1
|
He G, Long H, He J, Zhu C. The Immunomodulatory Effects and Applications of Probiotic Lactiplantibacillus plantarum in Vaccine Development. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10338-9. [PMID: 39101975 DOI: 10.1007/s12602-024-10338-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2024] [Indexed: 08/06/2024]
Abstract
Lactiplantibacillus plantarum (previously known as Lactobacillus plantarum) is a lactic acid bacterium that exists in various niches. L. plantarum is a food-grade microorganism that is commonly considered a safe and beneficial microorganism. It is widely used in food fermentation, agricultural enhancement, and environmental protection. L. plantarum is also part of the normal flora that can regulate the intestinal microflora and promote intestinal health. Some strains of L. plantarum are powerful probiotics that induce and modulate the innate and adaptive immune responses. Due to its outstanding immunoregulatory capacities, an increasing number of studies have examined the use of probiotic L. plantarum strains as natural immune adjuvants or alternative live vaccine carriers. The present review summarizes the main immunomodulatory characteristics of L. plantarum and discusses the preliminary immunological effects of L. plantarum as a vaccine adjuvant and delivery carrier. Different methods for improving the immune capacities of recombinant vector vaccines are also discussed.
Collapse
Affiliation(s)
- Guiting He
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, 421001, Hunan, China
| | - Huanbing Long
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, 421001, Hunan, China
| | - Jiarong He
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, 421001, Hunan, China
| | - Cuiming Zhu
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, 421001, Hunan, China.
| |
Collapse
|
2
|
Chen B, Yang Y, Wang Z, Dai X, Cao Y, Zhang M, Zhang D, Ni X, Zeng Y, Pan K. Surface Display of Duck Hepatitis A Virus Type 1 VP1 Protein on Bacillus subtilis Spores Elicits Specific Systemic and Mucosal Immune Responses on Mice. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10323-2. [PMID: 39002060 DOI: 10.1007/s12602-024-10323-2] [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] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
Duck viral hepatitis, primarily caused by duck hepatitis A virus type 1 (DHAV-1), poses a significant threat to the global duck industry. Bacillus subtilis is commonly utilized as a safe probiotic in the development of mucosal vaccines. In this study, a recombinant strain of B. subtilis, designated as B. subtilis RV, was constructed to display the DHAV-1 capsid protein VP1 on its spore surface using the outer coat protein B as an anchoring agent. The immunogenicity of this recombinant strain was evaluated in a mouse model through mixed feeding immunization. The results indicated that B. subtilis RV could elicit specific systemic and mucosal immune responses in mice, as evidenced by the high levels of serum IgG, intestinal secretory IgA, and potent virus-neutralizing antibodies produced. Furthermore, the recombinant strain significantly upregulated the expression levels of IL-2, IL-6, IL-10, TNF-α, and IFN-γ in the intestinal mucosa. Thus, the recombinant strain maintained the balance of the Th1/Th2 immune response and demonstrated an excellent mucosal immune adjuvant function. In summary, this study suggests that B. subtilis RV can be a novel alternative for effectively controlling DHAV-1 infection as a vaccine-based feed additive.
Collapse
Affiliation(s)
- Bin Chen
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Yang Yang
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Zhenhua Wang
- College of Animal Husbandry and Veterinary, Chengdu Agricultural College, Chengdu, 611130, China
| | - Xixi Dai
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang District, Chengdu, 611130, China
- Chongqing Three Gorges Vocational College, Chongqing, 404155, China
| | - Yuheng Cao
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Mengwei Zhang
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Dongmei Zhang
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Xueqin Ni
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Yan Zeng
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang District, Chengdu, 611130, China.
| | - Kangcheng Pan
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang District, Chengdu, 611130, China.
| |
Collapse
|
3
|
Zhang T, Qu H, Zheng W, Zhang Y, Li Y, Pan T, Li J, Yang W, Cao X, Jiang Y, Wang J, Zeng Y, Shi C, Huang H, Wang C, Yang G, Zhang J, Wang N. Oral vaccination with a recombinant Lactobacillus plantarum expressing the Eimeria tenella rhoptry neck 2 protein elicits protective immunity in broiler chickens infected with Eimeria tenella. Parasit Vectors 2024; 17:277. [PMID: 38943202 PMCID: PMC11212160 DOI: 10.1186/s13071-024-06355-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/15/2024] [Indexed: 07/01/2024] Open
Abstract
BACKGROUND Chicken coccidiosis is a protozoan disease that leads to considerable economic losses in the poultry industry. Live oocyst vaccination is currently the most effective measure for the prevention of coccidiosis. However, it provides limited protection with several drawbacks, such as poor immunological protection and potential reversion to virulence. Therefore, the development of effective and safe vaccines against chicken coccidiosis is still urgently needed. METHODS In this study, a novel oral vaccine against Eimeria tenella was developed by constructing a recombinant Lactobacillus plantarum (NC8) strain expressing the E. tenella RON2 protein. We administered recombinant L. plantarum orally at 3, 4 and 5 days of age and again at 17, 18 and 19 days of age. Meanwhile, each chick in the commercial vaccine group was immunized with 3 × 102 live oocysts of coccidia. A total of 5 × 104 sporulated oocysts of E. tenella were inoculated in each chicken at 30 days. Then, the immunoprotection effect was evaluated after E. tenella infection. RESULTS The results showed that the proportion of CD4+ and CD8+ T cells, the proliferative ability of spleen lymphocytes, inflammatory cytokine levels and specific antibody titers of chicks immunized with recombinant L. plantarum were significantly increased (P < 0.05). The relative body weight gains were increased and the number of oocysts per gram (OPG) was decreased after E. tenella challenge. Moreover, the lesion scores and histopathological cecum sections showed that recombinant L. plantarum can significantly relieve pathological damage in the cecum. The ACI was 170.89 in the recombinant L. plantarum group, which was higher than the 150.14 in the commercial vaccine group. CONCLUSIONS These above results indicate that L. plantarum expressing RON2 improved humoral and cellular immunity and enhanced immunoprotection against E. tenella. The protective efficacy was superior to that of vaccination with the commercial live oocyst vaccine. This study suggests that recombinant L. plantarum expressing the RON2 protein provides a promising strategy for vaccine development against coccidiosis.
Collapse
Affiliation(s)
- Tongxuan Zhang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Hangfan Qu
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Wei Zheng
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Yanan Zhang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Yanning Li
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Tianxu Pan
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Junyi Li
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Wentao Yang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Xin Cao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Yanlong Jiang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Jianzhong Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Yan Zeng
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Chunwei Shi
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Haibin Huang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Chunfeng Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Guilian Yang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, China.
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, China.
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| | - Jingwei Zhang
- College of Foreign Languages, Jilin Agricultural University, Changchun, 130118, China.
| | - Nan Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, China.
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, China.
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
4
|
Nolin SJ, Siegel PB, Ashwell CM. Differences in the microbiome of the small intestine of Leghorn lines divergently selected for antibody titer to sheep erythrocytes suggest roles for commensals in host humoral response. Front Physiol 2024; 14:1304051. [PMID: 38260103 PMCID: PMC10800846 DOI: 10.3389/fphys.2023.1304051] [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: 09/28/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
For forty generations, two lines of White Leghorn chickens have been selected for high (HAS) or low (LAS) antibody response to a low dose injection of sheep red blood cells (SRBCs). Their gut is home to billons of microorganisms and the largest number of immune cells in the body; therefore, the objective of this experiment was to gain understanding of the ways the microbiome may influence the differential antibody response observed in these lines. We achieved this by characterizing the small intestinal microbiome of HAS and LAS chickens, determining their functional microbiome profiles, and by using machine learning to identify microbes which best differentiate HAS from LAS and associating the abundance of those microbes with host gene expression. Microbiome sequencing revealed greater diversity in LAS but statistically higher abundance of several strains, particularly those of Lactobacillus, in HAS. Enrichment of microbial metabolites implicated in immune response such as lactic acid, short chain fatty acids, amino acids, and vitamins were different between HAS and LAS. The abundance of several microbial strains corresponds to enriched host gene expression pathways related to immune response. These data provide a compelling argument that the microbiome is both likely affected by host divergent genetic selection and that it exerts influence on host antibody response by various mechanisms.
