1
|
Singer M, Kandeel F, Husseiny MI. Salmonella-Based Vaccine: A Promising Strategy for Type 1 Diabetes. Vaccines (Basel) 2025; 13:405. [PMID: 40333284 PMCID: PMC12031388 DOI: 10.3390/vaccines13040405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 05/09/2025] Open
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease characterized by the progressive destruction of insulin-producing β-cells in the pancreas. Currently, no therapy exists to halt or cure T1D. Vaccination with diabetic autoantigens may offer protection against T1D development. Genetically modified, attenuated Salmonella utilizing the Salmonella-Pathogenicity Island 2 (SPI2)-encoded Type Three Secretion System (T3SS) can elicit robust immune responses, making it an attractive vaccine platform. Using SPI2-T3SS to deliver an autoantigen alongside immunomodulators and anti-CD3 antibodies induces antigen-specific regulatory T-cells. Our preclinical studies demonstrated the efficacy of a Salmonella-based vaccine in both preventing and reversing autoimmune diabetes in non-obese diabetic (NOD) mice while also exploring its genetic modifications, underlying mechanisms, and delivery strategies. This review evaluates the advantages of an oral T1D vaccine employing live, attenuated Salmonella for autoantigen delivery. We also discuss future directions for advancing this strategy in the treatment of other autoimmune diseases.
Collapse
Affiliation(s)
- Mahmoud Singer
- Department of Radiological Sciences, School of Medicine, University of California Irvine, Irvine, CA 92697, USA
| | - Fouad Kandeel
- Department of Translational Research and Cellular Therapeutics, Artur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Mohamed I. Husseiny
- Department of Translational Research and Cellular Therapeutics, Artur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
2
|
Liu Y, Gong H, Zhu J, Liu F. Oral Vaccination with Attenuated Salmonella Expressing Viral M25 Protein Effectively Protects Mice Against Murine Cytomegalovirus Infection. Pathogens 2025; 14:314. [PMID: 40333046 PMCID: PMC12030445 DOI: 10.3390/pathogens14040314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 05/09/2025] Open
Abstract
Attenuated Salmonella strains are promising oral vectors for vaccination against human infectious diseases. Human cytomegalovirus (CMV) is among the most common causes of disability in children, including intellectual disability and sensorineural hearing loss. Developing an anti-CMV vaccine is a major public health priority. We report in this study the construction of a new attenuated Salmonella strain to express murine cytomegalovirus (MCMV) M25 protein and its use for vaccination in mice against MCMV infection. In mice orally vaccinated with the constructed Salmonella vector carrying the M25 expression cassette, we revealed a substantial induction of anti-MCMV serum IgG and mucosal IgA humoral responses and a considerable elicitation of anti-MCMV T cell responses. When the vaccinated mice were challenged intraperitoneally and intranasally with MCMV, we observed a significant inhibition of virus infection and growth in various organs including spleens, livers, lungs, and salivary glands, compared to the non-vaccinated animals or those receiving a control vaccine without M25 protein expression. Moreover, we showed effective protection of these vaccinated mice from MCMV challenge. Our study provides the first direct evidence that an attenuated Salmonella-based vector with the MCMV M25 expression cassette can induce strong humoral and T cell responses and provide effective protection against MCMV infection. These results illustrate the feasibility of engineering Salmonella-based vectors expressing the M25 antigen for anti-CMV oral vaccine development.
Collapse
Affiliation(s)
- Yujun Liu
- School of Public Health, University of California, Berkeley, CA 94720, USA
| | - Hao Gong
- School of Public Health, University of California, Berkeley, CA 94720, USA
| | - Jiaming Zhu
- Program in Comparative Biochemistry, University of California, Berkeley, CA 94720, USA
| | - Fenyong Liu
- School of Public Health, University of California, Berkeley, CA 94720, USA
- Program in Comparative Biochemistry, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
3
|
Liu Y, Gong H, Zhu J, Liu F. Effective Immune Protection of Mice from Murine Cytomegalovirus Infection by Oral Salmonella-Based Vaccine Expressing Viral M78 Antigen. Vaccines (Basel) 2025; 13:137. [PMID: 40006684 PMCID: PMC11861581 DOI: 10.3390/vaccines13020137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Human cytomegalovirus (CMV) is the most common cause of viral congenital infections worldwide. The development of effective vaccines against human CMV infection and disease is a high priority. Attenuated Salmonella are attractive oral vaccine vectors against human diseases because they can be administrated orally. Methods: In this study, an attenuated Salmonella strain was generated as an oral vaccine vector for the delivery and expression of the M78 protein of murine cytomegalovirus (MCMV). Using the MCMV infection of mice as the CMV infection model, we characterized the immune responses and protection induced by the constructed Salmonella-based vaccine. Results: The generated Salmonella-based vaccine, v-M78, which contained an M78 expression plasmid construct, carried out gene transfer efficiently for M78 expression and showed little pathogenicity and virulence in mice. In orally vaccinated mice, v-M78 induced anti-MCMV serum IgG and mucosal IgA responses and also elicited anti-MCMV T cell responses. Furthermore, mice immunized with v-M78 were protected from intraperitoneal and intranasal challenges with MCMV. The v-M78 vaccination reduced the titers of the challenged viruses in spleens, livers, lungs, and salivary glands. Conclusions: These results provide the first direct evidence that a Salmonella-based vaccine expressing M78 elicits strong humoral and cellular immune responses and induces immune protection against MCMV infection. Furthermore, our study demonstrates the potential of using Salmonella-based oral vaccines against CMV infection.
Collapse
Affiliation(s)
- Yujun Liu
- School of Public Health, University of California, Berkeley, CA 94720, USA
| | - Hao Gong
- School of Public Health, University of California, Berkeley, CA 94720, USA
| | - Jiaming Zhu
- School of Public Health, University of California, Berkeley, CA 94720, USA
| | - Fenyong Liu
- School of Public Health, University of California, Berkeley, CA 94720, USA
- Program in Comparative Biochemistry, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
4
|
Yan S, Gan Y, Xu H, Piao H. Bacterial carrier-mediated drug delivery systems: a promising strategy in cancer therapy. Front Bioeng Biotechnol 2025; 12:1526612. [PMID: 39845371 PMCID: PMC11750792 DOI: 10.3389/fbioe.2024.1526612] [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: 11/12/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025] Open
Abstract
Cancer is a major killer threatening modern human health and a leading cause of death worldwide. Due to the heterogeneity and complexity of cancer, traditional treatments have limited effectiveness. To address this problem, an increasing number of researchers and medical professionals are working to develop new ways to treat cancer. Bacteria have chemotaxis that can target and colonize tumor tissue, as well as activate anti-tumor immune responses, which makes them ideal for biomedical applications. With the rapid development of nanomedicine and synthetic biology technologies, bacteria are extensively used as carriers for drug delivery to treat tumors, which holds the promise of overcoming the limitations of conventional cancer treatment regimens. This paper summarizes examples of anti-cancer drugs delivered by bacterial carriers, and their strengths and weaknesses. Further, we emphasize the promise of bacterial carrier delivery systems in clinical translation.
Collapse
Affiliation(s)
- Sizuo Yan
- Department of Neurosurgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Shenyang, China
| | - Yu Gan
- Department of Neurosurgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Shenyang, China
| | - Huizhe Xu
- Institute of Cancer Medicine, Dalian University of Technology, Dalian, China
- Central Laboratory, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Shenyang, China
| | - Haozhe Piao
- Department of Neurosurgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Shenyang, China
- Institute of Cancer Medicine, Dalian University of Technology, Dalian, China
| |
Collapse
|
5
|
Singer ZS, Pabón J, Huang H, Sun W, Luo H, Grant KR, Obi I, Coker C, Rice CM, Danino T. Engineered bacteria launch and control an oncolytic virus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.09.28.559873. [PMID: 37808855 PMCID: PMC10557668 DOI: 10.1101/2023.09.28.559873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
The ability of bacteria and viruses to selectively replicate in tumors has led to synthetic engineering of new microbial therapies. Here we design a cooperative strategy whereby S. typhimurium bacteria transcribe and deliver the Senecavirus A RNA genome inside host cells, launching a potent oncolytic viral infection. "Encapsidated" by bacteria, the viral genome can further bypass circulating antiviral antibodies to reach the tumor and initiate replication and spread within immune mice. Finally, we engineer the virus to require a bacterially delivered protease to achieve virion maturation, demonstrating bacterial control over the virus. This work extends bacterially delivered therapeutics to viral genomes, and shows how a consortium of microbes can achieve a cooperative aim.
Collapse
|
6
|
Heggie A, Thurston TLM, Ellis T. Microbial messengers: nucleic acid delivery by bacteria. Trends Biotechnol 2025; 43:145-161. [PMID: 39117490 DOI: 10.1016/j.tibtech.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024]
Abstract
The demand for diverse nucleic acid delivery vectors, driven by recent biotechnological breakthroughs, offers opportunities for continuous improvements in efficiency, safety, and delivery capacity. With their enhanced safety and substantial cargo capacity, bacterial vectors offer significant potential across a variety of applications. In this review, we explore methods to engineer bacteria for nucleic acid delivery, including strategies such as engineering attenuated strains, lysis circuits, and conjugation machinery. Moreover, we explore pioneering techniques, such as manipulating nanoparticle (NP) coatings and outer membrane vesicles (OMVs), representing the next frontier in bacterial vector engineering. We foresee these advancements in bacteria-mediated nucleic acid delivery, through combining bacterial pathogenesis with engineering biology techniques, as a pivotal step forward in the evolution of nucleic acid delivery technologies.
Collapse
Affiliation(s)
- Alison Heggie
- Centre for Bacterial Resistance Biology, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK; Imperial College Centre for Synthetic Biology, South Kensington Campus, London, SW7 2AZ, UK; Department of Bioengineering, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Teresa L M Thurston
- Centre for Bacterial Resistance Biology, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Tom Ellis
- Imperial College Centre for Synthetic Biology, South Kensington Campus, London, SW7 2AZ, UK; Department of Bioengineering, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK.
| |
Collapse
|
7
|
Eng SW, Muniandy V, Punniamoorthy L, Tew HX, Norazmi MN, Ravichandran M, Lee SY. Live Attenuated Bacterial Vectors as Vehicles for DNA Vaccine Delivery: A Mini Review. Malays J Med Sci 2024; 31:6-20. [PMID: 39830112 PMCID: PMC11740808 DOI: 10.21315/mjms2024.31.6.2] [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: 06/23/2024] [Accepted: 08/16/2024] [Indexed: 01/22/2025] Open
Abstract
DNA vaccines are third-generation vaccines composed of plasmids that encode vaccine antigens. Their advantages include fast development, safety, stability, and cost effectiveness, which make them an attractive vaccine platform for genetic and infectious diseases. However, the low transfection efficiency of DNA vaccines results in poor performance in both larger animals and humans, thereby limiting their clinical use. To overcome this issue, live attenuated bacterial vector (LABV) has been proposed as a DNA delivery vehicle. LABV is known to improve DNA vaccine transfection efficiency, thus enhancing the immune response. This article highlights recent advancements in the development of LABV DNA vaccines, the design of shuttle plasmids and adjuvants, and the potential applications of LABV candidates.
