approach from immunoinformatics to expression. J Biomol Struct Dyn 2023; 41:15460-15484. [PMID: 36927475 DOI: 10.1080/07391102.2023.2188962] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 03/02/2023] [Indexed: 03/18/2023]
Abstract
Brucellosis is a zoonotic caused by the Brucella which is a well-known infectious disease agent in domestic animals and if transmitted, it can cause infection in humans. Because brucellosis is contagious, its control depends on the eradication of the animal disease in farms. There are two vaccines based on the killed and/or weakened bacteria against B. melitensis and B. abortus, but no recombinant vaccine is available for preventing the disease. The present study was designed to develop a multi-epitope vaccine against of B. melitensis and B. abortus using virB10, Omp31 and Omp16 antigens by the prediction of T lymphocytes, T cell cytotoxicity and IFN-γ epitopes. 50S L7/L12 Ribosomal protein from Mycobacterium tuberculosis was used as a bovine TLR4 and TLR9 agonist. GPGPG, AAY and KK linkers were used as a linker. Brucella construct was well-integrated in the pET-32a Shuttle vector with BamHI and HindIII restriction enzymes. The final construct contained 769 amino acids, that it was soluble protein of about ∼82 kDa after expression in the Escherichia coli SHuffle host. Modeled protein analysis based on the tertiary structure validation, molecular docking studies, molecular dynamics simulations results like RMSD, Gyration and RMSF as well as MM/PBSA analysis showed that this protein has a stable construct and is capable being in interaction with bovine TLR4 and TLR9. Analysis of the data obtained suggests that the proposed vaccine can induce the immune response by stimulating T- and B-cells, and may be used for prevention and remedial purposes, against B. melitensis and B. abortus.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Somayyeh Rahimnahal
- Department of Animal Science, Faculty of Agriculture, Ilam University, Ilam, Iran
| | - Shahnaz Yousefizadeh
- Department of Laboratory and Clinical Sciences, Faculty of Para-Veterinary, Ilam University, Ilam, Iran
| | - Yahya Mohammadi
- Department of Animal Science, Faculty of Agriculture, Ilam University, Ilam, Iran
| |
Collapse
|
12
|
Ali Z, Cardoza JV, Basak S, Narsaria U, Singh VP, Isaac SP, França TCC, LaPlante SR, George SS. Computational design of candidate multi-epitope vaccine against SARS-CoV-2 targeting structural (S and N) and non-structural (NSP3 and NSP12) proteins. J Biomol Struct Dyn 2023; 41:13348-13367. [PMID: 36744449 DOI: 10.1080/07391102.2023.2173297] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 01/20/2023] [Indexed: 02/07/2023]
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 virus has created a global damage and has exposed the vulnerable side of scientific research towards novel diseases. The intensity of the pandemic is huge, with mortality rates of more than 6 million people worldwide in a span of 2 years. Considering the gravity of the situation, scientists all across the world are continuously attempting to create successful therapeutic solutions to combat the virus. Various vaccination strategies are being devised to ensure effective immunization against SARS-CoV-2 infection. SARS-CoV-2 spreads very rapidly, and the infection rate is remarkably high than other respiratory tract viruses. The viral entry and recognition of the host cell is facilitated by S protein of the virus. N protein along with NSP3 is majorly responsible for viral genome assembly and NSP12 performs polymerase activity for RNA synthesis. In this study, we have designed a multi-epitope, chimeric vaccine considering the two structural (S and N protein) and two non-structural proteins (NSP3 and NSP12) of SARS-CoV-2 virus. The aim is to induce immune response by generating antibodies against these proteins to target the viral entry and viral replication in the host cell. In this study, computational tools were used, and the reliability of the vaccine was verified using molecular docking, molecular dynamics simulation and immune simulation studies in silico. These studies demonstrate that the vaccine designed shows steady interaction with Toll like receptors with good stability and will be effective in inducing a strong and specific immune response in the body.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Zeeshan Ali
- Krupanidhi College of Physiotherapy, Bangalore, India
| | | | | | | | - Vijay Pratap Singh
- Department of Physiotherapy, Kasturba Medical College, Mangalore, Manipal academy of higher education, Mangalore, Manipal, India
| | | | - Tanos C C França
- Université de Québec, INRS - Centre Armand-Frappier Santé Biotechnologie, Laval, Québec, Canada
- Laboratory of Molecular Modeling Applied to Chemical and Biological Defense, Military Institute of Engineering, Rio de Janeiro, Brazil
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Steven R LaPlante
- Université de Québec, INRS - Centre Armand-Frappier Santé Biotechnologie, Laval, Québec, Canada
| | | |
Collapse
|
13
|
Jiang F, Liu Y, Xue Y, Cheng P, Wang J, Lian J, Gong W. Developing a multiepitope vaccine for the prevention of SARS-CoV-2 and monkeypox virus co-infection: A reverse vaccinology analysis. Int Immunopharmacol 2023; 115:109728. [PMID: 36652758 PMCID: PMC9832108 DOI: 10.1016/j.intimp.2023.109728] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/01/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023]
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and monkeypox virus (MPXV) severely threaten human health; however, currently, no vaccine can prevent a co-infection with both viruses. METHODS Five antigens were selected to predict dominant T and B cell epitopes screened for immunogenicity, antigenicity, toxicity, and sensitization. After screening, all antigens joined in the construction of a novel multiepitope vaccine. The physicochemical and immunological characteristics, and secondary and tertiary structures of the vaccine were predicted and analyzed using bio- and immunoinformatics. Finally, codon optimization and cloning in-silico were performed. RESULTS A new multiepitope vaccine, named S7M8, was constructed based on four helper T lymphocyte (HTL) epitopes, six cytotoxic T lymphocyte (CTL) epitopes, five B cell epitopes, as well as Toll-like receptor (TLR) agonists. The antigenicity and immunogenicity scores of the S7M8 vaccine were 0.907374 and 0.6552, respectively. The S7M8 vaccine was comprised of 26.96% α-helices, the optimized Z-value of the tertiary structure was -5.92, and the favored area after majorization in the Ramachandran plot was 84.54%. Molecular docking showed that the S7M8 vaccine could tightly bind to TLR2 (-1100.6 kcal/mol) and TLR4 (-950.3 kcal/mol). In addition, the immune stimulation prediction indicated that the S7M8 vaccine could activate T and B lymphocytes to produce high levels of Th1 cytokines and antibodies. CONCLUSION S7M8 is a promising biomarker with good antigenicity, immunogenicity, non-toxicity, and non-sensitization. The S7M8 vaccine can trigger significantly high levels of Th1 cytokines and antibodies and may be a potentially powerful tool in preventing SARS-CoV-2 and MPXV.
