1
|
Adams CS, Kim H, Burtner AE, Lee DS, Dobbins C, Criswell C, Coventry B, Kim HM, King NP. De novo design of protein minibinder agonists of TLR3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.17.589973. [PMID: 38659926 PMCID: PMC11042314 DOI: 10.1101/2024.04.17.589973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Toll-like Receptor 3 (TLR3) is a pattern recognition receptor that initiates antiviral immune responses upon binding double-stranded RNA (dsRNA). Several nucleic acid-based TLR3 agonists have been explored clinically as vaccine adjuvants in cancer and infectious disease, but present substantial manufacturing and formulation challenges. Here, we use computational protein design to create novel miniproteins that bind to human TLR3 with nanomolar affinities. Cryo-EM structures of two minibinders in complex with TLR3 reveal that they bind the target as designed, although one partially unfolds due to steric competition with a nearby N-linked glycan. Multimeric forms of both minibinders induce NF-κB signaling in TLR3-expressing cell lines, demonstrating that they may have therapeutically relevant biological activity. Our work provides a foundation for the development of specific, stable, and easy-to-formulate protein-based agonists of TLRs and other pattern recognition receptors.
Collapse
Affiliation(s)
- Chloe S. Adams
- Institute for Protein Design, University of Washington, Seattle, WA, 98195 USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195 USA
| | - Hyojin Kim
- Center for Biomolecular & Cellular Structure, Institute for Basic Science (IBS), Daejeon 34126, South Korea
| | - Abigail E. Burtner
- Institute for Protein Design, University of Washington, Seattle, WA, 98195 USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195 USA
| | - Dong Sun Lee
- Center for Biomolecular & Cellular Structure, Institute for Basic Science (IBS), Daejeon 34126, South Korea
| | - Craig Dobbins
- Institute for Protein Design, University of Washington, Seattle, WA, 98195 USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195 USA
| | - Cameron Criswell
- Institute for Protein Design, University of Washington, Seattle, WA, 98195 USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195 USA
| | - Brian Coventry
- Institute for Protein Design, University of Washington, Seattle, WA, 98195 USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195 USA
| | - Ho Min Kim
- Center for Biomolecular & Cellular Structure, Institute for Basic Science (IBS), Daejeon 34126, South Korea
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, South Korea
| | - Neil P. King
- Institute for Protein Design, University of Washington, Seattle, WA, 98195 USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195 USA
| |
Collapse
|
2
|
Abdelaziz K, Helmy YA, Yitbarek A, Hodgins DC, Sharafeldin TA, Selim MSH. Advances in Poultry Vaccines: Leveraging Biotechnology for Improving Vaccine Development, Stability, and Delivery. Vaccines (Basel) 2024; 12:134. [PMID: 38400118 PMCID: PMC10893217 DOI: 10.3390/vaccines12020134] [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: 01/01/2024] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
With the rapidly increasing demand for poultry products and the current challenges facing the poultry industry, the application of biotechnology to enhance poultry production has gained growing significance. Biotechnology encompasses all forms of technology that can be harnessed to improve poultry health and production efficiency. Notably, biotechnology-based approaches have fueled rapid advances in biological research, including (a) genetic manipulation in poultry breeding to improve the growth and egg production traits and disease resistance, (b) rapid identification of infectious agents using DNA-based approaches, (c) inclusion of natural and synthetic feed additives to poultry diets to enhance their nutritional value and maximize feed utilization by birds, and (d) production of biological products such as vaccines and various types of immunostimulants to increase the defensive activity of the immune system against pathogenic infection. Indeed, managing both existing and newly emerging infectious diseases presents a challenge for poultry production. However, recent strides in vaccine technology are demonstrating significant promise for disease prevention and control. This review focuses on the evolving applications of biotechnology aimed at enhancing vaccine immunogenicity, efficacy, stability, and delivery.
Collapse
Affiliation(s)
- Khaled Abdelaziz
- Department of Animal and Veterinary Science, College of Agriculture, Forestry and Life Sciences, Clemson University Poole Agricultural Center, Jersey Ln #129, Clemson, SC 29634, USA
- Clemson University School of Health Research (CUSHR), Clemson, SC 29634, USA
| | - Yosra A. Helmy
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food, and Environment, University of Kentucky, Lexington, KY 40546, USA;
| | - Alexander Yitbarek
- Department of Animal & Food Sciences, University of Delaware, 531 S College Ave, Newark, DE 19716, USA;
| | - Douglas C. Hodgins
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - Tamer A. Sharafeldin
- Department of Veterinary Biomedical Science, Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD 57007, USA; (T.A.S.); (M.S.H.S.)
| | - Mohamed S. H. Selim
- Department of Veterinary Biomedical Science, Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD 57007, USA; (T.A.S.); (M.S.H.S.)
| |
Collapse
|
3
|
Khim K, Puth S, Radhakrishnan K, Nguyen TD, Lee YS, Jung CH, Lee SE, Rhee JH. Deglycosylation of eukaryotic-expressed flagellin restores adjuvanticity. NPJ Vaccines 2023; 8:139. [PMID: 37752138 PMCID: PMC10522637 DOI: 10.1038/s41541-023-00738-3] [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: 11/24/2022] [Accepted: 09/15/2023] [Indexed: 09/28/2023] Open
Abstract
Flagellin, the TLR5 agonist, shows potent adjuvant activities in diverse vaccines and immunotherapies. Vibrio vulnificus flagellin B expressed in eukaryotic cells (eFlaB) could not stimulate TLR5 signaling. Enzymatic deglycosylation restored eFlaB's TLR5 stimulating functionality, suggesting that glycosylation interferes with eFlaB binding to TLR5. Site-directed mutagenesis of N-glycosylation residues restored TLR5 stimulation and adjuvanticity. Collectively, deglycosylated eFlaB may provide a built-in adjuvant platform for eukaryotic-expressed antigens and nucleic acid vaccines.
Collapse
Affiliation(s)
- Koemchhoy Khim
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
- Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, Republic of Korea
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
| | - Sao Puth
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea
- Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, Republic of Korea
| | - Kamalakannan Radhakrishnan
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
| | - Tien Duc Nguyen
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
| | - Youn Suhk Lee
- Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, Republic of Korea
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Che-Hun Jung
- Department of Chemistry, Chonnam National University, Gwangju, Republic of Korea
| | - Shee Eun Lee
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea.
- Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, Republic of Korea.
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea.
| | - Joon Haeng Rhee
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea.
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea.
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea.
| |
Collapse
|
4
|
Wang X, Qiu W, Liu H, He M, He W, Li Z, Wu Z, Xu X, Chen P. The inducible secreting TLR5 agonist, CBLB502, enhances the anti-tumor activity of CAR133-NK92 cells in colorectal cancer. Cancer Biol Med 2023; 20:j.issn.2095-3941.2023.0033. [PMID: 37731205 PMCID: PMC10546094 DOI: 10.20892/j.issn.2095-3941.2023.0033] [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: 01/29/2023] [Accepted: 07/17/2023] [Indexed: 09/22/2023] Open
Abstract
OBJECTIVE CAR-T/NK cells have had limited success in the treatment of solid tumors, such as colorectal cancer (CRC), in part because of the heterogeneous nature of tumor-associated antigens that lead to antigen-negative relapse after the initial response. This barrier might be overcome by enhancing the recruitment and durability of endogenous immune cells. METHODS Immunohistochemistry and flow cytometry were used to assess the expression of CD133 antigen in tissue microarrays and cell lines, respectively. Retroviral vector transduction was used to generate CBLB502-secreting CAR133-NK92 cells (CAR133-i502-NK92). The tumor killing capacity of CAR133-NK92 cells in vitro and in vivo were quantified via LDH release, the RTCA assay, and the degranulation test, as well as measuring tumor bioluminescence signal intensity in mice xenografts. RESULTS We engineered CAR133-i502-NK92 cells and demonstrated that those cells displayed enhanced proliferation (9.0 × 104 cells vs. 7.0 × 104 cells) and specific anti-tumor activities in vitro and in a xenogeneic mouse model, and were well-tolerated. Notably, CBLB502 secreted by CAR133-i502-NK92 cells effectively activated endogenous immune cells. Furthermore, in hCD133+/hCD133- mixed cancer xenograft models, CAR133-i502-NK92 cells suppressed cancer growth better than the counterparts (n = 5, P = 0.0297). Greater T-cell infiltration was associated with greater anti-tumor potency (P < 0.0001). CONCLUSIONS Armed with a CBLB502 TLR5 agonist, CAR133-NK92 cells were shown to be capable of specifically eliminating CD133-positive colon cancer cells in a CAR133-dependent manner and indirectly eradicating CD133-negative colon cancer cells in a CBLB502-specific endogenous immune response manner. This study describes a novel technique for optimizing CAR-T/NK cells for the treatment of antigenically-diverse solid tumors.
