1
|
Lau YT, Tan HS. Acinetobacter baumannii subunit vaccines: recent progress and challenges. Crit Rev Microbiol 2024; 50:434-449. [PMID: 37211625 DOI: 10.1080/1040841x.2023.2215303] [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: 01/16/2023] [Revised: 03/28/2023] [Accepted: 05/12/2023] [Indexed: 05/23/2023]
Abstract
Acinetobacter baumannii is a Gram-negative, opportunistic pathogen that causes nosocomial infection with a high mortality rate in immunocompromised individuals. With the frequent emergence of multidrug-resistant A. baumannii strains that have rapidly gained resistance to most antibiotics, an extensive search for an effective A. baumannii vaccine is ongoing. Over the decade, many subunit vaccine candidates were identified using reverse vaccinology and in vivo animal studies for validation. Nineteen subunit vaccine candidates with a wide range of efficacy, from 14% to 100% preclinical survival rates, were included in this review. This article provides an updated review of several outer membrane proteins (Omp) that emerged as vaccine candidates with great potential, including OmpA, Omp34, Omp22 and BamA, based on their high conservancy, antigenicity, and immune protection against A. baumannii infection. However, there is still no licenced A. baumannii vaccine currently due to several practical issues that have yet to be resolved, such as inconsistencies between validation studies, antigen variability and insolubility. Moving forward, much investigation and innovation are still required to tackle these challenges for the regulatory approval of an A. baumannii subunit vaccine, including standardisation of immunisation study parameters, improving antigen solubility and the incorporation of nucleic acid vaccine technology.
Collapse
Affiliation(s)
- Yi Teng Lau
- School of Science, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Hock Siew Tan
- School of Science, Monash University Malaysia, Bandar Sunway, Malaysia
- Tropical Medicine and Biology Multidisciplinary Platform, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
2
|
Chen HC, Pan YL, Chen Y, Yang TH, Hsu ET, Huang YT, Chiang MH. Monoclonal Antibodies as a Therapeutic Strategy against Multidrug-Resistant Bacterial Infections in a Post-COVID-19 Era. Life (Basel) 2024; 14:246. [PMID: 38398755 PMCID: PMC10890110 DOI: 10.3390/life14020246] [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: 12/13/2023] [Revised: 01/23/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
The development of severe multidrug-resistant bacterial infections has recently intensified because of the COVID-19 pandemic. According to the guidelines issued by the World Health Organization (WHO), routine antibiotic administration is not recommended for patients with supposed or confirmed mild SARS-CoV-2 infection or pneumonia, unless bacterial infection is clinically suspected. However, recent studies have pointed out that the proportion of non-essential antibiotic use in patients infected with SARS-CoV-2 remains high. Therefore, the silent pandemic of antibiotic resistance remains a pressing issue regardless of the present threats presented by the COVID-19 pandemic. To prevent or delay entry into the postulated post-antibiotic era, the long-term advocacy for the rational use of antibiotics, the optimization of infection control procedures, and the development of new antibacterial agents and vaccines should be underscored as vital practices of the antibacterial toolbox. Recently, the development of vaccines and monoclonal antibodies has gradually received attention following the advancement of biotechnology as well as enhanced drug discovery and development in cancer research. Although decent progress has been made in laboratory-based research and promising results have been obtained following clinical trials of some of these products, challenges still exist in their widespread clinical applications. This article describes the current advantages of antibacterial monoclonal antibodies, the development of associated clinical trials, and some perceived future perspectives and challenges. Further, we anticipate the development of more therapeutic agents to combat drug-resistant bacterial infections as well as to increase the resilience of current or novel agents/strategies.
Collapse
Affiliation(s)
- Hsiao-Chun Chen
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei City 11490, Taiwan;
| | - Yu-Ling Pan
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei City 11490, Taiwan; (Y.-L.P.); (Y.C.)
| | - Ying Chen
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei City 11490, Taiwan; (Y.-L.P.); (Y.C.)
| | - Tsung-Hsuan Yang
- School of Nursing, National Defense Medical Center, Taipei City 11490, Taiwan;
| | - Erh-Tung Hsu
- School of Medicine, National Defense Medical Center, Taipei City 11490, Taiwan; (E.-T.H.); (Y.-T.H.)
| | - Yu-Ting Huang
- School of Medicine, National Defense Medical Center, Taipei City 11490, Taiwan; (E.-T.H.); (Y.-T.H.)
| | - Ming-Hsien Chiang
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei City 11490, Taiwan; (Y.-L.P.); (Y.C.)
- mProbe Taiwan Inc., Taipei City 105037, Taiwan
| |
Collapse
|
3
|
Abdollahi S, Raoufi Z. A novel vaccine candidate against A. baumannii based on a new OmpW family protein (OmpW2); structural characterization, antigenicity and epitope investigation, and in-vivo analysis. Microb Pathog 2023; 183:106317. [PMID: 37611777 DOI: 10.1016/j.micpath.2023.106317] [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: 04/28/2023] [Revised: 06/06/2023] [Accepted: 08/20/2023] [Indexed: 08/25/2023]
Abstract
A. baumannii is an MDR pathogen whose SARS-CoV-2 has recently increased its mortality rate in hospitalized patients. So, the virulence factors investigation and design of a vaccine against this bacterium seem to be critical. In this regard, the OmpW2 protein was structurally characterized by this study, and its B-T cell epitopes were mapped by bioinformatic tools. In-vivo analyses were employed to verify the immunogenicity of this protein and its selected epitopes. The results indicated that OmpW2 is a conserved virulent antigen, not toxic for the host, and not similar to the human or mouse proteome. A putative interaction between OmpW2 and a Fe-S-cluster redox enzyme was detected. Based on the results, OmpW2 belongs to the OmpW superfamily and eight beta sheets have been predicted in its tight beta-barrel structure. Several exposed epitopes were detected in the OmpW2 sequence and structure, and a sub-unit potential vaccine was generated based on the epitopes. The ELISA results indicated that after the second booster vaccination of BALB/c mice with the whole OmpW2 protein or its sub-unit fragment, the IgG titer significantly raised (p < 0.05). The mortality rate and the bacterial burden in the lung, liver, kidney, and spleen in both passive and active immunized mice were significantly decreased (p ≤ 0.001). In-vivo experiments confirmed that the OmpW2 whole protein and its sub-unit fragment induce the host immune system and can be applied to design a commercial vaccine or diagnostic kit.
Collapse
Affiliation(s)
- Sajad Abdollahi
- Department of Biology, Faculty of Basic Science, Behbahan Khatam Alanbia University of Technology, Behbahan, Iran.
| | - Zeinab Raoufi
- Department of Biology, Faculty of Basic Science, Behbahan Khatam Alanbia University of Technology, Behbahan, Iran
| |
Collapse
|
4
|
Heidarinia H, Tajbakhsh E, Rostamian M, Momtaz H. Two peptides derivate from Acinetobacter baumannii outer membrane protein K as vaccine candidates: a comprehensive in silico study. BMC Res Notes 2023; 16:128. [PMID: 37391796 DOI: 10.1186/s13104-023-06409-9] [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: 02/11/2023] [Accepted: 06/20/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND The lack of appropriate vaccines is an obstacle to the effective management of A. baumannii infections. Peptide vaccines offer an attractive and promising preventive strategy against A. baumannii. OBJECTIVE In this study, we identified specific T cell epitopes of A. baumannii outer membrane protein K (OMPK) using comprehensive bioinformatics and detailed molecular docking analysis. METHODS Both class-I and class-II T cell epitopes of A. baumannii OMPK were predicted by three tools namely IEDB, SYFPEITHI, and ProPred. The predicted epitopes were shortlisted based on several analyses including prediction scoring, clustering, exclusion of human similarity, considering immunogenicity and cytokine production, and removal of toxic and/or allergen epitopes. The epitopic peptides with high prediction scores and appropriate properties containing both class-I and class-II T cell epitopes were selected. Two of these class I/II epitopic peptides were chosen for molecular docking studies and assessing their physicochemical properties as vaccine candidates. RESULTS The results showed many T-cell epitopes of OMPK that could be evaluated for possible immunogenicity. Two of these epitopes (containing both class-I and II epitopes) had high prediction scores, were predicted by several tools, attached to several HLAs, and had the best docking score. They had different physicochemical properties and were conserved among Acinetobacter species. DISCUSSION We identified the A. baumannii OMPK high immunogenic class-I and class-II T cell epitopes and introduced two promising high immunogenic peptides as vaccine candidates. It is recommended to perform in vitro/in vivo investigation of these peptides to determine their true efficacy and efficiency.
Collapse
Affiliation(s)
- Hana Heidarinia
- Department of Microbiology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Elahe Tajbakhsh
- Department of Microbiology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Mosayeb Rostamian
- Infectious Diseases Research Center, Health Institute, Kermanshah University of Medical Sciences, Imam Reza Hospital, Parastar Blvd, Kermanshah, 6714415333, Iran.
| | - Hassan Momtaz
- Department of Microbiology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| |
Collapse
|
5
|
MatRahim NA, Jones KM, Keegan BP, Strych U, Zhan B, Lee HY, AbuBakar S. TonB-Dependent Receptor Protein Displayed on Spores of Bacillus subtilis Stimulates Protective Immune Responses against Acinetobacter baumannii. Vaccines (Basel) 2023; 11:1106. [PMID: 37376495 DOI: 10.3390/vaccines11061106] [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: 04/19/2023] [Revised: 06/06/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
The emergence of antibiotic-resistant Acinetobacter baumannii strains with limited treatment options has become a significant global health concern. Efforts to develop vaccines against the bacteria have centred on several potential protein targets, including the TonB-dependent receptors (TBDRs). In the present study, TBDRs from A. baumannii were displayed on the surface of Bacillus subtilis spores. The immunogenicity of the recombinant spores was evaluated in orally vaccinated mice. None of the immunized mice demonstrated signs of illness and were observed to be healthy throughout the study. Sera and the intestinal secretions from the recombinant spores-treated mice demonstrated mucosal and humoral antibody responses to the vaccine antigen. In addition, bactericidal activities of the sera against A. baumannii clinical isolates were demonstrated. These observations suggest that the B. subtilis spore-displayed TBDRs should be further explored as much-needed potential oral vaccine candidates against A. baumannii.
Collapse
Affiliation(s)
- Nor-Aziyah MatRahim
- Tropical Infectious Diseases Research and Education Center (TIDREC), Universiti Malaya, Kuala Lumpur 50603, Malaysia
- Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX 77030, USA
- Virology Unit, Infectious Diseases Research Centre, Institute for Medical Research, National Institutes of Health, Shah Alam 40170, Malaysia
| | - Kathryn Marie Jones
- Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX 77030, USA
| | - Brian P Keegan
- Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ulrich Strych
- Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bin Zhan
- Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hai-Yen Lee
- Tropical Infectious Diseases Research and Education Center (TIDREC), Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Sazaly AbuBakar
- Tropical Infectious Diseases Research and Education Center (TIDREC), Universiti Malaya, Kuala Lumpur 50603, Malaysia
| |
Collapse
|
6
|
Solanki V, Tiwari M, Tiwari V. Investigation of Peptidoglycan-Associated Lipoprotein of Acinetobacter baumannii and Its Interaction with Fibronectin To Find Its Therapeutic Potential. Infect Immun 2023; 91:e0002323. [PMID: 37017535 PMCID: PMC10187120 DOI: 10.1128/iai.00023-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/07/2023] [Indexed: 04/06/2023] Open
Abstract
Acinetobacter baumannii causes hospital-acquired infections and is responsible for high mortality and morbidity. The interaction of this bacterium with the host is critical in bacterial pathogenesis and infection. Here, we report the interaction of peptidoglycan-associated lipoprotein (PAL) of A. baumannii with host fibronectin (FN) to find its therapeutic potential. The proteome of A. baumannii was explored in the host-pathogen interaction database to filter out the PAL of the bacterial outer membrane that interacts with the host's FN protein. This interaction was confirmed experimentally using purified recombinant PAL and pure FN protein. To investigate the pleiotropic role of PAL protein, different biochemical assays using wild-type PAL and PAL mutants were performed. The result showed that PAL mediates bacterial pathogenesis, adherence, and invasion in host pulmonary epithelial cells and has a role in the biofilm formation, bacterial motility, and membrane integrity of bacteria. All of the results suggest that PAL's interaction with FN plays a vital role in host-cell interaction. In addition, the PAL protein also interacts with Toll-like receptor 2 and MARCO receptor, which suggests the role of PAL protein in innate immune responses. We have also investigated the therapeutic potential of this protein for vaccine and therapeutic design. Using reverse vaccinology, PAL's potential epitopes were filtered out that exhibit binding potential with host major histocompatibility complex class I (MHC-I), MHC-II, and B cells, suggesting that PAL protein is a potential vaccine target. The immune simulation showed that PAL protein could elevate innate and adaptive immune response with the generation of memory cells and would have subsequent potential to eliminate bacterial infection. Therefore, the present study highlights the interaction ability of a novel host-pathogen interacting partner (PAL-FN) and uncovers its therapeutic potential to combat infection caused by A. baumannii.
