1
|
Panikulam S, Morgan H, Gutknecht M, Villiger TK, Lebesgue N, Karle AC. Host cell protein-mediated adjuvanticity and immunogenicity risks of biotherapeutics. Biotechnol Adv 2025; 81:108575. [PMID: 40180137 DOI: 10.1016/j.biotechadv.2025.108575] [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: 10/18/2024] [Revised: 02/18/2025] [Accepted: 03/30/2025] [Indexed: 04/05/2025]
Abstract
Host cell proteins (HCPs) are process-related impurities of biotherapeutic production that might affect product quality and/or patient safety. In a few cases, adverse events were attributed to HCPs present in the administered biotherapeutic. HCP-associated immune risks include adjuvanticity and immunogenicity with potential cross-reactivity. Based on the published data, some HCPs can act as adjuvants increasing the immunogenicity of the biotherapeutic as a bystander effect. HCPs may also induce immunogenicity against themselves, resulting in anti-HCP T cell responses and anti-HCP antibody formation. Depending on sequence similarities, these anti-HCP immune responses might theoretically be cross-reactive to the biotherapeutic or human endogenous proteins. In this review, we examine HCP-associated immune-related risks reported from non-clinical and clinical studies. We also discuss the potential and limitations of in vitro and in silico methods to evaluate the adjuvanticity and immunogenicity potential of HCPs. A risk-based assessment of the safety impact of HCPs may include the identity of the HCP and similarity to the biotherapeutic and human proteins, as well as product, treatment-, and patient-related factors.
Collapse
Affiliation(s)
- Sherin Panikulam
- Institute of Pharma Technology, University of Applied Sciences Northwestern Switzerland, Muttenz, Switzerland; Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Hannah Morgan
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | - Thomas K Villiger
- Institute of Pharma Technology, University of Applied Sciences Northwestern Switzerland, Muttenz, Switzerland
| | - Nicolas Lebesgue
- Technical Research and Development, Novartis Pharma AG, Basel, Switzerland
| | - Anette C Karle
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland.
| |
Collapse
|
2
|
Pang S, Wang L, Liu M, Shao M, Zhu G, Duan Q. Truncated flagellin lacking the hypervariable region: A structural basis for improved immune responses and adjuvanticity. Int J Biol Macromol 2025; 308:142742. [PMID: 40180103 DOI: 10.1016/j.ijbiomac.2025.142742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 03/02/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Bacterial flagellins are recognized for their potent immunomodulatory properties and potential as vaccine adjuvants. They activate innate and adaptive immune responses by interacting with Toll-like receptor 5 (TLR5) and the cytosolic NOD-like receptor protein 4 (NLRC4) inflammasome, thereby enhancing immune responses. This study investigates the impact of various truncated flagellin derivatives, derived from Escherichia coli (EHEC EDL933) and lacking specific domains, on TLR5 activation and their adjuvant properties. We generated several truncated flagellin mutants and assessed their ability to activate TLR5 in vitro and their immunoadjuvant effects in vivo. Our data show that only the FliCH7, FliCNC, FliCH7-FaeG, and FliCNC-FaeG proteins, which lack the hypervariable region (HVP) but retain both the amino- and carboxy-terminal regions, significantly enhanced TNF-α and IL-8 production compared to other flagellin derivatives. These findings underscore the essential roles of both conserved terminal regions in TLR5 activation. Notably, the FliCNC truncated mutant exhibited TLR5 activation comparable to that of native flagellin and induced higher antibody titers when co-administered with a model antigen or used as a fusion protein. Our results suggest that the HVR is not essential for flagellin's immunoadjuvant activity and that its removal enhances flagellin's ability to activate the innate immune system. This study provides valuable insights into optimizing flagellin derivatives for vaccine development, offering a more potent platform for enhancing immune responses against a range of pathogens.
Collapse
Affiliation(s)
- Shengmei Pang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint Laboratory for International Cooperation in Agriculture and Agricultural Product Safety, Ministry of Education, Yangzhou 225009, China; Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu, China
| | - Longlong Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint Laboratory for International Cooperation in Agriculture and Agricultural Product Safety, Ministry of Education, Yangzhou 225009, China; Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu, China
| | - Mei Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint Laboratory for International Cooperation in Agriculture and Agricultural Product Safety, Ministry of Education, Yangzhou 225009, China; Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu, China
| | - Mingqing Shao
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint Laboratory for International Cooperation in Agriculture and Agricultural Product Safety, Ministry of Education, Yangzhou 225009, China; Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu, China
| | - Guoqiang Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint Laboratory for International Cooperation in Agriculture and Agricultural Product Safety, Ministry of Education, Yangzhou 225009, China; Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu, China
| | - Qiangde Duan
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint Laboratory for International Cooperation in Agriculture and Agricultural Product Safety, Ministry of Education, Yangzhou 225009, China; Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
3
|
Nguyen TD, Le HD, Dang GC, Jung HS, Choi Y, Khim K, Kim Y, Lee SE, Rhee JH. A combined adjuvant and ferritin nanocage based mucosal vaccine against Streptococcus pneumoniae induces protective immune responses in a murine model. Nat Commun 2025; 16:2871. [PMID: 40128220 PMCID: PMC11933286 DOI: 10.1038/s41467-025-58115-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 03/13/2025] [Indexed: 03/26/2025] Open
Abstract
Protein nanocages are multimeric structures that can be engineered to mimic the molecular conformation of microorganisms. Based on previous findings showing that a mucosal FlaB-tPspA fusion (flagellin fused with truncated PspA antigen of Streptococcus pneumoniae) vaccine-induced protective immune response against S. pneumoniae, we develop a ferritin nanocage vaccine displaying multivalent presentation of both antigen and adjuvant on a nanocarrier using the SpyTag/SpyCatcher strategy. The 1:1 antigen/adjuvant nanocage is further used as a mucosal vaccine, which can translocate to draining lymph nodes with higher efficiency than fusion vaccine. Moreover, intranasal immunization with the nanocage vaccine significantly enhances mucosal immune responses with more efficient B-cell memory generation and antibody maturation, as well as more balanced (Th1/Th2) immune responses with increased IFN-γ and IL-17 production, comparing with fusion vaccine. Mice immunized with the nanocage vaccine exhibited enhanced protection against lethal infection compare to the FlaB-tPspA fusion group. Our study thus demonstrates the effectiveness of an all-in-one nanocage mucosal vaccine platform, which guarantees enhanced protection with balanced immune responses.
Collapse
Affiliation(s)
- 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
| | - Hoang Duy Le
- 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
| | - Giang Chau Dang
- 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
| | - Hyun Seok Jung
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
| | - Yoonjoo Choi
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
| | - 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
| | - Young Kim
- Department of Oral Pathology, Chonnam National University School of Dentistry, Gwangju, Republic of Korea
| | - Shee Eun Lee
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea.
- National Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, Republic of Korea.
- Department of Pharmacology and Dental Therapeutics, Chonnam National University School of Dentistry, 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
|
Huang S, Li X, Cao Y, Mou M, Li J, Zhuo K, Wang L, Zeng Z, Wei X, Tang C, Zhong M. TLR5 activation in respiratory epithelial cells orchestrate mucosal Th17 response through both indirect and direct pathways. Respir Res 2025; 26:104. [PMID: 40098159 PMCID: PMC11916947 DOI: 10.1186/s12931-025-03186-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/10/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Flagellin, a potent mucosal adjuvant administered via the intranasal route, has been widely recognized for its capacity to enhance immune responses against diverse pathogens. However, the effects and the underlying mechanisms by which flagellin modulates CD4+ T cell differentiation remain incompletely understood. METHODS Recombinant flagellin proteins, including full-length flagellin (SF) and a TLR5-binding deficient variant (SFΔ90-97), were produced and purified. An OT-II derived CD4+ T cell adoptive transfer model, a classical intranasal immunization model and dendritic cell (DC)-CD4+ T co-culturing system were used. The proliferation and differentiation of CD4+ T cells were analyzed using flow cytometry analysis. RNA sequencing and neutralizing antibody blocking experiments were performed to determine the essential cytokines involved in flagellin modulated Th17 differentiation. RESULTS Flagellin preferentially promotes Th17 cells differentiation. Respiratory epithelial cells (RECs), acting as sentinel cells, are the first to encounter exogenous stimuli during intranasal immunization. Flagellin stimulates the secretion of various soluble cytokines by binding to TLR5 on the surface of RECs, with GM-CSF facilitating the functional activation of airway DCs. GM-CSF-conditioned DCs exhibit upregulated IL-6 expression which in turn drives the polarization of naïve CD4+ T cells toward the Th17 phenotype. Furthermore, TLR5-regulated REC-derived IL-6 synergizes with TLR5-modulated DCs to amplify Th17 polarization signals, thereby enhancing the Th17 induction. CONCLUSION Flagellin preferentially induced a Th17-enhanced immune response and RECs were highlighted its essential roles during this process through both indirect and direct pathways. For indirect pathway, RECs modulate DC function through GM-CSF. Moreover, RECs directly contribute to Th17 differentiation by secreting IL-6.
Collapse
Affiliation(s)
- Sijian Huang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China
| | - Xu Li
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China
- Clinical Laboratory, Wuhan Asia General Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, 430056, China
| | - Yuan Cao
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China
- Analytical & Testing Center, Wuhan University of Science and Technology, Wuhan, Hubei, 430065, China
| | - Man Mou
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China
- Department of Blood Transfusion, Wuhan Asia General Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, 430056, China
| | - Jianlun Li
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China
| | - Kexing Zhuo
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China
| | - Lijuan Wang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China
| | - Zihang Zeng
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China
| | - Xianghong Wei
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China
| | - Chunlian Tang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China.
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, 430063, China.
| | - Maohua Zhong
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, # 2 Huangjiahu West Road, Wuhan, Hubei, 430065, China.
| |
Collapse
|
5
|
Yang K, Liu YJ, Zhang JN, Chen YJ, Yang J, Xiao JP, Lin HB, Yang HJ. Advances in the structural characterization and pharmacological activity of Salvia miltiorrhiza polysaccharides. Front Chem 2025; 13:1492533. [PMID: 40161004 PMCID: PMC11949878 DOI: 10.3389/fchem.2025.1492533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Background Salvia miltiorrhiza Bunge is the dried root and rhizome of Salvia miltiorrhiza Bunge, a labiatae plant. Salvia miltiorrhiza polysaccharide (SMP) is the main active component of Salvia miltiorrhiza Bunge. The extraction methods of SMP mainly include water extraction, ultrasonic extraction, enzyme extraction, microwave-assisted extraction and acid-base extraction. It is mainly composed of glucose, arabinose, rhamnose, galactose and other monosaccharides. SMP has a variety of biological activities, including immune regulation, anti-tum, anti-oxidation, myocardial protection, liver protection and so on. Purpose Salvia miltiorrhiza polysaccharide is widely used in nutraceuticals and pharmaceuticals, and has high research value. Natural polysaccharides are non-toxic, soluble in water, and have a wide range of biological activities, so they have broad research prospects. Methods The data was collected using different online resources including PubMed, Google Scholar, and Web of Science using keywords given below. Results In the past decades, various reports have shown that the pharmacological activities of Salvia miltiorrhiza polysaccharides have good effects, and the side effects are small. Conclusion This paper summarizes the extraction and purification methods, molecular weight, monosaccharide composition, glycosidic linkage, pharmacological activity, toxicity, product development, clinical research and other contents of Salvia miltiorrhiza polysaccharides in recent years, providing a theoretical basis for further study of Salvia miltiorrhiza polysaccharides.
Collapse
Affiliation(s)
- Ke Yang
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Science, China Jiliang University, Hangzhou, China
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yi-Jun Liu
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Science, China Jiliang University, Hangzhou, China
| | - Jia-Ning Zhang
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Science, China Jiliang University, Hangzhou, China
| | - Ya-Jing Chen
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Science, China Jiliang University, Hangzhou, China
| | - Jian Yang
- State Key Laboratory Breeding Base of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
- Dexing Research and Training Center of Chinese Medical Sciences, Dexing, China
| | - Jun-Ping Xiao
- Jiangxi Prozin Pharmaceutical Co., Ltd., Ji’an City, Jiangxi, China
| | - Han-Bin Lin
- Zhongshan Institute for Drug Discovery, Zhongke Zhongshan Pharmaceutical Innovation Research Institute (SIMM CAS), Zhongshan, Guangdong, China
| | - Hong-Jun Yang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
6
|
Jeon D, Hill E, McNeel DG. Toll-like receptor agonists as cancer vaccine adjuvants. Hum Vaccin Immunother 2024; 20:2297453. [PMID: 38155525 PMCID: PMC10760790 DOI: 10.1080/21645515.2023.2297453] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/16/2023] [Indexed: 12/30/2023] Open
Abstract
Cancer immunotherapy has emerged as a promising strategy to treat cancer patients. Among the wide range of immunological approaches, cancer vaccines have been investigated to activate and expand tumor-reactive T cells. However, most cancer vaccines have not shown significant clinical benefit as monotherapies. This is likely due to the antigen targets of vaccines, "self" proteins to which there is tolerance, as well as to the immunosuppressive tumor microenvironment. To help circumvent immune tolerance and generate effective immune responses, adjuvants for cancer vaccines are necessary. One representative adjuvant family is Toll-Like receptor (TLR) agonists, synthetic molecules that stimulate TLRs. TLRs are the largest family of pattern recognition receptors (PRRs) that serve as the sensors of pathogens or cellular damage. They recognize conserved foreign molecules from pathogens or internal molecules from cellular damage and propel innate immune responses. When used with vaccines, activation of TLRs signals an innate damage response that can facilitate the development of a strong adaptive immune response against the target antigen. The ability of TLR agonists to modulate innate immune responses has positioned them to serve as adjuvants for vaccines targeting infectious diseases and cancers. This review provides a summary of various TLRs, including their expression patterns, their functions in the immune system, as well as their ligands and synthetic molecules developed as TLR agonists. In addition, it presents a comprehensive overview of recent strategies employing different TLR agonists as adjuvants in cancer vaccine development, both in pre-clinical models and ongoing clinical trials.