Collapse
Affiliation(s)
- Shelly J. Nolin
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC, United States
| | - Paul B. Siegel
- School of Animal Science, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Christopher M. Ashwell
- Davis College of Agriculture, Natural Resources, and Design, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
5
|
Kobierecka P, Wyszyńska A, Aleksandrzak-Piekarczyk T, Sałańska A, Gawor J, Bardowski J, Jagusztyn Krynicka KE. Genomic and transcriptomic analysis of Ligilactobacillus salivarius IBB3154-in search of new promoters for vaccine construction. Microbiol Spectr 2023; 11:e0284423. [PMID: 37982628 PMCID: PMC10715006 DOI: 10.1128/spectrum.02844-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/16/2023] [Indexed: 11/21/2023] Open
Abstract
IMPORTANCE The genome of the strain Ligilactobacillus salivarius IBB3154 was sequenced, and transcriptome analysis was carried out at two different temperatures, allowing the determination of gene expression levels in response to environmental changes (temperature). Genes with higher expression at 42°C were identified. The use of a reporter gene (β- glucuronidase) did not confirm the transcriptomic results; it was found that the promoters of the genes sasA1 and sasA2 were active in the presence of bile salts. This opens up new opportunities for the overexpression of genes of other bacterial species in Ligilactobacillus cells in the intestinal environment.
Collapse
Affiliation(s)
- Patrycja Kobierecka
- Department of Bacterial Genetics, Institute of Microbiology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Agnieszka Wyszyńska
- Department of Bacterial Genetics, Institute of Microbiology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | | | - Agnieszka Sałańska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Jan Gawor
- DNA Sequencing and Synthesis Facility, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Jacek Bardowski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | |
Collapse
|
6
|
Xie W, Wang X, Cai J, Bai H, Shao Y, Li Z, Cai L, Zhang S, Li J, Cui W, Jiang Y, Tang L. Optimum Fermentation Conditions for Bovine Lactoferricin-Lactoferrampin-Encoding LimosiLactobacillus reuteri and Regulation of Intestinal Inflammation. Foods 2023; 12:4068. [PMID: 38002126 PMCID: PMC10670345 DOI: 10.3390/foods12224068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/02/2023] [Accepted: 11/04/2023] [Indexed: 11/26/2023] Open
Abstract
The multifunctional antibacterial peptide lactoferricin-lactoferrampin (LFCA) is derived from bovine lactoferrin. Optimization of the fermentation process should be studied since different microorganisms have their own favorable conditions and processes for growth and the production of metabolites. In this study, the culture conditions of a recombinant strain, pPG-LFCA-E/LR-CO21 (LR-LFCA), expressing LFCA was optimized, utilizing the high-density fermentation process to augment the biomass of LimosiLactobacillus reuteri and the expression of LFCA. Furthermore, an assessment of the protective effect of LR-LFCA on intestinal inflammation induced by lipopolysaccharide (LPS) was conducted to evaluate the impact of LR-LFCA on the disease resistance of piglets. The findings of this study indicate that LR-LFCA fermentation conditions optimally include 2% inoculation volume, 36.5 °C fermentation temperature, 9% dissolved oxygen concentration, 200 revolutions/minute stirring speed, pH 6, 10 mL/h glucose flow, and 50% glucose concentration. The inclusion of fermented LR-LFCA in the diet resulted in an elevation of immunoglobulin levels, significant upregulation of tight junction proteins ZO-1 and occludin, reinforcement of the intestinal barrier function, and significant amelioration of the aberrant alterations in blood physiological parameters induced by LPS. These results offer a theoretical framework for the implementation of this micro-ecological preparation in the field of piglet production to enhance intestinal well-being.
Collapse
Affiliation(s)
- Weichun Xie
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (W.X.); (X.W.); (J.C.); (H.B.); (Y.S.); (Z.L.); (L.C.); (S.Z.); (J.L.); (W.C.)
| | - Xueying Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (W.X.); (X.W.); (J.C.); (H.B.); (Y.S.); (Z.L.); (L.C.); (S.Z.); (J.L.); (W.C.)
| | - Jiyao Cai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (W.X.); (X.W.); (J.C.); (H.B.); (Y.S.); (Z.L.); (L.C.); (S.Z.); (J.L.); (W.C.)
| | - Huitao Bai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (W.X.); (X.W.); (J.C.); (H.B.); (Y.S.); (Z.L.); (L.C.); (S.Z.); (J.L.); (W.C.)
| | - Yilan Shao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (W.X.); (X.W.); (J.C.); (H.B.); (Y.S.); (Z.L.); (L.C.); (S.Z.); (J.L.); (W.C.)
| | - Zhuoran Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (W.X.); (X.W.); (J.C.); (H.B.); (Y.S.); (Z.L.); (L.C.); (S.Z.); (J.L.); (W.C.)
| | - Limeng Cai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (W.X.); (X.W.); (J.C.); (H.B.); (Y.S.); (Z.L.); (L.C.); (S.Z.); (J.L.); (W.C.)
| | - Senhao Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (W.X.); (X.W.); (J.C.); (H.B.); (Y.S.); (Z.L.); (L.C.); (S.Z.); (J.L.); (W.C.)
| | - Jiaxuan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (W.X.); (X.W.); (J.C.); (H.B.); (Y.S.); (Z.L.); (L.C.); (S.Z.); (J.L.); (W.C.)
| | - Wen Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (W.X.); (X.W.); (J.C.); (H.B.); (Y.S.); (Z.L.); (L.C.); (S.Z.); (J.L.); (W.C.)
| | - Yanping Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (W.X.); (X.W.); (J.C.); (H.B.); (Y.S.); (Z.L.); (L.C.); (S.Z.); (J.L.); (W.C.)
| | - Lijie Tang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (W.X.); (X.W.); (J.C.); (H.B.); (Y.S.); (Z.L.); (L.C.); (S.Z.); (J.L.); (W.C.)
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
7
|
Chau ECT, Kwong TC, Pang CK, Chan LT, Chan AML, Yao X, Tam JSL, Chan SW, Leung GPH, Tai WCS, Kwan YW. A Novel Probiotic-Based Oral Vaccine against SARS-CoV-2 Omicron Variant B.1.1.529. Int J Mol Sci 2023; 24:13931. [PMID: 37762235 PMCID: PMC10530581 DOI: 10.3390/ijms241813931] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
COVID-19 pandemic, caused by the SARS-CoV-2 virus, is still affecting the entire world via the rapid emergence of new contagious variants. Vaccination remains the most effective prevention strategy for viral infection, yet not all countries have sufficient access to vaccines due to limitations in manufacturing and transportation. Thus, there is an urgent need to develop an easy-to-use, safe, and low-cost vaccination approach. Genetically modified microorganisms, especially probiotics, are now commonly recognized as attractive vehicles for delivering bioactive molecules via oral and mucosal routes. In this study, Lactobacillus casei has been selected as the oral vaccine candidate based on its' natural immunoadjuvant properties and the ability to resist acidic gastric environment, to express antigens of SARS-CoV-2 Omicron variant B.1.1.529 with B-cell and T-cell epitopes. This newly developed vaccine, OMGVac, was shown to elicit a robust IgG systemic immune response against the spike protein of Omicron variant B.1.1.529 in Golden Syrian hamsters. No adverse effects were found throughout this study, and the overall safety was evaluated in terms of physiological and histopathological examinations of different organs harvested. In addition, this study illustrated the use of the recombinant probiotic as a live delivery vector in the initiation of systemic immunity, which shed light on the future development of next-generation vaccines to combat emerging infectious diseases.