Collapse
Affiliation(s)
- Sze Wei Eng
- Faculty of Applied Sciences, AIMST University, Kedah, Malaysia
- Centre of Excellence for Vaccine Development (CoEVD), Faculty of Applied Science, AIMST University, Kedah, Malaysia
| | - Vilassini Muniandy
- Faculty of Applied Sciences, AIMST University, Kedah, Malaysia
- Centre of Excellence for Vaccine Development (CoEVD), Faculty of Applied Science, AIMST University, Kedah, Malaysia
| | - Lohshinni Punniamoorthy
- Faculty of Applied Sciences, AIMST University, Kedah, Malaysia
- Centre of Excellence for Vaccine Development (CoEVD), Faculty of Applied Science, AIMST University, Kedah, Malaysia
| | - Hui Xian Tew
- Faculty of Applied Sciences, AIMST University, Kedah, Malaysia
- Centre of Excellence for Vaccine Development (CoEVD), Faculty of Applied Science, AIMST University, Kedah, Malaysia
| | - Mohd Nor Norazmi
- School of Health Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
- Malaysia Genome and Vaccine Institute, National Institutes of Biotechnology Malaysia, Selangor, Malaysia
| | - Manickam Ravichandran
- Faculty of Applied Sciences, AIMST University, Kedah, Malaysia
- MyGenome Sdn Bhd, Kuala Lumpur, Malaysia
| | - Su Yin Lee
- Faculty of Applied Sciences, AIMST University, Kedah, Malaysia
- Centre of Excellence for Vaccine Development (CoEVD), Faculty of Applied Science, AIMST University, Kedah, Malaysia
| |
Collapse
|
8
|
Zhang T, Tian Y, Zhang X, Wang W, He Y, Ge C, Jia F, Wang Z, Jiang Y. Improved cellular immune response induced by intranasal boost immunization with chitosan coated DNA vaccine against H9N2 influenza virus challenge. Microb Pathog 2024; 195:106871. [PMID: 39163919 DOI: 10.1016/j.micpath.2024.106871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 08/13/2024] [Accepted: 08/17/2024] [Indexed: 08/22/2024]
Abstract
The H9N2 avian influenza virus (AIV) is spreading worldwide. Presence of H9N2 virus tends to increase the chances of infection with other pathogens which can lead to more serious economic losses. In a previous study, a regulated delayed lysis Salmonella vector was used to deliver a DNA vaccine named pYL233 encoding M1 protein, mosaic HA protein and chicken GM-CSF adjuvant. To further increase its efficiency, chitosan as a natural adjuvant was applied in this study. The purified plasmid pYL233 was coated with chitosan to form a DNA containing nanoparticles (named CS233) by ionic gel method and immunized by intranasal boost immunization in birds primed by oral administration with Salmonella strain. The CS233 DNA nanoparticle has a particle size of about 150 nm, with an encapsulation efficiency of 93.2 ± 0.12 % which protected the DNA plasmid from DNase I digestion and could be stable for a period of time at 37°. After intranasal boost immunization, the CS233 immunized chickens elicited higher antibody response, elevated CD4+ T cells and CD8+ T cells activation and increased T-lymphocyte proliferation, as well as increased productions of IL-4 and IFN-γ. After challenge, chickens immunized with CS233 resulted in the lowest levels of pulmonary virus titer and viral shedding as compared to the other challenge groups. The results showed that the combination of intranasal immunization with chitosan-coated DNA vaccine and oral immunization with regulatory delayed lytic Salmonella strain could enhance the immune response and able to provide protection against H9N2 challenge.
Collapse
MESH Headings
- Animals
- Influenza A Virus, H9N2 Subtype/immunology
- Influenza A Virus, H9N2 Subtype/genetics
- Vaccines, DNA/immunology
- Vaccines, DNA/administration & dosage
- Chitosan
- Administration, Intranasal
- Influenza in Birds/prevention & control
- Influenza in Birds/immunology
- Chickens/immunology
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Antibodies, Viral/blood
- Immunity, Cellular
- Virus Shedding
- Plasmids/genetics
- Nanoparticles
- Immunization, Secondary
- CD8-Positive T-Lymphocytes/immunology
- Adjuvants, Immunologic/administration & dosage
- Interferon-gamma
- Interleukin-4
- Adjuvants, Vaccine
- Poultry Diseases/prevention & control
- Poultry Diseases/immunology
- Poultry Diseases/virology
- CD4-Positive T-Lymphocytes/immunology
- Salmonella/immunology
- Salmonella/genetics
Collapse
Affiliation(s)
- Tongyu Zhang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Yawen Tian
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Xiao Zhang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Wenfeng Wang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Yingkai He
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Chongbo Ge
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Futing Jia
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Zhannan Wang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| | - Yanlong Jiang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
9
|
Lloren KKS, Sivasankar C, Lee JH. Comparative immunogenic and immunoprotective activities of PCV2d Cap and Rep antigens delivered by an efficient eukaryotic expression system engineered into a Salmonella vaccine vector. Vet Microbiol 2024; 295:110151. [PMID: 38870752 DOI: 10.1016/j.vetmic.2024.110151] [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: 03/07/2024] [Revised: 06/05/2024] [Accepted: 06/09/2024] [Indexed: 06/15/2024]
Abstract
Porcine circovirus type 2 (PCV2) stands as a predominant etiological agent in porcine circovirus-associated diseases. To manage the spread of the disease, it is necessary to develop a next-generation vaccine expressing PCV2 antigens that target the prevailing genotype such as PCV2d. A bacterial-mediated vaccine delivery by live-attenuated Salmonella has attracted interest for its low-cost production and highly effective vaccine delivery. Thus, in this study, we utilized the advantages of the Salmonella-mediated vaccine delivery by cloning PCV2d cap and rep into a eukaryotic expression plasmid pJHL204 and electroporation into an engineered live-attenuated Salmonella Typhimurium JOL2500 (Δlon, ΔcpxR, ΔsifA, Δasd). The eukaryotic antigen expression by JOL2995 (p204:cap) and JOL2996 (p204:rep) was confirmed in vitro and in vivo which showed efficient antigen delivery. Furthermore, vaccination of mice model with the vaccine candidates elicited humoral and cell-mediated immune responses as depicted by high levels of PCV2-specific antibodies, CD4+ and CD8+ T cells, and neutralizing antibodies, especially by JOL2995 (p204:cap) which correlated with the significant decrease in the viral load in PCV2d-challenged mice. Interestingly, JOL2996 (p204:rep) may not have elicited high levels of neutralizing antibodies and protective efficacy, but it elicited considerably higher cell-mediated immune responses. This study demonstrated Salmonella-mediated vaccine delivery system coupled with the eukaryotic expression vector can efficiently deliver and express the target PCV2d antigens for strong induction of immune response and protective efficacy in mice model, further supporting the potential application of the Salmonella-mediated vaccine delivery system as an effective novel approach in vaccine strategies for PCV2d.
Collapse
Affiliation(s)
- Khristine Kaith S Lloren
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeollabuk-do 54596, Republic of Korea
| | - Chandran Sivasankar
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeollabuk-do 54596, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeollabuk-do 54596, Republic of Korea.
| |
Collapse
|
10
|
Zhou Y, Li Q, Wu Y, Zhang W, Ding L, Ji C, Li P, Chen T, Feng L, Tang BZ, Huang X. Synergistic Brilliance: Engineered Bacteria and Nanomedicine Unite in Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313953. [PMID: 38400833 DOI: 10.1002/adma.202313953] [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: 12/20/2023] [Revised: 02/21/2024] [Indexed: 02/26/2024]
Abstract
Engineered bacteria are widely used in cancer treatment because live facultative/obligate anaerobes can selectively proliferate at tumor sites and reach hypoxic regions, thereby causing nutritional competition, enhancing immune responses, and producing anticancer microbial agents in situ to suppress tumor growth. Despite the unique advantages of bacteria-based cancer biotherapy, the insufficient treatment efficiency limits its application in the complete ablation of malignant tumors. The combination of nanomedicine and engineered bacteria has attracted increasing attention owing to their striking synergistic effects in cancer treatment. Engineered bacteria that function as natural vehicles can effectively deliver nanomedicines to tumor sites. Moreover, bacteria provide an opportunity to enhance nanomedicines by modulating the TME and producing substrates to support nanomedicine-mediated anticancer reactions. Nanomedicine exhibits excellent optical, magnetic, acoustic, and catalytic properties, and plays an important role in promoting bacteria-mediated biotherapies. The synergistic anticancer effects of engineered bacteria and nanomedicines in cancer therapy are comprehensively summarized in this review. Attention is paid not only to the fabrication of nanobiohybrid composites, but also to the interpromotion mechanism between engineered bacteria and nanomedicine in cancer therapy. Additionally, recent advances in engineered bacteria-synergized multimodal cancer therapies are highlighted.
Collapse
Affiliation(s)
- Yaofeng Zhou
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Qianying Li
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Yuhao Wu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Wan Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, P. R. China
| | - Lu Ding
- Department of Cardiology, Jiangxi Hypertension Research Institute, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, P. R. China
| | - Chenlin Ji
- School of Engineering, Westlake University, Hangzhou, 310030, P. R. China
| | - Ping Li
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| | - Tingtao Chen
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330036, P. R. China
| | - Lili Feng
- Key Laboratory of Superlight Materials and Surface Technology Ministry of Education, College of Material Sciences and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Ben Zhong Tang
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, 518172, P. R. China
| | - Xiaolin Huang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang, 330047, P. R. China
| |
Collapse
|
11
|
Hahn J, Ding S, Im J, Harimoto T, Leong KW, Danino T. Bacterial therapies at the interface of synthetic biology and nanomedicine. NATURE REVIEWS BIOENGINEERING 2024; 2:120-135. [PMID: 38962719 PMCID: PMC11218715 DOI: 10.1038/s44222-023-00119-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/08/2023] [Indexed: 07/05/2024]
Abstract
Bacteria are emerging as living drugs to treat a broad range of disease indications. However, the inherent advantages of these replicating and immunostimulatory therapies also carry the potential for toxicity. Advances in synthetic biology and the integration of nanomedicine can address this challenge through the engineering of controllable systems that regulate spatial and temporal activation for improved safety and efficacy. Here, we review recent progress in nanobiotechnology-driven engineering of bacteria-based therapies, highlighting limitations and opportunities that will facilitate clinical translation.
Collapse
Affiliation(s)
- Jaeseung Hahn
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Suwan Ding
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Jongwon Im
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Tetsuhiro Harimoto
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Kam W. Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Systems Biology, Columbia University Medical Center, New York, NY, USA
| | - Tal Danino
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
- Data Science Institute, Columbia University, New York, NY, USA
| |
Collapse
|
12
|
Liu K, Li Z, Li Q, Wang S, Curtiss R, Shi H. Salmonella typhimurium Vaccine Candidate Delivering Infectious Bronchitis Virus S1 Protein to Induce Protection. Biomolecules 2024; 14:133. [PMID: 38275762 PMCID: PMC10813627 DOI: 10.3390/biom14010133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/18/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Infectious bronchitis (IB) is a highly infectious viral disease of chickens which causes significant economic losses in the poultry industry worldwide. An effective vaccine against IB is urgently needed to provide both biosafety and high-efficiency immune protection. In this study, the S1 protein of the infectious bronchitis virus was delivered by a recombinant attenuated Salmonella typhimurium vector to form the vaccine candidate χ11246(pYA4545-S1). S. typhimurium χ11246 carried a sifA- mutation with regulated delayed systems, striking a balance between host safety and immunogenicity. Here, we demonstrated that S1 protein is highly expressed in HD11 cells. Immunization with χ11246(pYA4545-S1) induced the production of antibody and cytokine, leading to an effective immune response against IB. Oral immunization with χ11246(pYA4545-S1) provided 72%, 56%, and 56% protection in the lacrimal gland, trachea, and cloaca against infectious bronchitis virus infection, respectively. Furthermore, it significantly reduced histopathological lesions in chickens. Together, this study provides a new idea for the prevention of IB.
Collapse
Affiliation(s)
- Kaihui Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (K.L.); (Z.L.); (Q.L.)
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Zewei Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (K.L.); (Z.L.); (Q.L.)
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Quan Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (K.L.); (Z.L.); (Q.L.)
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Shifeng Wang
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA; (S.W.)
| | - Roy Curtiss
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA; (S.W.)
| | - Huoying Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (K.L.); (Z.L.); (Q.L.)
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University (JIRLAAPS), Yangzhou 225009, China
| |
Collapse
|
13
|
Li Y, Sun Y, Zhang Y, Li Q, Wang S, Curtiss R, Shi H. A Bacterial mRNA-Lysis-Mediated Cargo Release Vaccine System for Regulated Cytosolic Surveillance and Optimized Antigen Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303568. [PMID: 37867213 PMCID: PMC10667801 DOI: 10.1002/advs.202303568] [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: 06/01/2023] [Revised: 09/14/2023] [Indexed: 10/24/2023]
Abstract
Engineered vector-based in vivo protein delivery platforms have made significant progress for both prophylactic and therapeutic applications. However, the lack of effective release strategies results in foreign cargo being trapped within the vector, restricting the provision of significant performance benefits and enhanced therapeutic results compared to traditional vaccines. Herein, the development of a Salmonella mRNA interferase regulation vector (SIRV) system is reported to overcome this challenge. The genetic circuits are engineered that (1) induce self-lysis to release foreign antigens into target cells and (2) activate the cytosolic surveillance cGAS-STING axis by releasing DNA into the cytoplasm. Delayed synthesis of the MazF interferase regulates differential mRNA cleavage, resulting in a 36-fold increase in the delivery of foreign antigens and modest activation of the inflammasome, which collectively contribute to the marked maturation of antigen-presenting cells (APCs). Bacteria delivering the protective antigen SaoA exhibits excellent immunogenicity and safety in mouse and pig models, significantly improving the survival rate of animals challenged with multiple serotypes of Streptococcus suis. Thus, the SIRV system enables the effective integration of various modular components and antigen cargos, allowing for the generation of an extensive range of intracellular protein delivery systems using multiple bacterial species in a highly efficient manner.