Collapse
Affiliation(s)
- Xiaohui Wang
- College of Biotechnology, Southwest University, Chongqing 400715, China
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Wei Qiu
- Department of Dermatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Haoyu Liu
- College of Biotechnology, Southwest University, Chongqing 400715, China
| | - Min He
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Wei He
- College of Biotechnology, Southwest University, Chongqing 400715, China
| | - Zhan Li
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Zhiqiang Wu
- Department of Biotherapeutics, The First Medical Center, Chinese PLA General Hospital, Beijing 100038, China
| | - Xiang Xu
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Ping Chen
- College of Biotechnology, Southwest University, Chongqing 400715, China
| |
Collapse
|
5
|
Jackson Hoffman BA, Pumford EA, Enueme AI, Fetah KL, Friedl OM, Kasko AM. Engineered macromolecular Toll-like receptor agents and assemblies. Trends Biotechnol 2023; 41:1139-1154. [PMID: 37068999 DOI: 10.1016/j.tibtech.2023.03.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/06/2023] [Accepted: 03/13/2023] [Indexed: 04/19/2023]
Abstract
Macromolecular Toll-like receptor (TLR) agents have been utilized as agonists and inhibitors in preclinical and clinical settings. These agents interface with the TLR class of innate immune receptors which recognize macromolecular ligands that are characteristic of pathogenic material. As such, many agents that have been historically investigated are derived from the natural macromolecules which activate or inhibit TLRs. This review covers recent research and clinically available TLR agents that are macromolecular or polymeric. Synthetic materials that have been found to interface with TLRs are also discussed. Assemblies of these materials are investigated in the context of improving stability or efficacy of ligands. Attention is given to strategies which modify or enhance the current agents and to future outlooks on the development of these agents.
Collapse
Affiliation(s)
| | - Elizabeth A Pumford
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Amaka I Enueme
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Kirsten L Fetah
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Olivia M Friedl
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Andrea M Kasko
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA; California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
6
|
Immunogenic Modification of Ligilactobacillus agilis by Specific Amino Acid Substitution of Flagellin. Appl Environ Microbiol 2022; 88:e0127722. [PMID: 36173204 PMCID: PMC9599256 DOI: 10.1128/aem.01277-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Ligilactobacillus agilis is a flagellated motile commensal microbe that resides in the gastrointestinal tract of mammals and birds. Flagellin, the major subunit protein of flagellar filament, from pathogenic bacteria is generally a proinflammatory molecule that stimulates immune cells via Toll-like receptor 5 (TLR5). Interestingly, the flagellins of L. agilis are known to be immunologically attenuated despite the fact that the structure of the proteins, including the TLR5 recognition site, is highly conserved among bacteria. The results of our previous study suggested that this is attributed to the differences in three specific amino acids within the conserved TLR5 recognition site; however, this hypothesis remains to be confirmed. In this study, a series of recombinant L. agilis flagellins, with amino acid substitutions at the TLR5 recognition site, were constructed, and their immunogenic activity was evaluated in vitro. Then, an L. agilis strain with an active immunogenic TLR5 recognition site was generated. In vitro and in vivo immunological studies revealed that the mutant L. agilis strain with the modified flagellin was more immunogenic than the wild-type strain. In conclusion, the specific amino acid residues in L. agilis flagellins likely contribute to the discrimination between pathogens and commensals by the host defense system. Additionally, the immunogenically potent L. agilis mutants may serve as a useful platform for oral vaccine delivery. IMPORTANCE The interactions between gut microbes and immune cells play an important role in the health and disease of hosts. Ligilactobacillus agilis is a flagellated commensal bacterium found in the gut of mammals and birds. However, the flagellin proteins of L. agilis are immunologically attenuated and barely induce TLR5-dependent inflammation, unlike the flagellins of several pathogenic bacteria. This study demonstrated that three specific amino acids in the flagellin protein are responsible for this low immunogenicity in L. agilis. The results obtained herein improve our understanding of the symbiosis between gut microbes and their hosts.
Collapse
|
7
|
Díaz-Dinamarca DA, Salazar ML, Castillo BN, Manubens A, Vasquez AE, Salazar F, Becker MI. Protein-Based Adjuvants for Vaccines as Immunomodulators of the Innate and Adaptive Immune Response: Current Knowledge, Challenges, and Future Opportunities. Pharmaceutics 2022; 14:1671. [PMID: 36015297 PMCID: PMC9414397 DOI: 10.3390/pharmaceutics14081671] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/05/2022] [Accepted: 08/07/2022] [Indexed: 12/03/2022] Open
Abstract
New-generation vaccines, formulated with subunits or nucleic acids, are less immunogenic than classical vaccines formulated with live-attenuated or inactivated pathogens. This difference has led to an intensified search for additional potent vaccine adjuvants that meet safety and efficacy criteria and confer long-term protection. This review provides an overview of protein-based adjuvants (PBAs) obtained from different organisms, including bacteria, mollusks, plants, and humans. Notably, despite structural differences, all PBAs show significant immunostimulatory properties, eliciting B-cell- and T-cell-mediated immune responses to administered antigens, providing advantages over many currently adopted adjuvant approaches. Furthermore, PBAs are natural biocompatible and biodegradable substances that induce minimal reactogenicity and toxicity and interact with innate immune receptors, enhancing their endocytosis and modulating subsequent adaptive immune responses. We propose that PBAs can contribute to the development of vaccines against complex pathogens, including intracellular pathogens such as Mycobacterium tuberculosis, those with complex life cycles such as Plasmodium falciparum, those that induce host immune dysfunction such as HIV, those that target immunocompromised individuals such as fungi, those with a latent disease phase such as Herpes, those that are antigenically variable such as SARS-CoV-2 and those that undergo continuous evolution, to reduce the likelihood of outbreaks.
Collapse
Affiliation(s)
- Diego A. Díaz-Dinamarca
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7750000, Chile
| | - Michelle L. Salazar
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
| | - Byron N. Castillo
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
| | - Augusto Manubens
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
- Biosonda Corporation, Santiago 7750000, Chile
| | - Abel E. Vasquez
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7750000, Chile
- Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Providencia, Santiago 8320000, Chile
| | - Fabián Salazar
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter EX4 4QD, UK
| | - María Inés Becker
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
- Biosonda Corporation, Santiago 7750000, Chile
| |
Collapse
|
8
|
Intranasal Immunization with Zika Virus Envelope Domain III-Flagellin Fusion Protein Elicits Systemic and Mucosal Immune Responses and Protection against Subcutaneous and Intravaginal Virus Challenges. Pharmaceutics 2022; 14:pharmaceutics14051014. [PMID: 35631599 PMCID: PMC9144594 DOI: 10.3390/pharmaceutics14051014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 11/25/2022] Open
Abstract
Zika virus (ZIKV) infections in humans are mainly transmitted by the mosquito vectors, but human-to-human sexual transmission is also another important route. Developing a ZIKV mucosal vaccine that can elicit both systemic and mucosal immune responses is of particular interest. In this study, we constructed a recombinant ZIKV envelope DIII (ZDIII) protein genetically fused with Salmonella typhimurium flagellin (FliC-ZDIII) as a novel mucosal antigen for intranasal immunization. The results indicated that the FliC-ZDIII fusion proteins formulated with E. coli heat-labile enterotoxin B subunit (LTIIb-B5) adjuvant greatly increased the ZDIII-specific IgG, IgA, and neutralizing titers in sera, and the ZDIII-specific IgA titers in bronchoalveolar lavage and vaginal fluids. Protective immunity was further assessed by subcutaneous and intravaginal ZIKV challenges. The second-generation FliCΔD3-2ZDIII was shown to result in a reduced titer of anti-FliC IgG antibodies in sera and still retained the same levels of serum IgG, IgA, and neutralizing antibodies and mucosal IgA antibodies without compromising the vaccine antigenicity. Therefore, intranasal immunization with FliCΔD3-2ZDIII fusion proteins formulated with LTIIb-B5 adjuvant elicited the greatest protective immunity against subcutaneous and intravaginal ZIKV challenges. Our findings indicated that the combination of FliCΔD3-2ZDIII fusion proteins and LTIIb-B5 adjuvant for intranasal immunization can be used for developing ZIKV mucosal vaccines.
Collapse
|
9
|
Wajanarogana S, Taylor WRJ, Kritsiriwuthinan K. Enhanced serodiagnosis of melioidosis by indirect ELISA using the chimeric protein rGroEL-FLAG300 as an antigen. BMC Infect Dis 2022; 22:387. [PMID: 35439967 PMCID: PMC9020111 DOI: 10.1186/s12879-022-07369-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 04/06/2022] [Indexed: 11/24/2022] Open
Abstract
Background The accurate and rapid diagnosis of melioidosis is challenging. Several serological approaches have been developed using recombinant antigens to improve the diagnostic indices of serological tests for melioidosis.
Methods Fusion proteins from Burkholderia pseudomallei (rGroEL-FLAG300) were evaluated as a potential target antigen for melioidosis antibodies. A total of 220 serum samples from 38 culture proven melioidosis patients (gold standard), 126 healthy individuals from endemic (n = 37) and non-endemic (n = 89) Thai provinces and 56 patients with other proven bacterial infections as negative controls were tested using indirect enzyme-linked immunosorbent assays (ELISA). Results Using an optical density (OD) cut-off of 0.299148, our assay had 94.74% sensitivity (95% confidence interval (CI) = 82.3–99.4%), 95.05% specificity (95% CI = 90.8–97.7%), and 95% accuracy, which was better than in our previous work (90.48% sensitivity, 87.14% specificity, and 87.63% accuracy). Conclusion Our results suggest that the application of chimeric antigens in ELISA could improve the serological diagnosis of melioidosis and should be reconfirmed with greater patient numbers.