Collapse
Affiliation(s)
- Donghwan Jeon
- Department of Oncology, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Ethan Hill
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Douglas G. McNeel
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| |
Collapse
|
7
|
He G, Long H, He J, Zhu C. The Immunomodulatory Effects and Applications of Probiotic Lactiplantibacillus plantarum in Vaccine Development. Probiotics Antimicrob Proteins 2024; 16:2229-2250. [PMID: 39101975 DOI: 10.1007/s12602-024-10338-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2024] [Indexed: 08/06/2024]
Abstract
Lactiplantibacillus plantarum (previously known as Lactobacillus plantarum) is a lactic acid bacterium that exists in various niches. L. plantarum is a food-grade microorganism that is commonly considered a safe and beneficial microorganism. It is widely used in food fermentation, agricultural enhancement, and environmental protection. L. plantarum is also part of the normal flora that can regulate the intestinal microflora and promote intestinal health. Some strains of L. plantarum are powerful probiotics that induce and modulate the innate and adaptive immune responses. Due to its outstanding immunoregulatory capacities, an increasing number of studies have examined the use of probiotic L. plantarum strains as natural immune adjuvants or alternative live vaccine carriers. The present review summarizes the main immunomodulatory characteristics of L. plantarum and discusses the preliminary immunological effects of L. plantarum as a vaccine adjuvant and delivery carrier. Different methods for improving the immune capacities of recombinant vector vaccines are also discussed.
Collapse
Affiliation(s)
- Guiting He
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, 421001, Hunan, China
| | - Huanbing Long
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, 421001, Hunan, China
| | - Jiarong He
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, 421001, Hunan, China
| | - Cuiming Zhu
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, 421001, Hunan, China.
| |
Collapse
|
8
|
Behrouz B, Rasooli I, Badmasti F. Inserting Omp22 into the flagellin protein, replacing its hypervariable region, results in stronger protection against lethal Acinetobacter baumannii infection. Sci Rep 2024; 14:27646. [PMID: 39533090 PMCID: PMC11557591 DOI: 10.1038/s41598-024-79013-x] [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: 07/01/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Acinetobacter baumannii, a common nosocomial pathogen, is known for its rapid acquisition of antimicrobial resistance, underscoring the urgent need to develop an effective vaccine against this pathogen. Outer membrane protein 22 (Omp22) regulates the biogenesis of outer membrane vesicles to transport virulence-promoting factors into the host cells and facilitates the progression of A. baumannii infection. In this study, we used a mouse model to assess a vaccine's immunogenicity and protective efficacy using recombinant Omp22 protein within the hypervariable region of flagellin (FliC-Omp22). FliC-Omp22 demonstrated superior protection following challenge with a lethal dose of multidrug-resistant (MDR) A. baumannii strain 58ST compared to Omp22 alone. In addition, it elicited increased IgG1/IgG2a and IL-4/IFN-γ ratios, indicating a predominant Th2 immune response. Furthermore, the FliC-Omp22 vaccination elicited strong specific antibodies that inhibited the adhesion and invasion of A. baumannii 58ST and enhanced the opsonic killing activity against the pathogen. FliC-Omp22 immunization significantly reduced bacterial loads in infected mice's spleen, lungs, and liver, thereby improving their survival against the lethal infection caused by MDR A. baumannii 58ST. This study suggests that integrating Omp22 into the hypervariable domain of flagellin holds promise for developing an effective vaccine against A. baumannii infections.
Collapse
Affiliation(s)
- Bahador Behrouz
- Department of Biology, Faculty of Basic Science, Shahed University, Tehran, Iran
| | - Iraj Rasooli
- Department of Biology, Faculty of Basic Science, Shahed University, Tehran, Iran.
- Molecular Microbiology Research Center, Department of Biology, Shahed University, Tehran-Qom Expressway, Tehran, 3319118651, Iran.
| | - Farzad Badmasti
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
9
|
Arshad F, Sarfraz A, Rubab A, Shehroz M, Moura AA, Sheheryar S, Ullah R, Shahat AA, Ibrahim MA, Nishan U, Shah M. Rational design of novel peptide-based vaccine against the emerging OZ virus. Hum Immunol 2024; 85:111162. [PMID: 39447523 DOI: 10.1016/j.humimm.2024.111162] [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: 08/27/2024] [Revised: 10/04/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024]
Abstract
Oz virus (OZV) belongs to the Orthomyxoviridae family which includes viruses with a negative-sense, single-stranded, and segmented RNA genome. OZV is a zoonotic pathogen, particularly since the virus can cause deadly illness when injected intracerebrally into nursing mice. OZV is an emerging pathogen with the potential to spark a pandemic as there is no preventive and licensed treatment against this virus. The goal of this study was to develop a novel multi-epitope vaccination against OZV proteins utilizing immunoinformatics and immunological simulation analysis. This work evaluated immunological epitopes (B cells, MHC-I, and MHC-II) to identify highly antigenic OZV target proteins. Shortlisted epitopes were joined together by using appropriate linkers and adjuvants to design multi-epitope vaccine constructs (MEVC). The vaccine models were designed, improved, validated, and the globular regions and post-translational modifications (PTMs) were also evaluated in the vaccine's structure. Molecular docking analysis with the Toll-like receptor (TLR4) showed strong interactions and appropriate binding energies. Molecular dynamics (MD) simulation confirmed stable interactions between the vaccines and TLR4. Bioinformatics tools helped optimize codons, resulting in successful cloning into appropriate host vectors. This study showed that the developed vaccines are stable and non-allergenic in the human body and successfully stimulated immunological responses against OZV. Finally, a mechanism of action for the designed vaccine construct was also proposed. Further experimental validations of the designed vaccine construct will pave the way to create a potentially effective vaccine against this emerging pathogen.
Collapse
Affiliation(s)
- Fizza Arshad
- Department of Biochemistry, Bahauddin Zakariya University, Multan 66000, Pakistan
| | - Asifa Sarfraz
- Department of Biochemistry, Bahauddin Zakariya University, Multan 66000, Pakistan
| | - Aleeza Rubab
- Department of Biochemistry, Bahauddin Zakariya University, Multan 66000, Pakistan
| | - Muhammad Shehroz
- Department of Bioinformatics, Kohsar University Murree, Murree 47150, Pakistan
| | - Arlindo A Moura
- Department of Animal Science, Federal University of Ceara, Fortaleza, Brazil
| | - Sheheryar Sheheryar
- Department of Animal Science, Federal University of Ceara, Fortaleza, Brazil
| | - Riaz Ullah
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdelaaty A Shahat
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed A Ibrahim
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Umar Nishan
- Department of Chemistry, Kohat University of Science & Technology, Kohat, Pakistan.
| | - Mohibullah Shah
- Department of Biochemistry, Bahauddin Zakariya University, Multan 66000, Pakistan; Department of Animal Science, Federal University of Ceara, Fortaleza, Brazil.
| |
Collapse
|
10
|
Pang S, Liu M, Wang L, Shao M, Zhu G, Duan Q. Differential Adjuvant Activity by Flagellins from Escherichia coli, Salmonella enterica Serotype Typhimurium, and Pseudomonas aeruginosa. Vaccines (Basel) 2024; 12:1212. [PMID: 39591115 PMCID: PMC11598095 DOI: 10.3390/vaccines12111212] [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: 09/19/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
(1) Background: The adjuvant properties of flagellin from various bacterial species have been extensively studied; however, a systematic comparison of the immunoadjuvant effects of flagellins from different bacterial species is lacking. This study aims to analyze the amino acid sequences and structural features of flagellins from Escherichia coli (FliCE.C), Salmonella enterica serotype Typhimurium (FliCS.T), and Pseudomonas aeruginosa (FliCP.A), and to evaluate their adjuvant activities in terms of Toll-like receptor 5 (TLR5) activation, antibody production, and cytokine responses in a murine model. (2) Methods: Bioinformatics analysis was conducted to compare the amino acid sequences and structural domains (D0, D1, D2, and D3) of flagellins from the three bacterial species. PyMol atomic models were used to confirm structural differences. Toll-like receptor 5 (TLR5) activation assays were performed to measure IL-8 and TNF-α production in vitro. The IgG antibody titers against the model antigen FaeG and cytokine responses, including IL-4 and TNF-α secretion were evaluated in a murine model. (3) Results: Bioinformatics analysis revealed that the D0 and D1 domains are highly conserved, whereas the D2 and D3 domains exhibit significant variability across the three species. Structural analysis via PyMol confirmed these differences, particularly in the D2 and D3 domains. TLR5 activation assays showed that FliCS.T and FliCP.A induced higher levels of IL-8 and TNF-α production compared to FliCE.C, indicating species-specific variations in TLR5 activation. In the murine model, FliCS.T as an adjuvant produced higher antibody titers against FaeG and increased IL-4 secretion in splenocytes compared to FliCE.C and FliCP.A. FliCP.A induced higher TNF-α expression than FliCS.T and FliCE.C, suggesting FliCS.T and FliCP.A are more effective at inducing T-cell responses. (4) Conclusions: This study highlights the potential of FliCS.T and FliCP.A as potent vaccine adjuvants. The results provide insights into the structure-function relationships of these flagellins and support their application in enhancing immune responses against diverse pathogens.
Collapse
Affiliation(s)
- Shengmei Pang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (S.P.); (M.L.); (L.W.); (M.S.); (G.Z.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Mei Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (S.P.); (M.L.); (L.W.); (M.S.); (G.Z.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Longlong Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (S.P.); (M.L.); (L.W.); (M.S.); (G.Z.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Mingqing Shao
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (S.P.); (M.L.); (L.W.); (M.S.); (G.Z.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Guoqiang Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (S.P.); (M.L.); (L.W.); (M.S.); (G.Z.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Qiangde Duan
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (S.P.); (M.L.); (L.W.); (M.S.); (G.Z.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
11
|
Liu Q, Huo X, Wang P, Zhao F, Yuan G, Yang C, Su J. Lactobacillus casei displaying MCP2α and FlaC delivered by PLA microspheres effectively enhances the immune protection of largemouth bass (Micropterus salmoides) against LMBV infection. FISH & SHELLFISH IMMUNOLOGY 2024; 153:109870. [PMID: 39218416 DOI: 10.1016/j.fsi.2024.109870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Largemouth bass ranavirus (LMBV) seriously affects the development of largemouth bass (Micropterus salmoides) industry and causes huge economic losses. Oral vaccine can be a promising method for viral disease precaution. In this study, MCP2α was identified as a valuable epitope region superior to MCP and MCP2 of LMBV by neutralizing antibody experiments. Then, recombinant Lactobacillus casei expressing the fusion protein MCP2αC (MCP2α as antigen, C represents flagellin C from Aeromonas hydrophila as adjuvant) on surface was constructed and verified. Further, PLA microsphere vaccine loading recombinant MCP2αC L. casei was prepared. The PLA microspheres vaccine were observed by scanning electron microscopy and showed a smooth, regular spherical surface with a particle size distribution between 100 and 200 μm. Furthermore, we evaluated the tolerance of PLA-MCP2αC vaccine in simulated gastric fluid and simulated intestinal fluid, and the results showed that PLA-MCP2αC can effectively resist the gastrointestinal environment. Moreover, the protective effect of PLA-MCP2αC against LMBV was evaluated after oral immunization and LMBV challenge. The results showed that PLA-MCP2αC effectively up-regulated the activity of serum biochemical enzymes (T-SOD, T-AOC, LZM, complement C3) and induced the mRNA expression of representative immune genes (IL-1β, TNF-α, IFN-γ, MHC-IIα, Mx, IgM) in spleen and head kidney tissues. The survival rate of largemouth bass vaccinated with PLA-MCP2αC increased from 24 % to 68 %. Meanwhile, PLA-MCP2αC inhibited the LMBV burden in spleen, head kidney and liver tissues and attenuated tissue damage in spleen. These results suggested that PLA-MCP2αC can be used as a candidate oral vaccine against LMBV infection in aquaculture.
Collapse
Affiliation(s)
- Qian Liu
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Xingchen Huo
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Pengxu Wang
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Fengxia Zhao
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Gailing Yuan
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Chunrong Yang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jianguo Su
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China.
| |
Collapse
|
12
|
Martin MU, Tay CM, Siew TW. Continuous Treatment with IncobotulinumtoxinA Despite Presence of BoNT/A Neutralizing Antibodies: Immunological Hypothesis and a Case Report. Toxins (Basel) 2024; 16:422. [PMID: 39453199 PMCID: PMC11510976 DOI: 10.3390/toxins16100422] [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: 06/24/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
Botulinum Neurotoxin A (BoNT/A) is a bacterial protein that has proven to be a valuable pharmaceutical in therapeutic indications and aesthetic medicine. One major concern is the formation of neutralizing antibodies (nAbs) to the core BoNT/A protein. These can interfere with the therapy, resulting in partial or complete antibody (Ab)-mediated secondary non-response (SNR) or immunoresistance. If titers of nAbs reach a level high enough that all injected BoNT/A molecules are neutralized, immunoresistance occurs. Studies have shown that continuation of treatment of neurology patients who had developed Ab-mediated partial SNR against complexing protein-containing (CPC-) BoNT/A was in some cases successful if patients were switched to complexing protein-free (CPF-) incobotulinumtoxinA (INCO). This seems to contradict the layperson's basic immunological understanding that repeated injection with the same antigen BoNT/A should lead to an increase in antigen-specific antibody titers. As such, we strive to explain how immunological memory works in general, and based on this, we propose a working hypothesis for this paradoxical phenomenon observed in some, but not all, neurology patients with immunoresistance. A critical factor is the presence of potentially immune-stimulatory components in CPC-BoNT/A products that can act as immunologic adjuvants and activate not only naïve, but also memory B lymphocyte responses. Furthermore, we propose that continuous injection of a BoN/TA formulation with low immunogenicity, e.g., INCO, may be a viable option for aesthetic patients with existing nAbs. These concepts are supported by a real-world case example of a patient with immunoresistance whose nAb levels declined with corresponding resumption of clinical response despite regular INCO injections.