Collapse
Affiliation(s)
- Fan Jiang
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China; The Second Brigade of Cadet, Basic Medical Science Academy of Air Force Medical University, Xi'an, China; Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Yinping Liu
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Yong Xue
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Peng Cheng
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Jie Wang
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Jianqi Lian
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, China.
| | - Wenping Gong
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China.
| |
Collapse
|
14
|
Reverse vaccinology assisted design of a novel multi-epitope vaccine to target Wuchereria bancrofti cystatin: An immunoinformatics approach. Int Immunopharmacol 2023; 115:109639. [PMID: 36586276 DOI: 10.1016/j.intimp.2022.109639] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/05/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022]
Abstract
Proteases are the critical mediators of immunomodulation exerted by the filarial parasites to bypass and divert host immunity. Cystatin is a small (∼15 kDa) immunomodulatory filarial protein and known to contribute in the immunomodulation strategy by inducing anti-inflammatory response through alternative activation of macrophages. Recently, Wuchereria bancrofti cystatin has been discovered as a ligand of human toll-like receptor 4 which is key behind the cystatin-induced anti-inflammatory response in major human antigen-presenting cells. Considering the pivotal role of cystatin in the immunobiology of filariasis, cystatin could be an efficacious target for developing vaccine. Herein, we present the design and in-silico analyses of a multi-epitope-based peptide vaccine to target W. bancrofti cystatin through immune-informatics approaches. The 262 amino acid long antigen construct comprises 9 MHC-I epitopes and MHC-II epitopes linked together by GPGPG peptide alongside an adjuvant (50S ribosomal protein L7/L12) at N terminus and 6 His tags at C terminus. Molecular docking study reveals that the peptide could trigger TLR4-MD2 to induce protective innate immune responses while the induced adaptive responses were found to be mediated by IgG, IgM and Th1 mediated responses. Notably, the designed vaccine exhibits high stability and no allergenicity in-silico. Furthermore, the muti epitope-vaccine was also predicted for its RNA structure and cloned in pET30ax for further experimental validation. Taken together, this study presents a novel multi-epitope peptide vaccine for triggering efficient innate and adaptive immune responses against W. bancrofti to intervene LF through immunotherapy.
Collapse
|
15
|
Mahmoudvand S, Esmaeili Gouvarchin Ghaleh H, Jalilian FA, Farzanehpour M, Dorostkar R. Design of a multi-epitope-based vaccine consisted of immunodominant epitopes of structural proteins of SARS-CoV-2 using immunoinformatics approach. Biotechnol Appl Biochem 2023:10.1002/bab.2431. [PMID: 36577011 PMCID: PMC9880719 DOI: 10.1002/bab.2431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 12/11/2022] [Indexed: 12/29/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has shown rapid global spread and has resulted in a significant death toll worldwide. In this study, we aimed to design a multi-epitope vaccine against SARS-CoV-2 based on structural proteins S, M, N, and E. We identified B- and T-cell epitopes and then the antigenicity, toxicity, allergenicity, and similarity of predicted epitopes were analyzed. T-cell epitopes were docked with corresponding HLA alleles. Consequently, the selected T- and B-cell epitopes were included in the final construct. All selected epitopes were connected with different linkers and flagellin and pan-HLA DR binding epitopes (PADRE) as an adjuvant were used in the vaccine construct. Furthermore, molecular docking was used to evaluate the complex between the final vaccine construct and two alleles, HLA-A*02:01 and HLA-DRB1*01:01. Finally, codons were optimized for in silico cloning into pET28a(+) vector using SnapGene. The final vaccine construct comprised 11 CTL, HTL, and B-cell epitopes corresponding to 394 amino acid residues. In silico evaluation showed that the designed vaccine might potentially promote an immune response. Further in vivo preclinical and clinical testing is required to determine the safety and efficacy of the designed vaccine.