Collapse
Affiliation(s)
- Sumet Wajanarogana
- Department of Basic Medical Science, Faculty of Medicine, Vajira Hospital, Navamindradhiraj University, Bangkok, 10300, Thailand.
| | - Water R J Taylor
- Mahidol Oxford Tropical Medicine Research Unit, Bangkok, 10400, Thailand.,Center for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | | |
Collapse
|
10
|
Tse Sum Bui B, Auroy T, Haupt K. Fighting Antibiotic‐Resistant Bacteria: Promising Strategies Orchestrated by Molecularly Imprinted Polymers. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202106493] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Bernadette Tse Sum Bui
- CNRS Laboratory for Enzyme and Cell Engineering Université de Technologie de Compiègne Rue du Docteur Schweitzer, CS 60319 60203 Compiègne Cedex France
| | - Tiffany Auroy
- CNRS Laboratory for Enzyme and Cell Engineering Université de Technologie de Compiègne Rue du Docteur Schweitzer, CS 60319 60203 Compiègne Cedex France
| | - Karsten Haupt
- CNRS Laboratory for Enzyme and Cell Engineering Université de Technologie de Compiègne Rue du Docteur Schweitzer, CS 60319 60203 Compiègne Cedex France
| |
Collapse
|
11
|
Côté-Cyr M, Gauthier L, Zottig X, Bourgault S, Archambault D. Recombinant Bacillus subtilis flagellin Hag is a potent immunostimulant with reduced proinflammatory properties compared to Salmonella enterica serovar Typhimurium FljB. Vaccine 2022; 40:11-17. [PMID: 34844822 DOI: 10.1016/j.vaccine.2021.11.049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 11/04/2021] [Accepted: 11/17/2021] [Indexed: 12/19/2022]
Abstract
Flagellin constitutes a potential adjuvant for vaccines owing to its robust immunostimulatory properties. However, clinical trials have revealed that flagellin derived from Salmonella enterica serovar Typhimurium induces high levels of proinflammatory markers and substantial adverse effects. The flagellin from Bacillus subtilis, Hag, shares high sequence homology with Salmonella FljB within the D0 and D1 domains responsible for TLR5 engagement, while the D2 and D3 domains associated with an off-target immune response are absent. Accordingly, we compared the immunostimulatory and proinflammatory properties of Hag with FljB by harnessing an epitope from the matrix 2 protein (M2e) of the influenza virus. Both flagellins engaged TLR5, with FljB showing a 2.5-fold higher potency than Hag. Mice inoculation showed a robust FljB- or Hag-induced M2e-specific antibody response, with Hag demonstrating a decreased secretion of proinflammatory markers and reduced weight loss. This study revealed that flagellin Hag is a potent immunoadjuvant with reduced proinflammatory properties.
Collapse
Affiliation(s)
- Mélanie Côté-Cyr
- Chemistry Department, Université du Québec à Montréal, Montréal, Canada; Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec, Canada; Department of Biological Sciences, Université du Québec à Montréal, Montréal, Canada; The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe, Canada
| | - Laurie Gauthier
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec, Canada; Department of Biological Sciences, Université du Québec à Montréal, Montréal, Canada; The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe, Canada
| | - Ximena Zottig
- Chemistry Department, Université du Québec à Montréal, Montréal, Canada; Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec, Canada; Department of Biological Sciences, Université du Québec à Montréal, Montréal, Canada; The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe, Canada
| | - Steve Bourgault
- Chemistry Department, Université du Québec à Montréal, Montréal, Canada; Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec, Canada; The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe, Canada.
| | - Denis Archambault
- Department of Biological Sciences, Université du Québec à Montréal, Montréal, Canada; The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe, Canada.
| |
Collapse
|
12
|
Tse Sum Bui B, Auroy T, Haupt K. Fighting Antibiotic-Resistant Bacteria : Promising Strategies Orchestrated by Molecularly Imprinted Polymers. Angew Chem Int Ed Engl 2021; 61:e202106493. [PMID: 34779567 DOI: 10.1002/anie.202106493] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Indexed: 11/09/2022]
Abstract
Infections caused by antibiotic-resistant bacteria are difficult and sometimes impossible to treat, making them one of the major public health problems of our time. We highlight how one unique material , molecularly imprinted polymers (MIPs), can orchestrate several strategies to fight this major societal issue. MIPs are tailor-made biomimetic supramolecular receptors that recognize and bind target molecules with a high affinity and selectivity, comparable to those of antibodies. While research on MIPs for combatting cancer has been constantly flourishing, comprehensive work on their involvement in combatting resistant superbugs has been rather scarce. This review aims at filling this gap. We will describe what are the causes of bacterial resistance and at which level MIPs can deploy their weapons. MIPs' targets can be biofilm constituents, quorum sensing messengers, bacterial surface proteins and antibiotic-deactivating enzymes, among others. We will conclude on the current challenges and future developments in this field.
Collapse
Affiliation(s)
- Bernadette Tse Sum Bui
- BUTC: Universite de Technologie de Compiegne Bibliotheques de l'Universite de Technologie de Compiegne, GEC, Rue du Docteur Schweitzer, 60203, Compiègne, FRANCE
| | - Tiffany Auroy
- Universite de Technologie de Compiegne, CNRS Laboratory for Enzyme and Cell Engineering, FRANCE
| | - Karsten Haupt
- Universite de Technologie de Compiegne, CNRS Laboratory for Enzyme and Cell Engineering, FRANCE
| |
Collapse
|
13
|
|
14
|
Akhtar N, Joshi A, Singh J, Kaushik V. Design of a novel and potent multivalent epitope based human cytomegalovirus peptide vaccine: An immunoinformatics approach. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.116586] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
15
|
Atapour A, Ghalamfarsa F, Naderi S, Hatam G. Designing of a Novel Fusion Protein Vaccine Candidate Against Human Visceral Leishmaniasis (VL) Using Immunoinformatics and Structural Approaches. Int J Pept Res Ther 2021; 27:1885-1898. [PMID: 33935610 PMCID: PMC8067785 DOI: 10.1007/s10989-021-10218-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2021] [Indexed: 11/25/2022]
Abstract
Leishmaniasis is caused by an obligate intracellular protozoan parasite. The clinical forms of leishmaniasis differ from cutaneous leishmaniasis, mucocutaneous leishmaniasis and visceral leishmaniasis (VL) which depend on the parasite species and the host's immune responses. There are significant challenges to the available anti-leishmanial drug therapy, particularly in severe forms of disease, and the rise of drug resistance has made it more difficult. Currently, no licensed vaccines have been introduced to the market for the control and elimination of VL. A potential target for use in candidate vaccines against leishmaniasis has been shown to be leishmania Kinetoplastid membrane protein-11 (KMP-11) antigen. In this study, we chose KMP-11 antigen as target antigen in our vaccine construct. In addition, B-type flagellin (fliC) was used as an adjuvant for enhancing vaccine immunogenicity. The GSGSGSGSGSG linker was applied to link the KMP-11 antigen and fliC (KMP-11-fliC) to construct our fusion protein. Bioinformatics approaches such as; 3D homology modeling, CTL, B-cell, MHC class I and II epitopes prediction, allergenicity, antigenicity evaluations, molecular docking, fast simulations of flexibility of docked complex and in silico cloning were employed to analysis and evaluation of various properties of the designed fusion construct. Computational results showed that our engineered structure has the potential for proper stimulation of cellular and humoral immune responses against VL. Consequently, it could be proposed as a candidate vaccine against VL according to these data and after verifying the efficacy of the candidate vaccine through in vivo and in vitro immunological tests.
Collapse
Affiliation(s)
- Amir Atapour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medical Biotechnology, Faculty of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, 71348-14336 Shiraz, Iran
| | - Farideh Ghalamfarsa
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samaneh Naderi
- Department of Diagnostic Laboratory Sciences and Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Gholamreza Hatam
- Basic Sciences in Infectious Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
16
|
In-silico design of a multivalent epitope-based vaccine against Candida auris. Microb Pathog 2021; 155:104879. [PMID: 33848597 DOI: 10.1016/j.micpath.2021.104879] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 12/04/2020] [Accepted: 03/08/2021] [Indexed: 12/15/2022]
Abstract
Candida auris is a rapidly emerging human pathogenic fungus with a high mortality rate. Recent report suggests that the new clinical isolates are showing resistance to the major classes of antifungal drugs. Due to the emergence of drug resistance, it becomes imperative to seek novel therapies for the treatment of C. auris. The potent vaccine could be one of the promising strategies for recalcitrant and multidrug-resistant pathogens. Using in silico approach we designed a novel multivalent vaccine against C. auris. We have selected the agglutinin-like sequence-3 (Als3) an adhesion protein, involved in virulence. The Als3p protein of C. auris was targeted to predict T cell and B cell epitopes. Epitopes which were found to be non-toxic, non-allergenic, highly conserved, and antigenic and could induce interferon-γ synthesis were selected for vaccine design. The selected epitopes were linked with suitable adjuvants to construct the final vaccine. The vaccine construct was predicted to be stable, soluble, antigenic, non-allergic with desirable physicochemical properties. We also constructed the 3D model of the vaccine and validated it with the Ramachandran plot. The ability of the vaccine construct to interact with Toll-like receptor (TLR) and major histocompatibility complex (MHC) was determined by molecular docking experiments. The binding energy of the vaccine construct with the TLR and MHC were found to be stable as predicted by molecular dynamics simulation. Further, in-silico cloning analysis showed that the vaccine construct can be successfully cloned and expressed in E. coli. Based on the results, we surmise that our candidate vaccine can be used as an alternative therapy for the treatment of C. auris. However, the efficacy and the safety of the vaccine model need to be determined by performing in vivo studies.