Collapse
Affiliation(s)
- Vandana Solanki
- Department of Biochemistry, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Monalisa Tiwari
- Department of Biochemistry, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Vishvanath Tiwari
- Department of Biochemistry, Central University of Rajasthan, Ajmer, Rajasthan, India
| |
Collapse
|
7
|
Fatehi Z, Doosti A, Jami MS. Oral vaccination with novel Lactococcus lactis mucosal live vector-secreting Brucella lumazine synthase (BLS) protein induces humoral and cellular immune protection against Brucella abortus. Arch Microbiol 2023; 205:122. [PMID: 36939918 PMCID: PMC10025791 DOI: 10.1007/s00203-023-03471-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/25/2023] [Accepted: 03/07/2023] [Indexed: 03/21/2023]
Abstract
This work aimed to provide recombinant Lactococcus lactis as a potential live vector for the manufacture of recombinant Brucella abortus (rBLS-Usp45). The sequences of the genes were collected from the GenBank database. Using Vaxijen and ccSOL, the proteins' immunogenicity and solubility were evaluated. Mice were given oral vaccinations with recombinant L. lactis. Anti-BLS-specific IgG antibodies were measured by ELISA assay. Cytokine reactions were examined using real-time PCR and the ELISA technique. The BLS protein was chosen for immunogenicity based on the vaccinology screening findings since it had maximum solubility and antigenic values of 99% and 0.75, respectively. The BLS gene, digested at 477 bp, was electrophoretically isolated to demonstrate that the recombinant plasmid was successfully produced. Protein-level antigen expression showed that the target group produced the 18 kDa-sized BLS protein, whereas the control group did not express any proteins. In the sera of mice given the L. lactis-pNZ8148-BLS-Usp45 vaccine 14 days after priming, there was a significant level of BLS-specific IgG1, IgG2a (P < 0.001) compared to the PBS control group. Vaccinated mice showed higher levels of IFN-γ, TNFα, IL-4, and IL-10 in samples obtained on days 14 and 28, after receiving the L. lactis-pNZ8148-BLS-Usp45 and IRBA vaccines (P < 0.001). The inflammatory reaction caused less severe spleen injuries, alveolar edema, lymphocyte infiltration, and morphological damage in the target group's spleen sections. Based on our findings, an oral or subunit-based vaccine against brucellosis might be developed using L. lactis-pNZ8148-BLS-Usp45 as a novel, promising, and safe alternative to the live attenuated vaccines now available.
Collapse
Affiliation(s)
- Zahra Fatehi
- Department of Biology, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Abbas Doosti
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Mohammad Saeid Jami
- Department of Biology, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
- Cellular and Molecular Research Center, Basic Health Sciences Research Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
8
|
Zhu F, Tan C, Li C, Ma S, Wen H, Yang H, Rao M, Zhang P, Peng W, Cui Y, Chen J, Pan P. Design of a multi-epitope vaccine against six Nocardia species based on reverse vaccinology combined with immunoinformatics. Front Immunol 2023; 14:1100188. [PMID: 36845087 PMCID: PMC9952739 DOI: 10.3389/fimmu.2023.1100188] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/13/2023] [Indexed: 02/12/2023] Open
Abstract
Background Nocardia genus, a complex group of species classified to be aerobic actinomycete, can lead to severe concurrent infection as well as disseminated infection, typically in immunocompromised patients. With the expansion of the susceptible population, the incidence of Nocardia has been gradually growing, accompanied by increased resistance of the pathogen to existing therapeutics. However, there is no effective vaccine against this pathogen yet. In this study, a multi-epitope vaccine was designed against the Nocardia infection using reverse vaccinology combined with immunoinformatics approaches. Methods First, the proteomes of 6 Nocardia subspecies Nocardia subspecies (Nocardia farcinica, Nocardia cyriacigeorgica, Nocardia abscessus, Nocardia otitidiscaviarum, Nocardia brasiliensis and Nocardia nova) were download NCBI (National Center for Biotechnology Information) database on May 1st, 2022 for the target proteins selection. The essential, virulent-associated or resistant-associated, surface-exposed, antigenic, non-toxic, and non-homologous with the human proteome proteins were selected for epitope identification. The shortlisted T-cell and B-cell epitopes were fused with appropriate adjuvants and linkers to construct vaccines. The physicochemical properties of the designed vaccine were predicted using multiple online servers. The Molecular docking and molecular dynamics (MD) simulation were performed to understand the binding pattern and binding stability between the vaccine candidate and Toll-like receptors (TLRs). The immunogenicity of the designed vaccines was evaluated via immune simulation. Results 3 proteins that are essential, virulent-associated or resistant-associated, surface-exposed, antigenic, non-toxic, and non-homologous with the human proteome were selected from 218 complete proteome sequences of the 6 Nocardia subspecies epitope identification. After screening, only 4 cytotoxic T lymphocyte (CTL) epitopes, 6 helper T lymphocyte (HTL) epitopes, and 8 B cell epitopes that were antigenic, non-allergenic, and non-toxic were included in the final vaccine construct. The results of molecular docking and MD simulation showed that the vaccine candidate has a strong affinity for TLR2 and TLR4 of the host and the vaccine-TLR complexes were dynamically stable in the natural environment. The results of the immune simulation indicated that the designed vaccine had the potential to induce strong protective immune responses in the host. The codon optimization and cloned analysis showed that the vaccine was available for mass production. Conclusion The designed vaccine has the potential to stimulate long-lasting immunity in the host, but further studies are required to validate its safety and efficacy.
Collapse
Affiliation(s)
- Fei Zhu
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China,Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China,Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China,Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Caixia Tan
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China,Department of Infection Control Center of Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chunhui Li
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China,Department of Infection Control Center of Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shiyang Ma
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China,Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China,Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China,Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Haicheng Wen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Hang Yang
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China,Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China,Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China,Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Mingjun Rao
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China,Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China,Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China,Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Peipei Zhang
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China,Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China,Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China,Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Wenzhong Peng
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China,Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China,Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China,Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Yanhui Cui
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China,Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China,Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China,Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Jie Chen
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China,Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China,Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China,Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China,*Correspondence: Jie Chen, ; Pinhua Pan,
| | - Pinhua Pan
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China,Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China,Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China,Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China,*Correspondence: Jie Chen, ; Pinhua Pan,
| |
Collapse
|
9
|
Acar MB, Ayaz-Güner Ş, Güner H, Dinç G, Ulu Kılıç A, Doğanay M, Özcan S. A subtractive proteomics approach for the identification of immunodominant Acinetobacter baumannii vaccine candidate proteins. Front Immunol 2022; 13:1001633. [DOI: 10.3389/fimmu.2022.1001633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/10/2022] [Indexed: 11/11/2022] Open
Abstract
BackgroundAcinetobacter baumannii is one of the most life-threatening multidrug-resistant pathogens worldwide. Currently, 50%–70% of clinical isolates of A. baumannii are extensively drug-resistant, and available antibiotic options against A. baumannii infections are limited. There is still a need to discover specific de facto bacterial antigenic proteins that could be effective vaccine candidates in human infection. With the growth of research in recent years, several candidate molecules have been identified for vaccine development. So far, no public health authorities have approved vaccines against A. baumannii.MethodsThis study aimed to identify immunodominant vaccine candidate proteins that can be immunoprecipitated specifically with patients’ IgGs, relying on the hypothesis that the infected person’s IgGs can capture immunodominant bacterial proteins. Herein, the outer-membrane and secreted proteins of sensitive and drug-resistant A. baumannii were captured using IgGs obtained from patient and healthy control sera and identified by Liquid Chromatography- Tandem Mass Spectrometry (LC-MS/MS) analysis.ResultsUsing the subtractive proteomic approach, we determined 34 unique proteins captured only in drug-resistant A. baumannii strain via patient sera. After extensively evaluating the predicted epitope regions, solubility, transverse membrane characteristics, and structural properties, we selected several notable vaccine candidates.ConclusionWe identified vaccine candidate proteins that triggered a de facto response of the human immune system against the antibiotic-resistant A. baumannii. Precipitation of bacterial proteins via patient immunoglobulins was a novel approach to identifying the proteins that could trigger a response in the patient immune system.
Collapse
|
10
|
Vaccinomics to Design a Multi-Epitopes Vaccine for Acinetobacter baumannii. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19095568. [PMID: 35564967 PMCID: PMC9104312 DOI: 10.3390/ijerph19095568] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/23/2022] [Accepted: 04/28/2022] [Indexed: 12/13/2022]
Abstract
Antibiotic resistance (AR) is the result of microbes’ natural evolution to withstand the action of antibiotics used against them. AR is rising to a high level across the globe, and novel resistant strains are emerging and spreading very fast. Acinetobacter baumannii is a multidrug resistant Gram-negative bacteria, responsible for causing severe nosocomial infections that are treated with several broad spectrum antibiotics: carbapenems, β-lactam, aminoglycosides, tetracycline, gentamicin, impanel, piperacillin, and amikacin. The A. baumannii genome is superplastic to acquire new resistant mechanisms and, as there is no vaccine in the development process for this pathogen, the situation is more worrisome. This study was conducted to identify protective antigens from the core genome of the pathogen. Genomic data of fully sequenced strains of A. baumannii were retrieved from the national center for biotechnological information (NCBI) database and subjected to various genomics, immunoinformatics, proteomics, and biophysical analyses to identify potential vaccine antigens against A. baumannii. By doing so, four outer membrane proteins were prioritized: TonB-dependent siderphore receptor, OmpA family protein, type IV pilus biogenesis stability protein, and OprD family outer membrane porin. Immuoinformatics predicted B-cell and T-cell epitopes from all four proteins. The antigenic epitopes were linked to design a multi-epitopes vaccine construct using GPGPG linkers and adjuvant cholera toxin B subunit to boost the immune responses. A 3D model of the vaccine construct was built, loop refined, and considered for extensive error examination. Disulfide engineering was performed for the stability of the vaccine construct. Blind docking of the vaccine was conducted with host MHC-I, MHC-II, and toll-like receptors 4 (TLR-4) molecules. Molecular dynamic simulation was carried out to understand the vaccine-receptors dynamics and binding stability, as well as to evaluate the presentation of epitopes to the host immune system. Binding energies estimation was achieved to understand intermolecular interaction energies and validate docking and simulation studies. The results suggested that the designed vaccine construct has high potential to induce protective host immune responses and can be a good vaccine candidate for experimental in vivo and in vitro studies.
Collapse
|
11
|
Singh R, Capalash N, Sharma P. Vaccine development to control the rising scourge of antibiotic-resistant Acinetobacter baumannii: a systematic review. 3 Biotech 2022; 12:85. [PMID: 35261870 PMCID: PMC8890014 DOI: 10.1007/s13205-022-03148-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 02/11/2022] [Indexed: 03/02/2023] Open
Abstract
Acinetobacter baumannii has emerged as one of major nosocomial pathogen and global emergence of multidrug-resistant strains has become a challenge for developing effective treatment options. A. baumannii has developed resistance to almost all the antibiotics viz. beta-lactams, carbapenems, tigecycline and now colistin, a last resort of antibiotics. The world is on the cusp of post antibiotic era and the evolution of multi-, extreme- and pan–drug-resistant A. baumannii strains is its obvious harbinger. Various combinations of antibiotics have been investigated but no successful treatment option is available. All these failed efforts have led researchers to develop and implement prophylactic vaccination for the prevention of infections caused by this pathogen. In this review, the advantages and disadvantages of active and passive immunization, the types of sub-unit and multi-component vaccine candidates investigated against A. baumannii viz. whole cell organism, outer membrane vesicles, outer membrane complexes, conjugate vaccines and sub-unit vaccines have been discussed. In addition, the benefits of Reverse vaccinology are emphasized here in which the potential vaccine candidates are predicted using bioinformatic online tools prior to in vivo validations.