Collapse
Affiliation(s)
- Eddie Chung Ting Chau
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China; (E.C.T.C.); (T.C.K.); (C.K.P.); (L.T.C.); (A.M.L.C.); (X.Y.)
| | - Tsz Ching Kwong
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China; (E.C.T.C.); (T.C.K.); (C.K.P.); (L.T.C.); (A.M.L.C.); (X.Y.)
| | - Chun Keung Pang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China; (E.C.T.C.); (T.C.K.); (C.K.P.); (L.T.C.); (A.M.L.C.); (X.Y.)
| | - Lee Tung Chan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China; (E.C.T.C.); (T.C.K.); (C.K.P.); (L.T.C.); (A.M.L.C.); (X.Y.)
| | - Andrew Man Lok Chan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China; (E.C.T.C.); (T.C.K.); (C.K.P.); (L.T.C.); (A.M.L.C.); (X.Y.)
| | - Xiaoqiang Yao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China; (E.C.T.C.); (T.C.K.); (C.K.P.); (L.T.C.); (A.M.L.C.); (X.Y.)
| | - John Siu Lun Tam
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China; (J.S.L.T.); (W.C.S.T.)
| | - Shun Wan Chan
- Department of Food and Health Sciences, Faculty of Science and Technology, Technological and Higher Education Institute of Hong Kong, Hong Kong, China;
| | - George Pak Heng Leung
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China;
| | - William Chi Shing Tai
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China; (J.S.L.T.); (W.C.S.T.)
| | - Yiu Wa Kwan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China; (E.C.T.C.); (T.C.K.); (C.K.P.); (L.T.C.); (A.M.L.C.); (X.Y.)
| |
Collapse
|
8
|
Ortiz Moyano R, Raya Tonetti F, Fukuyama K, Elean M, Tomokiyo M, Suda Y, Melnikov V, Kitazawa H, Villena J. The Respiratory Commensal Bacterium Corynebacterium pseudodiphtheriticum as a Mucosal Adjuvant for Nasal Vaccines. Vaccines (Basel) 2023; 11:vaccines11030611. [PMID: 36992195 PMCID: PMC10058227 DOI: 10.3390/vaccines11030611] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
Previously, we demonstrated that nasally administered Corynebacterium pseudodiphteriticum 090104 (Cp) or its bacterium-like particles (BLPs) increase the resistance of mice against bacterial and viral respiratory pathogens by modulating the innate immunity. In this work, we evaluated the ability of Cp and BLPs to stimulate alveolar macrophages, and to enhance the humoral immune response induced by a commercial vaccine against Streptococcus pneumoniae. In the first set of experiments, Cp or the BLPs were incubated with primary cultures of murine alveolar macrophages and the phagocytic activity, and the production of cytokines was evaluated. The results revealed that Cp and BLPs were efficiently phagocyted by respiratory macrophages and that both treatments triggered the production of TNF-α, IFN-γ, IL-6, and IL-1β. In the second set of experiments, 3-week-old Swiss mice were intranasally immunized at days 0, 14, and 28 with the pneumococcal vaccine Prevenar®13 (PCV), Cp + PCV, or BLPs + PCV. On day 33, samples of bronco-alveolar lavages (BAL) and serum were collected for the study of specific antibodies. In addition, immunized mice were challenged with S. pneumoniae serotypes 6B or 19F on day 33 and sacrificed on day 35 (day 2 post-infection) to evaluate the resistance to the infection. Both Cp + PCV and BLPs + PCV groups had higher specific serum IgG and BAL IgA antibodies than the PCV control mice. In addition, the mice that were immunized with Cp + PCV or BLPs + PCV had lower lung and blood pneumococcal cell counts as well as lower levels of BAL albumin and LDH, indicating a reduced lung damage compared to the control mice. Improved levels of anti-pneumococcal antibodies were also detected in the serum and BAL samples after the challenges with the pathogens. The results demonstrated that C. pseudodiphteriticum 090104 and its bacterium-like particles are capable of stimulating the respiratory innate immune system serving as adjuvants to potentiate the adaptive humoral immune response. Our study is a step forward in the positioning of this respiratory commensal bacterium as a promising mucosal adjuvant for vaccine formulations aimed at combating respiratory infectious diseases.
Collapse
Affiliation(s)
- Ramiro Ortiz Moyano
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucumán 4000, Argentina; (R.O.M.); (F.R.T.); (M.E.)
| | - Fernanda Raya Tonetti
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucumán 4000, Argentina; (R.O.M.); (F.R.T.); (M.E.)
| | - Kohtaro Fukuyama
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan; (K.F.); (M.T.)
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| | - Mariano Elean
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucumán 4000, Argentina; (R.O.M.); (F.R.T.); (M.E.)
| | - Mikado Tomokiyo
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan; (K.F.); (M.T.)
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| | - Yoshihito Suda
- Department of Food, Agriculture and Environment, Miyagi University, Sendai 980-8572, Japan;
| | - Vyacheslav Melnikov
- Gabrichevsky Research Institute for Epidemiology and Microbiology, 125212 Moscow, Russia;
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan; (K.F.); (M.T.)
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
- Correspondence: (H.K.); (J.V.)
| | - Julio Villena
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucumán 4000, Argentina; (R.O.M.); (F.R.T.); (M.E.)
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan; (K.F.); (M.T.)
- Correspondence: (H.K.); (J.V.)
| |
Collapse
|
9
|
Hendy DA, Haven A, Bachelder EM, Ainslie KM. Preclinical developments in the delivery of protein antigens for vaccination. Expert Opin Drug Deliv 2023; 20:367-384. [PMID: 36731824 PMCID: PMC9992317 DOI: 10.1080/17425247.2023.2176844] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 02/01/2023] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Vaccine technology has constantly advanced since its origin. One of these advancements is where purified parts of a pathogen are used rather than the whole pathogen. Subunit vaccines have no chance of causing disease; however, alone these antigens are often poorly immunogenic. Therefore, they can be paired with immune stimulating adjuvants. Further, subunits can be combined with delivery strategies such as nano/microparticles to enrich their delivery to organs and cells of interest as well as protect them from in vivo degradation. Here, we seek to highlight some of the more promising delivery strategies for protein antigens. AREAS COVERED We present a brief description of the different types of vaccines, clinically relevant examples, and their disadvantages when compared to subunit vaccines. Also, specific preclinical examples of delivery strategies for protein antigens. EXPERT OPINION Subunit vaccines provide optimal safety given that they have no risk of causing disease; however, they are often not immunogenic enough on their own to provide protection. Advanced delivery systems are a promising avenue to increase the immunogenicity of subunit vaccines, but scalability and stability can be improved. Further, more research is warranted on systems that promote a mucosal immune response to provide better protection against infection.
Collapse
Affiliation(s)
- Dylan A. Hendy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Alex Haven
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Eric M. Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Kristy M. Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA
- Department of Microbiology and Immunology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
10
|
Immunomodulatory action of Lactococcuslactis. J Biosci Bioeng 2023; 135:1-9. [PMID: 36428209 DOI: 10.1016/j.jbiosc.2022.10.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/07/2022] [Accepted: 10/24/2022] [Indexed: 11/24/2022]
Abstract
Fermented foods are gaining popularity due to health-promoting properties with high levels of nutrients, phytochemicals, bioactive compounds, and probiotic microorganisms. Due to its unique fermentation process, Lactococcus lactis plays a key role in the food business, notably in the manufacturing of dairy products. The superior biological activities of L. lactis in these functional foods include anti-inflammatory and immunomodulatory capabilities. L. lactis boosted growth performance, controlled amino acid profiles, intestinal immunology, and microbiota. Besides that, the administration of L. lactis increased the rate of infection clearance. Innate and acquired immune responses would be upregulated in both local and systemic compartments, resulting in these consequences. L. lactis is often employed in the food sector and is currently being exploited as a delivery vehicle for biological research. These bacteria are being eyed as potential candidates for biotechnological applications. With this in mind, we reviewed the immunomodulatory effects of different L. lactis strains.
Collapse
|
11
|
Endotoxin-free gram-negative bacterium as a system for production and secretion of recombinant proteins. Appl Microbiol Biotechnol 2022; 107:287-298. [DOI: 10.1007/s00253-022-12295-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 12/03/2022]
|
12
|
Accelerated water removal from frozen thin films containing bacteria. Int J Pharm 2022; 630:122408. [PMID: 36400132 DOI: 10.1016/j.ijpharm.2022.122408] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 10/31/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022]
Abstract
Freeze-drying, or lyophilization, is widely used to produce pharmaceutical solids that contain temperature-sensitive materials. Herein, using Escherichia coli as a model live organism, whose viability in dry powders is highly sensitive to the water content in the powders, we demonstrated that the drying rate from the frozen thin films generated by thin-film freezing (TFF) is significantly faster than from the bulk frozen solids in conventional shelf freeze-drying. This is likely because the loosely stacked frozen thin films provided a larger solid-air interface and the low thickness of the thin films provided a low mass transfer resistance. The highly porous microstructure and high specific surface area of the thin-film freeze-dried powders may also be related to the faster drying observed. Moreover, we demonstrated that TFF can be applied to produce dry powders of E. coli, a Gram-negative bacterium, or Lactobacillus acidophilus, a Gram-positive bacterium, with minimum bacterial viability loss (i.e., within one log reduction). It is concluded that the TFF technology is promising in accelerating water removal from frozen samples.