Collapse
|
17
|
McNeilly O, Mann R, Hamidian M, Gunawan C. Emerging Concern for Silver Nanoparticle Resistance in Acinetobacter baumannii and Other Bacteria. Front Microbiol 2021; 12:652863. [PMID: 33936010 PMCID: PMC8085274 DOI: 10.3389/fmicb.2021.652863] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
The misuse of antibiotics combined with a lack of newly developed ones is the main contributors to the current antibiotic resistance crisis. There is a dire need for new and alternative antibacterial options and nanotechnology could be a solution. Metal-based nanoparticles, particularly silver nanoparticles (NAg), have garnered widespread popularity due to their unique physicochemical properties and broad-spectrum antibacterial activity. Consequently, NAg has seen extensive incorporation in many types of products across the healthcare and consumer market. Despite clear evidence of the strong antibacterial efficacy of NAg, studies have raised concerns over the development of silver-resistant bacteria. Resistance to cationic silver (Ag+) has been recognised for many years, but it has recently been found that bacterial resistance to NAg is also possible. It is also understood that exposure of bacteria to toxic heavy metals like silver can induce the emergence of antibiotic resistance through the process of co-selection. Acinetobacter baumannii is a Gram-negative coccobacillus and opportunistic nosocomial bacterial pathogen. It was recently listed as the "number one" critical level priority pathogen because of the significant rise of antibiotic resistance in this species. NAg has proven bactericidal activity towards A. baumannii, even against strains that display multi-drug resistance. However, despite ample evidence of heavy metal (including silver; Ag+) resistance in this bacterium, combined with reports of heavy metal-driven co-selection of antibiotic resistance, little research has been dedicated to assessing the potential for NAg resistance development in A. baumannii. This is worrisome, as the increasingly indiscriminate use of NAg could promote the development of silver resistance in this species, like what has occurred with antibiotics.
Collapse
Affiliation(s)
- Oliver McNeilly
- iThree Institute, University of Technology Sydney, Ultimo, NSW, Australia
| | - Riti Mann
- iThree Institute, University of Technology Sydney, Ultimo, NSW, Australia
| | - Mohammad Hamidian
- iThree Institute, University of Technology Sydney, Ultimo, NSW, Australia
| | - Cindy Gunawan
- iThree Institute, University of Technology Sydney, Ultimo, NSW, Australia
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
18
|
In vitro Edwardsiella piscicida CK108 Transcriptome Profiles with Subinhibitory Concentrations of Phenol and Formalin Reveal New Insights into Bacterial Pathogenesis Mechanisms. Microorganisms 2020; 8:microorganisms8071068. [PMID: 32709101 PMCID: PMC7409036 DOI: 10.3390/microorganisms8071068] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 12/20/2022] Open
Abstract
Phenol and formalin are major water pollutants that are frequently discharged into the aquatic milieu. These chemicals can affect broad domains of life, including microorganisms. Aquatic pollutants, unlike terrestrial pollutants, are easily diluted in water environments and exist at a sub-inhibitory concentration (sub-IC), thus not directly inhibiting bacterial growth. However, they can modulate gene expression profiles. The sub-IC values of phenol and formalin were measured by minimal inhibitory concentration (MIC) assay to be 0.146% (1.3 mM) and 0.0039% (0.38 mM), respectively, in Edwardsiella piscicida CK108, a Gram-negative fish pathogen. We investigated the differentially expressed genes (DEG) by RNA-seq when the cells were exposed to the sub-ICs of phenol and formalin. DEG analyses revealed that genes involved in major virulence factors (type I fimbriae, flagella, type III and type VI secretion system) and various cellular pathways (energy production, amino acid synthesis, carbohydrate metabolism and two-component regulatory systems) were up- or downregulated by both chemicals. The genome-wide gene expression data corresponded to the results of a quantitative reverse complementary-PCR and motility assay. This study not only provides insight into how a representative fish pathogen, E. piscicida CK108, responds to the sub-ICs of phenol and formalin but also shows the importance of controlling chemical pollutants in aquatic environments.
Collapse
|
19
|
Gauthier L, Babych M, Segura M, Bourgault S, Archambault D. Identification of a novel TLR5 agonist derived from the P97 protein of Mycoplasma hyopneumoniae. Immunobiology 2020; 225:151962. [PMID: 32747018 DOI: 10.1016/j.imbio.2020.151962] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/22/2020] [Accepted: 05/15/2020] [Indexed: 02/08/2023]
Abstract
By modulating specific immune responses against antigens, adjuvants are used in many vaccine preparations to enhance protective immunity. The C-terminal domain of the protein P97 (P97c) of Mycoplasma hyopneumoniae, which is the etiologic agent of porcine enzootic pneumonia, has been shown to increase the specific humoral response against an antigen when this antigen is merged with P97c and delivered by adenovectors. However, the immunostimulating mechanism of this protein remains unknown. In the present study, recombinantly expressed P97c triggered a concentration-dependent TLR5 activation and stimulates the production of interleukin-8 from HEK-Blue mTLR5 cells. Circular dichroism spectroscopy and prediction of 3-dimensional conformation exposed a relevant secondary and tertiary structural homology between P97c and flagellin, the known potent TLR5 agonist. P97c adjuvanticity was evaluated by fusing the conserved epitope of the ectodomain matrix 2 protein (M2e) of the influenza A virus to the protein. Mice immunized with P97c-3M2e revealed a high antibody titer against the M2e epitope associated with a mixed Th1/Th2 immune response. Overall, this study identifies a novel agonist of the pattern recognition receptor TLR5 and reveals that P97c is a potential adjuvant through the activation of the innate immune system.
Collapse
Affiliation(s)
- Laurie Gauthier
- Department of Biological Sciences, Université du Québec à Montréal, Montréal, Canada; Department of Chemistry, Université du Québec à Montréal, Montréal, Canada; The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe, Canada; Quebec Network for Research on Protein Function, Engineering, and Applications, PROTEO, Québec, Canada; Centre d'Excellence en Recherche sur les Maladies Orphelines-Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, Canada
| | - Margaryta Babych
- Department of Chemistry, Université du Québec à Montréal, Montréal, Canada; Quebec Network for Research on Protein Function, Engineering, and Applications, PROTEO, Québec, Canada; Centre d'Excellence en Recherche sur les Maladies Orphelines-Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, Canada
| | - Mariela Segura
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe, Canada; Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Canada
| | - Steve Bourgault
- Department of Chemistry, Université du Québec à Montréal, Montréal, Canada; The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe, Canada; Quebec Network for Research on Protein Function, Engineering, and Applications, PROTEO, Québec, Canada; Centre d'Excellence en Recherche sur les Maladies Orphelines-Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, Canada.
| | - Denis Archambault
- Department of Biological Sciences, Université du Québec à Montréal, Montréal, Canada; The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe, Canada; Centre d'Excellence en Recherche sur les Maladies Orphelines-Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, Canada.
| |
Collapse
|
20
|
Zhao Y, Li Z, Zhu X, Cao Y, Chen X. Improving immunogenicity and safety of flagellin as vaccine carrier by high-density display on virus-like particle surface. Biomaterials 2020; 249:120030. [PMID: 32315864 DOI: 10.1016/j.biomaterials.2020.120030] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/30/2020] [Accepted: 04/03/2020] [Indexed: 12/17/2022]
Abstract
Flagellin is a protein-based adjuvant that activates toll-like receptor (TLR) 5. Flagellin has been actively explored as vaccine adjuvants and carriers. Preclinical and clinical studies find flagellin-based vaccines have a risk to induce systemic adverse reactions potentially due to its overt activation of TLR5. To improve safety and immunogenicity of flagellin as vaccine carriers, FljB was displayed at high densities on hepatitis b core (HBc) virus-like particle (VLP) surface upon c/e1 loop insertion. FljB-HBc (FH) VLPs showed significantly reduced ability to activate TLR5 or induce systemic interleukin-6 release as compared to FljB. FH VLPs also failed to significantly increase rectal temperature of mice, while FljB could significantly increase rectal temperature of mice. These data indicated systemic safety of FljB could be significantly improved by high-density display on HBc VLP surface. Besides improved safety, FH VLPs and FljB similarly boosted co-administered ovalbumin immunization and FH VLPs were found to induce two-fold higher anti-FljB antibody titer than FljB. These data indicated preserved adjuvant potency and improved immunogenicity after high-density display of FljB on HBc VLP surface. Consistent with the high immunogenicity, FH VLPs were found to be more efficiently taken up by bone marrow-derived dendritic cells and stimulate more potent dendritic cell maturation than FljB. Lastly, FH VLPs were found to be a more immunogenic carrier than FljB, HBc VLPs, or the widely used keyhole limpet hemocyanin for nicotine vaccine development with a good local and systemic safety. Our data support FH VLPs to be a potentially safer and more immunogenic carrier than FljB for vaccine development.