Collapse
|
12
|
Khan K, Jalal K, Uddin R. An integrated in silico based subtractive genomics and reverse vaccinology approach for the identification of novel vaccine candidate and chimeric vaccine against XDR Salmonella typhi H58. Genomics 2022; 114:110301. [PMID: 35149170 DOI: 10.1016/j.ygeno.2022.110301] [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: 05/31/2021] [Revised: 12/25/2021] [Accepted: 02/01/2022] [Indexed: 12/14/2022]
Abstract
Salmonella typhi is notorious for causing enteric fever which is also known as typhoid fever. It emerged as an extreme drug resistant strain that requires urgent attention to prevent its global spread. Statistically, about 11-17 million typhoid illnesses are reported worldwide annually. The only alternative approach for the control of this illness is proper vaccination. However, available typhoid vaccine has certain limitations such as poor long-term efficacy, and non-recommendation for below 6 years children, which opens the avenues for designing new vaccines to overcome such limitations. Computational-based reverse vaccinology along with subtractive genomics analysis is one of the robust approaches used for the prioritization of vaccine candidates through direct screening of genome sequence assemblies. In the current study, we have successfully designed a peptide-based novel antigen chimeric vaccine candidate against the XDR strain of S. typhi H58. The pipeline revealed four peptides from WP_001176621.1 i.e., peptidoglycan-associated lipoprotein Pal and two peptides from WP_000747548.1 i.e., OmpA family lipoprotein as promising target for the induction of immune response against S. typhi. The six epitopes from both proteins were found as immunogenic, antigenic, virulent, highly conserved, nontoxic, and non-allergenic among whole Salmonella H58 proteome. Furthermore, the binding interaction between a chimeric vaccine and human population alleles was unveiled through structure-based studies. So far, these proteins have never been characterized as vaccine targets against S. typhi. The current study proposed that construct V2 could be a significant vaccine candidate against S. typhi H58. However, to ascertain this, future experimental holistic studies are recommended as follow-up.
Collapse
Affiliation(s)
- Kanwal Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Pakistan
| | - Khurshid Jalal
- HEJ Research Institute of Chemistry International Center for Chemical and Biological Sciences, University of Karachi, Pakistan
| | - Reaz Uddin
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Pakistan.
| |
Collapse
|
13
|
Hagag YA, Said HS, Kenawy HI, Hassan R. A novel pentavalent vaccine candidate completely protects against Acinetobacter baumannii in a mouse model of peritonitis. Appl Microbiol Biotechnol 2022; 106:8151-8167. [PMID: 36401642 PMCID: PMC9676856 DOI: 10.1007/s00253-022-12231-3] [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: 08/03/2022] [Revised: 09/30/2022] [Accepted: 10/05/2022] [Indexed: 11/21/2022]
Abstract
Acinetobacter baumannii is considered as one of the most virulent and infectious organisms that have an increased ability to both evade host immune response and resist various classes of antibiotics, leading to life-threatening infections. Multiple virulence factors have been implicated in the high prevalence rate of A. baumannii in hospitalized and immunocompromised patients. Moreover, improper use of antibiotics has led to the emergence of extensive drug-resistant strains that urgently require alternative strategies to control this superbug. Unfortunately, the availability of a licensed vaccine against A. baumannii infections is still challenged by the vast diversity among A. baumannii strains. Here, we report the development of a novel pentavalent vaccine candidate composed of two recombinant proteins (Wza and YiaD) and a pool of capsular polysaccharides isolated from 3 clinical isolates. We tested this new vaccine in vivo in a mouse model of peritonitis against the standard strain ATCC 19606 in addition to 3 clinical isolates of A. baumannii. Immunization with this vaccine completely protected the challenged mice with 100% survival rate in the case of all the tested bacteria. Further clinical studies are urgently needed to evaluate the efficacy and safety of this proprietary vaccine to protect patients from A. baumannii lethal infections. KEY POINTS: • Recombinant proteins pool (Wza and YiaD) immunization led to a synergistic immune response. • Capsular polysaccharides pool induced up to 90% protection of tested clinical isolates. • The pentavalent pool showed superiority with 100% survival of immunized mice.
Collapse
Affiliation(s)
- Yomna A. Hagag
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516 Egypt
| | - Heba Shehta Said
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516 Egypt
| | - Hany I. Kenawy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516 Egypt
| | - Ramadan Hassan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516 Egypt
| |
Collapse
|
14
|
Meniaï I, Thibodeau A, Quessy S, Parreira VR, Fravalo P, Beauchamp G, Gaucher ML. Putative antigenic proteins identified by comparative and subtractive reverse vaccinology in necrotic enteritis-causing Clostridium perfringens isolated from broiler chickens. BMC Genomics 2021; 22:890. [PMID: 34903179 PMCID: PMC8666345 DOI: 10.1186/s12864-021-08216-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 11/24/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Avian necrotic enteritis (NE) caused by Clostridium perfringens is a disease with a major economic impact, generating losses estimated to 6 billion of dollars annually for the poultry industry worldwide. The incidence of the disease is particularly on the rise in broiler chicken flocks eliminating the preventive use of antibiotics. To date, no alternative allows for the efficient prevention of NE and a control of the disease using a vaccinal strategy would be mostly prized. For this purpose, comparative and subtractive reverse vaccinology identifying putative immunogenic bacterial surface proteins is one of the most promising approaches. RESULTS A comparative genomic study was performed on 16 C. perfringens strains isolated from healthy broiler chickens and from broilers affected with necrotic enteritis. Results showed that the analyzed genomes were composed of 155,700 distinct proteins from which 13% were identified as extracellular, 65% as cytoplasmic and 22% as part of the bacterial membrane. The evaluation of the immunogenicity of these proteins was determined using the prediction software VaxiJen®. CONCLUSIONS For the most part, proteins with the highest scores were associated with an extracellular localisation. For all the proteins analyzed, the combination of both the immunogenicity score and the localisation prediction led to the selection of 12 candidate proteins that were mostly annotated as hypothetical proteins. We describe 6 potential candidates of higher interest due to their antigenic potential, their extracellular localisation, and their possible role in virulence of C. perfringens.
Collapse
Affiliation(s)
- Ilhem Meniaï
- Département de Pathologie et Microbiologie, Faculté de médecine vétérinaire, Université de Montréal, 3200 Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada
| | - Alexandre Thibodeau
- Département de Pathologie et Microbiologie, Faculté de médecine vétérinaire, Université de Montréal, 3200 Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada.,Swine and Poultry Infectious Diseases Research Centre (CRIPA), Faculté de médecine vétérinaire, Université de Montréal, 3200 Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada
| | - Sylvain Quessy
- Département de Pathologie et Microbiologie, Faculté de médecine vétérinaire, Université de Montréal, 3200 Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada
| | - Valeria R Parreira
- Department of Food Science, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Philippe Fravalo
- Département de Pathologie et Microbiologie, Faculté de médecine vétérinaire, Université de Montréal, 3200 Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada.,Swine and Poultry Infectious Diseases Research Centre (CRIPA), Faculté de médecine vétérinaire, Université de Montréal, 3200 Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada
| | - Guy Beauchamp
- Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, J2S 2M2, Canada
| | - Marie-Lou Gaucher
- Département de Pathologie et Microbiologie, Faculté de médecine vétérinaire, Université de Montréal, 3200 Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada. .,Swine and Poultry Infectious Diseases Research Centre (CRIPA), Faculté de médecine vétérinaire, Université de Montréal, 3200 Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada.
| |
Collapse
|
15
|
DcaP porin and its epitope-based subunit promise effective vaccines against Acinetobacter baumannii; in-silico and in-vivo approaches. Microb Pathog 2021; 162:105346. [PMID: 34864145 DOI: 10.1016/j.micpath.2021.105346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/31/2021] [Accepted: 12/01/2021] [Indexed: 11/23/2022]
Abstract
A. baumannii is a multi-drug resistant pathogen with a relatively high mortality rate. To date, no vaccine has been approved against this bacterium. DcaP is a high abundance porin during infection that its structure has been recently determined, but no information about its immunogenic properties has been reported yet. So, in this study DcaP properties were analyzed and its vaccine potential was evaluated. The results showed this porin is an extremely conserved antigen with no allergenicity and toxicity that bears no resemblance to human proteins. Six potential immunogen areas in the DcaP sequence were detected based on in-silico B and T-cell epitope mapping and other approaches. A multiple-epitope potential vaccine was designed based on the predicted linear epitopes and amplified by overlap extension PCR technique. In-vivo results indicated that active and passive immunization of mice with the DcaP protein or its designed subunit vaccine raises the antibody titers and decreases the mortality rate of the immunized mice infected with A. baumannii. Based on the results, DcaP and its indicated immunogen regions can be considered as a peptide or subunit vaccine. The immunogen regions could also be applied in multivalent subunit vaccine candidates against A. baumannii and other bacteria.
Collapse
|
16
|
Abdollahi S, Raoufi Z, Fakoor MH. Physicochemical and structural characterization, epitope mapping and vaccine potential investigation of a new protein containing Tetratrico Peptide Repeats of Acinetobacter baumannii: An in-silico and in-vivo approach. Mol Immunol 2021; 140:22-34. [PMID: 34649027 DOI: 10.1016/j.molimm.2021.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/13/2021] [Accepted: 10/04/2021] [Indexed: 11/24/2022]
Abstract
Acinetobacter baumannii is an opportunistic multidrug-resistant pathogen that causes a significant mortality rate. The proteins containing Tetratrico Peptide Repeats (TPRs) are involved in the pathogenicity and virulence of bacteria and have different roles such as transfer of bacterial virulence factors to host cells, binding to the host cells and inhibition of phagolysosomal maturation. So, in this study, physicochemical properties of a new protein containing TPRs in A. baumannii which was named PcTPRs1 by this study were characterized and its 3D structure was predicted by in-silico tools. The protein B and T cell epitopes were mapped and its vaccine potential was in-silico and in-vivo investigated. Domain analysis indicated that the protein contains the Flp pilus assembly protein TadD domain which has three TPRs. The helix is dominant in the protein structure, and this protein is an outer membrane antigen which, is extremely conserved among A. baumannii strains; thus, has good properties to be applied as a recombinant vaccine. The best-predicted and refined model was applied in ligand-binding sites and conformational epitopes prediction. Based on epitope mapping results, several epitopes were characterized which could stimulate both immune systems. BLAST results showed the introduced epitopes are completely conserved among A. baumannii strains. The in-vivo analysis indicates that a 101 amino acid fragment of the protein which contains the best selected epitope, can produce a good protectivity against A. baumannii as well as the whole TPR protein and thus could be investigated as an effective subunit and potential vaccines.