Collapse
Affiliation(s)
- Yu‐an Li
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225000China
- Jiangsu Co‐innovation Center for the Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou225000China
| | - Yanni Sun
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225000China
- Jiangsu Co‐innovation Center for the Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou225000China
| | - Yuqin Zhang
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225000China
- Jiangsu Co‐innovation Center for the Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou225000China
| | - Quan Li
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225000China
- Jiangsu Co‐innovation Center for the Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou225000China
| | - Shifeng Wang
- Department of Infectious Diseases and ImmunologyCollege of Veterinary MedicineUniversity of FloridaGainesvilleFL32611‐0880USA
| | - Roy Curtiss
- Department of Infectious Diseases and ImmunologyCollege of Veterinary MedicineUniversity of FloridaGainesvilleFL32611‐0880USA
| | - Huoying Shi
- College of Veterinary MedicineYangzhou UniversityYangzhouJiangsu225000China
- Jiangsu Co‐innovation Center for the Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou225000China
- Joint International Research Laboratory of Agriculture and Agri‐Product SafetyYangzhou University (JIRLAAPS)Yangzhou225000China
| |
Collapse
|
14
|
Raman V, Deshpande CP, Khanduja S, Howell LM, Van Dessel N, Forbes NS. Build-a-bug workshop: Using microbial-host interactions and synthetic biology tools to create cancer therapies. Cell Host Microbe 2023; 31:1574-1592. [PMID: 37827116 DOI: 10.1016/j.chom.2023.09.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/16/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023]
Abstract
Many systemically administered cancer therapies exhibit dose-limiting toxicities that reduce their effectiveness. To increase efficacy, bacterial delivery platforms have been developed that improve safety and prolong treatment. Bacteria are a unique class of therapy that selectively colonizes most solid tumors. As delivery vehicles, bacteria have been genetically modified to express a range of therapies that match multiple cancer indications. In this review, we describe a modular "build-a-bug" method that focuses on five design characteristics: bacterial strain (chassis), therapeutic compound, delivery method, immune-modulating features, and genetic control circuits. We emphasize how fundamental research into gut microbe pathogenesis has created safe bacterial therapies, some of which have entered clinical trials. The genomes of gut microbes are fertile grounds for discovery of components to improve delivery and modulate host immune responses. Future work coupling these delivery vehicles with insights from gut microbes could lead to the next generation of microbial cancer therapy.
Collapse
Affiliation(s)
- Vishnu Raman
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA; Ernest Pharmaceuticals, LLC, Hadley, MA, USA
| | - Chinmay P Deshpande
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA
| | - Shradha Khanduja
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA
| | - Lars M Howell
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA
| | | | - Neil S Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA; Molecular and Cell Biology Program, University of Massachusetts, Amherst, Amherst, MA, USA; Institute for Applied Life Science, University of Massachusetts, Amherst, Amherst, MA, USA.
| |
Collapse
|
15
|
Wang Z, Zhang T, Jia F, Ge C, He Y, Tian Y, Wang W, Yang G, Huang H, Wang J, Shi C, Yang W, Cao X, Zeng Y, Wang N, Qian A, Wang C, Jiang Y. Homologous Sequential Immunization Using Salmonella Oral Administration Followed by an Intranasal Boost with Ferritin-Based Nanoparticles Enhanced the Humoral Immune Response against H1N1 Influenza Virus. Microbiol Spectr 2023; 11:e0010223. [PMID: 37154735 PMCID: PMC10269571 DOI: 10.1128/spectrum.00102-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023] Open
Abstract
The influenza virus continues to pose a great threat to public health due to the frequent variations in RNA viruses. Vaccines targeting conserved epitopes, such as the extracellular domain of the transmembrane protein M2 (M2e), a nucleoprotein, and the stem region of hemagglutinin proteins, have been developed, but more efficient strategies, such as nanoparticle-based vaccines, are still urgently needed. However, the labor-intensive in vitro purification of nanoparticles is still necessary, which could hinder the application of nanoparticles in the veterinary field in the future. To overcome this limitation, we used regulated lysis Salmonella as an oral vector with which to deliver three copies of M2e (3M2e-H1N1)-ferritin nanoparticles in situ and evaluated the immune response. Then, sequential immunization using Salmonella-delivered nanoparticles followed by an intranasal boost with purified nanoparticles was performed to further improve the efficiency. Compared with 3M2e monomer administration, Salmonella-delivered in situ nanoparticles significantly increased the cellular immune response. Additionally, the results of sequential immunization showed that the intranasal boost with purified nanoparticles dramatically stimulated the activation of lung CD11b dendritic cells (DCs) and elevated the levels of effector memory T (TEM) cells in both spleen and lung tissues as well as those of CD4 and CD8 tissue-resident memory T (TRM) cells in the lungs. The increased production of mucosal IgG and IgA antibody titers was also observed, resulting in further improvements to protection against a virus challenge, compared with the pure oral immunization group. Salmonella-delivered in situ nanoparticles efficiently increased the cellular immune response, compared with the monomer, and sequential immunization further improved the systemic immune response, as shown by the activation of DCs, the production of TEM cells and TRM cells, and the mucosal immune response, thereby providing us with a novel strategy by which to apply nanoparticle-based vaccines in the future. IMPORTANCE Salmonella-delivered in situ nanoparticle platforms may provide novel nanoparticle vaccines for oral administration, which would be beneficial for veterinary applications. The combination of administering Salmonella-vectored, self-assembled nanoparticles and an intranasal boost with purified nanoparticles significantly increased the production of effector memory T cells and lung resident memory T cells, thereby providing partial protection against an influenza virus challenge. This novel strategy could open a novel avenue for the application of nanoparticle vaccines for veterinary purposes.
Collapse
Affiliation(s)
- Zhannan Wang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Tongyu Zhang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Futing Jia
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chongbo Ge
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yingkai He
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yawen Tian
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Wenfeng Wang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Guilian Yang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Haibin Huang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Jianzhong Wang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chunwei Shi
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Wentao Yang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Xin Cao
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yan Zeng
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Nan Wang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Aidong Qian
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chunfeng Wang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yanlong Jiang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| |
Collapse
|
16
|
Cobb J, Soliman SSM, Retuerto M, Quijano JC, Orr C, Ghannoum M, Kandeel F, Husseiny MI. Changes in the gut microbiota of NOD mice in response to an oral Salmonella-based vaccine against type 1 diabetes. PLoS One 2023; 18:e0285905. [PMID: 37224176 DOI: 10.1371/journal.pone.0285905] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 05/03/2023] [Indexed: 05/26/2023] Open
Abstract
We developed an oral Salmonella-based vaccine that prevents and reverses diabetes in non-obese diabetic (NOD) mice. Related to this, the gastrointestinal tract harbors a complex dynamic population of microorganisms, the gut microbiome, that influences host homeostasis and metabolism. Changes in the gut microbiome are associated with insulin dysfunction and type 1 diabetes (T1D). Oral administration of diabetic autoantigens as a vaccine can restore immune balance. However, it was not known if a Salmonella-based vaccine would impact the gut microbiome. We administered a Salmonella-based vaccine to prediabetic NOD mice. Changes in the gut microbiota and associated metabolome were assessed using next-generation sequencing and gas chromatography-mass spectrometry (GC-MS). The Salmonella-based vaccine did not cause significant changes in the gut microbiota composition immediately after vaccination although at 30 days post-vaccination changes were seen. Additionally, no changes were noted in the fecal mycobiome between vaccine- and control/vehicle-treated mice. Significant changes in metabolic pathways related to inflammation and proliferation were found after vaccine administration. The results from this study suggest that an oral Salmonella-based vaccine alters the gut microbiome and metabolome towards a more tolerant composition. These results support the use of orally administered Salmonella-based vaccines that induced tolerance after administration.
Collapse
Affiliation(s)
- Jacob Cobb
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Sameh S M Soliman
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Mauricio Retuerto
- Center for Medical Mycology, Department of Dermatology, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Janine C Quijano
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Chris Orr
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Mahmoud Ghannoum
- Center for Medical Mycology, Department of Dermatology, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Fouad Kandeel
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Mohamed I Husseiny
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
- Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| |
Collapse
|
17
|
Ghasemi A, Wang S, Sahay B, Abbott JR, Curtiss R. Protective immunity enhanced Salmonella vaccine vectors delivering Helicobacter pylori antigens reduce H. pylori stomach colonization in mice. Front Immunol 2022; 13:1034683. [PMID: 36466847 PMCID: PMC9716130 DOI: 10.3389/fimmu.2022.1034683] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/31/2022] [Indexed: 08/18/2024] Open
Abstract
Helicobacter pylori is a major cause of gastric mucosal inflammation, peptic ulcers, and gastric cancer. Emerging antimicrobial-resistant H. pylori has hampered the effective eradication of frequent chronic infections. Moreover, a safe vaccine is highly demanded due to the absence of effective vaccines against H. pylori. In this study, we employed a new innovative Protective Immunity Enhanced Salmonella Vaccine (PIESV) vector strain to deliver and express multiple H. pylori antigen genes. Immunization of mice with our vaccine delivering the HpaA, Hp-NAP, UreA and UreB antigens, provided sterile protection against H. pylori SS1 infection in 7 out of 10 tested mice. In comparison to the control groups that had received PBS or a PIESV carrying an empty vector, immunized mice exhibited specific and significant cellular recall responses and antigen-specific serum IgG1, IgG2c, total IgG and gastric IgA antibody titers. In conclusion, an improved S. Typhimurium-based live vaccine delivering four antigens shows promise as a safe and effective vaccine against H. pylori infection.
Collapse
Affiliation(s)
- Amir Ghasemi
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, FL, United States
| | - Shifeng Wang
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, FL, United States
| | - Bikash Sahay
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, FL, United States
| | - Jeffrey R. Abbott
- Department of Comparative, Diagnostic and Population Medicine, University of Florida, Gainesville, FL, United States
| | - Roy Curtiss
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, FL, United States
| |
Collapse
|
18
|
Attenuated Salmonella Typhimurium with truncated LPS and outer membrane-displayed RGD peptide for cancer therapy. Biomed Pharmacother 2022; 155:113682. [PMID: 36095964 DOI: 10.1016/j.biopha.2022.113682] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/01/2022] [Accepted: 09/07/2022] [Indexed: 11/21/2022] Open
Abstract
Gram-negative, facultatively anaerobic bacteria Salmonella Typhimurium is a candidate agent or delivery vector for cancer therapy. Effective targeted therapies in addition to radiotherapy, chemotherapy and surgery have been urgently needed as an alternative or supplement. This study expected to further improve the tumor-targeting ability of Salmonella bacteria through genetic modifications. Based on an auxotrophic Salmonella bacterial strain (D2), we constructed Salmonella mutants with altered LPS length to facilitate displaying the RGD4C targeting peptide on the outer membrane surface of Salmonella. The expression of RGD4C peptide in fusion with OmpA was identified by outer membrane protein extraction and WB detection in different mutant strains. However, flow cytometry analysis following immunofluorescence staining demonstrated that the extracellular length of Salmonella LPS did affect the surface display of RGD4C peptide. The strain D2-RGD4C that synthesized intact LPS including lipid A, core oligosaccharides and O antigen polysaccharides could hardly display RGD4C peptide, showing the same fluorescence signal intensity as the strains not expressing RGD4C peptide. Among different strains, D2 ∆rfaJ-RGD4C that synthesized truncated LPS including lipid A and partial core oligosaccharides was capable of displaying RGD4C peptide most efficiently and showed the highest ability to target HUVECs expressing αV integrin and tumor tissue with abundant neovascularization. Animal experiments also demonstrated that this tumor-targeting attenuated Salmonella strain to simultaneously deliver endostatin and TRAIL, two agents with different anti-tumor activities, could significantly inhibit tumor growth and prolong mouse survival. Thus, our studies revealed that Salmonella could be genetically engineered to improve its tumor targeting via the truncation of LPS and surface display of targeting peptides, thereby eliciting superior anti-tumor effects through targeted delivery of drug molecules.