Collapse
Affiliation(s)
- Shahab Mahmoudvand
- Applied Virology Research CenterBaqiyatallah University of Medical SciencesTehranIran
| | | | - Farid Azizi Jalilian
- Department of Medical VirologyFaculty of MedicineHamadan University of Medical SciencesHamadanIran
| | - Mahdieh Farzanehpour
- Applied Virology Research CenterBaqiyatallah University of Medical SciencesTehranIran
| | - Ruhollah Dorostkar
- Applied Virology Research CenterBaqiyatallah University of Medical SciencesTehranIran
| |
Collapse
|
16
|
Zhou Y, Zhao S, Li Y, Yu M, Zheng J, Gong Q, Cao C, Ding J, Zhou X. Design and functional preliminary investigation of recombinant antigen EgG1Y162-EgG1Y162 against Echinococcus granulosus. Open Life Sci 2023; 18:20220558. [PMID: 36941829 PMCID: PMC10024343 DOI: 10.1515/biol-2022-0558] [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: 09/11/2022] [Revised: 12/02/2022] [Accepted: 01/02/2023] [Indexed: 03/15/2023] Open
Abstract
In the early stage, our research group cloned Echinococcus granulosus-specific antigen, EgG1Y162, from protoscolex and adult worms of E. granulosus. In order to enhance the immunogenicity of the vaccine, we prepared a recombinant vaccine by tandemly linking EgG1Y162, splicing the protein and linker at the gene level. This approach is expected to improve the immunogenicity of the vaccine by enhancing the molecular weight of the protein and increasing the antigenic epitopes. Bioinformatics was used to predict the physicochemical properties, transmembrane domain, protein structure, and T-/B-cell antigenic epitope of different recombinant proteins, EgG1Y162-linker-EgG1Y162. Finally, the linker sequence, "GGGGSGGG," which had the least influence on the migration of recombinant protein T/B epitope and can fold normally in series with EgG1Y162, was selected to design the recombinant vaccine. The plasmid was produced using genetic engineering techniques, and the recombinant protein, EGG1Y162-GGGGSGGG-EgG1Y162, was induced to be expressed and purified. EgG1Y162-GGGGSGGG-EgG1Y162 was identified to be correctly expressed with 100% specificity. Compared with EgG1Y162, EgG1Y162-GGGGSGGG-EgG1Y162 was more likely to promote dendritic cell maturation. EgG1Y162-GGGGSGGG-EgG1Y162 was speculated to have the potential to improve antigen immunogenicity by increasing the molecular weight and antigenic epitope.
Collapse
Affiliation(s)
- Yanxia Zhou
- Department of Immunology, Basic Medical College, Xinjiang Medical University, Xinjiang830011, Urumqi, China
- Department of Anesthesiology, Zhanjiang Central Hospital, Zhanjiang, 524037, Guangdong, China
| | - Shangqi Zhao
- Department of Immunology, Basic Medical College, Xinjiang Medical University, Xinjiang830011, Urumqi, China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Xinjiang830011, Urumqi, China
| | - Yanmin Li
- Department of Immunology, Basic Medical College, Xinjiang Medical University, Xinjiang830011, Urumqi, China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Xinjiang830011, Urumqi, China
| | - Mingkai Yu
- Department of Immunology, Basic Medical College, Xinjiang Medical University, Xinjiang830011, Urumqi, China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Xinjiang830011, Urumqi, China
| | - Jia Zheng
- Department of Immunology, Basic Medical College, Xinjiang Medical University, Xinjiang830011, Urumqi, China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Xinjiang830011, Urumqi, China
| | - Qiaoqiao Gong
- Department of Immunology, Basic Medical College, Xinjiang Medical University, Xinjiang830011, Urumqi, China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Xinjiang830011, Urumqi, China
| | - Chunbao Cao
- Department of Immunology, Basic Medical College, Xinjiang Medical University, Xinjiang830011, Urumqi, China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Xinjiang830011, Urumqi, China
| | - Jianbing Ding
- Department of Immunology, Basic Medical College, Xinjiang Medical University, Xinjiang830011, Urumqi, China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Xinjiang830011, Urumqi, China
| | - Xiaotao Zhou
- Department of Immunology, Basic Medical College, Xinjiang Medical University, Xinjiang830011, Urumqi, China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Xinjiang830011, Urumqi, China
| |
Collapse
|
17
|
Susithra Priyadarshni M, Isaac Kirubakaran S, Harish MC. In silico approach to design a multi-epitopic vaccine candidate targeting the non-mutational immunogenic regions in envelope protein and surface glycoprotein of SARS-CoV-2. J Biomol Struct Dyn 2022; 40:12948-12963. [PMID: 34528491 PMCID: PMC8477437 DOI: 10.1080/07391102.2021.1977702] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The novel corona virus (COVID-19) is a causative agent for severe acute respiratory syndrome (SARS-CoV-2) and responsible for the current human pandemic situation which has caused global social and economic commotion. The currently available vaccines use whole viruses whereas there is scope for peptide based vaccines. Thus, the global raise in statistics of this infection at an alarming rate evoked us to determine a novel and effective vaccine candidate against SARS-CoV-2. To find the potential vaccine candidate targets, immunoinformatics approaches were used to analyze the mutations in the envelope protein and surface glycoprotein and determine the conserved region; further specific T-cell epitopes VSLVKPSFY, SLVKPSFYV, RVKNLNSSR, SEETGTLIV, LVKPSFYVY, LTDEMIAQY, YLQPRTFLL, RLFRKSNLK, SPRRARSVA, AEIRASANL, TLLALHRSY, YSRVKNLNS and FELLHAPAT and B-cells epitopes TLAILTALRLCAYCCN and AGTITSGWTFGAGAAL were identified. The 3 D structure of epitope was predicted, refined and validated. The molecular docking analysis of multi-epitope vaccine candidates with TLR receptors, predicted effective binding. Overall, using bioinformatics approach this multi-epitopic target facilitates the proof of concept for SARS-CoV-2 conserved epitopic vaccine design.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - S. Isaac Kirubakaran
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, KS, USA
| | - M. C. Harish
- Department of Biotechnology, Thiruvalluvar University, Vellore, Tamil Nadu, India,CONTACT M. C. Harish Department of Biotechnology, Thiruvalluvar University, Serkkadu, Vellore632115, India
| |
Collapse
|
18
|
Epitope-based minigene vaccine targeting fibroblast activation protein α induces specific immune responses and anti-tumor effects in 4 T1 murine breast cancer model. Int Immunopharmacol 2022; 112:109237. [PMID: 36152535 DOI: 10.1016/j.intimp.2022.109237] [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: 04/26/2022] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 11/21/2022]