Collapse
Affiliation(s)
- Sajad Abdollahi
- Department of Biology, Faculty of Basic Science, Behbahan Khatam Alanbia University of Technology, Behbahan, Iran.
| | - Zeinab Raoufi
- Department of Biology, Faculty of Basic Science, Behbahan Khatam Alanbia University of Technology, Behbahan, Iran.
| | | |
Collapse
|
17
|
Beiranvand S, Doosti A, Mirzaei SA. Putative novel B-cell vaccine candidates identified by reverse vaccinology and genomics approaches to control Acinetobacter baumannii serotypes. INFECTION GENETICS AND EVOLUTION 2021; 96:105138. [PMID: 34793968 DOI: 10.1016/j.meegid.2021.105138] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/12/2021] [Accepted: 11/09/2021] [Indexed: 11/26/2022]
Abstract
In the last decade, Multi-drug resistance (MDR)-associated infections of Acinetobacter baumannii have grown worldwide. A cost-effective preventative strategy against this bacterium is vaccination. This study has presented five novel vaccine candidates against A. baumannii produced using the reverse vaccinology method. BLASTn was done to identify the most conserved antigens. PSORTb 3.0.2 was run to predict the subcellular localization of the proteins. The initial screening and antigenicity evaluation were performed using Vaxign. The ccSOL omics was also employed to predict protein solubility. The cross-membrane localization of the protein was predicted using PRED-TMBB. B cell epitope prediction was made for immunogenicity using the IEDB and BepiPred-2.0 database. Eventually, BLASTp was done to verify the extent of similarity to the human proteome to exclude the possibility of autoimmunity. Proteins failing to comply with the set parameters were filtered at each step. In silico, potential vaccines against 21 A. baumannii strains were identified using reverse vaccinology and subtractive genomic techniques. Based on the above criteria, out of the initial 15 A. baumannii proteins selected for screening, nine exposed/secreted/membrane proteins, i.e., Pfsr, LptE, OmpH, CarO, CsuB, CdiB, MlaA, FhuE, and were the most promising candidates. Their solubility and antigenicity were also examined and found to be more than 0.45 and 0.6, respectively. Based on the results, LptE was selected with the highest average antigenic score of 1.043 as the best protein, followed by FimF and Pfsr with scores of 1.022 and 1.014, respectively. In the end, five proteins were verified as promising candidates. Overall, the targets identified herein may be utilized in future strategies to control A. baumannii worldwide.
Collapse
Affiliation(s)
- Sheida Beiranvand
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Abbas Doosti
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Seyed Abbas Mirzaei
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran; Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
18
|
Mishra R, Chiang Tan Y, Adel Ahmed Abd El-Aal A, Lahiri C. Computational Identification of the Plausible Molecular Vaccine Candidates of Multidrug-Resistant Salmonella enterica. SALMONELLA SPP. - A GLOBAL CHALLENGE 2021. [DOI: 10.5772/intechopen.95856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Salmonella enterica serovars are responsible for the life-threatening, fatal, invasive diseases that are common in children and young adults. According to the most recent estimates, globally, there are approximately 11–20 million cases of morbidity and between 128,000 and 161,000 mortality per year. The high incidence rates of diseases like typhoid, caused by the serovars Typhi and Paratyphi, and gastroenteritis, caused by the non-typhoidal Salmonellae, have become worse, with the ever-increasing pathogenic strains being resistant to fluoroquinolones or almost even the third generation cephalosporins, such as ciprofloxacin and ceftriaxone. With vaccination still being one of the chosen methods of eradicating this disease, identification of candidate proteins, to be utilized for effective molecular vaccines, has probably remained a challenging issue. In our study here, we portray the usage of computational tools to analyze and predict potential vaccine candidate(s) for the multi-drug resistant serovars of S. enterica.
Collapse
|
19
|
Kamuyu G, Suen Cheng Y, Willcocks S, Kewcharoenwong C, Kiratisin P, Taylor PW, Wren BW, Lertmemongkolchai G, Stabler RA, Brown J. Sequential Vaccination With Heterologous Acinetobacter baumannii Strains Induces Broadly Reactive Antibody Responses. Front Immunol 2021; 12:705533. [PMID: 34394105 PMCID: PMC8363311 DOI: 10.3389/fimmu.2021.705533] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/08/2021] [Indexed: 12/11/2022] Open
Abstract
Antibody therapy may be an alternative treatment option for infections caused by the multi-drug resistant (MDR) bacterium Acinetobacter baumannii. As A. baumannii has multiple capsular serotypes, a universal antibody therapy would need to target conserved protein antigens rather than the capsular polysaccharides. We have immunized mice with single or multiple A. baumannii strains to induce antibody responses to protein antigens, and then assessed whether these responses provide cross-protection against a collection of genetically diverse clinical A. baumannii isolates. Immunized mice developed antibody responses to multiple protein antigens. Flow cytometry IgG binding assays and immunoblots demonstrated improved recognition of both homologous and heterologous clinical strains in sera from mice immunized with multiple strains compared to a single strain. The capsule partially inhibited bacterial recognition by IgG and the promotion of phagocytosis by human neutrophils. However, after immunization with multiple strains, serum antibodies to protein antigens promoted neutrophil phagocytosis of heterologous A. baumannii strains. In an infection model, mice immunized with multiple strains had lower bacterial counts in the spleen and liver following challenge with a heterologous strain. These data demonstrate that antibodies targeting protein antigens can improve immune recognition and protection against diverse A. baumannii strains, providing support for their use as an antibody therapy.
Collapse
Affiliation(s)
- Gathoni Kamuyu
- Centre for Inflammation and Tissue Repair, University College London (UCL) Respiratory, London, United Kingdom
| | - Yat Suen Cheng
- Centre for Inflammation and Tissue Repair, University College London (UCL) Respiratory, London, United Kingdom
| | - Sam Willcocks
- London School of Hygiene and Tropical Medicine, Infectious and Tropical Disease, Department of Infection Biology, London, United Kingdom
| | - Chidchamai Kewcharoenwong
- Cellular and Molecular Immunology Unit, Centre for Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Pattarachai Kiratisin
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-Noi, Bangkok, Thailand
| | - Peter W Taylor
- School of Pharmacy, University College London, London, United Kingdom
| | - Brendan W Wren
- London School of Hygiene and Tropical Medicine, Infectious and Tropical Disease, Department of Infection Biology, London, United Kingdom
| | - Ganjana Lertmemongkolchai
- Cellular and Molecular Immunology Unit, Centre for Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand.,Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Richard A Stabler
- London School of Hygiene and Tropical Medicine, Infectious and Tropical Disease, Department of Infection Biology, London, United Kingdom
| | - Jeremy Brown
- Centre for Inflammation and Tissue Repair, University College London (UCL) Respiratory, London, United Kingdom
| |
Collapse
|
20
|
The current state of immunization against Gram-negative bacteria in children: a review of the literature. Curr Opin Infect Dis 2021; 33:517-529. [PMID: 33044242 DOI: 10.1097/qco.0000000000000687] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW Gram-negative bacteria (GNB) are a major cause of infection worldwide and multidrug resistance in infants and children. The major pathogens include Klebsiella pneumoniae, Escherichia coli, Enterobacter spp., Pseudomonas aeruginosa and Acinetobacter baumannii. With new antibiotic options limited, immunization is likely to play a critical role in prevention. This review discusses their epidemiology, the current state of vaccine research and potential immunization strategies to protect children. A comprehensive review of the literature, conference abstracts along with web searches was performed to identify current and investigational vaccines against the major GNB in children. RECENT FINDINGS Phase I--III vaccine trials have been undertaken for the major Gram-negative bacteria but not in infants or children. E. coli is a common infection in immune-competent children, including neonatal sepsis. Several vaccines are in late-phase clinical trials, with some already licensed for recurrent urinary tract infections in women. Klebsiella spp. causes community-acquired and hospital-acquired infections, including sepsis in neonates and immunocompromised children although no vaccine trials have extended beyond early phase 2 trials. P. aeruginosa is a common pathogen in patients with cystic fibrosis. Phase 1--3 vaccine and monoclonal antibody trials are in progress, although candidates provide limited coverage against pathogenic strains. Enterobacter spp. and A. baumannii largely cause hospital-acquired infections with experimental vaccines limited to phase 1 research. SUMMARY The current immunization pipelines for the most prevalent GNB are years away from licensure. Similar to incentives for new antibiotics, global efforts are warranted to expedite the development of effective vaccines.
Collapse
|
21
|
Mat Rahim N, Lee H, Strych U, AbuBakar S. Facing the challenges of multidrug-resistant Acinetobacter baumannii: progress and prospects in the vaccine development. Hum Vaccin Immunother 2021; 17:3784-3794. [PMID: 34106809 PMCID: PMC8437540 DOI: 10.1080/21645515.2021.1927412] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
In 2017, the World Health Organization (WHO) named A. baumannii as one of the three antibiotic-resistant bacterial species on its list of global priority pathogens in dire need of novel and effective treatment. With only polymyxin and tigecycline antibiotics left as last-resort treatments, the need for novel alternative approaches to the control of this bacterium becomes imperative. Vaccines against numerous bacteria have had impressive records in reducing the burden of the respective diseases and addressing antimicrobial resistance; as in the case of Haemophilus influenzae type b . A similar approach could be appropriate for A. baumannii. Toward this end, several potentially protective antigens against A. baumannii were identified and evaluated as vaccine antigen candidates. A licensed vaccine for the bacteria, however, is still not in sight. Here we explore and discuss challenges in vaccine development against A. baumannii and the promising approaches for improving the vaccine development process.
Collapse
Affiliation(s)
- NorAziyah Mat Rahim
- Tropical Infectious Diseases Research and Education Center (TIDREC), Universiti Malaya, Kuala Lumpur, Malaysia.,Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA.,Virology Unit, Institute for Medical Research, National Institute of Health Complex, Setia Alam, Malaysia
| | - HaiYen Lee
- Tropical Infectious Diseases Research and Education Center (TIDREC), Universiti Malaya, Kuala Lumpur, Malaysia
| | - Ulrich Strych
- Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| | - Sazaly AbuBakar
- Tropical Infectious Diseases Research and Education Center (TIDREC), Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
22
|
López-Siles M, Corral-Lugo A, McConnell MJ. Vaccines for multidrug resistant Gram negative bacteria: lessons from the past for guiding future success. FEMS Microbiol Rev 2021; 45:fuaa054. [PMID: 33289833 DOI: 10.1093/femsre/fuaa054] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/18/2020] [Indexed: 02/07/2023] Open
Abstract
Antimicrobial resistance is a major threat to global public health. Vaccination is an effective approach for preventing bacterial infections, however it has not been successfully applied to infections caused by some of the most problematic multidrug resistant pathogens. In this review, the potential for vaccines to contribute to reducing the burden of disease of infections caused by multidrug resistant Gram negative bacteria is presented. Technical, logistical and societal hurdles that have limited successful vaccine development for these infections in the past are identified, and recent advances that can contribute to overcoming these challenges are assessed. A synthesis of vaccine technologies that have been employed in the development of vaccines for key multidrug resistant Gram negative bacteria is included, and emerging technologies that may contribute to future successes are discussed. Finally, a comprehensive review of vaccine development efforts over the last 40 years for three of the most worrisome multidrug resistant Gram negative pathogens, Acinetobacter baumannii, Klebsiella pneumoniae and Pseudomonas aeruginosa is presented, with a focus on recent and ongoing studies. Finally, future directions for the vaccine development field are highlighted.
Collapse
Affiliation(s)
- Mireia López-Siles
- Intrahospital Infections Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Andrés Corral-Lugo
- Intrahospital Infections Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Michael J McConnell
- Intrahospital Infections Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
23
|
McConnell MJ, Martín-Galiano AJ. Designing Multi-Antigen Vaccines Against Acinetobacter baumannii Using Systemic Approaches. Front Immunol 2021; 12:666742. [PMID: 33936107 PMCID: PMC8085427 DOI: 10.3389/fimmu.2021.666742] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/26/2021] [Indexed: 12/12/2022] Open
Abstract
Vaccines and monoclonal antibodies are promising approaches for preventing and treating infections caused by multidrug resistant Acinetobacter baumannii. However, only partial protection has been achieved with many previously tested protein antigens, which suggests that vaccines incorporating multiple antigens may be necessary in order to obtain high levels of protection. Several aspects that use the wealth of omic data available for A. baumannii have not been fully exploited for antigen identification. In this study, the use of fractionated proteomic and computational data from ~4,200 genomes increased the number of proteins potentially accessible to the humoral response to 8,824 non-redundant proteins in the A. baumannii panproteome. Among them, 59% carried predicted B-cell epitopes and T-cell epitopes recognized by two or more alleles of the HLA class II DP supertype. Potential cross-reactivity with human proteins was detected for 8.9% of antigens at the protein level and 2.7% at the B-cell epitope level. Individual antigens were associated with different infection types by genomic, transcriptomic or functional analyses. High intra-clonal genome density permitted the identification of international clone II as a “vaccitype”, in which 20% of identified antigens were specific to this clone. Network-based centrality measurements were used to identify multiple immunologic nodes. Data were formatted, unified and stored in a data warehouse database, which was subsequently used to identify synergistic antigen combinations for different vaccination strategies. This study supports the idea that integration of multi-omic data and fundamental knowledge of the pathobiology of drug-resistant bacteria can facilitate the development of effective multi-antigen vaccines against these challenging infections.