Collapse
|
19
|
Pérez Jorge G, Módolo DG, Jaimes-Florez YP, Fávaro WJ, Bispo de Jesus M, Brocchi M. p53 gene delivery via a recombinant Salmonella enterica Typhimurium leads to human bladder carcinoma cell death in vitro. Lett Appl Microbiol 2022; 75:1010-1020. [PMID: 35737820 DOI: 10.1111/lam.13777] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 06/13/2022] [Accepted: 06/17/2022] [Indexed: 11/28/2022]
Abstract
Numerous studies have attempted to restore the function of the tumour suppressor p53 as an anticancer strategy through gene delivery. However, most studies employed non-bacterial vectors to deliver p53. Various facultative and obligate anaerobic bacteria have been proposed as vectors because of their intrinsic tumour targeting ability and antitumour activity. Salmonella enterica Typhimurium is the most studied bacterial vector in anticancer therapy. We used the previously designed χ11218 strain of S. enterica Typhimurium, displaying regulated delayed lysis, as a vector for delivering p53 to human bladder carcinoma cells, restoring wild-type p53 protein function. We cloned p53 into pYA4545 (containing a eukaryotic expression system) to generate the χ11218 pYA4545p53 strain. Cloning of p53 did not affect the growth or interfere with the invasive and replicative capacity of χ11218 bacteria in tumour cells. Human bladder carcinoma cells (expressing mutated p53) transfected with pYA4545p53 showed a significant increase in the expression of p53 protein. We demonstrated that p53 supplied by χ11218 significantly decreased the viability of human bladder cancer cells in a dose-dependent manner. This study demonstrates the applicability of the attenuated χ11218 strain as a vector for DNA plasmids expressing tumour suppressor genes.
Collapse
Affiliation(s)
- Genesy Pérez Jorge
- Department of Genetics, Evolution, Microbiology, and Immunology, Tropical Disease Laboratory, Institute of Biology, University of Campinas - UNICAMP, 13083-970, Campinas, SP, Brazil
| | | | - Yessica Paola Jaimes-Florez
- Department of Genetics, Evolution, Microbiology, and Immunology, Tropical Disease Laboratory, Institute of Biology, University of Campinas - UNICAMP, 13083-970, Campinas, SP, Brazil
| | - Wagner José Fávaro
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas - UNICAMP, 13083-970, Campinas, SP, Brazil
| | - Marcelo Bispo de Jesus
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas - UNICAMP, 13083-970, Campinas, SP, Brazil
| | - Marcelo Brocchi
- Department of Genetics, Evolution, Microbiology, and Immunology, Tropical Disease Laboratory, Institute of Biology, University of Campinas - UNICAMP, 13083-970, Campinas, SP, Brazil
| |
Collapse
|
20
|
Protection against genotype VII Newcastle disease virus challenge by a minicircle DNA vaccine coexpressing F protein and chicken IL-18 adjuvant. Vet Microbiol 2022; 270:109474. [DOI: 10.1016/j.vetmic.2022.109474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 11/20/2022]
|
21
|
Garduño-González KA, Peña-Benavides SA, Araújo RG, Castillo-Zacarías C, Melchor-Martínez EM, Oyervides-Muñoz MA, Sosa-Hernández JE, Purton S, Iqbal HM, Parra-Saldívar R. Current challenges for modern vaccines and perspectives for novel treatment alternatives. J Drug Deliv Sci Technol 2022; 70:103222. [DOI: 10.1016/j.jddst.2022.103222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
22
|
A triple-sugar regulated Salmonella vaccine protects against Clostridium perfringens-induced necrotic enteritis in broiler chickens. Poult Sci 2021; 101:101592. [PMID: 34922043 PMCID: PMC8686071 DOI: 10.1016/j.psj.2021.101592] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 02/08/2023] Open
Abstract
Gram-positive Clostridium perfringens type G, the causative agent of necrotic enteritis (NE), has gained more attention in the poultry industry due to governmental restrictions on the use of growth-promoting antibiotics in poultry feed. Our previous work has proved that regulated delayed lysis Salmonella vaccines delivering a plasmid encoding an operon fusion of the nontoxic C-terminal adhesive part of alpha toxin and a GST-NetB toxin fusion were able to elicit significant protective immunity in broilers against C. perfringens challenge. We recently improved our S. Typhimurium antigen delivery vaccine strain by integrating a rhamnose-regulated O-antigen synthesis gene enabling a triple-sugar regulation system to control virulence, antigen-synthesis and lysis in vivo traits. The strain also includes a ΔsifA mutation that was previously shown to increase the immunogenicity of and level of protective immunity induced by Salmonella vectored influenza and Eimeria antigens. The new antigen-delivery vaccine vector system confers on the vaccine strain a safe profile and improved protection against C. perfringens challenge. The strain with the triple-sugar regulation system delivering a regulated lysis plasmid pG8R220 encoding the PlcC and GST-NetB antigens protected chickens at a similar level observed in antibiotic-treated chickens. Feed conversion and growth performance were also similar to antibiotic-treated chickens. These studies made use of a severe C. perfringens challenge with lesion formation and mortality enhanced by pre-exposure to Eimeria maxima oocysts. The vaccine achieved effectiveness through three different immunization routes, oral, spray and in drinking water. The vaccine has a potential for application in commercial hatcher and broiler-rearing conditions.
Collapse
|
23
|
Senevirathne A, Park JY, Hewawaduge C, Perumalraja K, Lee JH. Eukaryotic expression system complemented with expressivity of Semliki Forest Virus's RdRp and invasiveness of engineered Salmonella demonstrate promising potential for bacteria mediated gene therapy. Biomaterials 2021; 279:121226. [PMID: 34736150 DOI: 10.1016/j.biomaterials.2021.121226] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 09/29/2021] [Accepted: 10/22/2021] [Indexed: 10/20/2022]
Abstract
This study describes an efficient eukaryotic expression system (pJHL204) built into the Salmonella delivery system to enhance the essential efficacy and effectiveness of conventional DNA therapy. The expression system utilizes RNA-dependent RNA polymerase activity (RdRp) of Semiliki Forest Virus attributing to dramatic antigen expression by cytoplasmic mRNA amplification. Functional characterization of the pJHL204 by in vitro and in vivo transfection studies revealed the improved expression of mRNA at least 150 folds than the RdRp mutant plasmid under in vitro conditions. Using green fluorescence protein (GFP) and mCherry as bait proteins this system was extensively characterized for plasmid delivery capacity, antigen expression, and safety using in vivo and in vitro models by employing flow cytometry, fluorescence microscopy, and immunohistochemical staining. Employment of Salmonella as a carrier significantly extends plasmid in vivo survivability and prolongs the effective duration until the elimination of the Salmonella carrier strain in the host. The strategy can be easily adapted for P2A connected multiple antigen delivery in a single vector system due to the significantly high cargo capacity of Salmonella. A mouse challenge study was carried out utilizing P2A connected H1N1 hemagglutinin (HA) and neuraminidase (NA) via the Salmonella carrier strain JOL2500 significantly reduced viral activity and protected mice against the H1N1 challenge and demonstrates potential to redefine in vivo DNA therapy as a reliable and safe system to treat human diseases using useful microbes like Salmonella.
Collapse
Affiliation(s)
- Amal Senevirathne
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| | - Ji-Young Park
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| | - Chamith Hewawaduge
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| | - Kirthika Perumalraja
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea.
| |
Collapse
|
24
|
Senevirathne A, Hewawaduge C, Lee JH. Genetic interference exerted by Salmonella-delivered CRISPR/Cas9 significantly reduces the pathological burden caused by Marek's disease virus in chickens. Vet Res 2021; 52:125. [PMID: 34593043 PMCID: PMC8482593 DOI: 10.1186/s13567-021-00995-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 08/09/2021] [Indexed: 11/29/2022] Open
Abstract
Efficient in vivo delivery of a CRISPR/Cas9 plasmid is of paramount importance for effective therapy. Here, we investigated the usability of Salmonella as a plasmid carrier for in vivo therapy against virus-induced cancer using Marek’s disease virus (MDV) as a model for study in chickens. A green fluorescent protein-expressing CRISPR/Cas9 plasmid encoding the virulence gene pp38 was constructed against Marek’s disease virus. Therapeutic plasmids were transformed into Salmonella carrying lon and sifA gene deletions. The animals in 5 groups were intraperitoneally inoculated with phosphate-buffered saline, vector control, or Salmonella before or after MDV infection, or left uninfected as a naïve control. Therapeutic effectiveness was evaluated by observing disease outcomes and the viral copy number in peripheral blood mononuclear cells. The efficacy of plasmid delivery by Salmonella was 13 ± 1.7% in the spleen and 8.0 ± 1.8% in the liver on the 6th day post-infection. The Salmonella-treated groups showed significant resistance to MDV infection. The maximum effect was observed in the group treated with Salmonella before MDV infection. None of the chickens fully recovered; however, the results suggested that timely delivery of Salmonella could be effective for in vivo CRISPR/Cas9-mediated genetic interference against highly pathogenic MDV. The use of Salmonella in CRISPR systems provides a simpler and more efficient platform for in vivo therapy with CRISPR than the use of conventional in vivo gene delivery methods and warrants further development.
Collapse
Affiliation(s)
- Amal Senevirathne
- College of Veterinary Medicine, Chonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| | - Chamith Hewawaduge
- College of Veterinary Medicine, Chonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Chonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea.
| |
Collapse
|
25
|
Wang Z, Zhao X, Wang Y, Sun C, Sun M, Gao X, Jia F, Shan C, Yang G, Wang J, Huang H, Shi C, Yang W, Qian A, Wang C, Jiang Y. In Vivo Production of HN Protein Increases the Protection Rates of a Minicircle DNA Vaccine against Genotype VII Newcastle Disease Virus. Vaccines (Basel) 2021; 9:vaccines9070723. [PMID: 34358140 PMCID: PMC8310180 DOI: 10.3390/vaccines9070723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/26/2021] [Accepted: 06/26/2021] [Indexed: 01/09/2023] Open
Abstract
The Cre-recombinase mediated in vivo minicircle DNA vaccine platform (CRIM) provided a novel option to replace a traditional DNA vaccine. To further improve the immune response of our CRIM vaccine, we designed a dual promoter expression plasmid named pYL87 which could synthesize short HN protein under a prokaryotic in vivo promoter PpagC and full length HN protein of genotype VII Newcastle disease virus (NDV) under the previous eukaryotic CMV promoter at the same time. Making use of the self-lysed Salmonella strain as a delivery vesicle, chickens immunized with the pYL87 construction showed an increased serum haemagglutination inhibition antibody response, as well as an increased cell proliferation level and cellular IL-4 and IL-18 cytokines, compared with the previous CRIM vector pYL47. After the virus challenge, the pYL87 vector could provide 80% protection compared to 50% protection against genotype VII NDV in pYL47 immunized chickens, indicating a promising dual promoter strategy used in vaccine design.
Collapse
|
26
|
Chong A, Cooper KG, Kari L, Nilsson OR, Hillman C, Fleming BA, Wang Q, Nair V, Steele-Mortimer O. Cytosolic replication in epithelial cells fuels intestinal expansion and chronic fecal shedding of Salmonella Typhimurium. Cell Host Microbe 2021; 29:1177-1185.e6. [PMID: 34043959 DOI: 10.1016/j.chom.2021.04.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 03/30/2021] [Accepted: 04/29/2021] [Indexed: 12/20/2022]
Abstract
Persistent and intermittent fecal shedding, hallmarks of Salmonella infections, are important for fecal-oral transmission. In the intestine, Salmonella enterica serovar Typhimurium (STm) actively invades intestinal epithelial cells (IECs) and survives in the Salmonella-containing vacuole (SCV) and the cell cytosol. Cytosolic STm replicate rapidly, express invasion factors, and induce extrusion of infected epithelial cells into the intestinal lumen. Here, we engineered STm that self-destruct in the cytosol (STmCytoKill), but replicates normally in the SCV, to examine the role of cytosolic STm in infection. Intestinal expansion and fecal shedding of STmCytoKill are impaired in mouse models of infection. We propose a model whereby repeated rounds of invasion, cytosolic replication, and release of invasive STm from extruded IECs fuels the high luminal density required for fecal shedding.