Collapse
Affiliation(s)
| | | | - Tuck Wah Siew
- Radium Medical Aesthetics, 3 Temasek Boulevard #03-325/326/327/328, Suntec City Mall, Singapore 038983, Singapore
| |
Collapse
|
13
|
Prajapati A, Hemanth RA, Namrutha MR, Bindu S, Yogisharadhya R, Mohanty NN, Chanda MM, Shivachandra SB. Production of highly soluble and immuno-reactive recombinant flagellin protein of Clostridium chauvoei. Anaerobe 2024; 89:102899. [PMID: 39142535 DOI: 10.1016/j.anaerobe.2024.102899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/02/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024]
Abstract
OBJECTIVE Flagellin protein, an integral component of flagella, provides motility to several bacterial species and also acts as a candidate antigen in diagnostics and subunit vaccines. The bulk production of flagellin with retention of all conformational epitopes using recombinant protein technology is of paramount importance in the development of pathogen-specific immuno-assays and vaccines. We describe the production of highly soluble and immuno-reactive rFliA(C) protein of Clostridium chauvoei, a causative agent of blackleg or black quarter (BQ) affecting cattle and small ruminants worldwide. The bacterium is known to possess peritrichous flagella that provide motility and also act as a virulence factor with high protective antigenicity. METHODS Upon sequence and structural analysis, a partial fliA(C) gene from Clostridium chauvoei was cloned and the recombinant mature protein with N- and C- terminal truncation was over-expressed as a His-tagged fusion protein (∼25 kDa) in Escherichia coli. Subsequently, rFliA(C) protein was purified by single-step affinity chromatography and characterized for its immuno-reactivity in laboratory animals, Western blot, and indirect-ELISA format. RESULTS rFliA(C) was highly soluble and was purified in high quantity and quality. rFliA(C) elicited antigen-specific conformational polyclonal antibodies in rabbit and guinea pig models, as well as anti-Clostridium chauvoei-specific antibodies being specifically detected in BQ-vaccinated and convalescent sera of bovines in Western blot and in indirect-ELISA format. Further, no cross reactivity was noted with antibodies against major bovine diseases (e.g., foot-and-mouth disease, IBR, LSDV, hemorrhagic septicaemia, brucellosis, and leptospirosis). CONCLUSION The study indicated the production of conformational recombinant flagellin-rFliA(C)-antigen and its potential utility in development of diagnostics for detection of Clostridium chauvoei-specific antibodies in BQ-recovered and/or vaccinated animals.
Collapse
Affiliation(s)
- Awadhesh Prajapati
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics (NIVEDI), Post Box No. 6450, Yelahanka, Bengaluru, 560064, Karnataka, India
| | - Roopa Anandamurthy Hemanth
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics (NIVEDI), Post Box No. 6450, Yelahanka, Bengaluru, 560064, Karnataka, India
| | - Mandira Ramakrishna Namrutha
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics (NIVEDI), Post Box No. 6450, Yelahanka, Bengaluru, 560064, Karnataka, India
| | - Suresh Bindu
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics (NIVEDI), Post Box No. 6450, Yelahanka, Bengaluru, 560064, Karnataka, India
| | - Revanaiah Yogisharadhya
- ICAR-Krishi Vigyan Kendra (KVK), ICAR-Research Complex for NEH Region, Hailakandi, 788152, Assam, India
| | - Nihar Nalini Mohanty
- CCS-National Institute of Animal Health (NIAH), Baghpat, 250609, Uttar Pradesh (UP), India
| | - Mohammed Mudassar Chanda
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics (NIVEDI), Post Box No. 6450, Yelahanka, Bengaluru, 560064, Karnataka, India
| | - Sathish Bhadravati Shivachandra
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics (NIVEDI), Post Box No. 6450, Yelahanka, Bengaluru, 560064, Karnataka, India.
| |
Collapse
|
14
|
Han J, Mao K, Yang YG, Sun T. Impact of inorganic/organic nanomaterials on the immune system for disease treatment. Biomater Sci 2024; 12:4903-4926. [PMID: 39190428 DOI: 10.1039/d4bm00853g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
The study of nanomaterials' nature, function, and biocompatibility highlights their potential in drug delivery, imaging, diagnostics, and therapeutics. Advancements in nanotechnology have fostered the development and application of diverse nanomaterials. These materials facilitate drug delivery and influence the immune system directly. Yet, understanding of their impact on the immune system is incomplete, underscoring the need to select materials to achieve desired outcomes carefully. In this review, we outline and summarize the distinctive characteristics and effector functions of inorganic nanomaterials and organic materials in inducing immune responses. We highlight the role and advantages of nanomaterial-induced immune responses in the treatment of immune-related diseases. Finally, we briefly discuss the current challenges and future opportunities for disease treatment and clinical translation of these nanomaterials.
Collapse
Affiliation(s)
- Jing Han
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China.
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Kuirong Mao
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China.
- International Center of Future Science, Jilin University, Changchun, Jilin, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China.
- International Center of Future Science, Jilin University, Changchun, Jilin, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China.
- International Center of Future Science, Jilin University, Changchun, Jilin, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
15
|
Chung YC, Cheng LT, Chu CY, Afzal H, Doan TD. Flagellin Enhances the Immunogenicity of Pasteurella multocida Lipoprotein E Subunit Vaccine. Avian Dis 2024; 68:183-191. [PMID: 39400212 DOI: 10.1637/aviandiseases-d-24-00032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 04/22/2024] [Indexed: 10/15/2024]
Abstract
Fowl cholera, caused by Pasteurella multocida infection, poses challenges for prevention because of its many serotypes. Bacterins are currently widely used for vaccination against fowl cholera, but protection is limited to homologous strains. Live attenuated vaccines of P. multocida provide some heterologous protection, but side effects are considerable. More recently, protein-based antigens are promising subunit vaccines when their low immunogenicity has been addressed with effective adjuvants. Bacterial flagellin has been widely considered a promising adjuvant for vaccines. In this study, we tested the adjutancy of flagellin in a subunit vaccine against P. multocida in a mice and chicken models. For vaccine formulation, the antigen fPlpE (P. multocida liporotein E) was combined with fFliC (Salmonella Typhimurium flagellin). The recombinant proteins of fPlpE and fFliC were successfully expressed using the Escherichia coli system as the expected sizes of 55 kDa and 70 kDa, respectively. The fFliC elicited strong expression levels of proinflammatory cytokine (IL-1β, IL-8, and IL-6) when stimulated in native chicken peripheral blood mononuclear cells. Immunization of mice and chickens with the subunit vaccines containing fFliC accelerated the antibody response. In the challenge tests, fFliC increased vaccine protective efficacy against the heterologous strain P. multocida A1 and highly virulent strain Chu01 in mice and chickens, respectively. These data indicated potential possibilities of using fFliC as an immunostimulant adjuvant in developing a subunit vaccine against fowl cholera.
Collapse
Affiliation(s)
- Yao-Chi Chung
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Neipu, Pingtung 91201, Taiwan
- International Degree Program in Animal Vaccine Technology, International College, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Li-Ting Cheng
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Neipu, Pingtung 91201, Taiwan
- International Degree Program in Animal Vaccine Technology, International College, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Chun-Yen Chu
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Neipu, Pingtung 91201, Taiwan
- International Degree Program in Animal Vaccine Technology, International College, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Haroon Afzal
- International Degree Program in Animal Vaccine Technology, International College, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Thu-Dung Doan
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Neipu, Pingtung 91201, Taiwan,
- International Degree Program in Animal Vaccine Technology, International College, National Pingtung University of Science and Technology, Pingtung, Taiwan
| |
Collapse
|
16
|
Zhuang H, Wang R, Qi Y, Liu Y, Xiong H, Yao J. Nanocoated bacteria with H 2S generation-triggered self-amplified photothermal and photodynamic effect for breast cancer therapy. J Control Release 2024; 373:507-519. [PMID: 39025267 DOI: 10.1016/j.jconrel.2024.07.036] [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: 02/08/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Phototherapy utilizing bacterial carriers has demonstrated efficacy in anti-tumor therapy, while the poor delivery of phototherapeutic agents and immunogenicity of microbial substances remain problematic. Herein, we develop a nanocoated bacterial delivery system (IF-S.T) that in situ forms the efficient photothermal agents via biomineralization and improves the intracellular oxygenation, thus triggering the self-enhanced photothermal therapy (PTT) and photodynamic therapy (PDT) on tumor. We densely coat self-assembled IF (ICG-Fe2+) nanocomplex onto the surface of LT2, weakly virulent strain of Salmonella typhimurium (S.T), by bioadaptive nanocoating techniques, masking bacterial virulence factors and reducing the potential immune adverse effects. Upon penetrating into the tumor environment, IF-S.T responds to H2O2 to trigger the removal of the IF coating, where S.T produces excess hydrogen sulfide (H2S). H2S reacts with Fe2+, yielding ferrous sulfide (FeS) for PTT, and inhibits mitochondrial respiration to enhance tumor cell oxygenation for PDT. Consequently, IF-S.T plus laser irradiation exhibits direct tumor cells killing and elicits robust antitumor immune responses, leading to the complete tumor elimination. Thus, IF-S.T represents a promising platform for effective tumor delivery of photoactive agents for improved PTT/PDT efficacy.
Collapse
Affiliation(s)
- He Zhuang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Rui Wang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Yao Qi
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Yufei Liu
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Hui Xiong
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China.
| | - Jing Yao
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China.
| |
Collapse
|
17
|
Silvestre D, Moreno G, Argüelles MH, Tomás Fariña J, Biedma ME, Peri Ibáñez ES, Mandile MG, Glikmann G, Rumbo M, Castello AA, Temprana CF. Display of FliC131 on the Surface of Lactococcus lactis as a Strategy to Increase its Adjuvanticity for Mucosal Immunization. J Pharm Sci 2024; 113:1794-1803. [PMID: 38522753 DOI: 10.1016/j.xphs.2024.03.013] [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: 11/30/2023] [Revised: 03/15/2024] [Accepted: 03/15/2024] [Indexed: 03/26/2024]
Abstract
Research on innovative mucosal adjuvants is essential to develop new vaccines for safe mucosal application. In this work, we propose the development of a Lactococcus lactis that expresses a variant of flagellin on its surface (FliC131*), to increase the adjuvanticity of the living cell and cell wall-derived particles (CWDP). We optimized the expression of FliC131*, and confirmed its identity and localization by Western blot and flow cytometry. We also generated CWDP containing FliC131* (CDWP-FliC131*) and evaluated their storage stability. Lastly, we measured the human TLR5 stimulating activity in vitro and assessed the adjuvanticity in vivo using ovalbumin (OVA) as a model antigen. As a result, we generated L. lactis/pCWA-FliC131*, that expresses and displays FliC131* on its surface, obtained the corresponding CWDP-FliC131*, and showed that both activated hTLR5 in vitro in a dose-dependent manner. Furthermore, CWDP-FliC131* retained this biological activity after being lyophilized and stored for a year. Finally, intranasal immunization of mice with OVA plus live L. lactis/pCWA-FliC131* or CWDP-FliC131* induced OVA-specific IgG and IgA in serum, intestinal lavages, and bronchoalveolar lavages. Our work demonstrates the potential of this recombinant L. lactis with an enhanced adjuvant effect, prompting its further evaluation for the design of novel mucosal vaccines.
Collapse
Affiliation(s)
- Dalila Silvestre
- Laboratorio de Inmunología y Virología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Sáenz Peña 352, Bernal, 1876, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires, 1425, Argentina
| | - Griselda Moreno
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), Facultad de Ciencias Exactas, Universidad Nacional de La Plata - Consejo Nacional de Investigaciones Científicas y Técnicas (UNLP-CONICET), Boulevard 120 1489, La Plata, 1900, Argentina
| | - Marcelo H Argüelles
- Laboratorio de Inmunología y Virología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Sáenz Peña 352, Bernal, 1876, Argentina
| | - Julieta Tomás Fariña
- Laboratorio de Inmunología y Virología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Sáenz Peña 352, Bernal, 1876, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires, 1425, Argentina
| | - Marina E Biedma
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), Facultad de Ciencias Exactas, Universidad Nacional de La Plata - Consejo Nacional de Investigaciones Científicas y Técnicas (UNLP-CONICET), Boulevard 120 1489, La Plata, 1900, Argentina
| | - Estefanía S Peri Ibáñez
- Laboratorio de Inmunología y Virología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Sáenz Peña 352, Bernal, 1876, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires, 1425, Argentina
| | - Marcelo G Mandile
- Laboratorio de Inmunología y Virología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Sáenz Peña 352, Bernal, 1876, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires, 1425, Argentina
| | - Graciela Glikmann
- Laboratorio de Inmunología y Virología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Sáenz Peña 352, Bernal, 1876, Argentina
| | - Martín Rumbo
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), Facultad de Ciencias Exactas, Universidad Nacional de La Plata - Consejo Nacional de Investigaciones Científicas y Técnicas (UNLP-CONICET), Boulevard 120 1489, La Plata, 1900, Argentina
| | - Alejandro A Castello
- Laboratorio de Inmunología y Virología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Sáenz Peña 352, Bernal, 1876, Argentina; Instituto de Ciencias de la Salud, Universidad Nacional Arturo Jauretche, Av. Calchaquí 6200, Florencio Varela, 1888, Buenos Aires, Argentina
| | - C Facundo Temprana
- Laboratorio de Inmunología y Virología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Sáenz Peña 352, Bernal, 1876, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires, 1425, Argentina.
| |
Collapse
|
18
|
Oladejo M, Tijani AO, Puri A, Chablani L. Adjuvants in cutaneous vaccination: A comprehensive analysis. J Control Release 2024; 369:475-492. [PMID: 38569943 DOI: 10.1016/j.jconrel.2024.03.045] [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: 11/29/2023] [Revised: 03/15/2024] [Accepted: 03/26/2024] [Indexed: 04/05/2024]
Abstract
Skin is the body's largest organ and serves as a protective barrier from physical, thermal, and mechanical environmental challenges. Alongside, the skin hosts key immune system players, such as the professional antigen-presenting cells (APCs) like the Langerhans cells in the epidermis and circulating macrophages in the blood. Further, the literature supports that the APCs can be activated by antigen or vaccine delivery via multiple routes of administration through the skin. Once activated, the stimulated APCs drain to the associated lymph nodes and gain access to the lymphatic system. This further allows the APCs to engage with the adaptive immune system and activate cellular and humoral immune responses. Thus, vaccine delivery via skin offers advantages such as reliable antigen delivery, superior immunogenicity, and convenient delivery. Several preclinical and clinical studies have demonstrated the significance of vaccine delivery using various routes of administration via skin. However, such vaccines often employ adjuvant/(s), along with the antigen of interest. Adjuvants augment the immune response to a vaccine antigen and improve the therapeutic efficacy. Due to these reasons, adjuvants have been successfully used with infectious disease vaccines, cancer immunotherapy, and immune-mediated diseases. To capture these developments, this review will summarize preclinical and clinical study results of vaccine delivery via skin in the presence of adjuvants. A focused discussion regarding the FDA-approved adjuvants will address the experiences of using such adjuvant-containing vaccines. In addition, the challenges and regulatory concerns with these adjuvants will be discussed. Finally, the review will share the prospects of adjuvant-containing vaccines delivered via skin.