Collapse
|
13
|
Lactic Acid Bacteria as Mucosal Immunity Enhancers and Antivirals through Oral Delivery. Appl Microbiol 2022. [DOI: 10.3390/applmicrobiol2040064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Mucosal vaccination offer an advantage over systemic inoculation from the immunological viewpoint. The development of an efficient vaccine is now a priority for emerging diseases such as COVID-19, that was declared a pandemic in 2020 and caused millions of deaths globally. Lactic acid bacteria (LAB) especially Lactobacillus are the vital microbiota of the gut, which is observed as having valuable effects on animals’ and human health. LAB produce lactic acid as the major by-product of carbohydrate degradation and play a significant role in innate immunity enhancement. LAB have significant characteristics to mimic pathogen infections and intrinsically possess adjuvant properties to enhance mucosal immunity. Increasing demand and deliberations are being substantially focused on probiotic organisms that can enhance mucosal immunity against viral diseases. LAB can also strengthen their host’s antiviral defense system by producing antiviral peptides, and releasing metabolites that prevent viral infections and adhesion to mucosal surfaces. From the perspectives of “one health” and the use of probiotics, conventional belief has opened up a new horizon on the use of LAB as antivirals. The major interest of this review is to depict the beneficial use of LAB as antivirals and mucosal immunity enhancers against viral diseases.
Collapse
|
14
|
Levit R, Cortes-Perez NG, de Moreno de Leblanc A, Loiseau J, Aucouturier A, Langella P, LeBlanc JG, Bermúdez-Humarán LG. Use of genetically modified lactic acid bacteria and bifidobacteria as live delivery vectors for human and animal health. Gut Microbes 2022; 14:2110821. [PMID: 35960855 PMCID: PMC9377234 DOI: 10.1080/19490976.2022.2110821] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
There is now strong evidence to support the interest in using lactic acid bacteria (LAB)in particular, strains of lactococci and lactobacilli, as well as bifidobacteria, for the development of new live vectors for human and animal health purposes. LAB are Gram-positive bacteria that have been used for millennia in the production of fermented foods. In addition, numerous studies have shown that genetically modified LAB and bifodobacteria can induce a systemic and mucosal immune response against certain antigens when administered mucosally. They are therefore good candidates for the development of new mucosal delivery strategies and are attractive alternatives to vaccines based on attenuated pathogenic bacteria whose use presents health risks. This article reviews the most recent research and advances in the use of LAB and bifidobacteria as live delivery vectors for human and animal health.
Collapse
Affiliation(s)
- Romina Levit
- Centro de Referencia para Lactobacilos (CERELA-CONICET), Chacabuco 145, (T4000ILC) San Miguel de Tucumán, Tucumán, Argentina
| | - Naima G. Cortes-Perez
- Université Paris-Saclay, INRAE, AgroParisTech, UMR 0496, 78350 Jouy-en-Josas, France
| | - Alejandra de Moreno de Leblanc
- Centro de Referencia para Lactobacilos (CERELA-CONICET), Chacabuco 145, (T4000ILC) San Miguel de Tucumán, Tucumán, Argentina
| | - Jade Loiseau
- Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France
| | - Anne Aucouturier
- Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France
| | - Philippe Langella
- Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France
| | - Jean Guy LeBlanc
- Centro de Referencia para Lactobacilos (CERELA-CONICET), Chacabuco 145, (T4000ILC) San Miguel de Tucumán, Tucumán, Argentina
| | - Luis G. Bermúdez-Humarán
- Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France,CONTACT Luis G. Bermúdez-Humarán Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France
| |
Collapse
|
15
|
Jia Z, Pan X, Zhi W, Chen H, Bai B, Ma C, Ma D. Probiotics Surface-Delivering Fiber2 Protein of Fowl Adenovirus 4 Stimulate Protective Immunity Against Hepatitis-Hydropericardium Syndrome in Chickens. Front Immunol 2022; 13:919100. [PMID: 35837390 PMCID: PMC9273852 DOI: 10.3389/fimmu.2022.919100] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/27/2022] [Indexed: 12/19/2022] Open
Abstract
Background and ObjectivesHepatitis-hydropericardium syndrome (HHS) caused by Fowl adenoviruses serotype 4 (FAdV-4) leads to severe economic losses to the poultry industry. Although various vaccines are available, vaccines that effectively stimulate intestinal mucosal immunity are still deficient. In the present study, novel probiotics that surface-deliver Fiber2 protein, the major virulence determiner and efficient immunogen for FAdV-4, were explored to prevent this fecal–oral-transmitted virus, and the induced protective immunity was evaluated after oral immunization.MethodsThe probiotic Enterococcus faecalis strain MDXEF-1 and Lactococcus lactis NZ9000 were used as host strains to deliver surface-anchoring Fiber2 protein of FAdV-4. Then the constructed live recombinant bacteria were orally vaccinated thrice with chickens at intervals of 2 weeks. Following each immunization, immunoglobulin G (IgG) in sera, secretory immunoglobulin A (sIgA) in jejunum lavage, immune-related cytokines, and T-cell proliferation were detected. Following challenge with the highly virulent FAdV-4, the protective effects of the probiotics surface-delivering Fiber2 protein were evaluated by verifying inflammatory factors, viral load, liver function, and survival rate.ResultsThe results demonstrated that probiotics surface-delivering Fiber2 protein stimulated humoral and intestinal mucosal immune responses in chickens, shown by high levels of sIgA and IgG antibodies, substantial rise in mRNA levels of cytokines, increased proliferative ability of T cells in peripheral blood, improved liver function, and reduced viral load in liver. Accordingly, adequate protection against homologous challenges and a significant increase in the overall survival rate were observed. Notably, chickens orally immunized with E. faecalis/DCpep-Fiber2-CWA were completely protected from the FAdV-4 challenge, which is better than L. lactis/DCpep-Fiber2-CWA.ConclusionThe recombinant probiotics surface-expressing Fiber2 protein could evoke remarkable humoral and cellular immune responses, relieve injury, and functionally damage target organs. The current study indicates a promising method used for preventing FAdV-4 infection in chickens.
Collapse
Affiliation(s)
- Zhipeng Jia
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xinghui Pan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Wenjing Zhi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hang Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Bingrong Bai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Chunli Ma
- College of Food Science, Northeast Agricultural University, Harbin, China
- *Correspondence: Chunli Ma, ; Dexing Ma,
| | - Dexing Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Experimental Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
- *Correspondence: Chunli Ma, ; Dexing Ma,
| |
Collapse
|
16
|
Wiull K, Boysen P, Kuczkowska K, Moen LF, Carlsen H, Eijsink VGH, Mathiesen G. Comparison of the Immunogenic Properties of Lactiplantibacillus plantarum Carrying the Mycobacterial Ag85B-ESAT-6 Antigen at Various Cellular Localizations. Front Microbiol 2022; 13:900922. [PMID: 35722346 PMCID: PMC9204040 DOI: 10.3389/fmicb.2022.900922] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
The bacille Calmette-Guèrin (BCG) vaccine has been used for a century; nonetheless, tuberculosis (TB) remains one of the deadliest diseases in the world. Thus, new approaches to developing a new, more efficient vaccine are desirable. Mucosal vaccines are of particular interest, considering that Mycobacterium tuberculosis first enters the body through the mucosal membranes. We have previously demonstrated the immunogenicity of a recombinant Lactiplantibacillus plantarum delivery vector with TB hybrid antigen Ag85B-ESAT-6 anchored to the cell membrane. The goal of the present study was to analyze the impact of antigen localization in the immune response. Thus, we assessed two novel vaccine candidates, with the TB antigen either non-covalently anchored to the cell wall (LysMAgE6) or located intracellularly (CytAgE6). In addition, we compared two expression systems, using an inducible (LipoAgE6) or a constitutive promoter (cLipoAgE6) for expression of covalently anchored antigen to the cell membrane. Following administration to mice, antigen-specific CD4+ T-cell proliferation and IFN-γ and IL-17A secretion were analyzed for lung cell and splenocyte populations. Generally, the immune response in lung cells was stronger compared to splenocytes. The analyses showed that the type of expression system did not significantly affect the immunogenicity, while various antigen localizations resulted in markedly different responses. The immune response was considerably stronger for the surface-displaying candidate strains compared to the candidate with an intracellular antigen. These findings emphasize the significance of antigen exposure and further support the potential of L. plantarum as a mucosal vaccine delivery vehicle in the fight against TB.