Collapse
Affiliation(s)
- Audrey Chong
- Laboratory of Bacteriology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Kendal G Cooper
- Laboratory of Bacteriology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Laszlo Kari
- Laboratory of Bacteriology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Olof R Nilsson
- Laboratory of Bacteriology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Chad Hillman
- Laboratory of Bacteriology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Brittany A Fleming
- Laboratory of Bacteriology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Qinlu Wang
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20894, USA
| | - Vinod Nair
- Research Technologies Branch, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Olivia Steele-Mortimer
- Laboratory of Bacteriology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA.
| |
Collapse
|
27
|
Kong W. Development of Antiviral Vaccine Utilizing Self-Destructing Salmonella for Antigen and DNA Vaccine Delivery. Methods Mol Biol 2021; 2225:39-61. [PMID: 33108656 DOI: 10.1007/978-1-0716-1012-1_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Vaccines are the most effective means to prevent infectious diseases, especially for viral infection. The key to an excellent antiviral vaccine is the ability to induce long-term protective immunity against a specific virus. Bacterial vaccine vectors have been used to impart protection against self, as well as heterologous antigens. One significant benefit of using live bacterial vaccine vectors is their ability to invade and colonize deep effector lymphoid tissues after mucosal delivery. The bacterium Salmonella is considered the best at this deep colonization. This is critically essential for inducing protective immunity. This chapter describes the methodology for developing genetically modified self-destructing Salmonella (GMS) vaccine delivery systems targeting influenza infection. Specifically, the methods covered include the procedures for the development of GMSs for protective antigen delivery to induce cellular immune responses and DNA vaccine delivery to induce systemic immunity against the influenza virus. These self-destructing GMS could be modified to provide effective biological containment for genetically engineered bacteria used for a diversity of purposes in addition to vaccines.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/biosynthesis
- Antibodies, Viral/biosynthesis
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Female
- Genes, Lethal
- Genetic Engineering/methods
- Humans
- Immunity, Cellular/drug effects
- Immunity, Mucosal/drug effects
- Immunization/methods
- Influenza Vaccines/genetics
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Mice
- Mice, Inbred BALB C
- Nucleoproteins/genetics
- Nucleoproteins/immunology
- Organisms, Genetically Modified
- Plasmids/chemistry
- Plasmids/metabolism
- Salmonella typhimurium/genetics
- Salmonella typhimurium/immunology
- Transgenes
- Vaccines, DNA/genetics
Collapse
Affiliation(s)
- Wei Kong
- Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
28
|
Liang K, Liu Q, Kong Q. New technologies in developing recombinant-attenuated bacteria for cancer therapy. Biotechnol Bioeng 2020; 118:513-530. [PMID: 33038015 DOI: 10.1002/bit.27596] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/12/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022]
Abstract
Cancer has always been a global problem, with more cases of cancer patients being diagnosed every year. Conventional cancer treatments, including radiotherapy, chemotherapy, and surgery, are still unable to bypass their obvious limitations, and developing effective targeted therapies is still required. More than one century ago, the doctor William B. Coley discovered that cancer patients had tumor regression by injection of Streptococcus bacteria. The studies of cancer therapy using bacterial microorganisms are now very widespread. In particular, the facultative anaerobic bacteria Salmonella typhimurium is widely investigated as it can selectively colonize different types of tumors, locally deliver various antitumor drugs, and inhibit tumor growth. The exciting antitumor efficacy and safety observed in animal tumor models prompted the well-known attenuated Salmonella bacterial strain VNP20009 to be tested in human clinical trials in the early 21st century. Regrettably, no patients showed significant therapeutic effects and even bacterial colonization in tumor tissue was undetectable in most patients. Salmonella bacteria are still considered as a promising agent or vehicle for cancer therapy. Recent efforts have been focused on the generation of attenuated bacterial strains with higher targeting for tumor tissue, and optimization of the delivery of therapeutic antitumor cargoes into the tumor microenvironment. This review will summarize new technologies or approaches that may improve bacteria-mediated cancer therapy.
Collapse
Affiliation(s)
- Kang Liang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Qing Liu
- College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Qingke Kong
- College of Veterinary Medicine, Southwest University, Chongqing, China
| |
Collapse
|
29
|
Liu Q, Su H, Bian X, Wang S, Kong Q. Live attenuated Salmonella Typhimurium with monophosphoryl lipid A retains ability to induce T-cell and humoral immune responses against heterologous polysaccharide of Shigella flexneri 2a. Int J Med Microbiol 2020; 310:151427. [PMID: 32654768 DOI: 10.1016/j.ijmm.2020.151427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 05/03/2020] [Accepted: 05/10/2020] [Indexed: 12/19/2022] Open
Abstract
Shigella flexneri 2a (Sf2a) is one of the most frequently isolated Shigella strains that causes the endemic shigellosis in developing countries. In this study, we used recombinant attenuated Salmonella vaccine (RASV) strains to deliver Sf2a O-antigen and characterized the immune responses induced by the vectored O-antigen. First, we identified genes sufficient for biosynthesis of Sf2a O-antigen. A plasmid containing the identified genes was then introduced into the RASV strains, which were manipulated to produce only the heterologous O-antigen and modified lipid A. After oral immunization of mice, we demonstrated that RASV strains could induce potent humoral immune responses as well as robust CD4+ T-cell responses against Sf2a Lipopolysaccharide (LPS) and protect mice against virulent Sf2a challenge. The induced serum antibodies mediated high levels of Shigella-specific serum bactericidal activity and C3 deposition. Moreover, the IgG+ B220low/int BM cell and T follicular helper (Tfh) cell responses could also be triggered effectively. The live attenuated Salmonella with the modified lipid A delivering Sf2a O-antigen polysaccharide showed the same ability to induce immune responses against Sf2a LPS as the strain with the original lipid A. These findings underscore the potential of RASV delivered Sf2a O-antigen for induction of robust CD4+ T-cell and IgG responses and warrant further studies toward the development of Shigella vaccine candidates with RASV strains.
Collapse
Affiliation(s)
- Qing Liu
- College of Animal Science and Technology, Southwest University, 400715, Chongqing, China
| | - Huali Su
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, 32611, FL, USA
| | - Xiaoping Bian
- College of Animal Science and Technology, Southwest University, 400715, Chongqing, China
| | - Shifeng Wang
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, 32611, FL, USA
| | - Qingke Kong
- College of Animal Science and Technology, Southwest University, 400715, Chongqing, China; Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, 32611, FL, USA.
| |
Collapse
|
30
|
Won G, Senevirathne A, Lee JH. Salmonella Enteritidis ghost vaccine carrying the hemagglutinin globular head (HA1) domain from H1N1 virus protects against salmonellosis and influenza in chickens. Vaccine 2020; 38:4387-4394. [PMID: 32402750 DOI: 10.1016/j.vaccine.2020.04.077] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/26/2020] [Accepted: 04/29/2020] [Indexed: 01/29/2023]
Abstract
This study evaluated the attenuated Salmonella Enteritidis (SE) ghost strain JOL2114 (Δlon ΔcpxR Δasd), which displays on the bacterial surface the H1N1 hemagglutinin globular head portion (HA1; amino acid residues 63-286) on the bacterial surface for protective efficacy against Salmonella and H1N1 challenge in the chicken model, as the birds are the predominant reservoirs for both diseases. The ghost system enhanced the lysis process by converging two lysis processes found in bacteriophages: bacteriophage PhiX174 lysis gene E and holin-endolysin genes found in bacteriophage λ, complemented with accessory lysis-related proteins Rz/Rz1. The present lysis machinery resulted in complete lysis of host-attenuated SE strains in about 24 hrs of incubation under a non-permissible temperature of 42 °C in the absence of L-arabinose, an antisense inducer that blocks lysis gene expression during the growth phase. SE ghost JOL2114 surface display of HA1 was confirmed by Western blot analysis resulting in an immune-reactive band of 31 kDa in size. Chicken immunization via intramuscular and oral routes yielded both SE and HA1 antigen-specific immune responses. Protective humoral and cell-mediated immune responses were effectively elicited against both Salmonella and influenza challenge. This efficient strategy of ghost generation employs a dual system of phage lysis for biological generation of SE ghosts that preserves the surface antigenic architecture, offering a rapid and effective way to generate vaccines that could be deployed in urgent circumstances to protect against both Salmonella and influenza infection.
Collapse
Affiliation(s)
- Gayeon Won
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, Gobong-ro 79, Iksan 54596, Republic of Korea
| | - Amal Senevirathne
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, Gobong-ro 79, Iksan 54596, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, Gobong-ro 79, Iksan 54596, Republic of Korea.
| |
Collapse
|
31
|
Mbongue JC, Alhoshani A, Rawson J, Garcia PA, Gonzalez N, Ferreri K, Kandeel F, Husseiny MI. Tracking of an Oral Salmonella-Based Vaccine for Type 1 Diabetes in Non-obese Diabetic Mice. Front Immunol 2020; 11:712. [PMID: 32411136 PMCID: PMC7198770 DOI: 10.3389/fimmu.2020.00712] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/30/2020] [Indexed: 11/28/2022] Open
Abstract
Type 1 diabetes (T1D) arises secondary to immune-driven destruction of pancreatic β-cells and manifests as insulin-deficient hyperglycemia. We showed that oral vaccination with live attenuated Salmonella, which simultaneously delivers autoantigens and a TGFβ expression vector to immune cells in the gut mucosa, provides protection against the progression of T1D in non-obese diabetic (NOD) mice. In this study we employed the Sleeping Beauty (SB) transposon system that is composed of a transposase and transposon encoding the td-Tomato to express red fluorescent protein (RFP) to permanently mark the cells that take up the Salmonella vaccine. After animal vaccination, the transposon labeled-dendritic cells (DCs) with red fluorescence appeared throughout the secondary lymphoid tissues. Furthermore, Sleeping Beauty containing tgfβ1 gene (SB-tgfβ1) co-expressed TGFβ and RFP. The labeled DCs were detected predominantly in Peyer's patches (PP) and mesenteric lymph nodes (MLN) and expressed CD103 surface marker. CD103+ DCs induced tolerogenic effects and gut homing. TGFβ significantly increased programmed death-ligand-1 (PDL-1 or CD274) expression in the DCs in the MLN and PP of treated mice. Also, TGFβ increased cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) levels in CD4+ cells in MLN and PP. Interestingly, DCs increased in all lymphatic organs of mice vaccinated with oral live Salmonella-based vaccine expressing preproinsulin (PPI), in combination with TGFβ, IL10, and subtherapeutic-doses of anti-CD3 mAb compared with vehicle-treated mice. These DCs are mostly tolerogenic in MLN and PP. Furthermore the DCs obtained from vaccine-treated but not vehicle-treated mice suppressed in vitro T cell proliferation. These data suggest that the MLN and the PP are a central hub for the beneficial anti-diabetic effects of an oral Salmonella-based vaccine prevention of diabetes in rodents.
Collapse
Affiliation(s)
- Jacques C. Mbongue
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Ali Alhoshani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Jeffrey Rawson
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Pablo A. Garcia
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Nelson Gonzalez
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Kevin Ferreri
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Fouad Kandeel
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Mohamed I. Husseiny
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
- Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| |
Collapse
|
32
|
Inda ME, Broset E, Lu TK, de la Fuente-Nunez C. Emerging Frontiers in Microbiome Engineering. Trends Immunol 2019; 40:952-973. [PMID: 31601521 DOI: 10.1016/j.it.2019.08.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 08/15/2019] [Accepted: 08/15/2019] [Indexed: 02/07/2023]
Abstract
The gut microbiome has a significant impact on health and disease and can actively contribute to obesity, diabetes, inflammatory bowel disease, cardiovascular disease, and neurological disorders. We do not yet have the necessary tools to fine-tune the microbial communities that constitute the microbiome, though such tools could unlock extensive benefits to human health. Here, we provide an overview of the current state of technological tools that may be used for microbiome engineering. These tools can enable investigators to define the parameters of a healthy microbiome and to determine how gut bacteria may contribute to the etiology of a variety of diseases. These tools may also allow us to explore the exciting prospect of developing targeted therapies and personalized treatments for microbiome-linked diseases.