Collapse
Affiliation(s)
- Michael J McConnell
- Intrahospital Infections Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III (ISCIII), Majadahonda, Spain
| | - Antonio J Martín-Galiano
- Intrahospital Infections Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III (ISCIII), Majadahonda, Spain
| |
Collapse
|
24
|
Martín-Galiano AJ, Escolano-Martínez MS, Corsini B, de la Campa AG, Yuste J. Immunization with SP_1992 (DiiA) Protein of Streptococcus pneumoniae Reduces Nasopharyngeal Colonization and Protects against Invasive Disease in Mice. Vaccines (Basel) 2021; 9:vaccines9030187. [PMID: 33668195 PMCID: PMC7995960 DOI: 10.3390/vaccines9030187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/19/2021] [Accepted: 02/19/2021] [Indexed: 11/16/2022] Open
Abstract
Knowledge-based vaccinology can reveal uncharacterized antigen candidates for a new generation of protein-based anti-pneumococcal vaccines. DiiA, encoded by the sp_1992 locus, is a surface protein containing either one or two repeats of a 37mer N-terminal motif that exhibits low interstrain variability. DiiA belongs to the core proteome, contains several conserved B-cell epitopes, and is associated with colonization and pathogenesis. Immunization with DiiA protein via the intraperitoneal route induced a strong IgG response, including different IgG subtypes. Vaccination with DiiA increased bacterial clearance and induced protection against sepsis, conferring 70% increased survival at 48 h post-infection when compared to the adjuvant control. The immunogenic response and survival rates in mice immunized with a truncated DiiA version lacking 119 N-terminal residues were remarkably lower, confirming the relevance of the repeat zone in the immunoprotection by DiiA. Intranasal immunization of mice with the entire recombinant protein elicited mucosal IgG and IgA responses that reduced bacterial colonization of the nasopharynx, confirming that this protein might be a vaccine candidate for reducing the carrier rate. DiiA constitutes an example of how functionally unannotated proteins may still represent promising candidates that can be used in prophylactic strategies against the pneumococcal carrier state and invasive disease.
Collapse
Affiliation(s)
- Antonio J. Martín-Galiano
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), 28220 Madrid, Spain; (M.S.E.-M.); (B.C.); (A.G.d.l.C.)
- Correspondence: (A.J.M.-G.); (J.Y.); Tel.: +34-918223976 (A.J.M.-G.); +34-918223620 (J.Y.)
| | - María S. Escolano-Martínez
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), 28220 Madrid, Spain; (M.S.E.-M.); (B.C.); (A.G.d.l.C.)
| | - Bruno Corsini
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), 28220 Madrid, Spain; (M.S.E.-M.); (B.C.); (A.G.d.l.C.)
| | - Adela G. de la Campa
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), 28220 Madrid, Spain; (M.S.E.-M.); (B.C.); (A.G.d.l.C.)
- Presidencia Consejo Superior de Investigaciones Científicas, 28006 Madrid, Spain
| | - José Yuste
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), 28220 Madrid, Spain; (M.S.E.-M.); (B.C.); (A.G.d.l.C.)
- CIBER de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
- Correspondence: (A.J.M.-G.); (J.Y.); Tel.: +34-918223976 (A.J.M.-G.); +34-918223620 (J.Y.)
| |
Collapse
|
25
|
Ma C, Chen W. Where are we and how far is there to go in the development of an Acinetobacter vaccine? Expert Rev Vaccines 2021; 20:281-295. [PMID: 33554671 DOI: 10.1080/14760584.2021.1887735] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Healthcare-associated infections caused by multidrug-resistant Acinetobacter baumannii are becoming alarming worldwide. However, the pipeline of new antibiotics is very limited. Vaccination is one of the most cost effective and promising strategies to prevent infections and can play an important role in combat multidrug resistance A. baumannii and prevent the development of new drug resistance. AREA COVERED This review gives an overview of the research and development of A. baumannii vaccines during the past five years (2015-2020), discusses the key progresses and current challenges of the field, and speculates on the future of A. baumannii vaccine development. EXPERT OPINION Moderate progresses have been made in the research and development of A. baumannii vaccine in the last five years, in particular in the areas of identification of new protein targets, development of multicomponent vaccines, and use of vaccines and antibodies as adjuncts for antibiotics therapies. However, substantial scientific and logistic challenges, such as selection of lead vaccine candidates and formulation, vaccine clinical trials and targeted population, and financial incentives, remain. Thus, innovative strategies will be needed before an A. baumannii vaccine candidate can be brought into late stage of preclinical development in next five years.
Collapse
Affiliation(s)
- Crystal Ma
- Human Health Therapeutics Research Center (HHT), National Research Council Canada, Ottawa, Ontario Canada
| | - Wangxue Chen
- Human Health Therapeutics Research Center (HHT), National Research Council Canada, Ottawa, Ontario Canada.,Department of Biology, Brock University, St. Catharines, Ontario Canada
| |
Collapse
|
26
|
Hajialibeigi A, Amani J, Gargari SLM. Identification and evaluation of novel vaccine candidates against Shigella flexneri through reverse vaccinology approach. Appl Microbiol Biotechnol 2021; 105:1159-1173. [PMID: 33452891 PMCID: PMC7811352 DOI: 10.1007/s00253-020-11054-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/29/2020] [Accepted: 12/09/2020] [Indexed: 02/08/2023]
Abstract
Shigellosis is a significant type of diarrhea that causes 160,000 deaths annually in a global scale. The mortality occurs mainly in children less than 5 years of age. No licensed vaccine is available, and conventional efforts for developing an effective and safe vaccine against shigellosis have not been succeeded yet. The reverse vaccinology is a novel promising method that screens genome or proteome of an organism for finding new vaccine candidates. In this study, through reverse vaccinology approach, new vaccine candidates against Shigella flexneri were identified and experimentally evaluated. Proteomes of S. flexneri were obtained from UniProt, and then outer membrane and extracellular proteins were predicted and selected for the evaluation of transmembrane domains, protein conservation, host homology, antigenicity, and solubility. From 103 proteins, 7 high-scored proteins were introduced as novel vaccine candidates, and after B- and T-cell epitope prediction, the best protein was selected for experimental studies. Recombinant protein was expressed, purified, and injected to BALB/c mice. The adhesion inhibitory effect of sera was also studied. The immunized mice demonstrated full protection against the lethal dose challenge. The sera remarkably inhibited S. flexneri adhesion to Caco-2 epithelial cells. The results indicate that identified antigen can serve for vaccine development against shigellosis and support reverse vaccinology for discovering novel effective antigens. KEY POINTS: • Seven Shigella new antigens were identified by reverse vaccinology (RV) approach. • The best antigen experimented demonstrated full protection against lethal dose. • In vivo results verified RV analyses and suggest FimG as a new potent vaccine candidate.
Collapse
Affiliation(s)
- Abolfazl Hajialibeigi
- Faculty of Basic Sciences, Department of Cell Biology, Shahed University, Tehran, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
27
|
Taheri F, Nazarian S, Ahmadi TS, Gargari SLM. Protective effects of egg yolk immunoglobulins (IgYs) developed against recombinant immunogens CtxB, OmpW and TcpA on infant mice infected with Vibrio cholerae. Int Immunopharmacol 2020; 89:107054. [DOI: 10.1016/j.intimp.2020.107054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/25/2020] [Accepted: 09/25/2020] [Indexed: 12/11/2022]
|
28
|
Smiline Girija AS. Delineating the Immuno-Dominant Antigenic Vaccine Peptides Against gacS -Sensor Kinase in Acinetobacter baumannii: An in silico Investigational Approach. Front Microbiol 2020; 11:2078. [PMID: 33013757 PMCID: PMC7506167 DOI: 10.3389/fmicb.2020.02078] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 08/06/2020] [Indexed: 11/13/2022] Open
Abstract
Objectives To predict the novel vaccine peptide candidates against gacS protein involved with the citrate utilization in the two-component system of A. baumannii-associated virulence as an alternative strategy to combat the multi-drug resistant strains using an immuno-informatic approach. Methods The study is designed as an observational in silico study design with the application of BepiPred, AlgPred, VaxiJen, AntigenPro, SolPro, Expasy ProtParam server, IEDB database, and MHC cluster analytical tools and servers to predict the immuno-dominant B-cell and T-cell epitopes from gacS FASTA sequences retrieved from UNIPROT database. Further peptide interactions with TLR-4 was assessed based on the number of hydrogen bonds. Results Nine peptides (20aa) with the highest score of 1 were selected from the 137 epitopes, and five were predicted as antigenic epitopes (E1-E5). E3 was selected as the potent antigen (score: 0.939537) and E1 as the best vaccine candidate (score: 0.9803) under AntigenPro and Vaxijen server, respectively. SolPro predicted all epitopes as soluble peptides. ProtParam predictions showed E3 and E5 as stable proteins with a shelf life of 3.5 and 1.9 h and possessed negative GRAVY values. PsortB server predicted a final localization score of 7.88 for the gacS protein sequence as a cytoplasmic membrane protein. IEDB conservancy analysis showed 100% conserved sequences within the gacS sequence, and class I conservancy yielded positive values for all epitopes. Cluster analysis showed strong interactions, and the protein-peptide interactions with TLR-2 finally detected E5 as the best interacting peptide (H bonds = 14) followed by E3 (H bonds = 12). Conclusion The study suggests five antigenic peptides as promiscuous vaccine candidates to target the gacS of A. baumannii using immuno-informatic approach toward the peptide synthesis and in vitro analysis. However, the study recommends further experimental validation for immunological response and memory through in vivo studies.
Collapse
Affiliation(s)
- A S Smiline Girija
- Department of Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, India
| |
Collapse
|
29
|
Mehmood A, Naseer S, Ali A, Fatimah H, Rehman S, Kiani AK. Identification of novel vaccine candidates against carbapenem resistant Klebsiella pneumoniae: A systematic reverse proteomic approach. Comput Biol Chem 2020; 89:107380. [PMID: 32992120 DOI: 10.1016/j.compbiolchem.2020.107380] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 08/09/2020] [Accepted: 09/18/2020] [Indexed: 11/18/2022]
Abstract
Klebsiella pneumoniae is declared as antibiotic resistant by WHO, with the critical urgency of developing novel antimicrobial therapeutics as drug resistance is the second most dangerous threat after terrorism. Besides many attempts still, there is no effective vaccine available against K. pneumoniae. By utilizing all the available proteomic data we prioritized the novel proteins ideal for vaccine development using bioinformatics tools and techniques. Among the huge data, eight proteins passed all the barriers and were considered ideal candidates for vaccine development. These include: copper silver efflux system outer membrane protein (CusC), outer membrane porin protein (OmpN), Fe++ enterobactin transporter substrate binding protein (fepB), zinc transporter substrate binding protein (ZnuA), ribonuclease HI, tellurite resistant methyltransferase (the B), and two uncharacterized hypothetical proteins (WP_002918223 and WP_002892366). These proteins were also subjected to epitope analysis and were found best for developing subunit vaccine against K. pneumoniae. The study shows that the potential vaccine targets are sufficiently efficient being virulent, of outer membranous origin and can be proposed for the DNA third-generation vaccines development that would help to cope up infections caused by multidrug-resistant K. pneumoniae.