Abstract
Fibroblast activation protein (FAPα) is a tumor stromal antigen expressed by cancer-associated fibroblasts (CAFs) in more than 90 % of malignant epithelial carcinomas. FAPα-based immunotherapy has been reported and showed that FAPα-specific immune response can remold immune microenvironment and contribute to tumor regression. Many FAPα-based vaccines have been investigated in preclinical trials, which can elicit strong and durable cytolytic T lymphocytes (CTL) with good safety. However, epitope-based FAPα vaccines are rarely reported. To break tolerance against self-antigens, analogue epitopes with modified peptides at the anchor residues are typically used to improve epitope immunogenicity. To investigate the feasibility of a FAPα epitope-based vaccine for cancer immunotherapy in vivo, we conducted a preclinical study to identify a homologous CTL epitope of human and mouse FAPα and obtained its analogue epitope in BALB/c mice, and explored the anti-tumor activity of their minigene vaccines in 4 T1 tumor-bearing mice. By using in silico epitope prediction tools and immunogenicity assays, immunodominant epitope FAP.291 (YYFSWLTWV) and its analogue epitope FAP.291I9 (YYFSWLTWI) were identified. The FAP.291-based epitope minigene vaccine successfully stimulated CTLs targeting CAFs and exhibited anti-tumor activity in a 4 T1 murine breast cancer model. Furthermore, although the analogue epitope FAP.291I9 enhanced FAP.291-specific immune responses, improvement of anti-tumor immunity effects was not observed. Check of immunosuppressive factors revealed that the high levels of IL-10, IL-13, myeloid-derived suppressor cells and iNOS induced by FAP.291I9 increased, which considered the main cause of the failure of the analogue epitope-based vaccine. Thus, we demonstrated for the first time that the FAP.291 minigene vaccine could induce mouse CTLs and also function as a tumor regression antigen, providing the basis for future studies of FAPα epitope-based vaccines. This study may also be valuable for further improvement of the immunogenicity of analogue epitope vaccines.
Collapse
|
19
|
Zhang X, Ge X, Jiang T, Yang R, Li S. Research progress on immunotherapy in triple‑negative breast cancer (Review). Int J Oncol 2022; 61:95. [PMID: 35762339 PMCID: PMC9256074 DOI: 10.3892/ijo.2022.5385] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/15/2022] [Indexed: 11/16/2022] Open
Abstract
Triple‑negative breast cancer (TNBC) is a highly heterogeneous and aggressive malignancy. Due to the absence of estrogen receptors and progesterone receptors and the lack of overexpression of human epidermal growth factor receptor 2, TNBC responds poorly to endocrine and targeted therapies. As a neoadjuvant therapy, chemotherapy is usually the only option for TNBC; however, chemotherapy may induce tumor resistance. The emergence of immunotherapy as an adjuvant therapy is expected to make up for the deficiency of chemotherapy. Most of the research on immunotherapies has been performed on advanced metastatic TNBC, which has provided significant clinical benefits. In the present review, possible immunotherapy targets and ongoing immunotherapy strategies were discussed. In addition, progress in research on immune checkpoint inhibitors in early TNBC was outlined.
Collapse
Affiliation(s)
- Xiaoxiao Zhang
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin 130012, P.R. China
| | - Xueying Ge
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin 130012, P.R. China
| | - Tinghan Jiang
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin 130012, P.R. China
| | - Ruming Yang
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin 130012, P.R. China
| | - Sijie Li
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin 130012, P.R. China
| |
Collapse
|
20
|
Tan C, Zhu F, Xiao Y, Wu Y, Meng X, Liu S, Liu T, Chen S, Zhou J, Li C, Wu A. Immunoinformatics Approach Toward the Introduction of a Novel Multi-Epitope Vaccine Against Clostridium difficile. Front Immunol 2022; 13:887061. [PMID: 35720363 PMCID: PMC9204425 DOI: 10.3389/fimmu.2022.887061] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
Clostridium difficile (C.difficile) is an exclusively anaerobic, spore-forming, and Gram-positive pathogen that is the most common cause of nosocomial diarrhea and is becoming increasingly prevalent in the community. Because C. difficile is strictly anaerobic, spores that can survive for months in the external environment contribute to the persistence and diffusion of C. difficile within the healthcare environment and community. Antimicrobial therapy disrupts the natural intestinal flora, allowing spores to develop into propagules that colonize the colon and produce toxins, thus leading to antibiotic-associated diarrhea and pseudomembranous enteritis. However, there is no licensed vaccine to prevent Clostridium difficile infection (CDI). In this study, a multi-epitope vaccine was designed using modern computer methods. Two target proteins, CdeC, affecting spore germination, and fliD, affecting propagule colonization, were chosen to construct the vaccine so that it could simultaneously induce the immune response against two different forms (spore and propagule) of C. difficile. We obtained the protein sequences from the National Center for Biotechnology Information (NCBI) database. After the layers of filtration, 5 cytotoxic T-cell lymphocyte (CTL) epitopes, 5 helper T lymphocyte (HTL) epitopes, and 7 B-cell linear epitopes were finally selected for vaccine construction. Then, to enhance the immunogenicity of the designed vaccine, an adjuvant was added to construct the vaccine. The Prabi and RaptorX servers were used to predict the vaccine's two- and three-dimensional (3D) structures, respectively. Additionally, we refined and validated the structures of the vaccine construct. Molecular docking and molecular dynamics (MD) simulation were performed to check the interaction model of the vaccine-Toll-like receptor (TLR) complexes, vaccine-major histocompatibility complex (MHC) complexes, and vaccine-B-cell receptor (BCR) complex. Furthermore, immune stimulation, population coverage, and in silico molecular cloning were also conducted. The foregoing findings suggest that the final formulated vaccine is promising against the pathogen, but more researchers are needed to verify it.