Collapse
Affiliation(s)
- Yiwen Zhao
- Biomedical & Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Zhuofan Li
- Biomedical & Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Xiaoyue Zhu
- Biomedical & Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Yan Cao
- Biomedical & Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Xinyuan Chen
- Biomedical & Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA.
| |
Collapse
|
21
|
Gao W, Cho E, Liu Y, Lu Y. Advances and Challenges in Cell-Free Incorporation of Unnatural Amino Acids Into Proteins. Front Pharmacol 2019; 10:611. [PMID: 31191324 PMCID: PMC6549004 DOI: 10.3389/fphar.2019.00611] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/15/2019] [Indexed: 12/17/2022] Open
Abstract
Incorporation of unnatural amino acids (UNAAs) into proteins currently is an active biological research area for various fundamental and applied science. In this context, cell-free synthetic biology (CFSB) has been developed and recognized as a robust testing and biomanufacturing platform for highly efficient UNAA incorporation. It enables the orchestration of unnatural biological machinery toward an exclusive user-defined objective of unnatural protein synthesis. This review aims to overview the principles of cell-free unnatural protein synthesis (CFUPS) systems, their advantages, different UNAA incorporation approaches, and recent achievements. These have catalyzed cutting-edge research and diverse emerging applications. Especially, present challenges and future trends are focused and discussed. With the development of CFSB and the fusion with other advanced next-generation technologies, CFUPS systems would explicitly deliver their values for biopharmaceutical applications.
Collapse
Affiliation(s)
- Wei Gao
- Department of Chemical Engineering, Tsinghua University, Beijing, China
- College of Life Science, Shenyang Normal University, Shenyang, China
| | - Eunhee Cho
- Department of Chemical Engineering, Tsinghua University, Beijing, China
| | - Yingying Liu
- Department of Chemical Engineering, Tsinghua University, Beijing, China
- College of Life Science, Shenyang Normal University, Shenyang, China
| | - Yuan Lu
- Department of Chemical Engineering, Tsinghua University, Beijing, China
- Institute of Biochemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing, China
- Key Lab of Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing, China
| |
Collapse
|
22
|
Polymorphism in Toll-Like Receptors and Helicobacter Pylori Motility in Autoimmune Atrophic Gastritis and Gastric Cancer. Cancers (Basel) 2019; 11:cancers11050648. [PMID: 31083432 PMCID: PMC6562993 DOI: 10.3390/cancers11050648] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/03/2019] [Accepted: 05/07/2019] [Indexed: 12/17/2022] Open
Abstract
Autoimmune atrophic gastritis (AAG) is associated with an increased risk of certain types of gastric cancer (GC). Helicobacter pylori (H. pylori) infection may have a role in the induction and/or maintenance of AAG and GC. Toll-like receptors (TLR) are essential for H. pylori recognition and subsequent innate and adaptive immunity responses. This study therefore aimed to characterize TLR polymorphisms, and features of bacterial flagellin A in samples from patients with AAG (n = 67), GC (n = 114) and healthy donors (HD; n = 97). TLR5 rs5744174 C/C genotype was associated with GC, lower IgG anti H. pylori response and a higher H. pylori flagellin A abundance and motility. In a subset of patients with AAG, H. pylori strains showed a reduction of the flagellin A abundance and a moderate motility compared with strains from GC patients, a prerequisite for active colonization of the deeper layers of the mucosa, host immune response and inflammation. TLR9 rs5743836 T allele showed an association with serum gastrin G17. In conclusion, our study suggests that alterations of flaA protein, moderate motility in H. pylori and two polymorphisms in TLR5 and TLR9 may favor the onset of AAG and GC, at least in a subset of patients. These findings corroborate the function of pathogen–host cell interactions and responses, likely influencing the pathogenetic process.
Collapse
|
23
|
Barnowski C, Kadzioch N, Damm D, Yan H, Temchura V. Advantages and Limitations of Integrated Flagellin Adjuvants for HIV-Based Nanoparticle B-Cell Vaccines. Pharmaceutics 2019; 11:E204. [PMID: 31052410 PMCID: PMC6572692 DOI: 10.3390/pharmaceutics11050204] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/24/2019] [Accepted: 04/26/2019] [Indexed: 11/29/2022] Open
Abstract
The great advantage of virus-like particle (VLP) nano-vaccines is their structural identity to wild-type viruses, ensuring that antigen-specific B-cells encounter viral proteins in their natural conformation. "Wild-type" viral nanoparticles can be further genetically or biochemically functionalized with biomolecules (antigens and adjuvants). Flagellin is a potent inducer of innate immunity and it has demonstrated adjuvant effectiveness due to its affinity for toll-like receptor 5 (TLR5). In contrast to most TLR ligands, flagellin is a protein and can induce an immune response against itself. To avoid side-effects, we incorporated a less inflammatory and less immunogenic form of flagellin as an adjuvant into HIV-based nanoparticle B-cell-targeting vaccines that display either the HIV-1 envelope protein (Env) or a model antigen, hen egg lysozyme (HEL). While flagellin significantly enhanced HEL-specific IgG responses, anti-Env antibody responses were suppressed. We demonstrated that flagellin did not activate B-cells directly in vitro, but might compete for CD4+ T-cell help in vivo. Therefore, we hypothesize that in the context of VLP-based B-cell nano-vaccines, flagellin serves as an antigen itself and may outcompete a less immunogenic antigen with its antibody response. In contrast, in combination with a strong immunogen, the adjuvant activity of flagellin may dominate over its immunogenicity.
Collapse
Affiliation(s)
- Cornelia Barnowski
- Department of Molecular and Medical Virology, Ruhr-University Bochum, 44801 Bochum, Germany.
- Institute of Virology, Medical Faculty, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany.
| | - Nicole Kadzioch
- Department of Molecular and Medical Virology, Ruhr-University Bochum, 44801 Bochum, Germany.
- Division of Experimental Clinical Research, Department of Clinical Research and Veterinary Public Health, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland.
| | - Dominik Damm
- Institute of Clinical and Molecular Virology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | - Huimin Yan
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Vladimir Temchura
- Institute of Clinical and Molecular Virology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany.
| |
Collapse
|
24
|
Attachment of flagellin enhances the immunostimulatory activity of a hemagglutinin-ferritin nano-cage. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 17:223-235. [DOI: 10.1016/j.nano.2019.01.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 10/31/2018] [Accepted: 01/11/2019] [Indexed: 12/15/2022]
|
25
|
Introducing a cost-effective method for purification of bioactive flagellin from several flagellated gram-negative bacteria. Protein Expr Purif 2019; 155:48-53. [DOI: 10.1016/j.pep.2018.11.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 11/18/2018] [Accepted: 11/18/2018] [Indexed: 12/18/2022]
|
26
|
Recombinant flagellins with deletions in domains D1, D2, and D3: Characterization as novel immunoadjuvants. Vaccine 2018; 37:652-663. [PMID: 30583910 DOI: 10.1016/j.vaccine.2018.12.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 11/29/2018] [Accepted: 12/02/2018] [Indexed: 12/17/2022]
Abstract
Bacterial flagellin activates the innate immune system and ultimately the adaptive immune system through a Toll-like receptor 5 (TLR5)-dependent signaling mechanism. Given that TLR5 is widely distributed in epithelia, flagellin is currently being developed as a mucosal adjuvant. Flagellin FliC from Salmonella enterica has four domains: the conserved D0 and D1 domains and the hypervariable D2 and D3 domains. The deletion of D3 and partial deletion of D2 in the recombinant FliCΔ174-400 strongly impairs flagellin's intrinsic antigenicity but does not affect the TLR5-dependent immunostimulation activity, i.e., the capacity to promote innate responses and adaptive responses to co-administered antigens. Here, we describe the development of novel recombinant flagellins with various deletions encompassing all of D2 and D3, and part of D1. Most of the recombinant molecules conserved an α-helical secondary structure that was as resistant to heat denaturation as the native protein. Whereas the recombinant flagellins' ability to trigger TLR5 varied markedly in vitro, most gave equivalent in vivo TLR5-dependent innate immune responses following intranasal administration of 2 μg of flagellin to mice. Concordantly, the recombinant flagellins were also valuable respiratory adjuvants for eliciting antibody responses to the foreign antigen ovalbumin, although their intrinsic antigenicity was decreased compared to the native flagellin and not increased compared to FliCΔ174-400. Our results show that the additional deletions of D2 and the distal part of D1 of FliCΔ174-400 does not impact on antigenicity and does not significantly modify the immunostimulatory adjuvant activity. Altogether, this study generated a novel set of recombinant flagellin that constitutes a portfolio of TLR5-dependent candidate adjuvants for vaccination.
Collapse
|
27
|
Fagerquist CK, Zaragoza WJ. Proteolytic Surface-Shaving and Serotype-Dependent Expression of SPI-1 Invasion Proteins in Salmonella enterica Subspecies enterica. Front Nutr 2018; 5:124. [PMID: 30619870 PMCID: PMC6295468 DOI: 10.3389/fnut.2018.00124] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/23/2018] [Indexed: 12/15/2022] Open
Abstract
We performed proteolytic surface-shaving with trypsin on three strains/sevovars of Salmonella enterica enterica (SEE): Newport, Kentucky, and Thompson. Surfaced-exposed proteins of live bacterial cells were digested for 15 min. A separate 20 h re-digestion was also performed on the supernatant of each shaving experiment to more completely digest protein fragments into detectable peptides for proteomic analysis by nano-liquid chromatography-electrospray ionization-Orbitrap mass spectrometry. Control samples (i.e., no trypsin during surface-shaving step) were also performed in parallel. We detected peptides of flagella proteins: FliC (filament), FliD (cap), and FlgL (hook-filament junction) as well as peptides of FlgM (anti-σ28 factor), i.e., the negative regulator of flagella synthesis. For SEE Newport and Thompson, we detected Salmonella pathogenicity island 1 (SPI-1) secreted effector/invasion proteins: SipA, SipB, SipC, and SipD, whereas no Sip proteins were detected in control samples. No Sip proteins were detected for SEE Kentucky (or its control) although sip genes were confirmed to be present. Our results may suggest a biological response (<15 min) to proteolysis of live cells for these SEE strains and, in the case of Newport and Thompson, a possible invasion response.