Collapse
Affiliation(s)
- Asim Mehmood
- Department of Biology and Environmental Science, Faculty of Sciences, Allama Iqbal Open University, Islamabad, Pakistan
| | - Samar Naseer
- Department of Biology and Environmental Science, Faculty of Sciences, Allama Iqbal Open University, Islamabad, Pakistan.
| | - Amjad Ali
- Department of Industrial Biotechnology, Atta Ur Rahman School of Applied Biosciences (ASAB), National University of Sciences & Technology (NUST), Islamabad, Pakistan
| | - Hina Fatimah
- Department of Biology and Environmental Science, Faculty of Sciences, Allama Iqbal Open University, Islamabad, Pakistan
| | - Shazia Rehman
- Department of Botany, Rawalpindi Women University, Rawalpindi, Pakistan
| | - Aysha Karim Kiani
- Department of Biology and Environmental Science, Faculty of Sciences, Allama Iqbal Open University, Islamabad, Pakistan
| |
Collapse
|
30
|
Bahari D, Babamiri B, Salimi A, Salimizand H. Ratiometric fluorescence resonance energy transfer aptasensor for highly sensitive and selective detection of Acinetobacter baumannii bacteria in urine sample using carbon dots as optical nanoprobes. Talanta 2020; 221:121619. [PMID: 33076147 DOI: 10.1016/j.talanta.2020.121619] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 11/20/2022]
Abstract
Development of sensitive and selective analytical method for accurate diagnosis of Acinetobacter baumannii (Ab) bacteria in biological samples is a challenge. Herein, we developed an ingenious ratiometric fluorescent aptasensor for sensitive and selective detection of (Ab) bacteria based on fluorescence resonance energy transfer (FRET) between ortho-phenylenediamines carbon dot (o-CD), nitrogen-doped carbon nanodots (NCND) as donor's species and graphene oxide (GO) as acceptor. NCND that assembled onto the edge of graphene oxide (GO) exhibited quenched photoluminescence emission, and with the absorption of the modified o-CD with aptamer (o-CD-ssDNA) onto the graphene oxide surface the fluorescence of o-CD was efficiently quenched. The aptamer (ssDNA) as a biorecognition element is bound with A. baumannii specifically which releases the o-CD-ssDNA from GO and the recovery of the fluorescence signal of o-CD, while the fluorescence intensity of NCND only slightly altered and acted as the reference signal in ratiometric fluorescence assay. The fluorescence intensity ratio (I550 nm/I440nm) varied from 2.0 to 10.0 with the concentration of bacteria changing from 2.0 × 103 to 4.5 × 107 cfu/mL and the low detection limit of 3.0 × 102 cfu/mL (S/N = 3). The feasibility of the developed aptasensor for selective detection of A. baumannii in urine sample with satisfactory results was also demonstrated.
Collapse
Affiliation(s)
- Delnia Bahari
- Department of Chemistry, University of Kurdistan, 66177-15175, Sanandaj, Iran
| | - Bahareh Babamiri
- Department of Chemistry, University of Kurdistan, 66177-15175, Sanandaj, Iran
| | - Abdollah Salimi
- Department of Chemistry, University of Kurdistan, 66177-15175, Sanandaj, Iran; Research Center for Nanotechnology, University of Kurdistan, 66177-15175, Sanandaj, Iran.
| | - Himen Salimizand
- Department of Microbiology, Kurdistan University of Medical Sciences, 66177-13446, Sanandaj, Iran
| |
Collapse
|
31
|
Li S, Chen DQ, Ji L, Sun S, Jin Z, Jin ZL, Sun HW, Zeng H, Zhang WJ, Lu DS, Luo P, Zhao AN, Luo J, Zou QM, Li HB. Development of Different Methods for Preparing Acinetobacter baumannii Outer Membrane Vesicles Vaccine: Impact of Preparation Method on Protective Efficacy. Front Immunol 2020; 11:1069. [PMID: 32655550 PMCID: PMC7324643 DOI: 10.3389/fimmu.2020.01069] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 05/04/2020] [Indexed: 12/27/2022] Open
Abstract
Acinetobacter baumannii (A. baumannii) is becoming a common global concern due to the emergence of multi-drug or pan-drug resistant strains. Confronting the issue of antimicrobial resistance by developing vaccines against the resistant pathogen is becoming a common strategy. In this study, different methods for preparing A. baumannii outer membrane vesicles (AbOMVs) vaccines were developed. sOMV (spontaneously released AbOMV) was extracted from the culture supernatant, while SuOMV (sucrose-extracted AbOMV) and nOMV (native AbOMV) were prepared from the bacterial cells. Three AbOMVs exhibited significant differences in yield, particle size, protein composition, and LPS/DNA content. To compare the protective efficacy of the three AbOMVs, groups of mice were immunized either intramuscularly or intranasally with each AbOMV. Vaccination via both routes conferred significant protection against lethal and sub-lethal A. baumannii challenge. Moreover, intranasal vaccination provided more robust protection, which may be attributed to the induction of significant sIgA response in mucosal sites. Among the three AbOMVs, SuOMV elicited the highest level of protective immunity against A. baumannii infection, whether intramuscular or intranasal immunization, which was characterized by the expression of the most profound specific serum IgG or mucosal sIgA. Taken together, the preparation method had a significant effect on the yield, morphology, and composition of AbOMVs, that further influenced the protective effect against A. baumannii infection.
Collapse
Affiliation(s)
- Sun Li
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Da-Qun Chen
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Lu Ji
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Si Sun
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Zhe Jin
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Zi-Li Jin
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Hong-Wu Sun
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Hao Zeng
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Wei-Jun Zhang
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Dong-Shui Lu
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Ping Luo
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - An-Ni Zhao
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Jiao Luo
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Quan-Ming Zou
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Hai-Bo Li
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| |
Collapse
|
32
|
Mahmoudi Z, Rasooli I, Jahangiri A, Darvish Alipour Astaneh S. Prevention of nosocomial Acinetobacter baumannii infections with a conserved immunogenic fimbrial protein. APMIS 2020; 128:476-483. [PMID: 32445596 DOI: 10.1111/apm.13061] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 03/18/2020] [Indexed: 01/07/2023]
Abstract
Acinetobacter baumannii, one of the most life-threatening nosocomial drug-resistant pathogens, imposes high morbidity and mortality rates, thus highlighting immunization-based treatments or prevention measures. The selection of appropriate antigens can elicit protective immunity. The gene encoding a fimbrial protein introduced via reverse vaccinology was cloned, expressed and evaluated for immunogenicity in a murine model. Mice immunized with the recombinant protein were challenged with A. baumannii ATCC 19606. Adherence to A549 cell line of specific anti-sera treated A. baumannii was also assessed. Passive immunity was evaluated in a murine pneumonia model. Indirect ELISA showed a high specific antibody titre. Adherence of A. baumannii to A549 cell line decreased by 40% after incubation with 1:250 dilution of specific anti-sera. All the actively immunized mice survived. Bacterial load in the spleen and liver of the immunized mice was 3-fold lower than those of the control. The number of bacteria in the lungs of passively immunized mice was about 6-fold lower than the control mice. The fimbrial protein could be considered as a promising protective immunogen against A. baumannii.
Collapse
Affiliation(s)
| | - Iraj Rasooli
- Department of Biology, Shahed University, Tehran, Iran.,Molecular Microbiology Research Center, Shahed University, Tehran, Iran
| | - Abolfazl Jahangiri
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
33
|
Fereshteh S, Abdoli S, Shahcheraghi F, Ajdary S, Nazari M, Badmasti F. New putative vaccine candidates against Acinetobacter baumannii using the reverse vaccinology method. Microb Pathog 2020; 143:104114. [DOI: 10.1016/j.micpath.2020.104114] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 02/15/2020] [Accepted: 03/01/2020] [Indexed: 01/15/2023]
|
34
|
Chiang MH, Yang YS, Sun JR, Wang YC, Kuo SC, Lee YT, Chuang YP, Chen TL. Confronting Tigecycline-Resistant Acinetobacter baumannii via Immunization Against Conserved Resistance Determinants. Front Microbiol 2020; 11:536. [PMID: 32296411 PMCID: PMC7136391 DOI: 10.3389/fmicb.2020.00536] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 03/12/2020] [Indexed: 11/13/2022] Open
Abstract
Antimicrobial-resistant (AMR) bacterial infections, including those caused by Acinetobacter baumannii, have emerged as a clinical crisis worldwide. Immunization with AMR determinants has been suggested as a novel approach to combat AMR bacteria, but has not been validated. The present study targeted tigecycline (TGC) resistance determinants in A. baumannii to test the feasibility of this approach. Using bioinformatic tools, four candidates, AdeA, AdeI, AdeK, and TolC, belonging to the resistance-nodulation-division (RND) efflux pump were identified as highly conserved and exposed antigens from 15 A. baumannii genomes. Antisera generated from recombinant proteins showed the capability to reserve Hoechst 33342, a substrate of the efflux pump, in bacterial cells. The rTolC antisera had the highest complement-dependent killing and opsonophagocytosis effect compared to the sera from phosphate-buffered saline immunized mice. Among the antisera, anti-rAdeK-specific antisera decreased the minimal inhibitory concentration of TGC in 26.7% of the tested isolates. Immunization with rAdeK significantly potentiated TGC efficacy in treating TGC-resistant A. baumannii pneumonia in the murine model. The bacterial load (7.5 × 105 vs. 3.8 × 107, p < 0.01) and neutrophil infiltration in the peri-bronchial vasculature region of immunized mice was significantly lower compared to the PBS-immunized mice when TGC was administrated concomitantly. Collectively, these results suggest that active immunization against resistance determinants might be a feasible approach to combat multidrug-resistant pathogens in high risk population.
Collapse
Affiliation(s)
- Ming-Hsien Chiang
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Ya-Sung Yang
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Jun-Ren Sun
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Yung-Chih Wang
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Chen Kuo
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Yi-Tzu Lee
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Emergency Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Ping Chuang
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Te-Li Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
35
|
Lee SY, Yun SH, Lee H, Yi YS, Park EC, Kim W, Kim HY, Lee JC, Kim GH, Kim SI. Analysis of the Extracellular Proteome of Colistin-Resistant Korean Acinetobacter baumannii Strains. ACS OMEGA 2020; 5:5713-5720. [PMID: 32226849 PMCID: PMC7097930 DOI: 10.1021/acsomega.9b03723] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 02/27/2020] [Indexed: 05/08/2023]
Abstract
We analyzed the extracellular proteome of colistin-resistant Korean Acinetobacter baumannii (KAB) strains to identify proteome profiles that can be used to characterize extensively drug-resistant KAB strains. Four colistin-resistant KAB strains with colistin resistance associated with point mutations in pmrB and pmrC genes were analyzed. Analysis of the extracellular proteome of these strains revealed the presence of 506 induced common proteins, which were hence considered as the core extracellular proteome. Class C ADC-30 and class D OXA-23 β-lactamases were abundantly induced in these strains. Porins (CarO and CarO-like porin), outer membrane proteins (OmpH and BamABDE), transport protein (AdeK), receptor (TonB), and several proteins of unknown function were among the specifically induced proteins. Based on the sequence homology analysis of proteins from the core proteome and those of other A. baumannii strains and pathogenic bacterial species as well as further in silico screening, we propose that CarO-like porin is an A. baumannii-specific protein and that two tryptic peptides that originate from CarO-like porin detected by tandem mass spectrometry are peptide makers of this protein.