Collapse
Affiliation(s)
- Mariam Oladejo
- Department of Immunotherapeutics and Biotechnology, Jerry H Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Akeemat O Tijani
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, USA
| | - Ashana Puri
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, USA.
| | - Lipika Chablani
- Wegmans School of Pharmacy, St. John Fisher University, 3690 East Ave, Rochester, NY 14618, USA.
| |
Collapse
|
19
|
Mo C, Bi J, Li S, Lin Y, Yuan P, Liu Z, Jia B, Xu S. The influence and therapeutic effect of microbiota in systemic lupus erythematosus. Microbiol Res 2024; 281:127613. [PMID: 38232494 DOI: 10.1016/j.micres.2024.127613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/18/2023] [Accepted: 01/09/2024] [Indexed: 01/19/2024]
Abstract
Systemic erythematosus lupus (SLE) is an autoimmune disease involving multiple organs that poses a serious risk to the health and life of patients. A growing number of studies have shown that commensals from different parts of the body and exogenous pathogens are involved in SLE progression, causing barrier disruption and immune dysregulation through multiple mechanisms. However, they sometimes alleviate the symptoms of SLE. Many factors, such as genetic susceptibility, metabolism, impaired barriers, food, and sex hormones, are involved in SLE, and the microbiota drives the development of SLE either by depending on or interacting with these factors. Among these, the crosstalk between genetic susceptibility, metabolism, and microbiota is a hot topic of research and is expected to lay the groundwork for the amelioration of the mechanism, diagnosis, and treatment of SLE. Furthermore, the microbiota has great potential for the treatment of SLE. Ideally, personalised therapeutic approaches should be developed in combination with more specific diagnostic methods. Herein, we provide a comprehensive overview of the role and mechanism of microbiota in lupus of the intestine, oral cavity, skin, and kidney, as well as the therapeutic potential of the microbiota.
Collapse
Affiliation(s)
- Chuzi Mo
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiaming Bi
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Siwei Li
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Yunhe Lin
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Peiyan Yuan
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhongjun Liu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Shuaimei Xu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
20
|
Wei MZ, Chen L, Zhang R, Chen Z, Shen YJ, Zhou BJ, Wang KG, Shan CL, Zhu EP, Cheng ZT. Overview of the recent advances in porcine epidemic diarrhea vaccines. Vet J 2024; 304:106097. [PMID: 38479492 DOI: 10.1016/j.tvjl.2024.106097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 02/04/2024] [Accepted: 03/04/2024] [Indexed: 03/18/2024]
Abstract
Vaccination is the most effective means of preventing and controlling porcine epidemic diarrhea (PED). Conventional vaccines developed from porcine epidemic diarrhea virus (PEDV) GI-a subtypes (CV777 and SM98) have played a vital role in preventing classical PED. However, with the emergence of PEDV mutants in 2010, conventional PEDV GI-a subtype-targeting vaccines no longer provide adequate protection against PEDV GII mutants, thereby making novel-type PED vaccine development an urgent concern to be addressed. Novel vaccines, including nucleic acid vaccines, genetically engineered subunit vaccines, and live vector vaccines, are associated with several advantages, such as high safety and stability, clear targeting, high yield, low cost, and convenient usage. These vaccines can be combined with corresponding ELISA kits to differentiate infected from vaccinated animals, which is beneficial for disease confirmation. This review provides a detailed overview of the recent advancements in PED vaccines, emphasizing on the research and application evaluation of novel PED vaccines. It also considers the future directions and challenges in advancing these vaccines to widespread use in clinics.
Collapse
Affiliation(s)
- Miao-Zhan Wei
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Lan Chen
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Rong Zhang
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Ze Chen
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Yan-Juan Shen
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Bi-Jun Zhou
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Kai-Gong Wang
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Chun-Lan Shan
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Er-Peng Zhu
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang 550025, China.
| | - Zhen-Tao Cheng
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang 550025, China.
| |
Collapse
|
21
|
Kruglova M, Nikitin N, Evtushenko E, Matveeva I, Mazurov A, Pavlenko I, Popova V, Bogomolova O, Vasilyev S, Markova E, Fedorov Y. Inactivated Flagellin-Containing Vaccine Efficacy against Ovine Enzootic Abortion. Pathogens 2024; 13:277. [PMID: 38668231 PMCID: PMC11053442 DOI: 10.3390/pathogens13040277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/29/2024] Open
Abstract
Chlamydia abortus is the etiological agent of abortion and fetal loss in sheep, goats and bovine cattle in many countries. Even though commercially available vaccines can reduce the incidence in sheep, the development of new, safe, and effective vaccines remains high on the agenda. In this study, an evaluation was made of the efficacy of a vaccine candidate, an inactivated antigen based on the extract of outer membrane proteins of a C. abortus strain known as Chlamydia VNITIBP-21, in combination with recombinant flagellin as an adjuvant. Pregnant sheep (n = 43) were divided into three groups: an experimental vaccinated group, a control infected group and a control non-infected group. The sheep were vaccinated twice, with an interval of 3 weeks, then infected with the homologous virulent strain of Chlamydia abortus on pregnancy day 75. The vaccine candidate reduced C. abortus shedding in vaginal swabs considerably, in comparison with the control group. In addition, ewes in the experimental group experienced no abortions, while those in the control group experienced instances of abortion, as well as births of weak and nonviable lambs. The findings show that the vaccine candidate proved itself to be promising in combatting the agent of ovine abortion and fetal loss.
Collapse
Affiliation(s)
- Maria Kruglova
- All-Russian Scientific Research and Technological Institute of Biological Industry, Biocombinat, 141142 Moscow, Russia
| | - Nikolai Nikitin
- Department of Virology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Ekaterina Evtushenko
- Department of Virology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Irina Matveeva
- All-Russian Scientific Research and Technological Institute of Biological Industry, Biocombinat, 141142 Moscow, Russia
| | - Aleksandr Mazurov
- All-Russian Scientific Research and Technological Institute of Biological Industry, Biocombinat, 141142 Moscow, Russia
| | - Igor Pavlenko
- All-Russian Scientific Research and Technological Institute of Biological Industry, Biocombinat, 141142 Moscow, Russia
| | - Vera Popova
- All-Russian Scientific Research and Technological Institute of Biological Industry, Biocombinat, 141142 Moscow, Russia
| | - Olesya Bogomolova
- All-Russian Scientific Research and Technological Institute of Biological Industry, Biocombinat, 141142 Moscow, Russia
| | - Stepan Vasilyev
- All-Russian Scientific Research and Technological Institute of Biological Industry, Biocombinat, 141142 Moscow, Russia
| | - Evgeniya Markova
- All-Russian Scientific Research and Technological Institute of Biological Industry, Biocombinat, 141142 Moscow, Russia
| | - Yuri Fedorov
- All-Russian Scientific Research and Technological Institute of Biological Industry, Biocombinat, 141142 Moscow, Russia
| |
Collapse
|
22
|
Choi M, Shridhar S, Fox H, Luo K, Amin MN, Tennant SM, Simon R, Cross AS. The O-glycan is essential for the induction of protective antibodies against lethal infection by flagella A-bearing Pseudomonas aeruginosa. Infect Immun 2024; 92:e0042723. [PMID: 38391207 PMCID: PMC10929410 DOI: 10.1128/iai.00427-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/25/2024] [Indexed: 02/24/2024] Open
Abstract
To address the problem of increased antimicrobial resistance, we developed a glycoconjugate vaccine comprised of O-polysaccharides (OPS) of the four most prevalent serotypes of Klebsiella pneumoniae (KP) linked to recombinant flagellin types A and B (rFlaA and rFlaB) of Pseudomonas aeruginosa (PA). Flagellin is the major subunit of the flagellar filament. Flagella A and B, essential virulence factors for PA, are glycosylated with different glycans. We previously reported that while both rFlaA and rFlaB were highly immunogenic, only the rFlaB antisera reduced PA motility and protected mice from lethal PA infection in a mouse model of thermal injury. Since recombinant flagellin is not glycosylated, we examined the possibility that the glycan on native FlaA (nFlaA) might be critical to functional immune responses. We compared the ability of nFlaA to that of native, deglycosylated FlaA (dnFlaA) to induce functionally active antisera. O glycan was removed from nFlaA with trifluoromethanesulfonic acid. Despite the similar high-titered anti-FlaA antibody levels elicited by nFlaA, rFlaA, and dnFlaA, only the nFlaA antisera inhibited PA motility and protected mice following lethal intraperitoneal bacterial challenge. Both the protective efficacy and carrier protein function of nFlaA were retained when conjugated to KP O1 OPS. We conclude that unlike the case with FlaB O glycan, the FlaA glycan is an important epitope for the induction of functionally active anti-FlaA antibodies.
Collapse
Affiliation(s)
- Myeongjin Choi
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Surekha Shridhar
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Heather Fox
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Kun Luo
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mohammed N. Amin
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Sharon M. Tennant
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Raphael Simon
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Alan S. Cross
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
23
|
Sokol OO, Nikitin NA, Evtushenko EA, Karpova OV, Matveeva IN, Gryn SA, Popova VM, Ivanov IV, Fedorov YN, Litenkova IY. Protective Activity of Inactivated Rabies Vaccine Using Flagellin-Based Adjuvant. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:574-582. [PMID: 38648774 DOI: 10.1134/s0006297924030155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/19/2024] [Accepted: 03/01/2024] [Indexed: 04/25/2024]
Abstract
Rabies is a zoonotic disease with high lethality. Most human deaths are associated with the bites received from dogs and cats. Vaccination is the most effective method of preventing rabies disease in both animals and humans. In this study, the ability of an adjuvant based on recombinant Salmonella typhimurium flagellin to increase protective activity of the inactivated rabies vaccine in mice was evaluated. A series of inactivated dry culture vaccine for dogs and cats "Rabikan" (strain Shchelkovo-51) with addition of an adjuvant at various dilutions were used. The control preparation was a similar series of inactivated dry culture vaccine without an adjuvant. Protective activity of the vaccine preparations was evaluated by the NIH potency test, which is the most widely used and internationally recommended method for testing effectiveness of the inactivated rabies vaccines. The value of specific activity of the tested rabies vaccine when co-administered with the adjuvant was significantly higher (48.69 IU/ml) than that of the vaccine without the adjuvant (3.75 IU/ml). Thus, recombinant flagellin could be considered as an effective adjuvant in the composition of future vaccine preparations against rabies virus.
Collapse
Affiliation(s)
- Olga O Sokol
- All-Russian Scientific Research and Technological Institute of Biological Industry, Biocombinat, Moscow Region, 141142, Russia
| | - Nikolai A Nikitin
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
| | | | - Olga V Karpova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Irina N Matveeva
- All-Russian Scientific Research and Technological Institute of Biological Industry, Biocombinat, Moscow Region, 141142, Russia
- Shchelkovo Biocombinat Federal State Enterprise, Biocombinat, Moscow Region, 141142, Russia
| | - Svetlana A Gryn
- All-Russian Scientific Research and Technological Institute of Biological Industry, Biocombinat, Moscow Region, 141142, Russia
| | - Vera M Popova
- All-Russian Scientific Research and Technological Institute of Biological Industry, Biocombinat, Moscow Region, 141142, Russia
| | - Igor V Ivanov
- All-Russian Scientific Research and Technological Institute of Biological Industry, Biocombinat, Moscow Region, 141142, Russia
| | - Yuri N Fedorov
- All-Russian Scientific Research and Technological Institute of Biological Industry, Biocombinat, Moscow Region, 141142, Russia
| | - Irina Y Litenkova
- Shchelkovo Biocombinat Federal State Enterprise, Biocombinat, Moscow Region, 141142, Russia
| |
Collapse
|
24
|
Fu Z, Wang L, Guo H, Lin S, Huang W, Pang Y. Bacterial Flagellum-Drug Nanoconjugates for Carrier-Free Immunochemotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306303. [PMID: 37919854 DOI: 10.1002/smll.202306303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Indexed: 11/04/2023]
Abstract
The combination of immunotherapy and chemotherapy to ablate tumors has attracted substantial attention due to the ability to simultaneously elicit antitumor immune responses and trigger direct tumor cell death. However, conventional combinational strategies mainly focus on the employment of drug carriers to deliver immunomodulators, chemotherapeutics, or their combinations, always suffering from complicated preparation and carrier-relevant side effects. Here, the fabrication of bacterial flagellum-drug nanoconjugates (FDNCs) for carrier-free immunochemotherapy is described. FDNCs are simply prepared by attaching chemotherapeutics to amine residues of flagellin through an acid-sensitive and traceless cis-aconityl linker. By virtue of native nanofibrous structure and immunogenicity, bacterial flagella not only show long-term tumor retention and highly efficient cell internalization, but also provoke robust systemic antitumor immune responses. Meanwhile, conjugated chemotherapeutics exhibit an acid-mediated release profile and durable intratumoral exposure, which can induce potent tumor cell inhibition via direct killing. More importantly, this combination is able to augment immunoactivation effects associated with chemotherapy-enabled immunogenic tumor cell death to further enhance antitumor efficacy. By leveraging the innate response of the immune system to pathogens, the conjugation of therapeutic agents with self-adjuvant bacterial flagella provides an alternative approach to develop carrier-free nanotherapeutics for tumor immunochemotherapy.
Collapse
Affiliation(s)
- Zhenzhen Fu
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Lu Wang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Haiyan Guo
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Sisi Lin
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Wei Huang
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yan Pang
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Department of Ophthalmology, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, China
| |
Collapse
|
25
|
Wang G, Wang Y, Ma F. Exploiting bacterial-origin immunostimulants for improved vaccination and immunotherapy: current insights and future directions. Cell Biosci 2024; 14:24. [PMID: 38368397 PMCID: PMC10874560 DOI: 10.1186/s13578-024-01207-7] [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: 10/10/2023] [Accepted: 02/06/2024] [Indexed: 02/19/2024] Open
Abstract
Vaccination is a valid strategy to prevent and control newly emerging and reemerging infectious diseases in humans and animals. However, synthetic and recombinant antigens are poor immunogenic to stimulate efficient and protective host immune response. Immunostimulants are indispensable factors of vaccines, which can promote to trigger fast, robust, and long-lasting immune responses. Importantly, immunotherapy with immunostimulants is increasing proved to be an effective and promising treatment of cancer, which could enhance the function of the immune system against tumor cells. Pattern recognition receptors (PRRs) play vital roles in inflammation and are central to innate and adaptive immune responses. Toll-like receptors (TLRs)-targeting immunostimulants have become one of the hotspots in adjuvant research and cancer therapy. Bacterial-origin immunoreactive molecules are usually the ligands of PRRs, which could be fast recognized by PRRs and activate immune response to eliminate pathogens. Varieties of bacterial immunoreactive molecules and bacterial component-mimicking molecules have been successfully used in vaccines and clinical therapy so far. This work provides a comprehensive review of the development, current state, mechanisms, and applications of bacterial-origin immunostimulants. The exploration of bacterial immunoreactive molecules, along with their corresponding mechanisms, holds immense significance in deepening our understanding of bacterial pathogenicity and in the development of promising immunostimulants.