Collapse
Affiliation(s)
- Kamilla Wiull
- Faculty of Chemistry, Biotechnology and Food Science, NMBU - Norwegian University of Life Sciences, Ås, Norway
- *Correspondence: Kamilla Wiull,
| | - Preben Boysen
- Faculty of Veterinary Medicine, NMBU - Norwegian University of Life Sciences, Ås, Norway
| | - Katarzyna Kuczkowska
- Faculty of Chemistry, Biotechnology and Food Science, NMBU - Norwegian University of Life Sciences, Ås, Norway
| | - Lars Fredrik Moen
- Faculty of Chemistry, Biotechnology and Food Science, NMBU - Norwegian University of Life Sciences, Ås, Norway
| | - Harald Carlsen
- Faculty of Chemistry, Biotechnology and Food Science, NMBU - Norwegian University of Life Sciences, Ås, Norway
| | - Vincent G. H. Eijsink
- Faculty of Chemistry, Biotechnology and Food Science, NMBU - Norwegian University of Life Sciences, Ås, Norway
| | - Geir Mathiesen
- Faculty of Chemistry, Biotechnology and Food Science, NMBU - Norwegian University of Life Sciences, Ås, Norway
- Geir Mathiesen,
| |
Collapse
|
17
|
Kazi TA, Acharya A, Mukhopadhyay BC, Mandal S, Arukha AP, Nayak S, Biswas SR. Plasmid-Based Gene Expression Systems for Lactic Acid Bacteria: A Review. Microorganisms 2022; 10:1132. [PMID: 35744650 PMCID: PMC9229153 DOI: 10.3390/microorganisms10061132] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 01/27/2023] Open
Abstract
Lactic acid bacteria (LAB) play a very vital role in food production, preservation, and as probiotic agents. Some of these species can colonize and survive longer in the gastrointestinal tract (GIT), where their presence is crucially helpful to promote human health. LAB has also been used as a safe and efficient incubator to produce proteins of interest. With the advent of genetic engineering, recombinant LAB have been effectively employed as vectors for delivering therapeutic molecules to mucosal tissues of the oral, nasal, and vaginal tracks and for shuttling therapeutics for diabetes, cancer, viral infections, and several gastrointestinal infections. The most important tool needed to develop genetically engineered LABs to produce proteins of interest is a plasmid-based gene expression system. To date, a handful of constitutive and inducible vectors for LAB have been developed, but their limited availability, host specificity, instability, and low carrying capacity have narrowed their spectrum of applications. The current review discusses the plasmid-based vectors that have been developed so far for LAB; their functionality, potency, and constraints; and further highlights the need for a new, more stable, and effective gene expression platform for LAB.
Collapse
Affiliation(s)
- Tawsif Ahmed Kazi
- Department of Botany, Visva-Bharati University, Santiniketan 731235, West Bengal, India; (T.A.K.); (A.A.); (B.C.M.)
| | - Aparupa Acharya
- Department of Botany, Visva-Bharati University, Santiniketan 731235, West Bengal, India; (T.A.K.); (A.A.); (B.C.M.)
| | - Bidhan Chandra Mukhopadhyay
- Department of Botany, Visva-Bharati University, Santiniketan 731235, West Bengal, India; (T.A.K.); (A.A.); (B.C.M.)
| | - Sukhendu Mandal
- Laboratory of Molecular Bacteriology, Department of Microbiology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India;
| | - Ananta Prasad Arukha
- Researcher 5 Department of Neurosurgery, Medical School, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Subhendu Nayak
- Sr. Scientist, Clorox, Better Health VMS, Durham, NC 27701, USA;
| | - Swadesh Ranjan Biswas
- Department of Botany, Visva-Bharati University, Santiniketan 731235, West Bengal, India; (T.A.K.); (A.A.); (B.C.M.)
| |
Collapse
|
18
|
Fox BE, Vilander A, Abdo Z, Dean GA. NOD2 signaling in CD11c + cells is critical for humoral immune responses during oral vaccination and maintaining the gut microbiome. Sci Rep 2022; 12:8491. [PMID: 35589853 PMCID: PMC9119386 DOI: 10.1038/s41598-022-12469-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 05/11/2022] [Indexed: 11/09/2022] Open
Abstract
Nucleotide-binding oligomerization domain containing 2 (NOD2) is a critical regulator of immune responses within the gastrointestinal tract. This innate immune receptor is expressed by several cell types, including both hematopoietic and nonhematopoietic cells within the gastrointestinal tract. Vaccination targeting the gastrointestinal mucosal immune system is especially difficult due to both physical and mechanistic barriers to reaching inductive sites. The use of lactic acid bacteria is appealing due to their ability to persist within harsh conditions, expression of selected adjuvants, and manufacturing advantages. Recombinant Lactobacillus acidophilus (rLA) has shown great promise in activating the mucosal immune response with minimal impacts on the resident microbiome. To better classify the kinetics of mucosal vaccination with rLA, we utilized mice harboring knockouts of NOD2 expression specifically within CD11c + cells. The results presented here show that NOD2 signaling in CD11c + cells is necessary for mounting a humoral immune response against exogenous antigens expressed by rLA. Additionally, disruption of NOD2 signaling in these cells results in an altered bacterial microbiome profile in both control mice and mice receiving L. acidophilus strain NCK1895 and vaccine strain LaOVA.
Collapse
Affiliation(s)
- B E Fox
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA.
| | - A Vilander
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Z Abdo
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA.
| | - G A Dean
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA.
| |
Collapse
|
19
|
Porfiri L, Burtscher J, Kangethe RT, Verhovsek D, Cattoli G, Domig KJ, Wijewardana V. Irradiated Non-replicative Lactic Acid Bacteria Preserve Metabolic Activity While Exhibiting Diverse Immune Modulation. Front Vet Sci 2022; 9:859124. [PMID: 35664846 PMCID: PMC9158532 DOI: 10.3389/fvets.2022.859124] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/07/2022] [Indexed: 11/23/2022] Open
Abstract
In the recent years, safety concerns regarding the administration of probiotics led to an increased interest in developing inactivated probiotics, also called “paraprobiotics”. Gamma irradiation represents a promising tool that can be used to produce safe paraprobiotics by inhibiting replication while preserving the structure, the metabolic activity, and the immunogenicity of bacteria. In this study, we evaluated the ability of four strains of lactic acid bacteria (LAB: Lacticaseibacillus casei, Lactobacillus acidophilus, Lactiplantibacillus plantarum, and Lacticaseibacillus paracasei) in preserving the metabolic activity and the immune modulation of swine porcine peripheral blood mononuclear cells, after gamma irradiation or heat inactivation. Our results show that all four strains retained the metabolic activity following gamma irradiation but not after heat inactivation. In terms of immune-modulatory capacity, irradiated L. acidophilus and Lc. paracasei were able to maintain an overall gene expression pattern similar to their live state, as heat inactivation did with Lc. casei. Moreover, we show that the two inactivation methods applied to the same strain can induce an opposed expression of key genes involved in pro-inflammatory response (e.g., IFNα and interleukin-6 for Lc. casei), whereas gamma irradiation of L. acidophilus and Lc. paracasei was able to induce a downregulation of the anti-inflammatory TGFβ. Taken together, our data show that immune modulation can be impacted not only by different inactivation methods but also by the strain of LAB selected. This study highlights that gamma irradiation harbors the potential to produce safe non-replicative metabolically active LAB and identifies immunomodulatory capacities that may be applied as vaccine adjuvants.