Collapse
Affiliation(s)
- Sang-Yeop Lee
- Research
Center for Bioconvergence Analysis, Korea
Basic Science Institute, Ochang 28119, Korea
- Center
for Convegent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea
| | - Sung Ho Yun
- Center
for Research Equipment, Korea Basic Science
Institute, Ochang 28119, Korea
| | - Hayoung Lee
- Research
Center for Bioconvergence Analysis, Korea
Basic Science Institute, Ochang 28119, Korea
- Department
of Bio-Analytical Science, University of
Science and Technology (UST), Daejeon 34113, Korea
| | - Yoon-Sun Yi
- Center
for Research Equipment, Korea Basic Science
Institute, Ochang 28119, Korea
| | - Edmond Changkyun Park
- Research
Center for Bioconvergence Analysis, Korea
Basic Science Institute, Ochang 28119, Korea
- Center
for Convegent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea
- Department
of Bio-Analytical Science, University of
Science and Technology (UST), Daejeon 34113, Korea
| | - Wooyoung Kim
- Research
Center for Bioconvergence Analysis, Korea
Basic Science Institute, Ochang 28119, Korea
- Center
for Convegent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea
- Department
of Toxicology, College of Pharmacy, Chungnam
National University, Daejeon 34134, Korea
| | - Hye-Yeon Kim
- Research
Center for Bioconvergence Analysis, Korea
Basic Science Institute, Ochang 28119, Korea
- Center
for Convegent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea
| | - Je Chul Lee
- Department
of Microbiology, School of Medicine, Kyungpook
National University, Daegu 41944, Korea
| | - Gun-Hwa Kim
- Research
Center for Bioconvergence Analysis, Korea
Basic Science Institute, Ochang 28119, Korea
- Department
of Bio-Analytical Science, University of
Science and Technology (UST), Daejeon 34113, Korea
| | - Seung Il Kim
- Research
Center for Bioconvergence Analysis, Korea
Basic Science Institute, Ochang 28119, Korea
- Center
for Convegent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea
- Department
of Bio-Analytical Science, University of
Science and Technology (UST), Daejeon 34113, Korea
- E-mail: . Phone: +82-43-240-5422. Fax: +82-43-240-5416
| |
Collapse
|
36
|
Abstract
Acinetobacter baumannii is one of the most problematic pathogens in clinical settings. Emerging of its antibiotic-resistant strains persuade researchers to find alternative treatment options such as immunization against the notorious nosocomial pathogen. Oma87 has been introduced as an immunogenic outer membrane protein via reverse vaccinology. However, protectivity of A. baumannii Oma87 is not well known. The current research undertakes a study on the immunogenicity of recombinant Oma87 in a murine model. Some physico-chemical properties were assessed via in silico analyses. The corresponding gene was amplified and cloned into pET28a plasmid. The recombinant protein was purified and then was administered to immunize mice. Sera obtained from the immunized mice were assessed with respect to the triggered antibodies. Challenges were performed on actively or passively immunized mice. In silico analyses revealed that this protein is the same as BamA. A high titer of specific antibody was raised against rOma87 even after the first injection. The specific antibody recognized the whole cell of A. baumannii. Both active and passive immunizations confer 100 and 50% protection, respectively against ~ 2 × lethal dose (LD) of A. baumannii in the murine sepsis model. Although none of mice received ~ 5 × LD of A. baumannii survived in passive immunization, 25% of mice challenged with ~ 7 × LD of the bacteria survived and the dead mice exhibited a delayed death. Based on these results, Oma87 is the same as BamA which could be considered as a promising vaccine candidate against A. baumannii in the sepsis model.
Collapse
|
37
|
Fernández-Prada M, López Trigo JA, Bayas JM, Cambronero MDR. [Quo vadis in vaccines: From the empirical approach to the new wave of technology]. Rev Esp Geriatr Gerontol 2020; 55:160-168. [PMID: 32014265 DOI: 10.1016/j.regg.2019.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 09/03/2019] [Accepted: 09/09/2019] [Indexed: 01/02/2023]
Abstract
The development of vaccines is a multifactorial process that has evolved and expanded, particularly over the last decades. The search for immunogenic vaccines that are also acceptably safe and tolerable enacted continuous technological advances in this field. In this regard, the technology applied to vaccines can historically be divided into 3 approaches: the empirical approach, the modern approach, and the new technological wave. The empirical approach for vaccine development includes whole micro-organisms, attenuation, inactivation, cell cultures and sub-unit vaccines. The modern approach contributed to leaps and bounds to vaccine development using chemical conjugation, as well as recombinant protein DNA technology and reverse vaccinology. Lastly, the new technological wave includes, among others, bioconjugation, viral vectors, synthetic biology, self-amplification of messenger RNA, generalized modules for membrane antigens, structural vaccinology and the new adjuvants.
Collapse
Affiliation(s)
- María Fernández-Prada
- Servicio de Medicina Preventiva y Salud Pública, Hospital Vital Álvarez Buylla, Mieres, España
| | | | - José M Bayas
- Departamento Médico, GSK, Tres Cantos, Madrid, España
| | | |
Collapse
|
38
|
Mobarak Qamsari M, Rasooli I, Darvish Alipour Astaneh S. Identification and immunogenic properties of recombinant ZnuD protein loops of Acinetobacter baumannii. INFORMATICS IN MEDICINE UNLOCKED 2020. [DOI: 10.1016/j.imu.2020.100342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
|
39
|
Aghajani Z, Rasooli I, Mousavi Gargari SL. Exploitation of two siderophore receptors, BauA and BfnH, for protection against Acinetobacter baumannii infection. APMIS 2019; 127:753-763. [PMID: 31512768 DOI: 10.1111/apm.12992] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 08/19/2019] [Indexed: 01/30/2023]
Abstract
Iron uptake system is expressed in early stages of Acinetobacter baumannii infections under iron-restricted conditions. This study is aimed at the evaluation of immuno-protectivity of BfnH in comparison with BauA in both mature and selected fragmental proteins. The study was designed in single and combined forms of antigens. BfnH is presented in 3472 strains of A. baumannii with more than 97% identity. The preliminary immune-informatics analysis of this protein indicated a region from the β-barrel domain including exposed loops 2-5, with antigenic score comparable to that of BfnH. There was a significant rise in the specific IgG response in all test groups. The bacterial challenge with a lethal dose of A. baumannii demonstrated partial protection of whole proteins which coincides with a significant reduction in the bacterial population colonized in the main organs and an increase in the survival level. Passive immunization of the mice brought about 50% survival in the mice groups immunized with BfnH and with a combination of BfnH and BauA. The protectivity of siderophore receptors suggests their potential immunogenic role that could be considered as a component of multivalent subunit vaccine candidates against A. baumannii.
Collapse
Affiliation(s)
| | - Iraj Rasooli
- Department of Biology, Shahed University, Tehran, Iran.,Molecular Microbiology Research Center, Shahed University, Tehran, Iran
| | | |
Collapse
|
40
|
DNA vaccine encoding OmpA and Pal from Acinetobacter baumannii efficiently protects mice against pulmonary infection. Mol Biol Rep 2019; 46:5397-5408. [PMID: 31342294 DOI: 10.1007/s11033-019-04994-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 07/19/2019] [Indexed: 02/01/2023]
Abstract
Acinetobacter baumannii (A. baumannii) is an opportunistic pathogen that causes serious infections in the lungs, blood, and brain in critically ill hospital patients, resulting in considerable mortality rates every year. Due to the rapid appearance of multi-drug resistance or even pan-drug resistance isolates, it is becoming more and more difficult to cure A. baumannii infection by traditional antibiotic treatment, alternative strategies are urgently required to combat A. baumannii infection. In this study, we developed a DNA vaccine encoding two antigens from A. baumannii, OmpA and Pal, and the immunogenicity and protective efficacy was further evaluated. The results showed that the DNA vaccine exhibited significant immune protective efficacy against acute A. baumannii infection in a mouse pneumonia model, and cross protective efficacy was observed when immunized mice were challenged with clinical strains of A. baumannii. DNA vaccine immunization induced high level of humoral response and a mixed Th1/Th2/Th17 cellular response, which protect against lethal bacterial challenges by decreased bacterial loads and pathology in the lungs, and reduced level of inflammatory cytokines expression and inflammatory cell infiltration in BALF. These results demonstrated that it is possible to prevent A. baumannii infection by DNA vaccine and both OmpA and Pal could be serve as promising candidate antigens.
Collapse
|
41
|
Vilela Rodrigues TC, Jaiswal AK, de Sarom A, de Castro Oliveira L, Freire Oliveira CJ, Ghosh P, Tiwari S, Miranda FM, de Jesus Benevides L, Ariston de Carvalho Azevedo V, de Castro Soares S. Reverse vaccinology and subtractive genomics reveal new therapeutic targets against Mycoplasma pneumoniae: a causative agent of pneumonia. ROYAL SOCIETY OPEN SCIENCE 2019; 6:190907. [PMID: 31417766 PMCID: PMC6689572 DOI: 10.1098/rsos.190907] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 07/04/2019] [Indexed: 05/03/2023]
Abstract
Pneumonia is an infectious disease caused by bacteria, viruses or fungi that results in millions of deaths globally. Despite the existence of prophylactic methods against some of the major pathogens of the disease, there is no efficient prophylaxis against atypical agents such as Mycoplasma pneumoniae, a bacterium associated with cases of community-acquired pneumonia. Because of the morphological peculiarity of M. pneumoniae, which leads to an increased resistance to antibiotics, studies that prospectively investigate the development of vaccines and drug targets appear to be one of the best ways forward. Hence, in this paper, bioinformatics tools were used for vaccine and pharmacological prediction. We conducted comparative genomic analysis on the genomes of 88 M. pneumoniae strains, as opposed to a reverse vaccinology analysis, in relation to the capacity of M. pneumoniae proteins to bind to the major histocompatibility complex, revealing seven targets with immunogenic potential. Predictive cytoplasmic proteins were tested as potential drug targets by studying their structures in relation to other proteins, metabolic pathways and molecular anchorage, which identified five possible drug targets. These findings are a valuable addition to the development of vaccines and the selection of new in vivo drug targets that may contribute to further elucidating the molecular basis of M. pneumoniae-host interactions.
Collapse
Affiliation(s)
| | - Arun Kumar Jaiswal
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Minas Gerais, Brazil
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Alissa de Sarom
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Minas Gerais, Brazil
| | - Letícia de Castro Oliveira
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Minas Gerais, Brazil
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Carlo José Freire Oliveira
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Minas Gerais, Brazil
| | - Preetam Ghosh
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Sandeep Tiwari
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Fábio Malcher Miranda
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Leandro de Jesus Benevides
- Bioinformatics Laboratory - LABINFO, National Laboratory of Scientific Computation - LNCC/MCTI, Rio de Janeiro, Brazil
| | | | - Siomar de Castro Soares
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Minas Gerais, Brazil
- Author for correspondence: Siomar de Castro Soares e-mail:
| |
Collapse
|
42
|
Mujawar S, Mishra R, Pawar S, Gatherer D, Lahiri C. Delineating the Plausible Molecular Vaccine Candidates and Drug Targets of Multidrug-Resistant Acinetobacter baumannii. Front Cell Infect Microbiol 2019; 9:203. [PMID: 31281799 PMCID: PMC6596342 DOI: 10.3389/fcimb.2019.00203] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/23/2019] [Indexed: 12/22/2022] Open
Abstract
Nosocomial infections have become alarming with the increase of multidrug-resistant bacterial strains of Acinetobacter baumannii. Being the causative agent in ~80% of the cases, these pathogenic gram-negative species could be deadly for hospitalized patients, especially in intensive care units utilizing ventilators, urinary catheters, and nasogastric tubes. Primarily infecting an immuno-compromised system, they are resistant to most antibiotics and are the root cause of various types of opportunistic infections including but not limited to septicemia, endocarditis, meningitis, pneumonia, skin, and wound sepsis and even urinary tract infections. Conventional experimental methods including typing, computational methods encompassing comparative genomics, and combined methods of reverse vaccinology and proteomics had been proposed to differentiate and develop vaccines and/or drugs for several outbreak strains. However, identifying proteins suitable enough to be posed as drug targets and/or molecular vaccines against the multidrug-resistant pathogenic bacterial strains has probably remained an open issue to address. In these cases of novel protein identification, the targets either are uncharacterized or have been unable to confer the most coveted protection either in the form of molecular vaccine candidates or as drug targets. Here, we report a strategic approach with the 3,766 proteins from the whole genome of A. baumannii ATCC19606 (AB) to rationally identify plausible candidates and propose them as future molecular vaccine candidates and/or drug targets. Essentially, we started with mapping the vaccine candidates (VaC) and virulence factors (ViF) of A. baumannii strain AYE onto strain ATCC19606 to identify them in the latter. We move on to build small networks of VaC and ViF to conceptualize their position in the network space of the whole genomic protein interactome (GPIN) and rationalize their candidature for drugs and/or molecular vaccines. To this end, we propose new sets of known proteins unearthed from interactome built using key factors, KeF, potent enough to compete with VaC and ViF. Our method is the first of its kind to propose, albeit theoretically, a rational approach to identify crucial proteins and pose them for candidates of vaccines and/or drugs effective enough to combat the deadly pathogenic threats of A. baumannii.