Collapse
Affiliation(s)
- Caixia Tan
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, China
| | - Fei Zhu
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanyuan Xiao
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, China
| | - Yuqi Wu
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, China
| | - Xiujuan Meng
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, China
| | - Sidi Liu
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, China
| | - Ting Liu
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, China
| | - Siyao Chen
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, China
| | - Juan Zhou
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, China
| | - Chunhui Li
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, China
| | - Anhua Wu
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, China
| |
Collapse
|
21
|
Fu J, Yingying Ge, Qingmei Zhang, Lin Y, Liu C, Nong W, Luo X, Xiao S, Xie X, Luo B. Immunohistochemistry Study of OY-TES-1 Location in Fetal and Adult Human Tissues. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:7052830. [PMID: 35463688 PMCID: PMC9020931 DOI: 10.1155/2022/7052830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 11/18/2022]
Abstract
OY-TES-1 is reportedly involved in carcinogenesis and spermatogenesis. However, the tissue distribution of OY-TES-1 in the normal human body remains elusive. This study detected OY-TES-1 expression in human fetal and adult normal tissues by immunohistochemistry. We identified a general principle of OY-TES-1 expression. The expression of OY-TES-1 was found in neurons, smooth muscle cells, and cardiac muscle cells from both fetuses and adults. The connective tissue showed no specific staining throughout the fetal and adult samples. With OY-TES-1-positive staining of the epithelium irregular, OY-TES-1 was strongly expressed in the epithelium of the skin and bladder, as well as hepatocytes, pancreatic islets, and acinous cells during the fetal stage but was not detected in the postnatal period. In contrast to the epithelium of blood vessels, the fetal and adult central hepatic vein and glomeruli showed negative expression of the OY-TES-1 protein. Sex-dimorphism was observed in the distribution of OY-TES-1 in male and female germ cells. Collectively, our results indicate that OY-TES-1 is a member of the cancer-testis antigen and autoantigen, with tissue-specific and period-specific expression patterns, revealing potential contributions of OY-TES-1 to the diagnosis and therapeutic treatment for neoplasms and infertility.
Collapse
Affiliation(s)
- Jun Fu
- Department of Histology & Embryology, School of Pre-clinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Yingying Ge
- Department of Histology & Embryology, School of Pre-clinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Qingmei Zhang
- Guangxi Colleges and Universities Key Laboratory Research of Preclinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Yongda Lin
- Department of Histology & Embryology, School of Pre-clinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Chang Liu
- Department of Neurosurgery, First Affiliated Hospital, Guangxi Medical University, Nanning 530021, China
| | - Weixia Nong
- Department of Histology & Embryology, School of Pre-clinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Xin Luo
- Department of Histology & Embryology, School of Pre-clinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Shaowen Xiao
- Department of Neurosurgery, First Affiliated Hospital, Guangxi Medical University, Nanning 530021, China
| | - Xiaoxun Xie
- Guangxi Colleges and Universities Key Laboratory Research of Preclinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Bin Luo
- Guangxi Colleges and Universities Key Laboratory Research of Preclinical Medicine, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
22
|
Peng W, Chang L, Li W, Liu Y, Zhang M. OCT4 and SOX2 Specific Cytotoxic T Cells Exhibit Not Only Good Efficiency but Also Synergize PD-1 Inhibitor (Nivolumab) in Treating Breast Cancer Stem-Like Cells and Drug-Resistant Breast Cancer Mice. Front Oncol 2022; 12:781093. [PMID: 35402219 PMCID: PMC8987438 DOI: 10.3389/fonc.2022.781093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 02/21/2022] [Indexed: 12/24/2022] Open
Abstract
Purpose This study aimed to investigate the effect of OCT4&SOX2 specific cytotoxic T lymphocytes (CTLs) plus programmed cell death protein-1 (PD-1) inhibitor (nivolumab) on treating breast cancer stem-like cells (BCSCs) in vitro and drug-resistance breast cancer (DRBC) mice in vivo. Methods In total, 160 breast cancer patients were enrolled following the immunofluorescence assay to detect tumor OCT4 and SOX2 expressions. CD154-activated B cells were co-cultured with CD8+ T cells (from breast cancer patients) in the presence of OCT4&SOX2 peptides, CMV pp65 peptides (negative control), and no peptides (normal control). MCF7-BCSCs were constructed by drug-resistance experiment and sphere-formation assay, then DRBC mice were constructed by planting MCF7-BCSCs. Subsequently, different doses of OCT4&SOX2 CTLs and PD-1 inhibitor (nivolumab) were used to treat MCF7-BCSCs and DRBC mice. Results OCT4 and SOX2 correlated with poor differentiation, more advanced stage, and worse prognosis in breast cancer patients. In vitro, OCT4&SOX2 CTLs with effector-target ratio (ETR) 5:1, 10:1 and 20:1 presented with increased cytotoxic activity compared to CMV pp65 CTLs with ETR 20:1 (negative control) and Control CTLs with ETR 20:1 (normal control) on killing MCF7-BCSCs. Besides, PD-1 inhibitor (nivolumab) improved the cytotoxic activity of OCT4&SOX2 CTLs against MCF7-BCSCs in a dose-dependent manner. In vivo, OCT4&SOX2 CTLs plus PD-1 inhibitor (nivolumab) decreased tumor volume and tumor weight while increased tumor apoptosis rate compared to OCT4&SOX2 CTLs alone, PD-1 inhibitor (nivolumab) alone, and control. Conclusion OCT4&SOX2 CTLs exhibit good efficiency and synergize PD-1 inhibitor (nivolumab) in treating BCSCs and DRBC.