Collapse
Affiliation(s)
- Luca Porfiri
- Animal Production and Health Section, Joint Food and Agriculture Organization (FAO)/International Atomic Energy Agency (IAEA) Centre of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna, Austria
| | - Johanna Burtscher
- Department of Food Science and Technology, Institute of Food Science, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Richard T. Kangethe
- Animal Production and Health Section, Joint Food and Agriculture Organization (FAO)/International Atomic Energy Agency (IAEA) Centre of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna, Austria
| | - Doris Verhovsek
- VetFarm Medau, University of Veterinary Medicine Vienna, Berndorf, Austria
| | - Giovanni Cattoli
- Animal Production and Health Section, Joint Food and Agriculture Organization (FAO)/International Atomic Energy Agency (IAEA) Centre of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna, Austria
| | - Konrad J. Domig
- Department of Food Science and Technology, Institute of Food Science, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Viskam Wijewardana
- Animal Production and Health Section, Joint Food and Agriculture Organization (FAO)/International Atomic Energy Agency (IAEA) Centre of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna, Austria
- *Correspondence: Viskam Wijewardana
| |
Collapse
|
20
|
Petrova P, Arsov A, Tsvetanova F, Parvanova-Mancheva T, Vasileva E, Tsigoriyna L, Petrov K. The Complex Role of Lactic Acid Bacteria in Food Detoxification. Nutrients 2022; 14:2038. [PMID: 35631179 PMCID: PMC9147554 DOI: 10.3390/nu14102038] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/02/2022] [Accepted: 05/10/2022] [Indexed: 12/13/2022] Open
Abstract
Toxic ingredients in food can lead to serious food-related diseases. Such compounds are bacterial toxins (Shiga-toxin, listeriolysin, Botulinum toxin), mycotoxins (aflatoxin, ochratoxin, zearalenone, fumonisin), pesticides of different classes (organochlorine, organophosphate, synthetic pyrethroids), heavy metals, and natural antinutrients such as phytates, oxalates, and cyanide-generating glycosides. The generally regarded safe (GRAS) status and long history of lactic acid bacteria (LAB) as essential ingredients of fermented foods and probiotics make them a major biological tool against a great variety of food-related toxins. This state-of-the-art review aims to summarize and discuss the data revealing the involvement of LAB in the detoxification of foods from hazardous agents of microbial and chemical nature. It is focused on the specific properties that allow LAB to counteract toxins and destroy them, as well as on the mechanisms of microbial antagonism toward toxigenic producers. Toxins of microbial origin are either adsorbed or degraded, toxic chemicals are hydrolyzed and then used as a carbon source, while heavy metals are bound and accumulated. Based on these comprehensive data, the prospects for developing new combinations of probiotic starters for food detoxification are considered.
Collapse
Affiliation(s)
- Penka Petrova
- Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (P.P.); (A.A.)
| | - Alexander Arsov
- Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (P.P.); (A.A.)
| | - Flora Tsvetanova
- Institute of Chemical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (F.T.); (T.P.-M.); (E.V.); (L.T.)
| | - Tsvetomila Parvanova-Mancheva
- Institute of Chemical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (F.T.); (T.P.-M.); (E.V.); (L.T.)
| | - Evgenia Vasileva
- Institute of Chemical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (F.T.); (T.P.-M.); (E.V.); (L.T.)
| | - Lidia Tsigoriyna
- Institute of Chemical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (F.T.); (T.P.-M.); (E.V.); (L.T.)
| | - Kaloyan Petrov
- Institute of Chemical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (F.T.); (T.P.-M.); (E.V.); (L.T.)
| |
Collapse
|
21
|
Oral Immunization of Mice with Cell Extracts from Recombinant Lactococcus lactis Expressing SARS-CoV-2 Spike Protein. Curr Microbiol 2022; 79:167. [PMID: 35460453 PMCID: PMC9034443 DOI: 10.1007/s00284-022-02866-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 04/05/2022] [Indexed: 12/22/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread all over the world and became a pandemic that named coronavirus disease-2019 (COVID-19). At present, several intramuscular vaccines have been successfully developed and mass vaccination has progressed in many countries. The aim of the study is to develop and examine an oral vaccine against COVID-19 with recombinant Lactococcus lactis IL1403, a strain of lactic acid bacteria, expressing SARS-CoV-2 spike (S) protein receptor-binding domain (RBD) S1 subunit as an immunizing antigen. PBS or cell extracts from recombinant L. lactis were orally administered into mice (control VS treatment), and formation of antigen-specific antibodies and changes in the gut microbiome were analyzed. Intracellular antigen was detected, but its secretion was not successful. After immunization, antigen-specific serum IgG and fecal IgA levels were 1.5-fold (P = 0.002) and 1.4-fold (P = 0.016) higher in the immunized mice (treatment) than control, respectively. Gut microbiome profiles were clearly separated between the two groups when analyzed for beta diversity with overall similarity. At the genus level, while Coprococcus (P = 0.036) and unclassified genus of Ruminococcaceae (P = 0.037) in treatment were more abundant than control, rc4-4 (P = 0.013) and Stenotrophomonas (P = 0.021) were less abundant. Our results indicate that cell extract containing SARS-CoV-2 antigen can induce mice to produce antigen-specific antibodies without overall changes in the gut microbiome. This strategy may be useful for the development of other oral viral vaccines.
Collapse
|
22
|
Wang H, Li P, Zhang M, Bi J, He Y, Li F, Yu R, Gao F, Kong W, Yu B, Chen L, Yu X. Vaccine with bacterium-like particles displaying HIV-1 gp120 trimer elicits specific mucosal responses and neutralizing antibodies in rhesus macaques. Microb Biotechnol 2022; 15:2022-2039. [PMID: 35290714 PMCID: PMC9249329 DOI: 10.1111/1751-7915.14022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 02/06/2022] [Indexed: 11/27/2022] Open
Abstract
Preclinical studies have shown that the induction of secretory IgA (sIgA) in mucosa and neutralizing antibodies (NAbs) in sera is essential for designing vaccines that can effectively block the transmission of HIV-1. We previously showed that a vaccine consisting of bacterium-like particles (BLPs) displaying Protan-gp120AE-MTQ (PAM) could induce mucosal immune responses through intranasal (IN) immunization in mice and NAbs through intramuscular (IM) immunization in guinea pigs. Here, we evaluated the ability of this vaccine BLP-PAM to elicit HIV-1-specific mucosal and systemic immune responses through IN and IM immunization combination strategies in rhesus macaques. First, the morphology, antigenicity and epitope accessibility of the vaccine were analysed by transmission electron microscopy, bio-layer interferometry and ELISA. In BLP-PAM-immunized macaques, HIV-1-specific sIgA were rapidly induced through IN immunization in situ and distant mucosal sites, although the immune responses are relatively weak. Furthermore, the HIV-1-specific IgG and IgA antibody levels in mucosal secretions were enhanced and maintained, while production of serum NAbs against heterologous HIV-1 tier 1 and 2 pseudoviruses was elicited after IM boost. Additionally, situ mucosal responses and systemic T cell immune responses were improved by rAd2-gp120AE boost immunization via the IN and IM routes. These results suggested that BLP-based delivery in combination with the IN and IM immunization approach represents a potential vaccine strategy against HIV-1.
Collapse
Affiliation(s)
- Huaiyu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Pingchao Li
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Mo Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Jinpeng Bi
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Yizi He
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences, Guangzhou, 510530, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fangshen Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Rongzhen Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Feng Gao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, China.,Department of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, China.,Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Bin Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Ling Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences, Guangzhou, 510530, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510060, China
| | - Xianghui Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, China.,Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
| |
Collapse
|
23
|
Karczmarzyk K, Kęsik-Brodacka M. Attacking the Intruder at the Gate: Prospects of Mucosal Anti SARS-CoV-2 Vaccines. Pathogens 2022; 11:pathogens11020117. [PMID: 35215061 PMCID: PMC8876505 DOI: 10.3390/pathogens11020117] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/04/2022] [Accepted: 01/14/2022] [Indexed: 12/13/2022] Open
Abstract
The sudden outbreak of the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) pandemic in December 2019 caused crises and health emergencies worldwide. The rapid spread of the virus created an urgent need for the development of an effective vaccine and mass immunization to achieve herd immunity. Efforts of scientific teams at universities and pharmaceutical companies around the world allowed for the development of various types of preparations and made it possible to start the vaccination process. However, it appears that the developed vaccines are not effective enough and do not guarantee long-lasting immunity, especially for new variants of SARS-CoV-2. Considering this problem, it is promising to focus on developing a Coronavirus Disease 2019 (COVID-19) mucosal vaccine. Such a preparation applied directly to the mucous membranes of the upper respiratory tract might provide an immune barrier at the primary point of virus entry into the human body while inducing systemic immunity. A number of such preparations against SARS-CoV-2 are already in various phases of preclinical and clinical trials, and several of them are very close to being accepted for general use, constituting a milestone toward pandemic containment.