Collapse
Affiliation(s)
- Clifton K Fagerquist
- Produce Safety & Microbiology Research Unit, Western Regional Research Center, Agricultural Research Service, U.S. Department of Agriculture, Albany, CA, United States
| | - William J Zaragoza
- Produce Safety & Microbiology Research Unit, Western Regional Research Center, Agricultural Research Service, U.S. Department of Agriculture, Albany, CA, United States
| |
Collapse
|
28
|
Wampach L, Heintz-Buschart A, Fritz JV, Ramiro-Garcia J, Habier J, Herold M, Narayanasamy S, Kaysen A, Hogan AH, Bindl L, Bottu J, Halder R, Sjöqvist C, May P, Andersson AF, de Beaufort C, Wilmes P. Birth mode is associated with earliest strain-conferred gut microbiome functions and immunostimulatory potential. Nat Commun 2018; 9:5091. [PMID: 30504906 PMCID: PMC6269548 DOI: 10.1038/s41467-018-07631-x] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 11/13/2018] [Indexed: 01/07/2023] Open
Abstract
The rate of caesarean section delivery (CSD) is increasing worldwide. It remains unclear whether disruption of mother-to-neonate transmission of microbiota through CSD occurs and whether it affects human physiology. Here we perform metagenomic analysis of earliest gut microbial community structures and functions. We identify differences in encoded functions between microbiomes of vaginally delivered (VD) and CSD neonates. Several functional pathways are over-represented in VD neonates, including lipopolysaccharide (LPS) biosynthesis. We link these enriched functions to individual-specific strains, which are transmitted from mothers to neonates in case of VD. The stimulation of primary human immune cells with LPS isolated from early stool samples of VD neonates results in higher levels of tumour necrosis factor (TNF-α) and interleukin 18 (IL-18). Accordingly, the observed levels of TNF-α and IL-18 in neonatal blood plasma are higher after VD. Taken together, our results support that CSD disrupts mother-to-neonate transmission of specific microbial strains, linked functional repertoires and immune-stimulatory potential during a critical window for neonatal immune system priming.
Collapse
Affiliation(s)
- Linda Wampach
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, avenue des Hauts-Fourneaux 7, 4362, Esch-sur-Alzette, Luxembourg
- Laboratoire National de Santé, rue Louis Rech 1, 3555, Dudelange, Luxembourg
| | - Anna Heintz-Buschart
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, avenue des Hauts-Fourneaux 7, 4362, Esch-sur-Alzette, Luxembourg
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Deutscher Platz 5e, 04103, Leipzig, Germany
- Helmholtz Centre for Environmental Research GmbH - UFZ, Theodor-Lieser-Str. 4, 06120, Halle (Saale), Germany
| | - Joëlle V Fritz
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, avenue des Hauts-Fourneaux 7, 4362, Esch-sur-Alzette, Luxembourg
- Centre Hospitalier de Luxembourg, rue Nicolas Ernest Barblé 4, 1210, Luxembourg, Luxembourg
| | - Javier Ramiro-Garcia
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, avenue des Hauts-Fourneaux 7, 4362, Esch-sur-Alzette, Luxembourg
| | - Janine Habier
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, avenue des Hauts-Fourneaux 7, 4362, Esch-sur-Alzette, Luxembourg
| | - Malte Herold
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, avenue des Hauts-Fourneaux 7, 4362, Esch-sur-Alzette, Luxembourg
| | - Shaman Narayanasamy
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, avenue des Hauts-Fourneaux 7, 4362, Esch-sur-Alzette, Luxembourg
- Megeno S.A., avenue des Hauts-Fourneaux 9, 4362, Esch-sur-Alzette, Luxembourg
| | - Anne Kaysen
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, avenue des Hauts-Fourneaux 7, 4362, Esch-sur-Alzette, Luxembourg
- Centre Hospitalier de Luxembourg, rue Nicolas Ernest Barblé 4, 1210, Luxembourg, Luxembourg
| | - Angela H Hogan
- Integrated BioBank of Luxembourg, rue Louis Rech 1, 3555, Dudelange, Luxembourg
| | - Lutz Bindl
- Centre Hospitalier de Luxembourg, rue Nicolas Ernest Barblé 4, 1210, Luxembourg, Luxembourg
| | - Jean Bottu
- Centre Hospitalier de Luxembourg, rue Nicolas Ernest Barblé 4, 1210, Luxembourg, Luxembourg
| | - Rashi Halder
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, avenue des Hauts-Fourneaux 7, 4362, Esch-sur-Alzette, Luxembourg
| | - Conny Sjöqvist
- KTH Royal Institute of Technology, Science for Life Laboratory, School of Biotechnology, Division of Gene Technology, Tomtebodavägen 23a, 17165, Solna, Sweden
- Environmental and Marine Biology, Åbo Akademi University, Tykistökatu 6, 20520, Turku, Finland
| | - Patrick May
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, avenue des Hauts-Fourneaux 7, 4362, Esch-sur-Alzette, Luxembourg
| | - Anders F Andersson
- KTH Royal Institute of Technology, Science for Life Laboratory, School of Biotechnology, Division of Gene Technology, Tomtebodavägen 23a, 17165, Solna, Sweden
| | - Carine de Beaufort
- Centre Hospitalier de Luxembourg, rue Nicolas Ernest Barblé 4, 1210, Luxembourg, Luxembourg
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, avenue des Hauts-Fourneaux 7, 4362, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
29
|
Gornati L, Zanoni I, Granucci F. Dendritic Cells in the Cross Hair for the Generation of Tailored Vaccines. Front Immunol 2018; 9:1484. [PMID: 29997628 PMCID: PMC6030256 DOI: 10.3389/fimmu.2018.01484] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 06/14/2018] [Indexed: 12/14/2022] Open
Abstract
Vaccines represent the discovery of utmost importance for global health, due to both prophylactic action to prevent infections and therapeutic intervention in neoplastic diseases. Despite this, current vaccination strategies need to be refined to successfully generate robust protective antigen-specific memory immune responses. To address this issue, one possibility is to exploit the high efficiency of dendritic cells (DCs) as antigen-presenting cells for T cell priming. DCs functional plasticity allows shaping the outcome of immune responses to achieve the required type of immunity. Therefore, the choice of adjuvants to guide and sustain DCs maturation, the design of multifaceted vehicles, and the choice of surface molecules to specifically target DCs represent the key issues currently explored in both preclinical and clinical settings. Here, we review advances in DCs-based vaccination approaches, which exploit direct in vivo DCs targeting and activation options. We also discuss the recent findings for efficient antitumor DCs-based vaccinations and combination strategies to reduce the immune tolerance promoted by the tumor microenvironment.
Collapse
Affiliation(s)
- Laura Gornati
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Ivan Zanoni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy.,Division of Gastroenterology, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Francesca Granucci
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
30
|
Flores-Langarica A, Müller Luda K, Persson EK, Cook CN, Bobat S, Marshall JL, Dahlgren MW, Hägerbrand K, Toellner KM, Goodall MD, Withers DR, Henderson IR, Johansson Lindbom B, Cunningham AF, Agace WW. CD103 +CD11b + mucosal classical dendritic cells initiate long-term switched antibody responses to flagellin. Mucosal Immunol 2018; 11:681-692. [PMID: 29346347 PMCID: PMC5912514 DOI: 10.1038/mi.2017.105] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/23/2017] [Indexed: 02/04/2023]
Abstract
Antibody responses induced at mucosal and nonmucosal sites demonstrate a significant level of autonomy. Here, we demonstrate a key role for mucosal interferon regulatory factor-4 (IRF4)-dependent CD103+CD11b+ (DP), classical dendritic cells (cDCs) in the induction of T-dependent immunoglobulin G (IgG) and immunoglobulin A (IgA) responses in the mesenteric lymph node (MLN) following systemic immunization with soluble flagellin (sFliC). In contrast, IRF8-dependent CD103+CD11b- (SP) are not required for these responses. The lack of this response correlated with a complete absence of sFliC-specific plasma cells in the MLN, small intestinal lamina propria, and surprisingly also the bone marrow (BM). Many sFliC-specific plasma cells accumulating in the BM of immunized wild-type mice expressed α4β7+, suggesting a mucosal origin. Collectively, these results suggest that mucosal DP cDC contribute to the generation of the sFliC-specific plasma cell pool in the BM and thus serve as a bridge linking the mucosal and systemic immune system.