Collapse
Affiliation(s)
- María Eugenia Inda
- Synthetic Biology Group, MIT Synthetic Biology Center, Department of Biological Engineering and Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Esther Broset
- Machine Biology Group, Departments of Psychiatry and Microbiology, Perelman School of Medicine, and Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Grupo de Genética de Micobacterias, Departamento de Microbiología y Medicina Preventiva, Facultad de Medicina, Universidad de Zaragoza, 50009, Spain
| | - Timothy K Lu
- Synthetic Biology Group, MIT Synthetic Biology Center, Department of Biological Engineering and Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Perelman School of Medicine, and Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
33
|
A Novel Cre Recombinase-Mediated In Vivo Minicircle DNA (CRIM) Vaccine Provides Partial Protection against Newcastle Disease Virus. Appl Environ Microbiol 2019; 85:AEM.00407-19. [PMID: 31053588 DOI: 10.1128/aem.00407-19] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 04/29/2019] [Indexed: 02/07/2023] Open
Abstract
Minicircle DNA (mcDNA), which contains only the necessary components for eukaryotic expression and is thus smaller than traditional plasmids, has been designed for application in genetic manipulation. In this study, we constructed a novel plasmid containing both the Cre recombinase under the phosphoglycerate kinase (PGK) promoter and recombinant lox66 and lox71 sites located outside the cytomegalovirus (CMV) expression cassette. The strictly controlled synthesis of Cre recombinase in vivo maintained the complete form of the plasmid in vitro, whereas the in vivo production of Cre transformed the parental plasmid to mcDNA after transfection. The newly designed Cre recombinase-mediated in vivo mcDNA platform, named CRIM, significantly increased the nuclear entry of mcDNA, followed by increased production of mRNA and protein, using enhanced green fluorescent protein (EGFP) as a model. Similar results were also observed in chickens when the vaccine was delivered by the regulated-delayed-lysis Salmonella strain χ11218, where significantly increased production of EGFP was observed in chicken livers. Then, we used the HN gene of genotype VII Newcastle disease virus as an antigen model to construct the traditional plasmid pYL43 and the novel mcDNA plasmid pYL47. After immunization, our CRIM vaccine provided significantly increased protection against challenge compared with that of the traditional plasmid, providing us with a novel mcDNA vaccine platform.IMPORTANCE Minicircle DNA (mcDNA) has been considered an attractive alternative to DNA vaccines; however, the relatively high cost and complicated process of purifying mcDNA dramatically restricts the application of mcDNA in the veterinary field. We designed a novel in vivo mcDNA platform in which the complete plasmid could spontaneously transform into mcDNA in vivo In combination with the regulated-delayed-lysis Salmonella strain, the newly designed mcDNA vaccine provides us with an elegant platform for veterinary vaccine development.
Collapse
|
34
|
Gao X, Xu K, Yang G, Shi C, Huang H, Wang J, Yang W, Liu J, Liu Q, Kang Y, Jiang Y, Wang C. Construction of a novel DNA vaccine candidate targeting F gene of genotype VII Newcastle disease virus and chicken IL-18 delivered by Salmonella. J Appl Microbiol 2019; 126:1362-1372. [PMID: 30785663 DOI: 10.1111/jam.14228] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/01/2019] [Accepted: 02/14/2019] [Indexed: 11/30/2022]
Abstract
AIMS Genotype VII Newcastle disease (ND) is one of the most epidemic and serious infectious diseases in the poultry industry. A novel vaccine targeting VII Newcastle disease virus (NDV) is still proving elusive. METHODS AND RESULTS In this study, we constructed regulated delayed lysis Salmonella strains expressing either a fusion protein (F) alone under an eukaryotic CMV promoter or together with chicken IL-18 (chIL-18) as a molecular adjuvant under prokaryotic Ptrc promoter, named pYL1 and pYL23 respectively. Oral immunization with recombinant strains induced NDV-specific serum IgG antibodies in both pYL1- and pYL23-immunized chickens. The presence of chIL-18 significantly increased lymphocyte proliferation in immunized chickens, as well as the percentages of CD3+ CD4+ and CD3+ CD8+ T cells in serum, even if a statistically significant difference did not exist. After a virulent challenge, pYL23 immunization provided about 80% protection at day 10 postinfection, compared with 60% of protection offered by pYL1 immunization and 100% protection in the inactivated vaccine group, indicating the enhanced immune response provided by chIL-18, which was also confirmed by histochemical analysis. CONCLUSIONS Recombinant lysis Salmonella-vectored DNA vaccine could provide us a novel potential option for controlling NDV infection. SIGNIFICANCE AND IMPACT OF THE STUDY This study took use of a regulated delayed lysis Salmonella vector for the design of an orally administrated vaccine against NDV.
Collapse
Affiliation(s)
- X Gao
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - K Xu
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - G Yang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - C Shi
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - H Huang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - J Wang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - W Yang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - J Liu
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Q Liu
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Y Kang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Y Jiang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - C Wang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| |
Collapse
|
35
|
Arora D, Sharma C, Jaglan S, Lichtfouse E. Live-Attenuated Bacterial Vectors for Delivery of Mucosal Vaccines, DNA Vaccines, and Cancer Immunotherapy. ENVIRONMENTAL CHEMISTRY FOR A SUSTAINABLE WORLD 2019. [PMCID: PMC7123696 DOI: 10.1007/978-3-030-01881-8_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Vaccines save millions of lives each year from various life-threatening infectious diseases, and there are more than 20 vaccines currently licensed for human use worldwide. Moreover, in recent decades immunotherapy has become the mainstream therapy, which highlights the tremendous potential of immune response mediators, including vaccines for prevention and treatment of various forms of cancer. However, despite the tremendous advances in microbiology and immunology, there are several vaccine preventable diseases which still lack effective vaccines. Classically, weakened forms (attenuated) of pathogenic microbes were used as vaccines. Although the attenuated microbes induce effective immune response, a significant risk of reversion to pathogenic forms remains. While in the twenty-first century, with the advent of genetic engineering, microbes can be tailored with desired properties. In this review, I have focused on the use of genetically modified bacteria for the delivery of vaccine antigens. More specifically, the live-attenuated bacteria, derived from pathogenic bacteria, possess many features that make them highly suitable vectors for the delivery of vaccine antigens. Bacteria can theoretically express any heterologous gene or can deliver mammalian expression vectors harboring vaccine antigens (DNA vaccines). These properties of live-attenuated microbes are being harnessed to make vaccines against several infectious and noninfectious diseases. In this regard, I have described the desired features of live-attenuated bacterial vectors and the mechanisms of immune responses manifested by live-attenuated bacterial vectors. Interestingly anaerobic bacteria are naturally attracted to tumors, which make them suitable vehicles to deliver tumor-associated antigens thus I have discussed important studies investigating the role of bacterial vectors in immunotherapy. Finally, I have provided important discussion on novel approaches for improvement and tailoring of live-attenuated bacterial vectors for the generation of desired immune responses.
Collapse
Affiliation(s)
- Divya Arora
- Indian Institute of Integrative Medicine, CSIR, Jammu, India
| | - Chetan Sharma
- Guru Angad Dev Veterinary and Animal Science University, Ludhiana, Punjab India
| | - Sundeep Jaglan
- Indian Institute of Integrative Medicine, CSIR, Jammu, India
| | - Eric Lichtfouse
- Aix Marseille University, CNRS, IRD, INRA, Coll France, CEREGE, Aix en Provence, France
| |
Collapse
|
36
|
Sedlmayer F, Aubel D, Fussenegger M. Synthetic gene circuits for the detection, elimination and prevention of disease. Nat Biomed Eng 2018; 2:399-415. [PMID: 31011195 DOI: 10.1038/s41551-018-0215-0] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 03/05/2018] [Indexed: 12/13/2022]
Abstract
In living organisms, naturally evolved sensors that constantly monitor and process environmental cues trigger corrective actions that enable the organisms to cope with changing conditions. Such natural processes have inspired biologists to construct synthetic living sensors and signalling pathways, by repurposing naturally occurring proteins and by designing molecular building blocks de novo, for customized diagnostics and therapeutics. In particular, designer cells that employ user-defined synthetic gene circuits to survey disease biomarkers and to autonomously re-adjust unbalanced pathological states can coordinate the production of therapeutics, with controlled timing and dosage. Furthermore, tailored genetic networks operating in bacterial or human cells have led to cancer remission in experimental animal models, owing to the network's unprecedented specificity. Other applications of designer cells in infectious, metabolic and autoimmune diseases are also being explored. In this Review, we describe the biomedical applications of synthetic gene circuits in major disease areas, and discuss how the first genetically engineered devices developed on the basis of synthetic-biology principles made the leap from the laboratory to the clinic.
Collapse
Affiliation(s)
- Ferdinand Sedlmayer
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Dominique Aubel
- IUTA Département Génie Biologique, Université Claude Bernard Lyon 1, Lyon, France
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland. .,Faculty of Science, University of Basel, Basel, Switzerland.
| |
Collapse
|
37
|
Yurina V. Live Bacterial Vectors-A Promising DNA Vaccine Delivery System. Med Sci (Basel) 2018; 6:E27. [PMID: 29570602 PMCID: PMC6024733 DOI: 10.3390/medsci6020027] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/19/2018] [Accepted: 03/19/2018] [Indexed: 12/14/2022] Open
Abstract
Vaccination is one of the most successful immunology applications that has considerably improved human health. The DNA vaccine is a new vaccine being developed since the early 1990s. Although the DNA vaccine is promising, no human DNA vaccine has been approved to date. The main problem facing DNA vaccine efficacy is the lack of a DNA vaccine delivery system. Several studies explored this limitation. One of the best DNA vaccine delivery systems uses a live bacterial vector as the carrier. The live bacterial vector induces a robust immune response due to its natural characteristics that are recognized by the immune system. Moreover, the route of administration used by the live bacterial vector is through the mucosal route that beneficially induces both mucosal and systemic immune responses. The mucosal route is not invasive, making the vaccine easy to administer, increasing the patient's acceptance. Lactic acid bacterium is one of the most promising bacteria used as a live bacterial vector. However, some other attenuated pathogenic bacteria, such as Salmonella spp. and Shigella spp., have been used as DNA vaccine carriers. Numerous studies showed that live bacterial vectors are a promising candidate to deliver DNA vaccines.
Collapse
Affiliation(s)
- Valentina Yurina
- Department of Pharmacy, Medical Faculty, Universitas Brawijaya, East Java 65145, Malang, Indonesia.
| |
Collapse
|
38
|
Metagenomic mining of regulatory elements enables programmable species-selective gene expression. Nat Methods 2018; 15:323-329. [PMID: 30052624 PMCID: PMC6065261 DOI: 10.1038/nmeth.4633] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/25/2018] [Indexed: 01/21/2023]
Abstract
Robust and predictably performing synthetic circuits rely on the use of well-characterized regulatory parts across different genetic backgrounds and environmental contexts. Here we report the large-scale metagenomic mining of thousands of natural 5' regulatory sequences from diverse bacteria, and their multiplexed gene expression characterization in industrially relevant microbes. We identified sequences with broad and host-specific expression properties that are robust in various growth conditions. We also observed substantial differences between species in terms of their capacity to utilize exogenous regulatory sequences. Finally, we demonstrate programmable species-selective gene expression that produces distinct and diverse output patterns in different microbes. Together, these findings provide a rich resource of characterized natural regulatory sequences and a framework that can be used to engineer synthetic gene circuits with unique and tunable cross-species functionality and properties, and also suggest the prospect of ultimately engineering complex behaviors at the community level.
Collapse
|
39
|
Clark-Curtiss JE, Curtiss R. Salmonella Vaccines: Conduits for Protective Antigens. THE JOURNAL OF IMMUNOLOGY 2018; 200:39-48. [PMID: 29255088 DOI: 10.4049/jimmunol.1600608] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 11/06/2017] [Indexed: 11/19/2022]
Abstract
Vaccines afford a better and more cost-effective approach to combatting infectious diseases than continued reliance on antibiotics or antiviral or antiparasite drugs in the current era of increasing incidences of diseases caused by drug-resistant pathogens. Recombinant attenuated Salmonella vaccines (RASVs) have been significantly improved to exhibit the same or better attributes than wild-type parental strains to colonize internal lymphoid tissues and persist there to serve as factories to continuously synthesize and deliver rAgs. Encoded by codon-optimized pathogen genes, Ags are selected to induce protective immunity to infection by that pathogen. After immunization through a mucosal surface, the RASV attributes maximize their abilities to elicit mucosal and systemic Ab responses and cell-mediated immune responses. This article summarizes many of the numerous innovative technologies and discoveries that have resulted in RASV platforms that will enable development of safe efficacious RASVs to protect animals and humans against a diversity of infectious disease agents.