Collapse
Affiliation(s)
- Guangyu Wang
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, Nanjing, Jiangsu, 210023, China
| | - Yongkang Wang
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, Nanjing, Jiangsu, 210023, China
| | - Fang Ma
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China.
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, 212013, China.
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou, 225300, China.
| |
Collapse
|
26
|
Song Y, Mehl F, Zeichner SL. Vaccine Strategies to Elicit Mucosal Immunity. Vaccines (Basel) 2024; 12:191. [PMID: 38400174 PMCID: PMC10892965 DOI: 10.3390/vaccines12020191] [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: 12/01/2023] [Revised: 01/29/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Vaccines are essential tools to prevent infection and control transmission of infectious diseases that threaten public health. Most infectious agents enter their hosts across mucosal surfaces, which make up key first lines of host defense against pathogens. Mucosal immune responses play critical roles in host immune defense to provide durable and better recall responses. Substantial attention has been focused on developing effective mucosal vaccines to elicit robust localized and systemic immune responses by administration via mucosal routes. Mucosal vaccines that elicit effective immune responses yield protection superior to parenterally delivered vaccines. Beyond their valuable immunogenicity, mucosal vaccines can be less expensive and easier to administer without a need for injection materials and more highly trained personnel. However, developing effective mucosal vaccines faces many challenges, and much effort has been directed at their development. In this article, we review the history of mucosal vaccine development and present an overview of mucosal compartment biology and the roles that mucosal immunity plays in defending against infection, knowledge that has helped inform mucosal vaccine development. We explore new progress in mucosal vaccine design and optimization and novel approaches created to improve the efficacy and safety of mucosal vaccines.
Collapse
Affiliation(s)
- Yufeng Song
- Department of Pediatrics, University of Virginia, Charlottesville, VA 22908, USA; (Y.S.)
| | - Frances Mehl
- Department of Pediatrics, University of Virginia, Charlottesville, VA 22908, USA; (Y.S.)
| | - Steven L. Zeichner
- Department of Pediatrics, University of Virginia, Charlottesville, VA 22908, USA; (Y.S.)
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
27
|
Li Y, Liu C, Sun Y, Wang R, Wu C, Zhao H, Zhang L, Song D, Gao Q. Construction of the flagellin F mutant of Vibrio parahaemolyticus and its toxic effects on silver pomfret (Pampus argenteus) cells. Int J Biol Macromol 2024; 259:129395. [PMID: 38218285 DOI: 10.1016/j.ijbiomac.2024.129395] [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: 11/20/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/15/2024]
Abstract
Vibrio parahaemolyticus causes diseases in aquatic organisms, leading to substantial financial losses to the aquaculture industry; its flagellin F (flaF) protein triggers severe inflammation in host cells. To enhance the understanding of the function of flaF in V. parahaemolyticus infection, in this study, a flaF-deficient mutant was constructed by employing two-step homologous recombination. The flaF-deficient mutant induced a significantly lower toll-like receptor 5 (TLR5) expression and apoptosis in fish intestinal epithelial cells than the wild-type V. parahaemolyticus. Furthermore, fluorescence labelling and microscopy analysis of TLR5 showed that V. parahaemolyticus and its mutant strain significantly enhanced TLR5 expression. Additionally, the findings suggest that flaF deletion did not significantly affect the expression of myeloid differentiation factor 88 (MyD88) and interleukin-8 (IL-8) induced by V.parahaemolyticus. In summary, V. parahaemolyticus induced a TLR5-dependent inflammatory response and apoptosis through MyD88, which was observed to be influenced by flaF deletion. In this study, we obtained stable mutants of V. parahaemolyticus via target gene deletion-which is a rapid and effective approach-and compared the induction of inflammatory response and apoptosis by V. parahaemolyticus and its mutant strain, providing novel perspectives for functional gene research in V. parahaemolyticus.
Collapse
Affiliation(s)
- Yang Li
- College of Life Science, Huzhou University, Huzhou 313000, PR China
| | - Chao Liu
- Songjiang Hospital Affiliated to Shanghai Jiaotong University, School of Medicine, Shanghai 201699, PR China
| | - Yuechen Sun
- Haidian Foreign Language Academy, Beijing 100195, PR China
| | - Ruijun Wang
- College of Life Science, Huzhou University, Huzhou 313000, PR China
| | - Choufei Wu
- College of Life Science, Huzhou University, Huzhou 313000, PR China
| | - Hanqu Zhao
- College of Life Science, Huzhou University, Huzhou 313000, PR China
| | - Liqin Zhang
- College of Life Science, Huzhou University, Huzhou 313000, PR China
| | - Dawei Song
- College of Life Science, Huzhou University, Huzhou 313000, PR China.
| | - Quanxin Gao
- College of Life Science, Huzhou University, Huzhou 313000, PR China.
| |
Collapse
|
28
|
Liu Q, Dai Y, Wu X, Zhang Q, An X, Lai F. Lawsonia intracellularis flagellin protein LfliC stimulates NF-κB and MAPK signaling pathways independently of TLR5 interaction. Vet Microbiol 2024; 289:109960. [PMID: 38176089 DOI: 10.1016/j.vetmic.2023.109960] [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: 02/22/2023] [Revised: 12/05/2023] [Accepted: 12/19/2023] [Indexed: 01/06/2024]
Abstract
Lawsonia intracellularis, a Gram-negative obligate intracellular bacterium and etiologic agent of porcine proliferative enteropathy, was observed to have a long, single, and unipolar flagellum. Bacterial flagellar filament comprises thousands of copies of the protein flagellin (FliC), and has been reported to be recognized by Toll-like receptor (TLR5) to activate the NF-κB and MAPK signaling pathways, thereby inducing the expression of proinflammatory genes. Recently, two L. intracellularis flagellin proteins, LfliC and LFliC, were reported to be involved in bacterial-host interaction and immune response. Here, to further explore the role of LfliC in proinflammatory response, we purified LfliC, and found that its exposure could activate NF-κB signaling pathway in both HEK293T and IPI-FX cells, as well as activate MAPK p38 and ERK1/2 in HEK293T cells but not in IPI-FX cells. However, our yeast two-hybrid and co-immunoprecipitation assay results revealed that LfliC has no interaction with the porcine TLR5 ECD domain though it harbors the conserved D1-like motif required for the interaction. Moreover, LfliC was identified as a substrate of the virulence-associated type III secretion system (T3SS) by using the heterologous Y. enterocolitica system. Transient expression of LfliC also activated the NF-κB and MAPK signaling pathway in HEK293T cells. Collectively, our results suggest that both the exposure and expression of L. intracellularis LfliC can induce the NF-κB and MAPK signaling pathway in mammalian cells. Our findings may provide important implications and resources for the development of diagnostic tools or vaccines and dissection of the pathogenesis of L. intracellularis.
Collapse
Affiliation(s)
- Qianru Liu
- School of Bioscience and Bioengineering, Jiangxi Key Laboratory for Conservation and Utilization of Fungal Resources, Jiangxi Agricultural University, Nanchang 330045, China
| | - Yimin Dai
- School of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| | - Xiaoyu Wu
- Collaborative Innovation Center of Postharvest Key Technology and Quality Safety of Fruits and Vegetables in Jiangxi Province, Nanchang, Jiangxi, China
| | - Qinghua Zhang
- School of Bioscience and Bioengineering, Jiangxi Key Laboratory for Conservation and Utilization of Fungal Resources, Jiangxi Agricultural University, Nanchang 330045, China
| | - Xuejiao An
- School of Bioscience and Bioengineering, Jiangxi Key Laboratory for Conservation and Utilization of Fungal Resources, Jiangxi Agricultural University, Nanchang 330045, China
| | - Fenju Lai
- School of Bioscience and Bioengineering, Jiangxi Key Laboratory for Conservation and Utilization of Fungal Resources, Jiangxi Agricultural University, Nanchang 330045, China.
| |
Collapse
|
29
|
Saravanan D, Mohan M. Immunoinformatics-driven approach for development of potential multi-epitope vaccine against the secreted protein FlaC of Campylobacter jejuni. J Biomol Struct Dyn 2024:1-12. [PMID: 38287490 DOI: 10.1080/07391102.2024.2308766] [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: 07/03/2023] [Accepted: 01/15/2024] [Indexed: 01/31/2024]
Abstract
Campylobacter jejuni causes a leading human gastrointestinal infection which is associated with foodborne diarrhea, stomach cramping, and fever. In the recent years, numerous multidrug-resistant strains of C. jejuni has evolved and is considered in the priority pathogens category. Therefore, an increasing demand exists to develop an effective vaccine against Campylobacteriosis. The T cell and B cell epitopes from the FlaC protein were predicted using comprehensive immunoinformatics tools. The predicted epitopes were chosen based on their antigenicity, allergenicity, and toxicity profiles. Using the bioinformatics approach various physicochemical properties of the constructed vaccine were determined. The molecular docking analysis of the vaccine with the TLRs demonstrated that TLR5 has a higher binding affinity of -1159.0 kcal/mol. Molecular dynamics simulation has confirmed the stable association of the vaccine with TLR5. The immune response of the constructed vaccine was validated using immunostimulation. Based on this study, we recommend the formulation of a multi-epitope vaccine as a promising agent to effectively combat the dreadful campylobacteriosis infection.
Collapse
Affiliation(s)
- Deepak Saravanan
- School of Interdisciplinary Design and Innovation, Indian Institute of Information Technology, Design and Manufacturing, Kancheepuram, Tamil Nadu, India
| | - Monisha Mohan
- School of Interdisciplinary Design and Innovation, Indian Institute of Information Technology, Design and Manufacturing, Kancheepuram, Tamil Nadu, India
| |
Collapse
|
30
|
Temchura V, Wagner JT, Damm D. Immunogenicity of Recombinant Lipid-Based Nanoparticle Vaccines: Danger Signal vs. Helping Hand. Pharmaceutics 2023; 16:24. [PMID: 38258035 PMCID: PMC10818441 DOI: 10.3390/pharmaceutics16010024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
Infectious diseases are a predominant problem in human health. While the incidence of many pathogenic infections is controlled by vaccines, some pathogens still pose a challenging task for vaccine researchers. In order to face these challenges, the field of vaccine development has changed tremendously over the last few years. For non-replicating recombinant antigens, novel vaccine delivery systems that attempt to increase the immunogenicity by mimicking structural properties of pathogens are already approved for clinical applications. Lipid-based nanoparticles (LbNPs) of different natures are vesicles made of lipid layers with aqueous cavities, which may carry antigens and other biomolecules either displayed on the surface or encapsulated in the cavity. However, the efficacy profile of recombinant LbNP vaccines is not as high as that of live-attenuated ones. This review gives a compendious picture of two approaches that affect the immunogenicity of recombinant LbNP vaccines: (i) the incorporation of immunostimulatory agents and (ii) the utilization of pre-existing or promiscuous cellular immunity, which might be beneficial for the development of tailored prophylactic and therapeutic LbNP vaccine candidates.
Collapse
Affiliation(s)
- Vladimir Temchura
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | | | - Dominik Damm
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| |
Collapse
|
31
|
Pei C, Dong H, Teng Z, Wei S, Zhang Y, Yin S, Tang J, Sun S, Guo H. Self-Assembling Nanovaccine Fused with Flagellin Enhances Protective Effect against Foot-and-Mouth Disease Virus. Vaccines (Basel) 2023; 11:1675. [PMID: 38006007 PMCID: PMC10675102 DOI: 10.3390/vaccines11111675] [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: 09/26/2023] [Revised: 10/26/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
Nanovaccines based on self-assembling nanoparticles (NPs) can show conformational epitopes of antigens and they have high immunogenicity. In addition, flagellin, as a biological immune enhancer, can be fused with an antigen to considerably enhance the immune effect of antigens. In improving the immunogenicity and stability of a foot-and-mouth disease virus (FMDV) antigen, novel FMDV NP antigens were prepared by covalently coupling the VP1 protein and truncated flagellin containing only N-terminus D0 and D1 (N-terminal aa 1-99, nFLiC) with self-assembling NPs (i301). The results showed that the fusion proteins VP1-i301 and VP1-i301-nFLiC can assemble into NPs with high thermal tolerance and stability, obtain high cell uptake efficiency, and upregulate marker molecules and immune-stimulating cytokines in vitro. In addition, compared with monomeric VP1 antigen, high-level cytokines were stimulated with VP1-i301 and VP1-i301-nFLiC nanovaccines in guinea pigs, to provide clinical protection against viral infection comparable to an inactivated vaccine. This study provides new insight for the development of a novel FMD vaccine.
Collapse
Affiliation(s)
- Chenchen Pei
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Hu Dong
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Zhidong Teng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Sumin Wei
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Yun Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Shuanghui Yin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Jianli Tang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Shiqi Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Huichen Guo
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
- College of Animal Science, Yangtze University, Jingzhou 434023, China
| |
Collapse
|
32
|
Liu Q, Huo X, Tian Q, Wang P, Zhao F, Yang C, Su J. The oral antigen-adjuvant fusion vaccine P-MCP-FlaC provides effective protective effect against largemouth bass ranavirus infection. FISH & SHELLFISH IMMUNOLOGY 2023; 142:109179. [PMID: 37863125 DOI: 10.1016/j.fsi.2023.109179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 10/22/2023]
Abstract
Largemouth bass ranavirus (LMBV) is highly contagious and lethal to largemouth bass, causing significant economic losses to the aquaculture industry. Oral vaccination is generally considered the most ideal strategy for protecting fish from viral infection. In this study, the fusion protein MCP-FlaC, consisting of the main capsid protein (MCP) as the antigen and flagellin C (FlaC) as the adjuvant, was intracellularly expressed in Pichia pastoris. Subsequently, the recombinant P. pastoris was freeze-dried to prepare the oral vaccine P-MCP-FlaC. Transmission electron microscopy and scanning electron microscopy analysis showed that the morphology and structure of the freeze-dried recombinant P. pastoris vaccine remained intact. The experiment fish (n = 100) was divided into five groups (P-MCP-FlaC, P-MCP, P-FlaC, P-pPIC3.5K, control) to evaluate the protective efficacy of the recombinant vaccine. Oral P-MCP-FlaC vaccine effectively up-regulated the serum enzymes activity (total superoxide dismutase, lysozyme, total antioxidant capacity, and complement component 3). The survival rate of P-MCP-FlaC group was significantly higher than that of the other groups. The mRNA expression of crucial immune genes (IL-1β, TNF-α, MHC-II, IFN-γ, Mx, IgM, IgT) was also signally elevated in P-MCP-FlaC group. Vaccine P-MCP-FlaC markedly inhibited the replication of LMBV in the spleen, head kidney, and intestine, while reducing the degree of lesion in the spleen. These results suggest that the oral P-MCP-FlaC vaccine could effectively control LMBV infection, proving an effective strategy for viral diseases prevention in aquaculture.