Collapse
Affiliation(s)
- A Flores-Langarica
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - K Müller Luda
- Immunology Section, Lund University, BMC D14 Sölvegatan 19, S-221 84. Lund 22184, Sweden
| | - E K Persson
- Immunology Section, Lund University, BMC D14 Sölvegatan 19, S-221 84. Lund 22184, Sweden
| | - C N Cook
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - S Bobat
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - J L Marshall
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - M W Dahlgren
- Immunology Section, Lund University, BMC D14 Sölvegatan 19, S-221 84. Lund 22184, Sweden
| | - K Hägerbrand
- Immunology Section, Lund University, BMC D14 Sölvegatan 19, S-221 84. Lund 22184, Sweden
| | - K M Toellner
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - M D Goodall
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - D R Withers
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - I R Henderson
- Institute of Microbiology and Infection, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - B Johansson Lindbom
- Immunology Section, Lund University, BMC D14 Sölvegatan 19, S-221 84. Lund 22184, Sweden
- Division of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark (DTU). Kongens Lyngby, Denmark
| | - A F Cunningham
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
- Institute of Microbiology and Infection, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - W W Agace
- Immunology Section, Lund University, BMC D14 Sölvegatan 19, S-221 84. Lund 22184, Sweden
- Division of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark (DTU). Kongens Lyngby, Denmark
| |
Collapse
|
31
|
Chassaing B, Gewirtz AT. Mice harboring pathobiont-free microbiota do not develop intestinal inflammation that normally results from an innate immune deficiency. PLoS One 2018; 13:e0195310. [PMID: 29617463 PMCID: PMC5884553 DOI: 10.1371/journal.pone.0195310] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 03/20/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Inability to maintain a stable and beneficial microbiota is associated with chronic gut inflammation, which classically manifests as colitis but may more commonly exist as low-grade inflammation that promotes metabolic syndrome. Alterations in microbiota, and associated inflammation, can originate from dysfunction in host proteins that manage the microbiota, such as the flagellin receptor TLR5. That the complete absence of a microbiota (i.e. germfree conditions) eliminates all evidence of inflammation in TLR5-deficient mice demonstrates that this model of gut inflammation is microbiota-dependent. We hypothesize that such microbiota dependency reflects an inability to manage pathobionts, such as Adherent-Invasive E. coli (AIEC). Herein, we examined the extent to which microbiota mismanagement and associated inflammation in TLR5-deficient mice would manifest in a limited and pathobiont-free microbiota. For this purpose, WT and TLR5-deficient mice were generated and maintained with the 8-member consortium of bacteria referred to as "Altered Schaedler Flora" (ASF). Such ASF animals were subsequently inoculated with AIEC reference strain LF82. Feces were assayed for bacterial loads, fecal lipopolysaccharide and flagellin loads, fecal inflammatory marker lipocalin-2 and microbiota composition. RESULTS Relative to similarly maintained WT mice, mice lacking TLR5 (T5KO) did not display low-grade intestinal inflammation nor metabolic syndrome under ASF conditions. Concomitantly, the ASF microbial community was similar between WT and T5KO mice, while inoculation with AIEC strain LF82 resulted in alteration of the ASF community in T5KO mice compared to WT control animals. AIEC LF82 inoculation in ASF T5KO mice resulted in microbiota components having elevated levels of bioactive lipopolysaccharide and flagellin, a modest level of low-grade inflammation and increased adiposity. CONCLUSIONS In a limited-complexity pathobiont-free microbiota, loss of the flagellin receptor TLR5 does not impact microbiota composition nor its ability to promote inflammation. Addition of AIEC to this ecosystem perturbs microbiota composition, increases levels of lipopolysaccharide and flagellin, but only modestly promotes gut inflammation and adiposity, suggesting that the phenotypes previously associated with loss of this innate immune receptor require disruption of complex microbiota.
Collapse
Affiliation(s)
- Benoit Chassaing
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States of America
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States of America
| | - Andrew T. Gewirtz
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States of America
| |
Collapse
|
32
|
Peng CJ, Chen HL, Chiu CH, Fang JM. Site-Selective Functionalization of Flagellin by Steric Self-Protection: A Strategy To Facilitate Flagellin as a Self-Adjuvanting Carrier in Conjugate Vaccine. Chembiochem 2018; 19:805-814. [PMID: 29377518 DOI: 10.1002/cbic.201700634] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Indexed: 01/18/2023]
Abstract
Flagellin (FliC) can act as a carrier protein in the preparation of conjugate vaccines to elicit a T-cell-dependent immune response and as an intrinsic adjuvant to activate the toll-like receptor 5 (TLR5) to enhance vaccine potency. To enable the use of FliC as a self-adjuvanting carrier, an effective method for site-selective modification (SSM) of pertinent amino-acid residues in the D2 and D3 domains of FliC is explored without excessive modification of the D0 and D1 domains, which are responsible for activating and binding with TLR5. In highly concentrated Na2 SO4 solution, FliC monomers form flagellar filaments, in which the D0 and D1 domains are situated inside the tubular structure. Thus, the lysine residues (K219, K224, K324, and K331) in the D2 and D3 domains of flagellin are selectively modified by a diazo-transfer reaction with imidazole-1-sulfonyl azide. The sites with azido modification are confirmed by MALDI-TOF-MS, ESI-TOF-MS, and LC-MS/MS analyses along with label-free quantitation. The azido-modified filament dissolves to give FliC monomers, which can conjugate with alkyne-hinged saccharides by the click reaction. Transmission electron microscopy imaging, dynamic light scattering measurements, and the secreted embryonic alkaline phosphatase reporter assay indicate that the modified FliC monomers retain the ability either to bind with TLR5 or to reassemble into filaments. Overall, this study establishes a feasible method for the SSM of FliC by steric self-protection of the D0 and D1 domains.
Collapse
Affiliation(s)
- Chi-Jiun Peng
- Department of Chemistry, National Taiwan University, 1, Sec. 4, Roosevelt Road, Taipei, 10617, Taiwan
| | - Hsiu-Ling Chen
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, 5, Fuxing Street, Guishan District, Taoyuan, 33302, Taiwan
| | - Cheng-Hsun Chiu
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, 5, Fuxing Street, Guishan District, Taoyuan, 33302, Taiwan
- Department of Pediatrics, Chang Gung Children's Hospital, 5, Fuxing Street, Guishan District, Taoyuan, 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, 259 Wenhua 1st Road, Guishan District, Taoyuan, 33302, Taiwan
| | - Jim-Min Fang
- Department of Chemistry, National Taiwan University, 1, Sec. 4, Roosevelt Road, Taipei, 10617, Taiwan
- The Genomics Research Center, Academia Sinica, 128, Sec. 2, Academia Road, Taipei, 11529, Taiwan
| |
Collapse
|
33
|
Negahdaripour M, Eslami M, Nezafat N, Hajighahramani N, Ghoshoon MB, Shoolian E, Dehshahri A, Erfani N, Morowvat MH, Ghasemi Y. A novel HPV prophylactic peptide vaccine, designed by immunoinformatics and structural vaccinology approaches. INFECTION GENETICS AND EVOLUTION 2017; 54:402-416. [DOI: 10.1016/j.meegid.2017.08.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 07/19/2017] [Accepted: 08/01/2017] [Indexed: 12/19/2022]
|
34
|
Wang C, Zhu W, Wang BZ. Dual-linker gold nanoparticles as adjuvanting carriers for multivalent display of recombinant influenza hemagglutinin trimers and flagellin improve the immunological responses in vivo and in vitro. Int J Nanomedicine 2017; 12:4747-4762. [PMID: 28740382 PMCID: PMC5503497 DOI: 10.2147/ijn.s137222] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Vaccination is the most cost-effective means of infectious disease control. Although current influenza vaccines are effective in battling closely matched strains, such vaccines have major limitations such as the requirement to produce new vaccines every season, an egg-dependent production system, long production periods, uncertainty in matching the vaccine to circulating strains, and the inability to react to new influenza pandemics resulting from genetic drift or shift. To overcome the intrinsic limitations of the conventional influenza vaccine, we have designed dual-linker gold nanoparticles (AuNPs) conjugated with both recombinant trimetric A/Aichi/2/68 (H3N2), hemagglutinin (HA) and TLR5 agonist flagellin (FliC) as a novel vaccine approach. Click chemistry and metal-chelating reactions were used to couple the two proteins. The conjugated proteins were found to possess high coupling specificity, high stability in harsh environments, high conjugation efficiency, and the ability to keep the appropriate protein conformations for immunogenicity and immunostimulation. Both AuNPs-HA/FliC and AuNPs-HA formulations induced higher levels of antibody responses than a mixture of soluble HA and FliC proteins when administered via a single intranasal immunization in mice. To further investigate the adjuvancy of these nanoparticles, in vitro experiments were conducted in both the JAWS II dendritic cell (DC) line and bone marrow-derived DC (BMDC) models. The results showed that dual-conjugated AuNPs were rapidly targeted and taken up by DCs. Consequently, DCs were induced toward maturation, as demonstrated by high levels of cytokine secretions and membrane costimulatory molecule expression. T cell proliferation was observed when splenic T cells were cocultured with AuNPs-HA/FliC-primed BMDCs. These results suggest that dual-conjugated AuNPs are effective at simultaneously displaying antigens and adjuvants in an oriented, multivalent format and can promote a strong immune response by activating DCs and T cells.