Collapse
Affiliation(s)
- Wei Peng
- Department of General Surgery, Guangdong Provincial People's Hospital Zhuhai Hospital (Zhuhai Golden Bay Center Hospital), Zhuhai, China
| | - Liang Chang
- Department of General Surgery, Guangdong Provincial People's Hospital Zhuhai Hospital (Zhuhai Golden Bay Center Hospital), Zhuhai, China
| | - Wenqiang Li
- Department of General Surgery, Guangdong Provincial People's Hospital Zhuhai Hospital (Zhuhai Golden Bay Center Hospital), Zhuhai, China
| | - Yanan Liu
- Department of Intensive Care Unit, Guangdong Provincial People's Hospital Zhuhai Hospital (Zhuhai Golden Bay Center Hospital), Zhuhai, China
- *Correspondence: Yanan Liu, ; Min Zhang,
| | - Min Zhang
- Department of General Surgery, Cixi People’s Hospital, Ningbo, China
- *Correspondence: Yanan Liu, ; Min Zhang,
| |
Collapse
|
23
|
Qiu D, Zhang G, Yan X, Xiao X, Ma X, Lin S, Wu J, Li X, Wang W, Liu J, Ma Y, Ma M. Prospects of Immunotherapy for Triple-Negative Breast Cancer. Front Oncol 2022; 11:797092. [PMID: 35111680 PMCID: PMC8801574 DOI: 10.3389/fonc.2021.797092] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/30/2021] [Indexed: 01/22/2023] Open
Abstract
In the classification and typing of breast cancer, triple-negative breast cancer (TNBC) is one type of refractory breast cancer, while chemotherapy stays in the traditional treatment methods. However, the impact of chemotherapy is short-lived and may lead to recurrence due to incomplete killing of tumor cells. The occurrence, development, and relapse of breast cancer are relevant to T cell dysfunction, multiplied expression of related immune checkpoint molecules (ICIs) such as programmed death receptor 1 (PD-1), programmed cell death 1 ligand 1 (PD-L1), and cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) produce immunosuppressive effect. Immunotherapy (namely, immune checkpoint inhibitors, adoptive cellular immunotherapy, CAR-T immunotherapy and some potential treatments) provides new hope in TNBC. This review focuses on the new immune strategies of TNBC patients.
Collapse
Affiliation(s)
- Dan Qiu
- School of Traditional Chinese Medicine of Jinan University, Jinan University, Guangzhou, China
| | - Guijuan Zhang
- School of Nursing of Jinan University, Jinan University, Guangzhou, China
| | - Xianxin Yan
- School of Traditional Chinese Medicine of Jinan University, Jinan University, Guangzhou, China
| | - Xinqin Xiao
- School of Traditional Chinese Medicine of Jinan University, Jinan University, Guangzhou, China
| | - Xinyi Ma
- School Public Health, Southern Medical University (No: 3210090112), Guangzhou, China
| | - Shujun Lin
- School of Traditional Chinese Medicine of Jinan University, Jinan University, Guangzhou, China
| | - Jieyan Wu
- School of Traditional Chinese Medicine of Jinan University, Jinan University, Guangzhou, China
| | - Xinyuan Li
- School of Medicine, Jinan University, Guangzhou, China
| | - Wandi Wang
- School of Medicine, Jinan University, Guangzhou, China
| | - Junchen Liu
- School of Medicine, Jinan University, Guangzhou, China
| | - Yi Ma
- Department of Cellular Biology, Institute of Biomedicine, National Engineering, Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, The National Demonstration Center for Experimental Education of Life Science and Technology, Jinan University, Guangzhou, China
| | - Min Ma
- School of Traditional Chinese Medicine of Jinan University, Jinan University, Guangzhou, China.,The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| |
Collapse
|
24
|
Roberto Raúl SG, Damaris IA, Ángel de Jesús JC, Leticia MF. Cry1Ac Protoxin Confers Antitumor Adjuvant Effect in a Triple-Negative Breast Cancer Mouse Model by Improving Tumor Immunity. BREAST CANCER: BASIC AND CLINICAL RESEARCH 2022; 16:11782234211065154. [PMID: 35002244 PMCID: PMC8738886 DOI: 10.1177/11782234211065154] [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] [Received: 11/22/2021] [Accepted: 11/17/2021] [Indexed: 12/07/2022] Open
Abstract
The Cry1Ac protoxin from Bacillus thuringiensis is a systemic
and mucosal adjuvant, able to confer protective immunity in different infection
murine models and induce both Th1 and TCD8+ cytotoxic lymphocyte responses,
which are required to induce antitumor immunity. The Cry1Ac toxin, despite
having not being characterized as an adjuvant, has also proved to be immunogenic
and able to activate macrophages. Here, we investigated the potential antitumor
adjuvant effect conferred by the Cry1Ac protoxin and Cry1Ac toxin in a triple
negative breast cancer (TNBC) murine model. First, we evaluated the ability of
Cry1Ac proteins to improve dendritic cell (DC) activation and cellular response
through intraperitoneal (i.p.) coadministration with the 4T1 cellular lysate.