Collapse
Affiliation(s)
- Qian Liu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, 266237, China; Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xingchen Huo
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Qingqing Tian
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Pengxu Wang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Fengxia Zhao
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Chunrong Yang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jianguo Su
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, 266237, China; Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
33
|
Liang H, Zhang L, Liu Z, Hoden B, DeRubei D, Zhang Y, Wang F, Zhang D. Upregulation of TLR5 indicates a favorable prognosis in prostate cancer. Prostate 2023; 83:1035-1045. [PMID: 37118933 PMCID: PMC10330358 DOI: 10.1002/pros.24545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/03/2023] [Accepted: 04/18/2023] [Indexed: 04/30/2023]
Abstract
BACKGROUND Toll-like receptors (TLRs) are the key sensors of innate immunity for triggering immune responses against infections. TLRs are well known to be expressed and activated in innate immune cells, such as macrophage and dendritic cells, but we and others have found that some TLRs are also functional in epithelial cells. However, the role of an epithelial TLR in prostate cancer remains elusive. METHODS TLR5 expression in messenger RNA and protein level in prostate cancer was determined by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and immunohistochemistry (IHC). The activation of TLR5 signaling in epithelial cells was detected upon nuclear factor-κB activation by luciferase assay and western blot analysis, and proinflammatory cytokine activation by RT-qPCR. Distinguishing between the TLR5 and NLRC4 pathways, both recognizing flagellin, is determined by small interfering RNA and proinflammatory cytokine activation. The role of TLR5 in prostate cancer was analyzed by IHC and bioinformatics using a general and single-cell database. RESULTS In the present study, we show that TLR5, among other TLRs, is exceedingly expressed in human prostate cancer cells. This cancer epithelial cell TLR5 functions to activate the TLR5 signaling pathway in human prostate cancer cells, as it does with innate immune cell TLR5. The bacterial protein flagellin induces a robust immune response in prostate cancer cells in a TLR5-dependent but NLRC4-independent manner. TLR5 is highly expressed in prostate cancer patient specimens, and high TLR5 expression in prostate cancer patients indicates a favorable prognosis. CONCLUSIONS TLR5, as an innate immunity receptor, is a functional TLR in human prostate cancer epithelial cells. TLR5 plays an important role in prostate cancer development and is a new potential prognosis biomarker. TLR5 may represent a novel immunotherapy target against prostate cancer.
Collapse
Affiliation(s)
- Hongbin Liang
- Center for Infectious and Inflammatory Diseases, Institute of Bioscience and Technology, Department of Translational Medical Sciences, Texas A&M University, Houston, TX, USA
| | - Lin Zhang
- Center for Infectious and Inflammatory Diseases, Institute of Bioscience and Technology, Department of Translational Medical Sciences, Texas A&M University, Houston, TX, USA
| | - Ziying Liu
- Center for Translational Cancer Research, Institute of Bioscience and Technology, Department of Translational Medical Sciences, Texas A&M University, Houston, TX, USA
| | - Bettina Hoden
- Center for Infectious and Inflammatory Diseases, Institute of Bioscience and Technology, Department of Translational Medical Sciences, Texas A&M University, Houston, TX, USA
| | - David DeRubei
- Center for Infectious and Inflammatory Diseases, Institute of Bioscience and Technology, Department of Translational Medical Sciences, Texas A&M University, Houston, TX, USA
| | - Yifan Zhang
- Center for Infectious and Inflammatory Diseases, Institute of Bioscience and Technology, Department of Translational Medical Sciences, Texas A&M University, Houston, TX, USA
| | - Fen Wang
- Center for Translational Cancer Research, Institute of Bioscience and Technology, Department of Translational Medical Sciences, Texas A&M University, Houston, TX, USA
| | - Dekai Zhang
- Center for Infectious and Inflammatory Diseases, Institute of Bioscience and Technology, Department of Translational Medical Sciences, Texas A&M University, Houston, TX, USA
| |
Collapse
|
34
|
Ren H, Jia W, Xie Y, Yu M, Chen Y. Adjuvant physiochemistry and advanced nanotechnology for vaccine development. Chem Soc Rev 2023; 52:5172-5254. [PMID: 37462107 DOI: 10.1039/d2cs00848c] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
Vaccines comprising innovative adjuvants are rapidly reaching advanced translational stages, such as the authorized nanotechnology adjuvants in mRNA vaccines against COVID-19 worldwide, offering new strategies to effectively combat diseases threatening human health. Adjuvants are vital ingredients in vaccines, which can augment the degree, extensiveness, and longevity of antigen specific immune response. The advances in the modulation of physicochemical properties of nanoplatforms elevate the capability of adjuvants in initiating the innate immune system and adaptive immunity, offering immense potential for developing vaccines against hard-to-target infectious diseases and cancer. In this review, we provide an essential introduction of the basic principles of prophylactic and therapeutic vaccination, key roles of adjuvants in augmenting and shaping immunity to achieve desired outcomes and effectiveness, and the physiochemical properties and action mechanisms of clinically approved adjuvants for humans. We particularly focus on the preclinical and clinical progress of highly immunogenic emerging nanotechnology adjuvants formulated in vaccines for cancer treatment or infectious disease prevention. We deliberate on how the immune system can sense and respond to the physicochemical cues (e.g., chirality, deformability, solubility, topology, and chemical structures) of nanotechnology adjuvants incorporated in the vaccines. Finally, we propose possible strategies to accelerate the clinical implementation of nanotechnology adjuvanted vaccines, such as in-depth elucidation of nano-immuno interactions, antigen identification and optimization by the deployment of high-dimensional multiomics analysis approaches, encouraging close collaborations among scientists from different scientific disciplines and aggressive exploration of novel nanotechnologies.
Collapse
Affiliation(s)
- Hongze Ren
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Wencong Jia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Yujie Xie
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| |
Collapse
|
35
|
Li Y, Yao H, Liu S, Song D, Wu C, Zhang S, Gao Q, Zhang L. The role of flagellin F in Vibrio Parahaemolyticus-induced intestinal immunity and functional domain identification. Int J Biol Macromol 2023; 244:125404. [PMID: 37327919 DOI: 10.1016/j.ijbiomac.2023.125404] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 06/18/2023]
Abstract
The marine pathogen Vibrio parahaemolyticus has caused huge economic losses to aquaculture. Flagellin is a key bacterial virulence factor that induces an inflammatory response via activation of Toll-like receptor 5 (TLR5) signaling. Herein, to explore the inflammatory activity of V. parahaemolyticus flagellins (flaA, flaB, flaC, flaD, flaE, and flaF), we investigated their ability to induce apoptosis in a fish cell line. All six flagellins induced severe apoptosis. Moreover, treatment with V. parahaemolyticus flagellins increased TLR5 and myeloid differentiation factor 88 (MyD88) expression and the production of TNF-α and IL-8 significantly. This indicated that flagellins might induce a TLR5-meditated immune response via an MyD88-dependent pathway. FlaF exhibited the strongest immunostimulatory effect; therefore, the interaction between TLR5 and flaF was screened using the yeast two-hybrid system. A significant interaction between the two proteins was observed, indicating that flaF binds directly to TLR5. Finally, the amino acids that participate in the TLR5-flaF interaction were identified using molecular simulation, which indicated three binding sites. These results deepen our understanding of the immunogenic properties of flagellins from V. parahaemolyticus, which could be used for vaccine development in the future.
Collapse
Affiliation(s)
- Yang Li
- College of Life Science, Huzhou University, Huzhou 313000, PR China
| | - Hongqing Yao
- Songjiang Hospital Affiliated to Shanghai Jiaotong University, School of Medicine, Shanghai 201699, PR China
| | - Sushuang Liu
- Department of Life Sciences and Health, School of Science and Engineering, Huzhou College, PR China
| | - Dawei Song
- College of Life Science, Huzhou University, Huzhou 313000, PR China
| | - Choufei Wu
- College of Life Science, Huzhou University, Huzhou 313000, PR China
| | - Shaoyong Zhang
- College of Life Science, Huzhou University, Huzhou 313000, PR China
| | - Quanxin Gao
- College of Life Science, Huzhou University, Huzhou 313000, PR China.
| | - Liqin Zhang
- College of Life Science, Huzhou University, Huzhou 313000, PR China.
| |
Collapse
|
36
|
Brai A, Poggialini F, Pasqualini C, Trivisani CI, Vagaggini C, Dreassi E. Progress towards Adjuvant Development: Focus on Antiviral Therapy. Int J Mol Sci 2023; 24:9225. [PMID: 37298177 PMCID: PMC10253057 DOI: 10.3390/ijms24119225] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/12/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
In recent decades, vaccines have been extraordinary resources to prevent pathogen diffusion and cancer. Even if they can be formed by a single antigen, the addition of one or more adjuvants represents the key to enhance the response of the immune signal to the antigen, thus accelerating and increasing the duration and the potency of the protective effect. Their use is of particular importance for vulnerable populations, such as the elderly or immunocompromised people. Despite their importance, only in the last forty years has the search for novel adjuvants increased, with the discovery of novel classes of immune potentiators and immunomodulators. Due to the complexity of the cascades involved in immune signal activation, their mechanism of action remains poorly understood, even if significant discovery has been recently made thanks to recombinant technology and metabolomics. This review focuses on the classes of adjuvants under research, recent mechanism of action studies, as well as nanodelivery systems and novel classes of adjuvants that can be chemically manipulated to create novel small molecule adjuvants.
Collapse
Affiliation(s)
- Annalaura Brai
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Federica Poggialini
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Claudia Pasqualini
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Claudia Immacolata Trivisani
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
- Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria
| | - Chiara Vagaggini
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Elena Dreassi
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| |
Collapse
|
37
|
Zhang Y, Tan W, Sultonova RD, Nguyen DH, Zheng JH, You SH, Rhee JH, Kim SY, Khim K, Hong Y, Min JJ. Synergistic cancer immunotherapy utilizing programmed Salmonella typhimurium secreting heterologous flagellin B conjugated to interleukin-15 proteins. Biomaterials 2023; 298:122135. [PMID: 37148758 DOI: 10.1016/j.biomaterials.2023.122135] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 04/12/2023] [Accepted: 04/26/2023] [Indexed: 05/08/2023]
Abstract
The use of appropriately designed immunotherapeutic bacteria is an appealing approach to tumor therapy because the bacteria specifically target tumor tissue and deliver therapeutic payloads. The present study describes the engineering of an attenuated strain of Salmonella typhimurium deficient in ppGpp biosynthesis (SAM) that could secrete Vibrio vulnificus flagellin B (FlaB) conjugated to human (hIL15/FlaB) and mouse (mIL15/FlaB) interleukin-15 proteins in the presence of L-arabinose (L-ara). These strains, named SAMphIF and SAMpmIF, respectively, secreted fusion proteins that retained bioactivity of both FlaB and IL15. SAMphIF and SAMpmIF inhibited the growth of MC38 and CT26 subcutaneous (sc) tumors in mice and increased mouse survival rate more efficiently than SAM expressing FlaB alone (SAMpFlaB) or IL15 alone (SAMpmIL15 and SAMphIL15), although SAMpmIF had slightly greater antitumor activity than SAMphIF. The mice treated with these bacteria showed enhanced macrophage phenotype shift, from M2-like to M1-like, as well as greater proliferation and activation of CD4+ T, CD8+ T, NK, and NKT cells in tumor tissues. After tumor eradication by these bacteria, ≥50% of the mice show no evidence of tumor recurrence upon rechallenge with the same tumor cells, indicating that they had acquired long-term immune memory. Treatment of mice of 4T1 and B16F10 highly malignant sc tumors with a combination of these bacteria and an immune checkpoint inhibitor, anti-PD-L1 antibody, significantly suppressed tumor metastasis and increased mouse survival rate. Taken together, these findings suggest that SAM secreting IL15/FlaB is a novel therapeutic candidate for bacterial-mediated cancer immunotherapy and that its antitumor activity is enhanced by combination with anti-PD-L1 antibody.
Collapse
Affiliation(s)
- Ying Zhang
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea; Department of Molecular Medicine (BrainKorea21 Plus), Chonnam National University Graduate School, Gwangju, 61469, Republic of Korea
| | - Wenzhi Tan
- School of Food Science and Bioengineering, Changsha University of Science & Technology, Changsha, Hunan, 410114, China
| | - Rukhsora D Sultonova
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea; Department of Molecular Medicine (BrainKorea21 Plus), Chonnam National University Graduate School, Gwangju, 61469, Republic of Korea
| | - Dinh-Huy Nguyen
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea; Department of Molecular Medicine (BrainKorea21 Plus), Chonnam National University Graduate School, Gwangju, 61469, Republic of Korea
| | - Jin Hai Zheng
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | | | - Joon Haeng Rhee
- Department of Microbiology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - So-Young Kim
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Koemchhoy Khim
- Department of Microbiology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Yeongjin Hong
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea; Department of Microbiology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.
| | - Jung-Joon Min
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea; Department of Molecular Medicine (BrainKorea21 Plus), Chonnam National University Graduate School, Gwangju, 61469, Republic of Korea; Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, 58128, Republic of Korea.
| |
Collapse
|
38
|
Gebauer J, Tesařík R, Králová N, Havlíčková H, Matiašovic J. Salmonella Typhimurium-based inactivated vaccine containing a wide spectrum of bacterial antigens which mimics protein expression changes during different stages of an infection process. Vet Microbiol 2023; 282:109756. [PMID: 37141806 DOI: 10.1016/j.vetmic.2023.109756] [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: 10/18/2022] [Revised: 03/21/2023] [Accepted: 04/22/2023] [Indexed: 05/06/2023]
Abstract
Salmonella infections are still considered a persistent problem in veterinary medicine. Vaccination is one of the tools for decreasing the burden of many pathogens on animals. However, the efficiency of available commercial or experimental vaccines against non-typhoid Salmonella strains is not yet sufficient. We followed the path of an inactivated vaccine that is safe and well accepted, but whose presented antigen spectrum is limited. We improved this issue by using diverse cultivation conditions mimicking bacterial protein expression during the natural infection process. The cultivation process was set up to simulate the host environment to enhance the expression of SPI-1 (Salmonella pathogenicity island) proteins, SPI-2 proteins, siderophore-related proteins, and flagellar proteins. Three different cultivation media were used and subsequent cultures were mixed together, inactivated, and used for the immunization of post-weaned piglets. A mixture of recombinant Salmonella proteins was also used as a recombinant vaccine for comparison. The clinical symptoms during the subsequent experimental infection, antibody response, and organ bacterial loads were examined. One day after the infection, we observed an increased rectal temperature in the group of unvaccinated animals and the animals vaccinated with the recombinant vaccine. The increase in the temperature of the pigs vaccinated with the inactivated Salmonella mixture was significantly lower. In the same group, we also found lower bacterial loads in the ileum content and the colon wall. The IgG response to several Salmonella antigens was enhanced in this group, but it did not reach the titers of the group vaccinated with the recombinant vaccine. To summarize, the pigs vaccinated with an inactivated mixture of Salmonella cultures mimicking protein expression changes during the natural infection exhibited less serious clinical symptoms and lower bacterial load in the body after the experimental infection compared to the unvaccinated pigs and the pigs vaccinated with a mixture of recombinant Salmonella proteins.