Collapse
Affiliation(s)
- Kacper Karczmarzyk
- Department of Bacterial Genetics, Institute of Microbiology, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland
- Correspondence:
| | | |
Collapse
|
24
|
Lee PT, Yamamoto FY, Low CF, Loh JY, Chong CM. Gut Immune System and the Implications of Oral-Administered Immunoprophylaxis in Finfish Aquaculture. Front Immunol 2022; 12:773193. [PMID: 34975860 PMCID: PMC8716388 DOI: 10.3389/fimmu.2021.773193] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/23/2021] [Indexed: 12/13/2022] Open
Abstract
The gastrointestinal immune system plays an important role in immune homeostasis regulation. It regulates the symbiotic host-microbiome interactions by training and developing the host's innate and adaptive immunity. This interaction plays a vital role in host defence mechanisms and at the same time, balancing the endogenous perturbations of the host immune homeostasis. The fish gastrointestinal immune system is armed with intricate diffused gut-associated lymphoid tissues (GALTs) that establish tolerance toward the enormous commensal gut microbiome while preserving immune responses against the intrusion of enteric pathogens. A comprehensive understanding of the intestinal immune system is a prerequisite for developing an oral vaccine and immunostimulants in aquaculture, particularly in cultured fish species. In this review, we outline the remarkable features of gut immunity and the essential components of gut-associated lymphoid tissue. The mechanistic principles underlying the antigen absorption and uptake through the intestinal epithelial, and the subsequent immune activation through a series of molecular events are reviewed. The emphasis is on the significance of gut immunity in oral administration of immunoprophylactics, and the different potential adjuvants that circumvent intestinal immune tolerance. Comprehension of the intestinal immune system is pivotal for developing effective fish vaccines that can be delivered orally, which is less labour-intensive and could improve fish health and facilitate disease management in the aquaculture industry.
Collapse
Affiliation(s)
- Po-Tsang Lee
- Department of Aquaculture, National Taiwan Ocean University, Keelung, Taiwan
| | - Fernando Y Yamamoto
- Thad Cochran National Warmwater Aquaculture Center, Mississippi Agriculture and Forestry Experiment Station, Mississippi State University, Stoneville, MS, United States
| | - Chen-Fei Low
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, Bangi, Malaysia
| | - Jiun-Yan Loh
- Centre of Research for Advanced Aquaculture (CORAA), UCSI University, Cheras, Malaysia
| | - Chou-Min Chong
- Aquatic Animal Health and Therapeutics Laboratory (AquaHealth), Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
25
|
Ren Y, Lu X, Yang Z, Lei H. Protective immunity induced by oral vaccination with a recombinant Lactococcus lactis vaccine against H5Nx in chickens. BMC Vet Res 2022; 18:3. [PMID: 34980121 PMCID: PMC8720943 DOI: 10.1186/s12917-021-03109-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 12/13/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The development of an influenza vaccine for poultry that provides broadly protective immunity against influenza H5Nx viruses is a challenging goal. RESULTS Lactococcus lactis (L. lactis)/pNZ8149-HA1-M2 expressing hemagglutinin-1 (HA1) of A/chicken/Vietnam/NCVD-15A59/2015 (H5N6) and the conserved M2 gene of A/Vietnam/1203/2004 (H5N1) was generated. L. lactis/pNZ8149-HA1-M2 could induce significant humoral, mucosal and cell-mediated immune responses, as well as neutralization antibodies. Importantly, L. lactis/pNZ8149-HA1-M2 could prevent disease symptoms without significant weight loss and confer protective immunity in a chicken model against lethal challenge with divergent influenza H5Nx viruses, including H5N6 and H5N1. CONCLUSIONS L. lactis/pNZ8149-HA1-M2 can serve as a promising vaccine candidate in poultry industry for providing protection against H5Nx virus infection in the field application.
Collapse
Affiliation(s)
- Yi Ren
- College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Xin Lu
- College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Zhonghe Yang
- College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Han Lei
- College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
| |
Collapse
|
26
|
Debnath N, Thakur M, Khushboo, Negi NP, Gautam V, Kumar Yadav A, Kumar D. Insight of oral vaccines as an alternative approach to health and disease management: An innovative intuition and challenges. Biotechnol Bioeng 2021; 119:327-346. [PMID: 34755343 DOI: 10.1002/bit.27987] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/06/2021] [Accepted: 11/03/2021] [Indexed: 12/11/2022]
Abstract
Vaccination is the most suitable and persuasive healthcare program for the prohibition of various deadly diseases. However, the higher production cost and purification strategies are out of reach for the developing nations. In this scenario, development of edible vaccine turns out to be the most promising alternative for remodeling the pharmaceutical industry with reduced production and purification costs. Generally, oral route of vaccination is mostly preferred due to its safety, compliance, low manufacturing cost and most importantly the ability to induce immunity in both systemic and mucosal sites. Genetically modified microorganisms and plants could efficiently be used as vehicles for edible vaccines. Edible vaccines are supposed to reduce the risk associated with traditional vaccines. Currently, oral vaccines are available in the market for several viral and bacterial diseases like cholera, hepatitis B, malaria, rabies etc. Herein, the review focuses on the breakthrough events in the area of edible vaccines associated with dietary microbes and plants for better control over diseases.
Collapse
Affiliation(s)
- Nabendu Debnath
- Centre for Molecular Biology, Central University of Jammu, Samba, Jammu & Kashmir (UT), India
| | - Mony Thakur
- Department of Microbiology, Central University of Haryana, Mahendergarh, Haryana, India
| | - Khushboo
- Department of Biotechnology, Central University of Haryana, Mahendergarh, Haryana, India
| | - Neelam P Negi
- Department of Biotechnology, University Institute of Biotechnology, Chandigarh University, Mohali, Punjab, India
| | - Vibhav Gautam
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ashok Kumar Yadav
- Centre for Molecular Biology, Central University of Jammu, Samba, Jammu & Kashmir (UT), India
| | - Deepak Kumar
- Department of Botany, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
27
|
Oral vaccination with recombinant Lactobacillus plantarum encoding Trichinella spiralis inorganic pyrophosphatase elicited a protective immunity in BALB/c mice. PLoS Negl Trop Dis 2021; 15:e0009865. [PMID: 34699522 PMCID: PMC8547688 DOI: 10.1371/journal.pntd.0009865] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 09/29/2021] [Indexed: 12/11/2022] Open
Abstract
Background Trichinellosis is a serious zoonotic disease distributed around the world. It is needed to develop a safe, effective and feasible anti-Trichinella vaccine for prevention and control of trichinellosis. The aim of this study was to construct a recombinant Lactobacillus plantarum encoding Trichinella spiralis inorganic pyrophosphatase (TsPPase) and investigate its immune protective effects against T. spiralis infection. Methodology/Principal findings The growth of recombinant L. plantarum was not affected by TsPPase/pSIP409-pgsA′ plasmid, and the recombinant plasmid was inherited stably in bacteria. Western blot and immunofluorescence assay (IFA) indicated that the rTsPPase was expressed on the surface of recombinant L. plantarum. Oral vaccination with rTsPPase induced higher levels of specific serum IgG, IgG1, IgG2a and mucosal secretory IgA (sIgA) in BALB/c mice. ELISA analysis revealed that the levels of IFN-γ and IL-4 released from spleen, mesenteric lymph nodes and Peyer’s patches were evidently increased at 2–4 weeks following vaccination, compared to MRS (De Man, Rogosa, Sharpe) medium control group (P < 0.05). Immunization of mice with rTsPPase exhibited a 67.18, 54.78 and 51.91% reduction of intestinal infective larvae, adult worms and muscle larvae at 24 hours post infection (hpi), 6 days post infection (dpi) and 35 dpi, respectively (P < 0.05), and the larval molting and development was significantly inhibited by 45.45% at 24 hpi, compared to the MRS group. Conclusions TsPPase plays a crucial role in T. spiralis molting and development, oral vaccination with rTsPPase induced a significant local mucosal sIgA response and systemic Th1/Th2 immune response, and immune protection against T. spiralis infection in BALB/c mice. In the previous study, a Trichinella spiralis inorganic pyrophosphatase (TsPPase) was expressed and its role in larval molting and development was observed. In this study, a recombinant TsPPase/pSIP409-pgsA′ plasmid was constructed and transferred into Lactobacillus plantarum NC8, the rTsPPase was expressed on the surface of recombinant L. plantarum NC8. Oral immunization of mice with rTsPPase DNA vaccine elicited a high level of specific serum IgG, IgG1, IgG2a and mucosal secretory IgA (sIgA). The levels of IFN-γ and IL-4 released from spleen, mesenteric lymph nodes and Peyer’s patches were evidently increased at 2–4 weeks following vaccination. Immunization of mice with rTsPPase showed a significant reduction of intestinal infective larvae, adult worms and muscle larvae, and intestinal larval molting and development was significantly suppressed. The results indicated that oral vaccination with rTsPPase elicited a significant local mucosal sIgA response and specific systemic Th1/Th2 immune response, and an obvious protective immunity against T. spiralis infection.