Collapse
Affiliation(s)
- Chao Wang
- Center for Inflammation, Immunity and Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA
| | - Wandi Zhu
- Center for Inflammation, Immunity and Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity and Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA
| |
Collapse
|
35
|
Kitzmüller C, Kalser J, Mutschlechner S, Hauser M, Zlabinger GJ, Ferreira F, Bohle B. Fusion proteins of flagellin and the major birch pollen allergen Bet v 1 show enhanced immunogenicity, reduced allergenicity, and intrinsic adjuvanticity. J Allergy Clin Immunol 2017; 141:293-299.e6. [PMID: 28456624 DOI: 10.1016/j.jaci.2017.02.044] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 02/03/2017] [Accepted: 02/22/2017] [Indexed: 11/28/2022]
Abstract
BACKGROUND Recombinant fusion proteins of flagellin and antigens have been demonstrated to induce strong innate and adaptive immune responses. Such fusion proteins can enhance the efficacy of allergen-specific immunotherapy. OBJECTIVE We sought to characterize different fusion proteins of flagellin and the major birch pollen allergen Bet v 1 for suitability as allergy vaccines. METHODS A truncated version of flagellin (NtCFlg) was genetically fused to the N- or C-terminus of Bet v 1. Toll-like receptor (TLR) 5 binding was assessed with HEK293 cells expressing TLR5. Upregulation of CD40, CD80, CD83, and CD86 on monocyte-derived dendritic cells from allergic patients was analyzed by using flow cytometry. The T cell-stimulatory capacity of the fusion proteins was assessed with naive and Bet v 1-specific T cells. IgE binding was tested in inhibition ELISAs and basophil activation tests. Mice were immunized with the fusion proteins in the absence and presence of aluminum hydroxide. Cellular and antibody responses were monitored. Murine antibodies were tested for blocking capacity in basophil activation tests. RESULTS Both fusion proteins matured monocyte-derived dendritic cells through TLR5. Compared with Bet v 1, the fusion proteins showed stronger T cell-stimulatory and reduced IgE-binding capacity and induced murine Bet v 1-specific antibodies in the absence of aluminum hydroxide. However, only antibodies induced by means of immunization with NtCFlg fused to the C-terminus of Bet v 1 inhibited binding of patients' IgE antibodies to Bet v 1. CONCLUSION Bet v 1-flagellin fusion proteins show enhanced immunogenicity, reduced allergenicity, and intrinsic adjuvanticity and thus represent promising vaccines for birch pollen allergen-specific immunotherapy. However, the sequential order of allergen and adjuvant within a fusion protein determines its immunologic characteristics.
Collapse
Affiliation(s)
- Claudia Kitzmüller
- Christian Doppler Laboratory for Immunomodulation, Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Julia Kalser
- Christian Doppler Laboratory for Immunomodulation, Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Sonja Mutschlechner
- Christian Doppler Laboratory for Immunomodulation, Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Michael Hauser
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | | | - Fatima Ferreira
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Barbara Bohle
- Christian Doppler Laboratory for Immunomodulation, Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
36
|
Biotechnology approaches to produce potent, self-adjuvanting antigen-adjuvant fusion protein subunit vaccines. Biotechnol Adv 2017; 35:375-389. [PMID: 28288861 DOI: 10.1016/j.biotechadv.2017.03.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 03/08/2017] [Accepted: 03/08/2017] [Indexed: 01/07/2023]
Abstract
Traditional vaccination approaches (e.g. live attenuated or killed microorganisms) are among the most effective means to prevent the spread of infectious diseases. These approaches, nevertheless, have failed to yield successful vaccines against many important pathogens. To overcome this problem, methods have been developed to identify microbial components, against which protective immune responses can be elicited. Subunit antigens identified by these approaches enable the production of defined vaccines, with improved safety profiles. However, they are generally poorly immunogenic, necessitating their administration with potent immunostimulatory adjuvants. Since few safe and effective adjuvants are currently used in vaccines approved for human use, with those available displaying poor potency, or an inability to stimulate the types of immune responses required for vaccines against specific diseases (e.g. cytotoxic lymphocytes (CTLs) to treat cancers), the development of new vaccines will be aided by the availability of characterized platforms of new adjuvants, improving our capacity to rationally select adjuvants for different applications. One such approach, involves the addition of microbial components (pathogen-associated molecular patterns; PAMPs), that can stimulate strong immune responses, into subunit vaccine formulations. The conjugation of PAMPs to subunit antigens provides a means to greatly increase vaccine potency, by targeting immunostimulation and antigen to the same antigen presenting cell. Thus, methods that enable the efficient, and inexpensive production of antigen-adjuvant fusions represent an exciting mean to improve immunity towards subunit antigens. Herein we review four protein-based adjuvants (flagellin, bacterial lipoproteins, the extra domain A of fibronectin (EDA), and heat shock proteins (Hsps)), which can be genetically fused to antigens to enable recombinant production of antigen-adjuvant fusion proteins, with a focus on their mechanisms of action, structural or sequence requirements for activity, sequence modifications to enhance their activity or simplify production, adverse effects, and examples of vaccines in preclinical or human clinical trials.
Collapse
|
37
|
Lu Y. Cell-free synthetic biology: Engineering in an open world. Synth Syst Biotechnol 2017; 2:23-27. [PMID: 29062958 PMCID: PMC5625795 DOI: 10.1016/j.synbio.2017.02.003] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 02/06/2017] [Indexed: 10/26/2022] Open
Abstract
Cell-free synthetic biology emerges as a powerful and flexible enabling technology that can engineer biological parts and systems for life science applications without using living cells. It provides simpler and faster engineering solutions with an unprecedented freedom of design in an open environment than cell system. This review focuses on recent developments of cell-free synthetic biology on biological engineering fields at molecular and cellular levels, including protein engineering, metabolic engineering, and artificial cell engineering. In cell-free protein engineering, the direct control of reaction conditions in cell-free system allows for easy synthesis of complex proteins, toxic proteins, membrane proteins, and novel proteins with unnatural amino acids. Cell-free systems offer the ability to design metabolic pathways towards the production of desired products. Buildup of artificial cells based on cell-free systems will improve our understanding of life and use them for environmental and biomedical applications.
Collapse
Affiliation(s)
- Yuan Lu
- Key Lab of Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China.,Institute of Biochemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| |
Collapse
|
38
|
Radomska KA, Vaezirad MM, Verstappen KM, Wösten MMSM, Wagenaar JA, van Putten JPM. Chicken Immune Response after In Ovo Immunization with Chimeric TLR5 Activating Flagellin of Campylobacter jejuni. PLoS One 2016; 11:e0164837. [PMID: 27760175 PMCID: PMC5070796 DOI: 10.1371/journal.pone.0164837] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 09/30/2016] [Indexed: 01/02/2023] Open
Abstract
Campylobacter jejuni is the main cause of bacterial food-borne diseases in developed countries. Chickens are the most important source of human infection. Vaccination of poultry is an attractive strategy to reduce the number of C. jejuni in the intestinal tract of chickens. We investigated the immunogenicity and protective efficacy of a recombinant C. jejuni flagellin-based subunit vaccine with intrinsic adjuvant activity. Toll-like receptor activation assays demonstrated the purity and TLR5 stimulating (adjuvant) activity of the vaccine. The antigen (20–40 μg) was administered in ovo to 18 day-old chicken embryos. Serum samples and intestinal content were assessed for antigen-specific systemic and mucosal humoral immune responses. In ovo vaccination resulted in the successful generation of IgY and IgM serum antibodies against the flagellin-based subunit vaccine as determined by ELISA and Western blotting. Vaccination did not induce significant amounts of flagellin-specific secretory IgA in the chicken intestine. Challenge of chickens with C. jejuni yielded similar intestinal colonization levels for vaccinated and control animals. Our results indicate that in ovo delivery of recombinant C. jejuni flagellin subunit vaccine is a feasible approach to yield a systemic humoral immune response in chickens but that a mucosal immune response may be needed to reduce C. jejuni colonization.
Collapse
Affiliation(s)
- Katarzyna A. Radomska
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - Mahdi M. Vaezirad
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - Koen M. Verstappen
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - Marc M. S. M. Wösten
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - Jaap A. Wagenaar
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
- Central Veterinary Institute of Wageningen UR, Lelystad, The Netherlands
| | - Jos P. M. van Putten
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
39
|
Liu WC, Liu YY, Chen TH, Liu CC, Jan JT, Wu SC. Multi-subtype influenza virus-like particles incorporated with flagellin and granulocyte-macrophage colony-stimulating factor for vaccine design. Antiviral Res 2016; 133:110-8. [PMID: 27491439 DOI: 10.1016/j.antiviral.2016.07.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/19/2016] [Accepted: 07/26/2016] [Indexed: 01/17/2023]
Abstract
Virus-like particle (VLP) technology is an attractive platform for seasonal and pandemic influenza vaccine development. We previously showed that influenza VLPs can be modified using M2 fusion with molecular adjuvants such as Salmonella typhimurium flagellin (FliC) to enhance VLP immunogenicity. For this study, three types of chimeric VLPs were incorporated with FliC, granulocyte-macrophage colony-stimulating factor (GM-CSF), or both GM-CSF and FliC (GM-CSF/FliC) to enhance anti-influenza immunogenicity. Our results indicate that immunizations with the chimeric FliC VLPs and GM-CSF/FliC H5N1 VLPs elicited more potent and broadly neutralizing antibodies and neuraminidase-inhibiting antibodies in sera, and induced higher numbers of hemagglutinin-specific antibody-secreting cells and germinal center B cell subsets in splenoctyes. Immunization with the chimeric GM-CSF H5N1 VLPs induced stronger Th1 and Th2 cellular responses. The chimeric GM-CSF/FliC H5N1 VLP constructs were further obtained to include H7 or H1H7 bi- or tri-subtype. It is our hope that these findings provide useful information for developing multi-subtype influenza vaccines.
Collapse
Affiliation(s)
- Wen-Chun Liu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ying-Yu Liu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ting-Hsuan Chen
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Chia-Chyi Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Jia-Tsrong Jan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Suh-Chin Wu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan; Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|