Collapse
Affiliation(s)
- Jan Gebauer
- Veterinary Research Institute, Hudcova 296/70, Brno, 62100, Czech Republic.
| | - Radek Tesařík
- Veterinary Research Institute, Hudcova 296/70, Brno, 62100, Czech Republic
| | - Natálie Králová
- Veterinary Research Institute, Hudcova 296/70, Brno, 62100, Czech Republic; Institute of Experimental Biology, Faculty of Science, Masaryk University, Kotlářská 2, Brno, 60200, Czech Republic
| | - Hana Havlíčková
- Veterinary Research Institute, Hudcova 296/70, Brno, 62100, Czech Republic
| | - Ján Matiašovic
- Veterinary Research Institute, Hudcova 296/70, Brno, 62100, Czech Republic
| |
Collapse
|
39
|
Rhee JH, Khim K, Puth S, Choi Y, Lee SE. Deimmunization of flagellin adjuvant for clinical application. Curr Opin Virol 2023; 60:101330. [PMID: 37084463 DOI: 10.1016/j.coviro.2023.101330] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 04/23/2023]
Abstract
Flagellin is the cognate ligand for host pattern recognition receptors, toll-like receptor 5 (TLR5) in the cell surface, and NAIP5/NLRC4 inflammasome in the cytosol. TLR5-binding domain is located in D1 domain, where crucial amino acid sequences are conserved among diverse bacteria. The highly conserved C-terminal 35 amino acids of flagellin were proved to be responsible for the inflammasome activation by binding to NAIP5. D2/D3 domains, located in the central region and exposed to the outside surface of flagellar filament, are heterogeneous across bacterial species and highly immunogenic. Taking advantage of TLR5- and NLRC4-stimulating activities, flagellin has been actively developed as a vaccine adjuvant and immunotherapeutic. Because of its immunogenicity, there exist worries concerning diminished efficacy and possible reactogenicity after repeated administration. Deimmunization of flagellin derivatives while preserving the TLR5/NLRC4-mediated immunomodulatory activity should be the most reasonable option for clinical application. This review describes strategies and current achievements in flagellin deimmunization.
Collapse
Affiliation(s)
- 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.
| | - 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
| | - Sao Puth
- 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
| | - Yoonjoo Choi
- 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
| | - 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
| |
Collapse
|
40
|
Kim J, Rosenberger MG, Chen S, IP CKM, Bahmani A, Chen Q, Shen J, Tang Y, Wang A, Kenna E, Son M, Tay S, Ferguson AL, Esser-Kahn AP. Discovery of New States of Immunomodulation for Vaccine Adjuvants via High Throughput Screening: Expanding Innate Responses to PRRs. ACS CENTRAL SCIENCE 2023; 9:427-439. [PMID: 36968540 PMCID: PMC10037445 DOI: 10.1021/acscentsci.2c01351] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Indexed: 06/18/2023]
Abstract
Stimulation of the innate immune system is crucial in both effective vaccinations and immunotherapies. This is often achieved through adjuvants, molecules that usually activate pattern recognition receptors (PRRs) and stimulate two innate immune signaling pathways: the nuclear factor kappa-light-chain-enhancer of activated B-cells pathway (NF-κB) and the interferon regulatory factors pathway (IRF). Here, we demonstrate the ability to alter and improve adjuvant activity via the addition of small molecule "immunomodulators". By modulating signaling activity instead of receptor binding, these molecules allow the customization of select innate responses. We demonstrate both inhibition and enhancement of the products of the NF-κB and IRF pathways by several orders of magnitude. Some modulators apply generally across many receptors, while others focus specifically on individual receptors. Modulators boost correlates of a protective immune responses in a commercial flu vaccine model and reduced correlates of reactogenicity in a typhoid vaccine model. These modulators have a range of applications: from adjuvanticity in prophylactics to enhancement of immunotherapy.
Collapse
Affiliation(s)
| | | | - Siquan Chen
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Carman KM IP
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Azadeh Bahmani
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Qing Chen
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Jinjing Shen
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Yifeng Tang
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Andrew Wang
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Emma Kenna
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Minjun Son
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Savaş Tay
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Andrew L. Ferguson
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| | - Aaron P. Esser-Kahn
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, Illinois 60637, United States
| |
Collapse
|
41
|
Desai N, Hasan U, K J, Mani R, Chauhan M, Basu SM, Giri J. Biomaterial-based platforms for modulating immune components against cancer and cancer stem cells. Acta Biomater 2023; 161:1-36. [PMID: 36907233 DOI: 10.1016/j.actbio.2023.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/02/2023] [Accepted: 03/02/2023] [Indexed: 03/12/2023]
Abstract
Immunotherapy involves the therapeutic alteration of the patient's immune system to identify, target, and eliminate cancer cells. Dendritic cells, macrophages, myeloid-derived suppressor cells, and regulatory T cells make up the tumor microenvironment. In cancer, these immune components (in association with some non-immune cell populations like cancer-associated fibroblasts) are directly altered at a cellular level. By dominating immune cells with molecular cross-talk, cancer cells can proliferate unchecked. Current clinical immunotherapy strategies are limited to conventional adoptive cell therapy or immune checkpoint blockade. Targeting and modulating key immune components presents an effective opportunity. Immunostimulatory drugs are a research hotspot, but their poor pharmacokinetics, low tumor accumulation, and non-specific systemic toxicity limit their use. This review describes the cutting-edge research undertaken in the field of nanotechnology and material science to develop biomaterials-based platforms as effective immunotherapeutics. Various biomaterial types (polymer-based, lipid-based, carbon-based, cell-derived, etc.) and functionalization methodologies for modulating tumor-associated immune/non-immune cells are explored. Additionally, emphasis has been laid on discussing how these platforms can be used against cancer stem cells, a fundamental contributor to chemoresistance, tumor relapse/metastasis, and failure of immunotherapy. Overall, this comprehensive review strives to provide up-to-date information to an audience working at the juncture of biomaterials and cancer immunotherapy. STATEMENT OF SIGNIFICANCE: Cancer immunotherapy possesses incredible potential and has successfully transitioned into a clinically lucrative alternative to conventional anti-cancer therapies. With new immunotherapeutics getting rapid clinical approval, fundamental problems associated with the dynamic nature of the immune system (like limited clinical response rates and autoimmunity-related adverse effects) have remained unanswered. In this context, treatment approaches that focus on modulating the compromised immune components within the tumor microenvironment have garnered significant attention amongst the scientific community. This review aims to provide a critical discussion on how various biomaterials (polymer-based, lipid-based, carbon-based, cell-derived, etc.) can be employed along with immunostimulatory agents to design innovative platforms for selective immunotherapy directed against cancer and cancer stem cells.
Collapse
Affiliation(s)
- Nimeet Desai
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Uzma Hasan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India; Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Jeyashree K
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Rajesh Mani
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Meenakshi Chauhan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Suparna Mercy Basu
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Jyotsnendu Giri
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, India.
| |
Collapse
|
42
|
Yi Y, Yu M, Li W, Zhu D, Mei L, Ou M. Vaccine-like nanomedicine for cancer immunotherapy. J Control Release 2023; 355:760-778. [PMID: 36822241 DOI: 10.1016/j.jconrel.2023.02.015] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/25/2023]
Abstract
The successful clinical application of immune checkpoint blockade (ICB) and chimeric antigen receptor T cells (CAR-T) therapeutics has attracted extensive attention to immunotherapy, however, their drawbacks such as limited specificity, persistence and toxicity haven't met the high expectations on efficient cancer treatments. Therapeutic cancer vaccines which instruct the immune system to capture tumor specific antigens, generate long-term immune memory and specifically eliminate cancer cells gradually become the most promising strategies to eradicate tumor. However, the disadvantages of some existing vaccines such as weak immunogenicity and in vivo instability have restricted their development. Nanotechnology has been recently incorporated into vaccine fabrication and exhibited promising results for cancer immunotherapy. Nanoparticles promote the stability of vaccines, as well as enhance antigen recognition and presentation owing to their nanometer size which promotes internalization of antigens by phagocytic cells. The surface modification with targeting units further permits the delivery of vaccines to specific cells. Meanwhile, nanocarriers with adjuvant effect can improve the efficacy of vaccines. In addition to classic vaccines composed of antigens and adjuvants, the nanoparticle-mediated chemotherapy, radiotherapy and certain other therapeutics could induce the release of tumor antigens in situ, which therefore effectively simulate antitumor immune responses. Such vaccine-like nanomedicine not only kills primary tumors, but also prevents tumor recurrence and helps eliminate metastatic tumors. Herein, we introduce recent developments in nanoparticle-based delivery systems for antigen delivery and in situ antitumor vaccination. We will also discuss the remaining opportunities and challenges of nanovaccine in clinical translation towards cancer treatment.
Collapse
Affiliation(s)
- Yunfei Yi
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China; School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Mian Yu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Wen Li
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Dunwan Zhu
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| | - Lin Mei
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| | - Meitong Ou
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| |
Collapse
|
43
|
Ou B, Yang Y, Lv H, Lin X, Zhang M. Current Progress and Challenges in the Study of Adjuvants for Oral Vaccines. BioDrugs 2023; 37:143-180. [PMID: 36607488 PMCID: PMC9821375 DOI: 10.1007/s40259-022-00575-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2022] [Indexed: 01/07/2023]
Abstract
Over the past 20 years, a variety of potential adjuvants have been studied to enhance the effect of oral vaccines in the intestinal mucosal immune system; however, no licensed adjuvant for clinical application in oral vaccines is available. In this review, we systematically updated the research progress of oral vaccine adjuvants over the past 2 decades, including biogenic adjuvants, non-biogenic adjuvants, and their multi-type composite adjuvant materials, and introduced their immune mechanisms of adjuvanticity, aiming at providing theoretical basis for developing feasible and effective adjuvants for oral vaccines. Based on these insights, we briefly discussed the challenges in the development of oral vaccine adjuvants and prospects for their future development.
Collapse
Affiliation(s)
- Bingming Ou
- School of Life Sciences, Zhaoqing University, Zhaoqing, China
| | - Ying Yang
- College of Animal Science, Guizhou University, Guiyang, China
| | - Haihui Lv
- School of Life Sciences, Zhaoqing University, Zhaoqing, China
| | - Xin Lin
- School of Life Sciences, Zhaoqing University, Zhaoqing, China
| | - Minyu Zhang
- School of Life Sciences, Zhaoqing University, Zhaoqing, China. .,School of Physical Education and Sports Science, South China Normal University, Guangzhou, China.
| |
Collapse
|
44
|
Jiang J, Huang Y, Zeng Z, Zhao C. Harnessing Engineered Immune Cells and Bacteria as Drug Carriers for Cancer Immunotherapy. ACS NANO 2023; 17:843-884. [PMID: 36598956 DOI: 10.1021/acsnano.2c07607] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Immunotherapy continues to be in the spotlight of oncology therapy research in the past few years and has been proven to be a promising option to modulate one's innate and adaptive immune systems for cancer treatment. However, the poor delivery efficiency of immune agents, potential off-target toxicity, and nonimmunogenic tumors significantly limit its effectiveness and extensive application. Recently, emerging biomaterial-based drug carriers, including but not limited to immune cells and bacteria, are expected to be potential candidates to break the dilemma of immunotherapy, with their excellent natures of intrinsic tumor tropism and immunomodulatory activity. More than that, the tiny vesicles and physiological components derived from them have similar functions with their source cells due to the inheritance of various surface signal molecules and proteins. Herein, we presented representative examples about the latest advances of biomaterial-based delivery systems employed in cancer immunotherapy, including immune cells, bacteria, and their derivatives. Simultaneously, opportunities and challenges of immune cells and bacteria-based carriers are discussed to provide reference for their future application in cancer immunotherapy.
Collapse
Affiliation(s)
- Jingwen Jiang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, People's Republic of China
| | - Yanjuan Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, People's Republic of China
| | - Zishan Zeng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, People's Republic of China
| | - Chunshun Zhao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, People's Republic of China
| |
Collapse
|
45
|
Zeng Y, Zou F, Xia N, Li S. In-depth review of delivery carriers associated with vaccine adjuvants: current status and future perspectives. Expert Rev Vaccines 2023; 22:681-695. [PMID: 37496496 DOI: 10.1080/14760584.2023.2238807] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 07/17/2023] [Indexed: 07/28/2023]
Abstract
INTRODUCTION Vaccines are powerful tools for controlling microbial infections and preventing epidemics. To enhance the immune response to antigens, effective subunit vaccines or mRNA vaccines often require the combination of adjuvants or delivery carriers. In recent years, with the rapid development of immune mechanism research and nanotechnology, various studies based on the optimization of traditional adjuvants or various novel carriers have been intensified, and the construction of vaccine adjuvant delivery systems (VADS) with both adjuvant activity and antigen delivery has become more and more important in vaccine research. AREAS COVERED This paper reviews the common types of vaccine adjuvant delivery carriers, classifies the VADS according to their basic carrier types, introduces the current research status and future development trend, and emphasizes the important role of VADS in novel vaccine research. EXPERT OPINION As the number of vaccine types increases, conventional aluminum adjuvants show limitations in effectively stimulating cellular immune responses, limiting their use in therapeutic vaccines for intracellular infections or tumors. In contrast, the use of conventional adjuvants as VADS to carry immunostimulatory molecules or deliver antigens can greatly enhance the immune boosting effect of classical adjuvants. A comprehensive understanding of the various delivery vehicles will further facilitate the development of vaccine adjuvant research.