Collapse
Affiliation(s)
- Shama Mujawar
- Department of Biological Sciences, Sunway University, Petaling Jaya, Malaysia
| | - Rohit Mishra
- Department of Bioinformatics, University of Mumbai, Mumbai, India
| | - Shrikant Pawar
- Department of Computer Science, Georgia State University, Atlanta, GA, United States.,Department of Biology, Georgia State University, Atlanta, GA, United States
| | - Derek Gatherer
- Division of Biomedical and Life Sciences, Lancaster University, Lancaster, United Kingdom
| | - Chandrajit Lahiri
- Department of Biological Sciences, Sunway University, Petaling Jaya, Malaysia
| |
Collapse
|
43
|
Bolourchi N, Shahcheraghi F, Shirazi AS, Janani A, Bahrami F, Badmasti F. Immunogenic reactivity of recombinant PKF and AbOmpA proteins as serum resistance factors against sepsis of Acinetobacter baumannii. Microb Pathog 2019; 131:9-14. [DOI: 10.1016/j.micpath.2019.03.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 03/23/2019] [Accepted: 03/25/2019] [Indexed: 01/20/2023]
|
44
|
Combating tigecycline resistant Acinetobacter baumannii: A leap forward towards multi-epitope based vaccine discovery. Eur J Pharm Sci 2019; 132:1-17. [DOI: 10.1016/j.ejps.2019.02.023] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/31/2019] [Accepted: 02/16/2019] [Indexed: 01/27/2023]
|
45
|
Salvador E, Pires de Souza G, Cotta Malaquias L, Wang T, Leomil Coelho L. Identification of relevant regions on structural and nonstructural proteins of Zika virus for vaccine and diagnostic test development: an in silico approach. New Microbes New Infect 2019; 29:100506. [PMID: 30858979 PMCID: PMC6396434 DOI: 10.1016/j.nmni.2019.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/11/2019] [Accepted: 01/15/2019] [Indexed: 01/07/2023] Open
Abstract
Zika virus (ZIKV) is an arbovirus belonging to the Flaviviridae family and the genus Flavivirus. Infection with ZIKV causes a mild, self-limiting febrile illness called Zika fever. However, ZIKV infection has been recently associated with microcephaly and Guillain-Barré syndrome. Vaccines for the disease are a high priority of World Health Organization. Several studies are currently being conducted to develop a vaccine against ZIKV, but until now there is no licensed ZIKV vaccine. This study used a novel immunoinformatics approach to identify potential T-cell immunogenic epitopes present in the structural and nonstructural proteins of ZIKV. Fourteen T-cell candidate epitopes were identified on ZIKV structural and nonstructural proteins: pr36-50; C61-75; C103-117; E374-382; E477-491; NS2a90-104; NS2a174-188; NS2a179-193; NS2a190-204; NS2a195-209; NS2a200-214; NS3175-189; and NS4a82-96; NS4a99-113. Among these epitopes, only E374-382 is a human leukocyte antigen (HLA) type I restricted epitope. All identified epitopes showed a low similarity with other important flaviviruses but had a high conservation rate among the ZIKV strains and a high population coverage rate. Therefore, these predicted T-cell epitopes are potential candidates targets for development of vaccines to prevent ZIKV infection.
Collapse
Affiliation(s)
- E.A. Salvador
- Institute of Biomedical Sciences, Department of Microbiology and Immunology, Federal University of Alfenas, Minas Gerais, Brazil
| | - G.A. Pires de Souza
- Institute of Biomedical Sciences, Department of Microbiology and Immunology, Federal University of Alfenas, Minas Gerais, Brazil
| | - L.C. Cotta Malaquias
- Institute of Biomedical Sciences, Department of Microbiology and Immunology, Federal University of Alfenas, Minas Gerais, Brazil
| | - T. Wang
- Department of Microbiology & Immunology, Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - L.F. Leomil Coelho
- Institute of Biomedical Sciences, Department of Microbiology and Immunology, Federal University of Alfenas, Minas Gerais, Brazil
- Corresponding author: L. F. Leomil Coelho, Laboratório de Vacinas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Rua Gabriel Monteiro, 700 Centro, Alfenasm Minas Gerais, 37130-001, Brazil.
| |
Collapse
|
46
|
Bidmos FA, Siris S, Gladstone CA, Langford PR. Bacterial Vaccine Antigen Discovery in the Reverse Vaccinology 2.0 Era: Progress and Challenges. Front Immunol 2018; 9:2315. [PMID: 30349542 PMCID: PMC6187972 DOI: 10.3389/fimmu.2018.02315] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 09/17/2018] [Indexed: 11/13/2022] Open
Abstract
The ongoing, and very serious, threat from antimicrobial resistance necessitates the development and use of preventative measures, predominantly vaccination. Polysaccharide-based vaccines have provided a degree of success in limiting morbidity from disseminated bacterial infections, including those caused by the major human obligate pathogens, Neisseria meningitidis, and Streptococcus pneumoniae. Limitations of these polysaccharide vaccines, such as partial coverage and induced escape leading to persistence of disease, provide a compelling argument for the development of protein vaccines. In this review, we briefly chronicle approaches that have yielded licensed vaccines before highlighting reverse vaccinology 2.0 and its potential application in the discovery of novel bacterial protein vaccine candidates. Technical challenges and research gaps are also discussed.
Collapse
Affiliation(s)
- Fadil A Bidmos
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Sara Siris
- Department of Medicine, Imperial College London, London, United Kingdom
| | | | - Paul R Langford
- Department of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
47
|
Nazir Z, Afridi SG, Shah M, Shams S, Khan A. Reverse vaccinology and subtractive genomics-based putative vaccine targets identification for Burkholderia pseudomallei Bp1651. Microb Pathog 2018; 125:219-229. [PMID: 30243554 DOI: 10.1016/j.micpath.2018.09.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/16/2018] [Accepted: 09/18/2018] [Indexed: 12/16/2022]
Abstract
The Burkholderia pseudomallei is a unique bio-threat and causative agent of melioidosis. The B. pseudomallei Bp1651 strain has been isolated from a chronic cystic fibrosis patient. The genome-level DNA sequences information of this strain has recently been published. Unfortunately, there is no commercial vaccine available till date to combat B. pseudomallei infection. The genome-wide prioritization approaches are widely used for the identification of potential therapeutic candidates against pathogens. In the present study, we utilized the recently available annotated genomic information of B. pseudomallei Bp1651 through subtractive genomics and reverse-vaccinology strategies to identify its potential vaccine targets. The analyses identified more than 60 pathogen-specific, human host non-homologous proteins that may prioritize in future studies to investigate therapeutic targets for B. pseudomallei Bp1651. The potential B and T-cells antigenic determinant peptides from these pathogen-specific proteins were cataloged using antigenicity and epitope prediction tools. The analyses unveiled a promising antigenic peptide "FQWEFSLSV" from protein-export membrane protein (SecF) of Bp1651 strain, which was predicted to interact with multiple class I and class II MHC alleles with IC50 value < 100 nM. The molecular docking analysis verified favorable molecular interaction of this lead antigenic peptide with the ligand-binding pocket residues of HLA A*02:06 human host immune cell surface receptor. This peptide is predicted to be a suitable epitope capable to elicit the cell-mediated immune response against the B. pseudomallei pathogen. The putative epitopes and proteins identified in this study may be promising vaccine targets against Bp1651 as well as other pathogenic strains of B. pseudomallei.
Collapse
Affiliation(s)
- Zarghoona Nazir
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, 23200, Khyber Pakhtunkhwa, Pakistan
| | - Sahib Gul Afridi
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, 23200, Khyber Pakhtunkhwa, Pakistan
| | - Mohibullah Shah
- Department of Biochemistry, Bahauddin Zakariya University Multan, Punjab, Pakistan
| | - Sulaiman Shams
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, 23200, Khyber Pakhtunkhwa, Pakistan
| | - Asifullah Khan
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, 23200, Khyber Pakhtunkhwa, Pakistan.
| |
Collapse
|
48
|
Watkins RR. A formidable foe: carbapenem-resistant Acinetobacter baumannii and emerging nonantibiotic therapies. Expert Rev Anti Infect Ther 2018; 16:591-593. [PMID: 30024320 DOI: 10.1080/14787210.2018.1503054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Richard R Watkins
- a Division of Infectious Diseases , Cleveland Clinic Akron General , Akron , OH , USA.,b Department of Internal Medicine , Northeast Ohio Medical University , Rootstown , Ohio , USA
| |
Collapse
|
49
|
Gagneux-Brunon A, Lucht F, Launay O, Berthelot P, Botelho-Nevers E. Vaccines for healthcare-associated infections: present, future, and expectations. Expert Rev Vaccines 2018; 17:421-433. [DOI: 10.1080/14760584.2018.1470507] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Amandine Gagneux-Brunon
- Inserm, CIC 1408, I-REIVAC, University Hospital of Saint-Etienne, Saint-Etienne, France
- GIMAP EA 3064, University of Lyon, Saint-Etienne, France
| | - Frédéric Lucht
- Inserm, CIC 1408, I-REIVAC, University Hospital of Saint-Etienne, Saint-Etienne, France
- GIMAP EA 3064, University of Lyon, Saint-Etienne, France
| | - Odile Launay
- Inserm CIC 1417, I-REIVAC, University of Paris-Descartes, University Hospital of Cochin-Broca-Hôtel-Dieu, Paris, France
| | - Philippe Berthelot
- GIMAP EA 3064, University of Lyon, Saint-Etienne, France
- Infection control unit, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Elisabeth Botelho-Nevers
- Inserm, CIC 1408, I-REIVAC, University Hospital of Saint-Etienne, Saint-Etienne, France
- GIMAP EA 3064, University of Lyon, Saint-Etienne, France
| |
Collapse
|
50
|
Ahmad S, Azam SS. A novel approach of virulome based reverse vaccinology for exploring and validating peptide-based vaccine candidates against the most troublesome nosocomial pathogen: Acinetobacter baumannii. J Mol Graph Model 2018; 83:1-11. [PMID: 29753164 DOI: 10.1016/j.jmgm.2018.04.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 04/27/2018] [Accepted: 04/30/2018] [Indexed: 12/12/2022]
Abstract
Acinetobacter baumannii is one of the major cause of nosocomial infections around the globe. The emergence of hyper-virulent strains of the pathogen greatly narrows down therapeutic options for patients infected with this red alert superbug. Development of a peptide-based vaccine can offers an alternative, attractive, and cost-effective remedy for multidrug-resistant A. baumannii associated complications. Herein, we introduced a novel virulome based Reverse Vaccinology for screening peptide based vaccine candidates against A. baumannii and its validation using a negative control. The pipeline screened "FYLNDQPVS" of polysaccharide export outer membrane protein (EpsA) and "LQNNTRRMK" of chaperone-usher pathway protein B (CsuB) as broad-spectrum peptides for induction of targeted immune responses. The 9-mer epitope of both proteins was rendered virulent, antigenic, non-allergen, and highly conserved among thirty-four completely annotated strains. Interactome examination unravels peptides protein direct and indirect interactions with biological significant pathways, essential for A. baumannii pathogenesis and survival. Protein-peptide docking aids in addition by unveiling deep binding of the epitopes in the active site of the most prevalent binding allele in the human population-the DRB1*0101. Both the proteins till to date are not characterized for immunoprotective efficacy and desirable to be deciphered experimentally. The designed series of in silico filters rejected few recently reported peptide and non-peptide vaccine targets and has delivered outcomes, which we believe will enrich the existing knowledge of vaccinology against this life-threatening human pathogen.
Collapse
Affiliation(s)
- Sajjad Ahmad
- Computational Biology Lab, National Center for Bioinformatics (NCB), Quaid-i-Azam University, Islamabad, Pakistan.
| | - Syed Sikander Azam
- Computational Biology Lab, National Center for Bioinformatics (NCB), Quaid-i-Azam University, Islamabad, Pakistan.
| |
Collapse
|