Collapse
Affiliation(s)
- Josephine E Clark-Curtiss
- Division of Infectious Diseases and Global Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610.,Emerging Pathogens Institute, University of Florida, Gainesville, FL 32611
| | - Roy Curtiss
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611; and .,Department of Comparative, Diagnostic and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611
| |
Collapse
|
40
|
Lau YH, Stirling F, Kuo J, Karrenbelt MAP, Chan YA, Riesselman A, Horton CA, Schäfer E, Lips D, Weinstock MT, Gibson DG, Way JC, Silver PA. Large-scale recoding of a bacterial genome by iterative recombineering of synthetic DNA. Nucleic Acids Res 2017; 45:6971-6980. [PMID: 28499033 PMCID: PMC5499800 DOI: 10.1093/nar/gkx415] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 05/02/2017] [Indexed: 01/02/2023] Open
Abstract
The ability to rewrite large stretches of genomic DNA enables the creation of new organisms with customized functions. However, few methods currently exist for accumulating such widespread genomic changes in a single organism. In this study, we demonstrate a rapid approach for rewriting bacterial genomes with modified synthetic DNA. We recode 200 kb of the Salmonella typhimurium LT2 genome through a process we term SIRCAS (stepwise integration of rolling circle amplified segments), towards constructing an attenuated and genetically isolated bacterial chassis. The SIRCAS process involves direct iterative recombineering of 10–25 kb synthetic DNA constructs which are assembled in yeast and amplified by rolling circle amplification. Using SIRCAS, we create a Salmonella with 1557 synonymous leucine codon replacements across 176 genes, the largest number of cumulative recoding changes in a single bacterial strain to date. We demonstrate reproducibility over sixteen two-day cycles of integration and parallelization for hierarchical construction of a synthetic genome by conjugation. The resulting recoded strain grows at a similar rate to the wild-type strain and does not exhibit any major growth defects. This work is the first instance of synthetic bacterial recoding beyond the Escherichia coli genome, and reveals that Salmonella is remarkably amenable to genome-scale modification.
Collapse
Affiliation(s)
- Yu Heng Lau
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, 5th Floor, Boston, MA 02115, USA.,Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Alpert 536, Boston, MA 02115, USA
| | - Finn Stirling
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, 5th Floor, Boston, MA 02115, USA.,Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Alpert 536, Boston, MA 02115, USA
| | - James Kuo
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, 5th Floor, Boston, MA 02115, USA.,Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Alpert 536, Boston, MA 02115, USA
| | - Michiel A P Karrenbelt
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, 5th Floor, Boston, MA 02115, USA.,Systems and Synthetic Biology, Wageningen University, Dreijenplein 10, 6703 HB Wageningen, The Netherlands
| | - Yujia A Chan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, 5th Floor, Boston, MA 02115, USA.,Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Alpert 536, Boston, MA 02115, USA
| | - Adam Riesselman
- Program in Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Connor A Horton
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, 5th Floor, Boston, MA 02115, USA.,Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Alpert 536, Boston, MA 02115, USA
| | - Elena Schäfer
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, 5th Floor, Boston, MA 02115, USA.,Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Alpert 536, Boston, MA 02115, USA
| | - David Lips
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, 5th Floor, Boston, MA 02115, USA.,Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Alpert 536, Boston, MA 02115, USA
| | - Matthew T Weinstock
- Synthetic Genomics, Inc., 11149 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Daniel G Gibson
- Synthetic Genomics, Inc., 11149 North Torrey Pines Road, La Jolla, CA 92037, USA.,Synthetic Biology and Bioenergy Group, J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, CA 92037, USA
| | - Jeffrey C Way
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, 5th Floor, Boston, MA 02115, USA.,Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Alpert 536, Boston, MA 02115, USA
| | - Pamela A Silver
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, 5th Floor, Boston, MA 02115, USA.,Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Alpert 536, Boston, MA 02115, USA
| |
Collapse
|
41
|
Kim MS, Choi SH, Yang JI, Kim KH. Production of RNase III-knockout, auxotrophic Edwardsiella tarda mutant for delivery of long double-stranded RNA and evaluation of its immunostimulatory potential. FISH & SHELLFISH IMMUNOLOGY 2017; 68:474-478. [PMID: 28756288 DOI: 10.1016/j.fsi.2017.07.053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 06/23/2017] [Accepted: 07/25/2017] [Indexed: 06/07/2023]
Abstract
The artificially synthesized polyinosinic-polycytidylic acid (poly IC) has been widely used to induce type I IFN responses in various vertebrates including fish. However, as poly IC is too expensive to use in aquaculture, the development of another economical long dsRNA producing method is needed to practically use long dsRNAs in aquaculture farms for the control of infectious diseases. In the present study, to produce long dsRNAs economically, we developed a novel long dsRNA production system based on the RNase III gene deleted auxotrophic mutant E. tarda (ΔalrΔrncΔasd E. tarda) and a long dsRNA-producing vector that was equipped with two modified λ phage PR promoters arranged in a head-to-head fashion. As the present genetically engineered E. tarda cannot live without supplementation of d-alanine and DAP, environmental and medicinal risks are minimized. Olive flounder (Paralichthys olivaceus) fingerlings administered the long dsRNA-producing auxotrophic E. tarda mutant (Δalr ΔrncΔasd E. tarda) showed significantly higher expressions of TLR22, Mx1, and ISG15 genes, indicating a potential to increase type I interferon responses.
Collapse
Affiliation(s)
- Min Sun Kim
- Graduate School of Integrated Bioindustry, Sejong University, Seoul 05006, South Korea
| | - Seung Hyuk Choi
- Ministry of Science and ICT, Gwacheon-si, Gyeonggi-do, 13809, South Korea
| | - Jeong In Yang
- Department of Aquatic Life Medicine, Pukyong National University, Busan 48513, South Korea
| | - Ki Hong Kim
- Department of Aquatic Life Medicine, Pukyong National University, Busan 48513, South Korea.
| |
Collapse
|
42
|
Jiang Y, Yang G, Wang Q, Wang Z, Yang W, Gu W, Shi C, Wang J, Huang H, Wang C. Molecular mechanisms underlying protection against H9N2 influenza virus challenge in mice by recombinant Lactobacillus plantarum with surface displayed HA2-LTB. J Biotechnol 2017; 259:6-14. [PMID: 28811215 DOI: 10.1016/j.jbiotec.2017.08.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/24/2017] [Accepted: 08/09/2017] [Indexed: 12/29/2022]
Abstract
It has been considered that the Avian influenza virus (AIV) causes severe threats to poultry industry. In this study, we constructed a series of recombinant Lactobacillus plantarum (L. plantarum) with surface displayed hemagglutinin subunit 2 (HA2) alone or together with heat-labile toxin B subunit (LTB) from enterotoxigenic Escherichia coli. Balb/c mice were used as model to evaluate the protective effects of recombinant L. plantarum strains against H9N2 subtype challenge. The results showed that the presence of LTB significantly increased the percentages of CD3+CD4+IL-4+, CD3+CD4+IFN-γ+ and CD3+CD4+IL-17+ T cells, as well as CD3+CD8+IFN-γ+ T cells in spleen and MLNs determined by Fluorescence-Activated Cell Sorting assay. Similar increased production of serum IFN-γ was also confirmed by enzyme linked immunosorbent assay (ELISA). The L. plantarum with surface displayed HA2-LTB also dramatically increased the percentages of B220+ IgA+ B cells in peyer patch, in consistent with elevated production of mucosal SIgA antibody determined by ELISA. Finally, the orally administrated HA2-LTB expressing strain efficiently protected mice against H9N2 subtype AIV challenge shown by increased survival percentages, body weight gains and decreased lung lesions in histopathologic analysis. In conclusion, this study provides more detail mechanisms underlying the adjuvant effects of LTB on heterologous antigen produced in recombinant lactic acid bacteria.
Collapse
Affiliation(s)
- Yanlong Jiang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Guilian Yang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Qi Wang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Zhannan Wang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Wentao Yang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Wei Gu
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China; Shandong Baolai-Leelai Bio-Tech Co., LTD, Taian, Shandong Province, 171000, China
| | - Chunwei Shi
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Jianzhong Wang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Haibin Huang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Chunfeng Wang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
43
|
Kamble NM, Hyoung KJ, Lee JH. Live-attenuated auxotrophic mutant of Salmonella Typhimurium expressing immunogenic HA1 protein enhances immunity and protective efficacy against H1N1 influenza virus infection. Future Microbiol 2017; 12:739-752. [PMID: 28594235 DOI: 10.2217/fmb-2016-0190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To evaluate the efficacy of attenuated Salmonella Typhimurium (JOL912) as a live bacterial vaccine vector. MATERIALS & METHODS The JOL912 engineered to deliver HA1 protein from influenza A/Puerto Rico/8/1934 (H1N1; PR8) virus was coined as JOL1635 and further evaluated for immunogenicity and protective efficacy. RESULTS The JOL1635 stably harbored the HA1 gene within pMMP65 plasmid with periplasmic expression and effective delivery of HA1 protein to RAW264.7 cells. The JOL1635 immunized chickens showed the significant increase in HA1-specific IgG, sIgA antibody, IFN-γ, IL-6 cytokine and cellular immune responses. The postoral challenge, the JOL1635-immunized chickens showed a faster clearance of PR8 virus cloacal shedding than the control group. CONCLUSION Generated JOL1635 can establish specific immunogenicity and protection against the PR8 virus in chickens.
Collapse
Affiliation(s)
- Nitin Machindra Kamble
- Veterinary Public Health, College of Veterinary Medicine, Chonbuk National University, Iksan Campus, Jeonju 570-752, Republic of Korea
| | - Kim Je Hyoung
- Veterinary Public Health, College of Veterinary Medicine, Chonbuk National University, Iksan Campus, Jeonju 570-752, Republic of Korea
| | - John Hwa Lee
- Veterinary Public Health, College of Veterinary Medicine, Chonbuk National University, Iksan Campus, Jeonju 570-752, Republic of Korea
| |
Collapse
|
44
|
Hu M, Zhao W, Gao W, Li W, Meng C, Yan Q, Wang Y, Zhou X, Geng S, Pan Z, Cui G, Jiao X. Recombinant Salmonella expressing SspH2-EscI fusion protein limits its colonization in mice. BMC Immunol 2017; 18:21. [PMID: 28468643 PMCID: PMC5415771 DOI: 10.1186/s12865-017-0203-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 04/24/2017] [Indexed: 12/20/2022] Open
Abstract
Background Activation of inflammasome contributes to the clearance of intracellular bacteria. C-terminus of E. coli EscI protein can activate NLRC4 (NLR family, CARD domain containing-4) inflammasome in macrophages. The purpose of this study was to determine if activation of NLRC4 inflammasome by EscI can reduce the colonization of Salmonella in mice. Results A recombinant S. typhimurium strain expressing fusion protein of the N-terminal SspH2 (a Salmonella type III secretion system 2 effector) and C-terminal EscI was constructed and designated as X4550(pYA3334-SspH2-EscI). In vitro assay showed that X4550(pYA3334-SspH2-EscI) significantly enhanced IL-1β and IL-18 secretion (P < 0.05) and pyroptotic cell death of mouse peritoneal macrophages, compared with those infected with control strain, X4550(pYA3334-SspH2). In vivo studies showed that colonization of X4550(pYA3334-SspH2-EscI) in both spleen and liver were significantly lower than that of X4550(pYA3334-SspH2) (P < 0.05). The bacterial counts of X4550(pYA3334-SspH2-EscI) in mice decreased, while those of X4550(pYA3334-SspH2) increased over the time after infection. Additionally, X4550(pYA3334-SspH2-EscI) induced a less pathological alteration in spleen and liver than X4550(pYA3334-SspH2). Conclusion Fusion protein SspH2-EscI may be translocated into macrophages and activate NLRC4 inflammasome, which limits Salmonella colonization in spleen and liver of mice.