Collapse
Affiliation(s)
- Yarong Zeng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, China
| | - Feihong Zou
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, China
- The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, China
| | - Shaowei Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, China
| |
Collapse
|
46
|
Wong-Benito V, Barraza F, Trujillo-Imarai A, Ruiz-Higgs D, Montero R, Sandino AM, Wang T, Maisey K, Secombes CJ, Imarai M. Infectious pancreatic necrosis virus (IPNV) recombinant viral protein 1 (VP1) and VP2-Flagellin fusion protein elicit distinct expression profiles of cytokines involved in type 1, type 2, and regulatory T cell response in rainbow trout (Oncorhynchus mykiss). FISH & SHELLFISH IMMUNOLOGY 2022; 131:785-795. [PMID: 36323384 DOI: 10.1016/j.fsi.2022.10.060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 06/16/2023]
Abstract
In this study, we examined the cytokine immune response against two proteins of infectious pancreatic necrosis virus (IPNV) in rainbow trout (Oncorhynchus mykiss), the virion-associated RNA polymerase VP1 and VP2-Flagellin (VP2-Flg) fusion protein. Since VP1 is not a structural protein, we hypothesize it can induce cellular immunity, an essential mechanism of the antiviral response. At the same time, the fusion construction VP2-Flg could be highly immunogenic due to the presence of the flagellin used as an adjuvant. Fish were immunized with the corresponding antigen in Montanide™, and the gene expression of a set of marker genes of Th1, Th2, and the immune regulatory response was quantified in the head kidney of immunized and control fish. Results indicate that VP1 induced upregulation of ifn-γ, il-12p40c, il-4/13a, il-4/13b2, il-10a, and tgf-β1 in immunized fish. Expression of il-2a did not change in treated fish at the times tested. The antigen-dependent response was analysed by in vitro restimulation of head kidney leukocytes. In this assay, the group of cytokines upregulated after VP1-restimulation was consistent with those upregulated in the head kidney in vivo. Interestingly, VP1 induced il-2a expression after in vitro restimulation. The analysis of sorted lymphocytes showed that the increase of cytokines occurred in CD4-1+ T cells suggesting that Th differentiation happens in response to VP1. This is also consistent with the expression of t-bet and gata3, the master regulators for Th1/Th2 differentiation in the kidneys of immunized animals. A different cytokine expression profile was found after VP2-Flg administration, i.e., upregulation occurs for ifn-γ, il-4/13a, il-10a, and tgf-β1, while down-regulation was observed in il-4/13b2 and il-2a. The cytokine response was due to flagellin; only the il-2a effect was dependent upon VP2 in the fusion protein. To the best of our knowledge this study reports for the first-time characteristics of the adaptive immune response induced in response to IPNV VP1 and the fusion protein VP2-Flg in fish. VP1 induces cytokines able to trigger the humoral and cell-mediated immune response in rainbow trout. The analysis of the fish response against VP2-Flg revealed the immunogenic properties of Aeromonas salmonicida flagellin, which can be further tested for adjuvanticity. The novel immunogenic effects of VP1 in rainbow trout open new opportunities for further IPNV vaccine development using this viral protein.
Collapse
Affiliation(s)
- Valentina Wong-Benito
- Laboratorio de Inmunología. Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Alameda, 3363, Santiago, Chile.
| | - Felipe Barraza
- Laboratorio de Inmunología. Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Alameda, 3363, Santiago, Chile.
| | - Agustín Trujillo-Imarai
- Laboratorio de Inmunología. Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Alameda, 3363, Santiago, Chile.
| | - Daniela Ruiz-Higgs
- Laboratorio de Inmunología. Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Alameda, 3363, Santiago, Chile.
| | - Ruth Montero
- Laboratorio de Inmunología Comparativa. Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Alameda, 3363, Santiago, Chile.
| | - Ana María Sandino
- Laboratorio de Virología. Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Alameda, 3363, Santiago, Chile.
| | - Tiehui Wang
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Aberdeen AB24 2TZ, United Kingdom.
| | - Kevin Maisey
- Laboratorio de Inmunología Comparativa. Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Alameda, 3363, Santiago, Chile.
| | - Christopher J Secombes
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Aberdeen AB24 2TZ, United Kingdom.
| | - Mónica Imarai
- Laboratorio de Inmunología. Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Alameda, 3363, Santiago, Chile.
| |
Collapse
|
47
|
Gong X, Gao Y, Shu J, Zhang C, Zhao K. Chitosan-Based Nanomaterial as Immune Adjuvant and Delivery Carrier for Vaccines. Vaccines (Basel) 2022; 10:1906. [PMID: 36423002 PMCID: PMC9696061 DOI: 10.3390/vaccines10111906] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/05/2022] [Accepted: 11/08/2022] [Indexed: 08/26/2023] Open
Abstract
With the support of modern biotechnology, vaccine technology continues to iterate. The safety and efficacy of vaccines are some of the most important areas of development in the field. As a natural substance, chitosan is widely used in numerous fields-such as immune stimulation, drug delivery, wound healing, and antibacterial procedures-due to its good biocompatibility, low toxicity, biodegradability, and adhesion. Chitosan-based nanoparticles (NPs) have attracted extensive attention with respect to vaccine adjuvants and delivery systems due to their excellent properties, which can effectively enhance immune responses. Here, we list the classifications and mechanisms of action of vaccine adjuvants. At the same time, the preparation methods of chitosan, its NPs, and their mechanism of action in the delivery system are introduced. The extensive applications of chitosan and its NPs in protein vaccines and nucleic acid vaccines are also introduced. This paper reviewed the latest research progress of chitosan-based NPs in vaccine adjuvant and drug delivery systems.
Collapse
Affiliation(s)
- Xiaochen Gong
- Institute of Nanobiomaterials and Immunology, School of Pharmaceutical Sciences & School of Life Science, Taizhou University, Taizhou 318000, China
- School of Medical Technology, Qiqihar Medical University, Qiqihar 161006, China
| | - Yuan Gao
- Institute of Nanobiomaterials and Immunology, School of Pharmaceutical Sciences & School of Life Science, Taizhou University, Taizhou 318000, China
| | - Jianhong Shu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
- Zhejiang Hom-Sun Biotechnology Co., Ltd., Shaoxing 312366, China
| | - Chunjing Zhang
- School of Medical Technology, Qiqihar Medical University, Qiqihar 161006, China
| | - Kai Zhao
- Institute of Nanobiomaterials and Immunology, School of Pharmaceutical Sciences & School of Life Science, Taizhou University, Taizhou 318000, China
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
- Zhejiang Hom-Sun Biotechnology Co., Ltd., Shaoxing 312366, China
| |
Collapse
|
48
|
Yang J, Liu MQ, Liu L, Li X, Xu M, Lin H, Liu S, Hu Y, Li B, Liu B, Li M, Sun Y, Chen YQ, Shi ZL, Yan H. A triple-RBD-based mucosal vaccine provides broad protection against SARS-CoV-2 variants of concern. Cell Mol Immunol 2022; 19:1279-1289. [PMID: 36220993 PMCID: PMC9552159 DOI: 10.1038/s41423-022-00929-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/12/2022] [Indexed: 11/06/2022] Open
Abstract
The rapid mutation and spread of SARS-CoV-2 variants urge the development of effective mucosal vaccines to provide broad-spectrum protection against the initial infection and thereby curb the transmission potential. Here, we designed a chimeric triple-RBD immunogen, 3Ro-NC, harboring one Delta RBD and two Omicron RBDs within a novel protein scaffold. 3Ro-NC elicits potent and broad RBD-specific neutralizing immunity against SARS-CoV-2 variants of concern. Notably, intranasal immunization with 3Ro-NC plus the mucosal adjuvant KFD (3Ro-NC + KFDi.n) elicits coordinated mucosal IgA and higher neutralizing antibody specificity (closer antigenic distance) against the Omicron variant. In Omicron-challenged human ACE2 transgenic mice, 3Ro-NC + KFDi.n immunization significantly reduces the tissue pathology in the lung and lowers the viral RNA copy numbers in both the lung (85.7-fold) and the nasal turbinate (13.6-fold). Nasal virologic control is highly correlated with RBD-specific secretory IgA antibodies. Our data show that 3Ro-NC plus KFD is a promising mucosal vaccine candidate for protection against SARS-CoV-2 Omicron infection, pathology and transmission potential.
Collapse
Affiliation(s)
- Jingyi Yang
- Vaccine and Immunology Research Center, Translational Medical Research Institute, Shanghai Public Health Clinical Center, Fudan University, 201508, Shanghai, China
| | - Mei-Qin Liu
- Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Lin Liu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Xian Li
- Vaccine and Immunology Research Center, Translational Medical Research Institute, Shanghai Public Health Clinical Center, Fudan University, 201508, Shanghai, China
- Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Mengxin Xu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Haofeng Lin
- Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Shuning Liu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Yunqi Hu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Bei Li
- Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
| | - Bowen Liu
- Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Min Li
- Vaccine and Immunology Research Center, Translational Medical Research Institute, Shanghai Public Health Clinical Center, Fudan University, 201508, Shanghai, China
| | - Ying Sun
- Aerosol Bio-Tech (Suzhou) Co., LTD, Suzhou, 215123, Jiangsu, China
| | - Yao-Qing Chen
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, Guangdong, China.
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.
| | - Zheng-Li Shi
- Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China.
| | - Huimin Yan
- Vaccine and Immunology Research Center, Translational Medical Research Institute, Shanghai Public Health Clinical Center, Fudan University, 201508, Shanghai, China.
| |
Collapse
|
49
|
Yang H, Qu Y, Gao Y, Sun S, Wu R, Wu J. Research Progress on the Correlation between the Intestinal Microbiota and Food Allergy. Foods 2022; 11:foods11182913. [PMID: 36141041 PMCID: PMC9498665 DOI: 10.3390/foods11182913] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/21/2022] Open
Abstract
The increasing incidence of food allergy is becoming a substantial public health concern. Increasing evidence suggests that alterations in the composition of the intestinal microbiota play a part in the development of food allergy. Additionally, the application of probiotics to correct gut microbiota imbalances and regulate food allergy has become a research hotspot. However, the mechanism by which the gut microbiota regulates food allergy and the efficacy of probiotics are still in the preliminary exploration stage, and there are no clear and specific conclusions. The aim of this review is to provide information regarding the immune mechanism underlying food allergy, the correlation between the intestinal microbiota and food allergy, a detailed description of causation, and mechanisms by which the intestinal microbiota regulates food allergy. Subsequently, we highlight how probiotics modulate the gut microbiome–immune axis to alleviate food allergy. This study will contribute to the dovetailing of bacterial therapeutics with immune system in allergic individuals to prevent food allergy and ameliorate food allergy symptoms.
Collapse
Affiliation(s)
| | | | | | | | - Rina Wu
- Correspondence: or ; Tel./Fax: +86-24-88487161
| | | |
Collapse
|
50
|
Nuñez-Ortiz N, Díaz-Rosales P, García JA, Serra CR, Enes P, Tafalla C, Gomez-Casado E. Immunostimulant properties of full-length and truncated Marinobacter algicola flagellins, and their effects against viral hemorrhagic septicemia virus (VHSV) in trout. FISH & SHELLFISH IMMUNOLOGY 2022; 128:695-702. [PMID: 35981702 DOI: 10.1016/j.fsi.2022.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 06/15/2023]
Abstract
Adjuvants that would help optimize fish vaccines against bacterial and viral pathogens are highly demanded by the aquaculture sector. Flagellin has been proposed as an immunostimulant and an adjuvant for more than a decade. However, the adjuvant ability of flagellins with hypervariable region deleted is still unclear in fish. In this study, we evaluated the immune-stimulating capacity of two recombinant flagellins, the wild-type flagellin F from Marinobacter algicola and a version with the hypervariable region deleted (FredV2), to induce the transcription of a wide range of immune genes using two rainbow trout cell lines: a monocyte/macrophage-cell line (RTS-11) and an epithelial cell line from intestine (RTgutGC). Additionally, we studied the capacity of both flagellins to limit the replication of viral hemorrhagic septicemia virus (VHSV) on the RTgutGC cell line. Our results demonstrated that both recombinant flagellins can significantly increase the transcription of IL-1β1, IL-6, and IL-8 in both cell lines. However, other cytokines such as IFNγ1, and TNFα or antimicrobial peptides such as hepcidin were induced by both flagellins in RTgutGC but not in RTS-11 cells. Furthermore, both flagellins were capable of reducing the replication of VHSV in RTgutGC cells. Although the immunostimulatory and the antiviral capacities exerted by F were slightly more potent than those obtained with FredV2, the effects were retained after losing the hypervariable region. Our results provide new information on the immunostimulating and antiviral capacities of flagellins that point to their potential as suitable adjuvants for the future optimization of vaccines in aquaculture.
Collapse
Affiliation(s)
- Noelia Nuñez-Ortiz
- Animal Health Research Center (CISA), INIA-CSIC, Valdeolmos-Alalpardo, 28130, Madrid, Spain; Department of Biotechnology, INIA-CSIC, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040, Madrid, Spain
| | - Patricia Díaz-Rosales
- Animal Health Research Center (CISA), INIA-CSIC, Valdeolmos-Alalpardo, 28130, Madrid, Spain
| | - Jose A García
- Animal Health Department, Faculty of Veterinary, Complutense University of Madrid (UCM), 28040, Madrid, Spain
| | - Claudia R Serra
- CIIMAR, Centro Interdisciplinar de Investigação Marinha e Ambiental, Av. General Norton de Matos s/n, 4450-208, Matosinhos, Portugal
| | - Paula Enes
- CIIMAR, Centro Interdisciplinar de Investigação Marinha e Ambiental, Av. General Norton de Matos s/n, 4450-208, Matosinhos, Portugal
| | - Carolina Tafalla
- Animal Health Research Center (CISA), INIA-CSIC, Valdeolmos-Alalpardo, 28130, Madrid, Spain
| | - Eduardo Gomez-Casado
- Department of Biotechnology, INIA-CSIC, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040, Madrid, Spain.
| |
Collapse
|