Mice coadministered with the Cry1Ac protoxin showed an increase in the number
and activation of CD11c+MHCII- and CD11c+MHCII+low in the peritoneal
cavity and an increase in DC activation (CD11c+MHCII+) in the spleen. Cry1Ac
protoxin increased the proliferation of TCD4+ and TCD8+ lymphocytes in the
spleen and mesenteric lymph nodes (MLN), while the Cry1Ac toxin only increased
the proliferation of TCD4+ and TCD8+ in the MLN. Remarkably, when tested in the
in vivo TNBC mouse model, prophylactic immunizations with 4T1 lysates plus the
Cry1Ac protoxin protected mice from developing tumors. The antitumor effect
conferred by the Cry1Ac protoxin also increased specific cytotoxic T cell
responses, and prevented the typical tumor-related decrease of T cells
(TCD3+ and TCD4+) as well the increase of myeloid-derived suppressor cells
(MDSC) in spleen. Also in the tumor microenvironment of mice coadministered
twice with Cry1Ac protoxin immunological improvements were found such as
reductions in immunosupressive populations (T regulatory lymphocytes and MDSC)
along with increases in macrophages upregulating CD86. These results show a
differential antitumor adjuvant capability of Cry1Ac proteins, highlighting the
ability of Cry1Ac protoxin to enhance local and systemic tumor immunity in TNBC.
Finally, using a therapeutic approach, we evaluated the coadministration of
Cry1Ac protoxin with doxorubicin. A significant reduction in tumor volume and
lung metastasis was found, with increased intratumoral levels of tumor necrosis
factor-α and IL-6 with respect to the vehicle group, further supporting its
antitumor applicability.
Collapse
Affiliation(s)
- Servin-Garrido Roberto Raúl
- Laboratorio de Inmunidad en Mucosas, Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1 Los Reyes Iztacala CP 54090, Tlalnepantla, Estado de México, México
| | - Ilhuicatzi-Alvarado Damaris
- Laboratorio de Inmunidad en Mucosas, Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1 Los Reyes Iztacala CP 54090, Tlalnepantla, Estado de México, México
| | - Jiménez-Chávez Ángel de Jesús
- Laboratorio de Inmunidad en Mucosas, Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1 Los Reyes Iztacala CP 54090, Tlalnepantla, Estado de México, México
| | - Moreno-Fierros Leticia
- Laboratorio de Inmunidad en Mucosas, Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1 Los Reyes Iztacala CP 54090, Tlalnepantla, Estado de México, México
| |
Collapse
|
25
|
Mugunthan SP, Mani Chandra H. A Computational Reverse Vaccinology Approach for the Design and Development of Multi-Epitopic Vaccine Against Avian Pathogen Mycoplasma gallisepticum. Front Vet Sci 2021; 8:721061. [PMID: 34765664 PMCID: PMC8577832 DOI: 10.3389/fvets.2021.721061] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 09/21/2021] [Indexed: 01/28/2023] Open
Abstract
Avian mycoplasma is a bacterial disease causing chronic respiratory disease (CRD) in poultry industries with high economic losses. The eradication of this disease still remains as a challenge. A multi-epitope prophylactic vaccine aiming the antigenic proteins of Mycoplasma gallisepticum can be a capable candidate to eradicate this infection. The present study is focused to design a multi-epitope vaccine candidate consisting of cytotoxic T-cell (CTL), helper T-cell (HTL), and B-cell epitopes of antigenic proteins, using immunoinformatics strategies. The multi-epitopic vaccine was designed, and its tertiary model was predcited, which was further refined and validated by computational tools. After initial validation, molecular docking was performed between multi-epitope vaccine construct and chicken TLR-2 and 5 receptors, which predicted effective binding. The in silico results specify the structural stability, precise specificity, and immunogenic response of the designed multi-epitope vaccine, and it could be an appropriate vaccine candidate for the M. gallisepticum infection.
Collapse
Affiliation(s)
| | - Harish Mani Chandra
- Plant Genetic Engineering and Molecular Farming Lab, Department of Biotechnology, Thiruvalluvar University, Vellore, India
| |
Collapse
|
26
|
Naqvi STQ, Yasmeen M, Ismail M, Muhammad SA, Nawazish-i-Husain S, Ali A, Munir F, Zhang Q. Designing of Potential Polyvalent Vaccine Model for Respiratory Syncytial Virus by System Level Immunoinformatics Approaches. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9940010. [PMID: 34136576 PMCID: PMC8177976 DOI: 10.1155/2021/9940010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/18/2021] [Accepted: 05/10/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) infection is a public health epidemic, leading to around 3 million hospitalization and about 66,000 deaths each year. It is a life-threatening condition exclusive to children with no effective treatment. METHODS In this study, we used system-level and vaccinomics approaches to design a polyvalent vaccine for RSV, which could stimulate the immune components of the host to manage this infection. Our framework involves data accession, antigenicity and subcellular localization analysis, T cell epitope prediction, proteasomal and conservancy evaluation, host-pathogen-protein interactions, pathway studies, and in silico binding affinity analysis. RESULTS We found glycoprotein (G), fusion protein (F), and small hydrophobic protein (SH) of RSV as potential vaccine candidates. Of these proteins (G, F, and SH), we found 9 epitopes for multiple alleles of MHC classes I and II bear significant binding affinity. These potential epitopes were linked to form a polyvalent construct using AAY, GPGPG linkers, and cholera toxin B adjuvant at N-terminal with a 23.9 kDa molecular weight of 224 amino acid residues. The final construct was a stable, immunogenic, and nonallergenic protein containing cleavage sites, TAP transport efficiency, posttranslation shifts, and CTL epitopes. The molecular docking indicated the optimum binding affinity of RSV polyvalent construct with MHC molecules (-12.49 and -10.48 kcal/mol for MHC classes I and II, respectively). This interaction showed that a polyvalent construct could manage and control this disease. CONCLUSION Our vaccinomics and system-level investigation could be appropriate to trigger the host immune system to prevent RSV infection.