Collapse
Affiliation(s)
- Maozhi Hu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, Jiangsu, China. .,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, Jiangsu, China.
| | - Weixin Zhao
- College of Tourism & Cuisine (College of Food Science and Engineering), Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Wei Gao
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Wenhua Li
- College of Tourism & Cuisine (College of Food Science and Engineering), Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Chuang Meng
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, Jiangsu, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Qiuxiang Yan
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Yuyang Wang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Xiaohui Zhou
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, Jiangsu, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, Jiangsu, China.,Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, 06269-3089, CT, USA
| | - Shizhong Geng
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, Jiangsu, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Zhiming Pan
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, Jiangsu, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Guiyou Cui
- College of Tourism & Cuisine (College of Food Science and Engineering), Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Xinan Jiao
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, Jiangsu, China. .,Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, Jiangsu, China.
| |
Collapse
|
45
|
Latasa C, Echeverz M, García B, Gil C, García-Ona E, Burgui S, Casares N, Hervás-Stubbs S, Lasarte JJ, Lasa I, Solano C. Evaluation of a Salmonella Strain Lacking the Secondary Messenger C-di-GMP and RpoS as a Live Oral Vaccine. PLoS One 2016; 11:e0161216. [PMID: 27537839 PMCID: PMC4990191 DOI: 10.1371/journal.pone.0161216] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 08/02/2016] [Indexed: 01/29/2023] Open
Abstract
Salmonellosis is one of the most important bacterial zoonotic diseases transmitted through the consumption of contaminated food, with chicken and pig related products being key reservoirs of infection. Although numerous studies on animal vaccination have been performed in order to reduce Salmonella prevalence, there is still a need for an ideal vaccine. Here, with the aim of constructing a novel live attenuated Salmonella vaccine candidate, we firstly analyzed the impact of the absence of cyclic-di-GMP (c-di-GMP) in Salmonella virulence. C-di-GMP is an intracellular second messenger that controls a wide range of bacterial processes, including biofilm formation and synthesis of virulence factors, and also modulates the host innate immune response. Our results showed that a Salmonella multiple mutant in the twelve genes encoding diguanylate cyclase proteins that, as a consequence, cannot synthesize c-di-GMP, presents a moderate attenuation in a systemic murine infection model. An additional mutation of the rpoS gene resulted in a synergic attenuating effect that led to a highly attenuated strain, referred to as ΔXIII, immunogenic enough to protect mice against a lethal oral challenge of a S. Typhimurium virulent strain. ΔXIII immunogenicity relied on activation of both antibody and cell mediated immune responses characterized by the production of opsonizing antibodies and the induction of significant levels of IFN-γ, TNF-α, IL-2, IL-17 and IL-10. ΔXIII was unable to form a biofilm and did not survive under desiccation conditions, indicating that it could be easily eliminated from the environment. Moreover, ΔXIII shows DIVA features that allow differentiation of infected and vaccinated animals. Altogether, these results show ΔXIII as a safe and effective live DIVA vaccine.
Collapse
Affiliation(s)
- Cristina Latasa
- Laboratory of Microbial Biofilms, Instituto de Agrobiotecnología (Idab), Universidad Pública de Navarra-CSIC-Gobierno de Navarra, Pamplona, Spain
- Recombina S. L. Mutilva, Navarra, Spain
| | - Maite Echeverz
- Laboratory of Microbial Biofilms, Instituto de Agrobiotecnología (Idab), Universidad Pública de Navarra-CSIC-Gobierno de Navarra, Pamplona, Spain
| | - Begoña García
- Laboratory of Microbial Biofilms, Instituto de Agrobiotecnología (Idab), Universidad Pública de Navarra-CSIC-Gobierno de Navarra, Pamplona, Spain
| | - Carmen Gil
- Laboratory of Microbial Biofilms, Instituto de Agrobiotecnología (Idab), Universidad Pública de Navarra-CSIC-Gobierno de Navarra, Pamplona, Spain
| | - Enrique García-Ona
- Laboratory of Microbial Biofilms, Instituto de Agrobiotecnología (Idab), Universidad Pública de Navarra-CSIC-Gobierno de Navarra, Pamplona, Spain
| | - Saioa Burgui
- Laboratory of Microbial Biofilms, Instituto de Agrobiotecnología (Idab), Universidad Pública de Navarra-CSIC-Gobierno de Navarra, Pamplona, Spain
| | - Noelia Casares
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdISNA), University of Navarra, Pamplona, Spain
| | - Sandra Hervás-Stubbs
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdISNA), University of Navarra, Pamplona, Spain
| | - Juan José Lasarte
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdISNA), University of Navarra, Pamplona, Spain
| | - Iñigo Lasa
- Laboratory of Microbial Biofilms, Instituto de Agrobiotecnología (Idab), Universidad Pública de Navarra-CSIC-Gobierno de Navarra, Pamplona, Spain
- Navarrabiomed-Universidad Pública de Navarra, Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
- * E-mail: (CS); (IL)
| | - Cristina Solano
- Laboratory of Microbial Biofilms, Instituto de Agrobiotecnología (Idab), Universidad Pública de Navarra-CSIC-Gobierno de Navarra, Pamplona, Spain
- * E-mail: (CS); (IL)
| |
Collapse
|
46
|
Live attenuated Salmonella displaying HIV-1 10E8 epitope on fimbriae: systemic and mucosal immune responses in BALB/c mice by mucosal administration. Sci Rep 2016; 6:29556. [PMID: 27411313 PMCID: PMC4944174 DOI: 10.1038/srep29556] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 06/17/2016] [Indexed: 01/06/2023] Open
Abstract
The HIV-1 membrane proximal external region (MPER) that is targeted by several broadly neutralizing antibodies (BNAbs) has been considered a potential immunogen for vaccine development. However, to date the immunogenicity of these BNAb epitopes has not been made sufficiently adequate. In the present work, we used live attenuated Salmonella as a platform to present the HIV-1 MPER 10E8 epitope in the fimbriae. The insertion of the 10E8 epitope into the fimbriae had no significant influence on the expression and the absorption capacity of bacterial fimbriae, nor on the virulence and invasiveness of the attenuated Salmonella. After oral administration of the vaccine construct to mice followed by 10E8 epitope peptide boost, specific antibody responses in serum and mucosa as well as memory lymphocytes in spleen and plasma cells in bone marrow were induced. We also found that the live attenuated Salmonella vector directed the immunity toward Th1 bias, induced Th1 and Th2 cytokine responses and stimulated significant B cell differentiation into GC B, memory B and plasma cells. Therefore, we propose that the live attenuated Salmonella constitutively expressing HIV-1 BNAb epitopes on the fimbriae will be an effective approach to improving immune microenvironment and enhancing the immunogenicity of HIV-1 epitope vaccines.
Collapse
|
47
|
Bridge DR, Whitmire JM, Makobongo MO, Merrell DS. Heterologous Pseudomonas aeruginosa O-antigen delivery using a Salmonella enterica serovar Typhimurium wecA mutant strain. Int J Med Microbiol 2016; 306:529-540. [PMID: 27476047 DOI: 10.1016/j.ijmm.2016.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 06/22/2016] [Accepted: 06/27/2016] [Indexed: 01/30/2023] Open
Abstract
There is a broad interest in adapting live vaccine strains (LVS) for use as recombinant vaccines that can deliver heterologous antigens. The Salmonella enterica serovar Typhimurium SL1344 ΔwecA LVS contains a mutation in wecA that abrogates production of Enterobacterial common antigen. This ΔwecA strain is attenuated in vivo, persistently colonizes the host, and protects against both wild type and cross-Salmonella serovar lethal challenge in a murine model of salmonellosis. Given these characteristics, we hypothesized that the SL1344 ΔwecA strain could be used as a carrier for heterologous antigen expression. To test this hypothesis, SL1344 ΔwecA was engineered to express the Pseudomonas aeruginosa O11 O-antigen gene cluster. Intraperitoneal (IP) but not oral immunization of BALB/c mice with the heterologous expression strain protected against lethal P. aeruginosa intranasal (IN) challenge. Furthermore, IP immunization resulted in P. aeruginosa O11-specific Ig and IgG antibody production. Functional analysis of sera collected from the IP immunized mice showed antibody-mediated agglutination and opsonophagocytic activity against P. aeruginosa. En masse, these results indicate that the S. Typhimurium SL1344 ΔwecA strain expressing the P. aeruginosa O11 O-antigen gene cluster is able to induce a humoral immune response and to protect against lethal P. aeruginosa challenge. As such, the S. Typhimurium SL1344 ΔwecA LVS can likely serve as a vehicle for expression of a wide variety of heterologous antigens as a means to create recombinant vaccines.
Collapse
Affiliation(s)
- Dacie R Bridge
- Department of Microbiology and Immunology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, United States.
| | - Jeannette M Whitmire
- Department of Microbiology and Immunology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, United States.
| | - Morris O Makobongo
- Department of Microbiology and Immunology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, United States.
| | - D Scott Merrell
- Department of Microbiology and Immunology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, United States.
| |
Collapse
|
48
|
Felgner S, Kocijancic D, Frahm M, Weiss S. Bacteria in Cancer Therapy: Renaissance of an Old Concept. Int J Microbiol 2016; 2016:8451728. [PMID: 27051423 PMCID: PMC4802035 DOI: 10.1155/2016/8451728] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 02/03/2016] [Accepted: 02/11/2016] [Indexed: 01/20/2023] Open
Abstract
The rising incidence of cancer cases worldwide generates an urgent need of novel treatment options. Applying bacteria may represent a valuable therapeutic variant that is intensively investigated nowadays. Interestingly, the idea to apply bacteria wittingly or unwittingly dates back to ancient times and was revived in the 19th century mainly by the pioneer William Coley. This review summarizes and compares the results of the past 150 years in bacteria mediated tumor therapy from preclinical to clinical studies. Lessons we have learned from the past provide a solid foundation on which to base future efforts. In this regard, several perspectives are discussed by which bacteria in addition to their intrinsic antitumor effect can be used as vector systems that shuttle therapeutic compounds into the tumor. Strategic solutions like these provide a sound and more apt exploitation of bacteria that may overcome limitations of conventional therapies.
Collapse
Affiliation(s)
- Sebastian Felgner
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Dino Kocijancic
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Michael Frahm
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Siegfried Weiss
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
49
|
Felgner S, Kocijancic D, Frahm M, Curtiss R, Erhardt M, Weiss S. Optimizing Salmonella enterica serovar Typhimurium for bacteria-mediated tumor therapy. Gut Microbes 2016; 7:171-7. [PMID: 26939530 PMCID: PMC4856459 DOI: 10.1080/19490976.2016.1155021] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Bacteria-mediated tumor therapy using Salmonella enterica serovar Typhimurium is a therapeutic option with great potential. Numerous studies explored the potential of Salmonella Typhimurium for therapeutic applications, however reconciling safety with vectorial efficacy remains a major issue. Recently we have described a conditionally attenuated Salmonella vector that is based on genetic lipopolysaccharide modification. This vector combines strong attenuation with appropriate anti-tumor properties by targeting various cancerous tissues in vivo. Therefore, it was promoted as an anti-tumor agent. In this addendum, we summarize these findings and demonstrate additional optimization steps that may further improve the therapeutic efficacy of our vector strain.
Collapse
Affiliation(s)
- Sebastian Felgner
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Dino Kocijancic
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Michael Frahm
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Roy Curtiss
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, USA
| | - Marc Erhardt
- Junior Research Group Infection Biology of Salmonella, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Siegfried Weiss
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany,Institute of Immunology, Medical School Hannover, Hannover, Germany
| |
Collapse
|
50
|
Zhang H, El Zowalaty ME. DNA-based influenza vaccines as immunoprophylactic agents toward universality. Future Microbiol 2015; 11:153-64. [PMID: 26673424 DOI: 10.2217/fmb.15.110] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Influenza is an illness of global public health concern. Influenza viruses have been responsible for several pandemics affecting humans. Current influenza vaccines have proved satisfactory safety; however, they have limitations and do not provide protection against unexpected emerging influenza virus strains. Therefore, there is an urgent need for alternative approaches to conventional influenza vaccines. The development of universal influenza vaccines will help alleviate the severity of influenza pandemics. Influenza DNA vaccines have been the subject of many studies over the past decades due to their ability to induce broad-based protective immune responses in various animal models. The present review highlights the recent advances in influenza DNA vaccine research and its potential as an affordable universal influenza vaccine.
Collapse
Affiliation(s)
- Han Zhang
- Department of Microbiology & Immunology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Mohamed E El Zowalaty
- Biomedical Research Center, Vice President Office for Research, Qatar University, Doha 2713, Qatar
| |
Collapse
|