Collapse
Affiliation(s)
| | - Mamoona Yasmeen
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University Multan, Pakistan
| | - Mehreen Ismail
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University Multan, Pakistan
| | - Syed Aun Muhammad
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University Multan, Pakistan
| | | | - Amjad Ali
- ASAB, National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Fahad Munir
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, China
- Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - QiYu Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, China
| |
Collapse
|
27
|
Mahdevar E, Safavi A, Abiri A, Kefayat A, Hejazi SH, Miresmaeili SM, Iranpur Mobarakeh V. Exploring the cancer-testis antigen BORIS to design a novel multi-epitope vaccine against breast cancer based on immunoinformatics approaches. J Biomol Struct Dyn 2021; 40:6363-6380. [PMID: 33599191 DOI: 10.1080/07391102.2021.1883111] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Recently, cancer immunotherapy has gained lots of attention to replace the current chemoradiation approaches and multi-epitope cancer vaccines are manifesting as the next generation of cancer immunotherapy. Therefore, in this study, we used multiple immunoinformatics approaches along with other computational approaches to design a novel multi-epitope vaccine against breast cancer. The most immunogenic regions of the BORIS cancer-testis antigen were selected according to the binding affinity to MHC-I and II molecules as well as containing multiple cytotoxic T lymphocyte (CTL) epitopes by multiple immunoinformatics servers. The selected regions were linked together by GPGPG linker. Also, a T helper epitope (PADRE) and the TLR-4/MD-2 agonist (L7/L12 ribosomal protein from mycobacterium) were incorporated by A(EAAAK)3A linker to form the final vaccine construct. Then, its physicochemical properties, cleavage sites, TAP transport efficiency, B cell epitopes, IFN-γ inducing epitopes and population coverage were predicted. The final vaccine construct was reverse translated, codon-optimized and inserted into pcDNA3.1 to form the DNA vaccine. The final vaccine construct was a stable, immunogenic and non-allergenic protein that contained numerous CTL epitopes, IFN-γ inducing epitopes and several linear and conformational B cell epitopes. Also, the final vaccine construct formed stable and significant interactions with TLR-4/MD-2 complex according to molecular docking and dynamics simulations. Moreover, its world population coverage for HLA-I and HLA-II were about 93% and 96%, respectively. Taking together, these preliminary results can be used as an appropriate platform for further experimental investigations.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Elham Mahdevar
- Department of Biology, Faculty of Science and Engineering, Science and Arts University, Yazd, Iran
| | - Ashkan Safavi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ardavan Abiri
- Department of Medicinal Chemistry, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Amirhosein Kefayat
- Department of Oncology, Cancer Prevention Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyed Hossein Hejazi
- Department of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyed Mohsen Miresmaeili
- Department of Biology, Faculty of Science and Engineering, Science and Arts University, Yazd, Iran
| | | |
Collapse
|
28
|
Safavi A, Kefayat A, Mahdevar E, Abiri A, Ghahremani F. Exploring the out of sight antigens of SARS-CoV-2 to design a candidate multi-epitope vaccine by utilizing immunoinformatics approaches. Vaccine 2020; 38:7612-7628. [PMID: 33082015 PMCID: PMC7546226 DOI: 10.1016/j.vaccine.2020.10.016] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/25/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022]
Abstract
SARS-CoV-2 causes a severe respiratory disease called COVID-19. Currently, global health is facing its devastating outbreak. However, there is no vaccine available against this virus up to now. In this study, a novel multi-epitope vaccine against SARS-CoV-2 was designed to provoke both innate and adaptive immune responses. The immunodominant regions of six non-structural proteins (nsp7, nsp8, nsp9, nsp10, nsp12 and nsp14) of SARS-CoV-2 were selected by multiple immunoinformatic tools to provoke T cell immune response. Also, immunodominant fragment of the functional region of SARS-CoV-2 spike (400-510 residues) protein was selected for inducing neutralizing antibodies production. The selected regions' sequences were connected to each other by furin-sensitive linker (RVRR). Moreover, the functional region of β-defensin as a well-known agonist for the TLR-4/MD complex was added at the N-terminus of the vaccine using (EAAAK)3 linker. Also, a CD4 + T-helper epitope, PADRE, was used at the C-terminal of the vaccine by GPGPG and A(EAAAK)2A linkers to form the final vaccine construct. The physicochemical properties, allergenicity, antigenicity, functionality and population coverage of the final vaccine construct were analyzed. The final vaccine construct was an immunogenic, non-allergen and unfunctional protein which contained multiple CD8 + and CD4 + overlapping epitopes, IFN-γ inducing epitopes, linear and conformational B cell epitopes. It could form stable and significant interactions with TLR-4/MD according to molecular docking and dynamics simulations. Global population coverage of the vaccine for HLA-I and II were estimated 96.2% and 97.1%, respectively. At last, the final vaccine construct was reverse translated to design the DNA vaccine. Although the designed vaccine exhibited high efficacy in silico, further experimental validation is necessary.
Collapse
Affiliation(s)
- Ashkan Safavi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Amirhosein Kefayat
- Department of Oncology, Cancer Prevention Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elham Mahdevar
- Department of Biology, Faculty of Science and Engineering, Science and Arts University, Yazd, Iran
| | - Ardavan Abiri
- Department of Medicinal Chemistry, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Fatemeh Ghahremani
- Department of Medical Physics and Radiotherapy, Arak School of Paramedicine, Arak University of Medical Sciences, Arak, Iran.
| |
Collapse
|