Collapse
|
28
|
Tang P, Cui E, Song Y, Yan R, Wang J. Porcine deltacoronavirus and its prevalence in China: a review of epidemiology, evolution, and vaccine development. Arch Virol 2021; 166:2975-2988. [PMID: 34524535 PMCID: PMC8440736 DOI: 10.1007/s00705-021-05226-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 07/16/2021] [Indexed: 11/29/2022]
Abstract
Porcine deltacoronavirus (PDCoV) is one of the most important enteropathogenic pathogens, and it causes enormous economic losses to the global commercial pork industry. PDCoV was initially reported in Hong Kong (China) in 2012 and subsequently emerged in swine herds with diarrhea in Ohio (USA) in 2014. Since then, it has spread to Canada, South Korea, mainland China, and several Southeast Asian countries. Information about the epidemiology, evolution, prevention, and control of PDCoV and its prevalence in China has not been comprehensively reported, especially in the last five years. This review is an update of current information on the general characteristics, epidemiology, geographical distribution, and evolutionary relationships, and the status of PDCoV vaccine development, focusing on the prevalence of PDCoV in China and vaccine research in particular. Together, this information will provide us with a greater understanding of PDCoV infection and will be helpful for establishing new strategies for controlling this virus worldwide.
Collapse
Affiliation(s)
- Pan Tang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Enhui Cui
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yihong Song
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Ruoqian Yan
- Henan Centre for Animal Diseases Control and Prevention, Zhengzhou, China.
| | - Jingyu Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China.
| |
Collapse
|
29
|
Abouloifa H, Rokni Y, Bellaouchi R, Ghabbour N, Karboune S, Brasca M, Ben Salah R, Chihib NE, Saalaoui E, Asehraou A. Characterization of Probiotic Properties of Antifungal Lactobacillus Strains Isolated from Traditional Fermenting Green Olives. Probiotics Antimicrob Proteins 2021; 12:683-696. [PMID: 30929140 DOI: 10.1007/s12602-019-09543-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The aim of this work is to characterize the potential probiotic properties of 14 antifungal Lactobacillus strains isolated from traditional fermenting Moroccan green olives. The molecular identification of strains indicated that they are composed of five Lactobacillus brevis, two Lactobacillus pentosus, and seven Lactobacillus plantarum. In combination with bile (0.3%), all the strains showed survival rates (SRs) of 83.19-56.51% at pH 3, while 10 strains showed SRs of 31.67-64.44% at pH 2.5. All the strains demonstrated high tolerance to phenol (0.6%) and produced exopolysaccharides. The autoaggregation, hydrophobicity, antioxidant activities, and surface tension value ranges of the strains were 10.29-41.34%, 15.07-34.67%, 43.11-52.99%, and 36.23-40.27 mN/m, respectively. Bacterial cultures exhibited high antifungal activity against Penicillium sp. The cell-free supernatant (CFS) of the cultures showed important inhibition zones against Candida pelliculosa (18.2-24.85 mm), as well as an antibacterial effect against some gram-positive and gram-negative bacteria (10.1-14.1 mm). The neutralized cell-free supernatant of the cultures displayed considerable inhibitory activity against C. pelliculosa (11.2-16.4 mm). None of the strains showed acquired or horizontally transferable antibiotic resistance or mucin degradation or DNase, hemolytic, or gelatinase activities. Lactobacillus brevis S82, Lactobacillus pentosus S75, and Lactobacillus plantarum S62 showed aminopeptidase, β-galactosidase, and β-glucosidase activities, while the other enzymes of API-ZYM were not detected. The results obtained revealed that the selected antifungal Lactobacillus strains are considered suitable candidates for use both as probiotic cultures for human consumption and for starters and as biopreservative cultures in agriculture, food, and pharmaceutical industries.
Collapse
Affiliation(s)
- Houssam Abouloifa
- Laboratory of Biochemistry and Biotechnology, Faculty of Sciences, Mohammed Premier University, 60000, Oujda, Morocco.
| | - Yahya Rokni
- Laboratory of Biochemistry and Biotechnology, Faculty of Sciences, Mohammed Premier University, 60000, Oujda, Morocco
| | - Reda Bellaouchi
- Laboratory of Biochemistry and Biotechnology, Faculty of Sciences, Mohammed Premier University, 60000, Oujda, Morocco
| | - Nabil Ghabbour
- Laboratory of Biochemistry and Biotechnology, Faculty of Sciences, Mohammed Premier University, 60000, Oujda, Morocco
| | - Salwa Karboune
- Department of Food Science and Agricultural Chemistry, Macdonald Campus, McGill University, 21,111 Lakeshore, Ste Anne de Bellevue, Montreal, Quebec, H9X 3V9, Canada
| | - Milena Brasca
- Institute of Sciences of Food Production, National Research Council, Via Celoria 2, 20133, Milan, Italy
| | - Riadh Ben Salah
- Laboratory of Microorganisms and Biomolecules, Centre of Biotechnology of Sfax, BP: 1177, 3018, Sfax, Tunisia
| | - Nour Eddine Chihib
- INRA-UMR UMET 8207- PIHM team, CNRS-INRA, University of Lille, 369 rue Jules Guesde, BP20039, 59651, Villeneuve d'Ascq Cedex, France
| | - Ennouamane Saalaoui
- Laboratory of Biochemistry and Biotechnology, Faculty of Sciences, Mohammed Premier University, 60000, Oujda, Morocco
| | - Abdeslam Asehraou
- Laboratory of Biochemistry and Biotechnology, Faculty of Sciences, Mohammed Premier University, 60000, Oujda, Morocco
| |
Collapse
|
30
|
Oh SH, Kim SH, Jeon JH, Kim EB, Lee NK, Beck S, Choi YJ, Kang SK. Cytoplasmic expression of a model antigen with M Cell-Targeting moiety in lactic acid bacteria and implication of the mechanism as a mucosal vaccine via oral route. Vaccine 2021; 39:4072-4081. [PMID: 34127296 DOI: 10.1016/j.vaccine.2021.06.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/27/2021] [Accepted: 06/03/2021] [Indexed: 11/15/2022]
Abstract
Lactic acid bacteria (LAB) have been widely studied as mucosal vaccine delivery carriers against many infectious diseases for heterologous expression of protein antigens. There are three antigen expression strategies for LAB: cytoplasmic expression (CE), cell surface display (SD), and extracellular secretion (ES). Despite the generally higher protein expression level and many observations of antigen-specific immunogenicity in CE, its application as a mucosal vaccine has been overlooked relative to SD and ES because of the antigens enclosed by the LAB cell wall. We hypothesized that the antigens in CE could be released from the LAB into the intestinal lumen before host bacterial access to gut-associated lymphoid tissue (GALT), which could contribute to antigen-specific immune responses after oral administration. To elucidate this hypothesis, three recombinant Lactobacillus plantarum (LP) strains were constructed to produce a model antigen, BmpB, with or without an M cell-targeting moiety, and their immunogenicities were analyzed comparatively as oral vaccines in mouse model. The data indicated that the recombinant LPs producing BmpBs with different conformations could induce mucosal immunity differentially. This suggests that the cytoplasmic antigens in LAB could be released into the intestinal lumen, subsequently translocated through M cells, and stimulate the GALT to generate antigen-specific immune responses. Therefore, the CE strategy has great potential, especially in the application of oral LAB vaccines as well as SD and ES strategies. This research provides a better understanding of the mechanism for recombinant oral LAB vaccines and gives insight to the future design of LAB vaccines and oral delivery applications for useful therapeutic proteins.
Collapse
Affiliation(s)
- Seo-Ho Oh
- Institute of Green-Bio Science & Technology, Seoul National University, Pyeongchang-gun 25354, Republic of Korea
| | - Sung-Hee Kim
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Ji-Hye Jeon
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun Bae Kim
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Nam-Kyung Lee
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Samuel Beck
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Yun-Jaie Choi
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Sang-Kee Kang
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang-gun 25354, Republic of Korea; Institute of Green-Bio Science & Technology, Seoul National University, Pyeongchang-gun 25354, Republic of Korea; Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
31
|
Oral Probiotic Vaccine Expressing Koi Herpesvirus (KHV) ORF81 Protein Delivered by Chitosan-Alginate Capsules Is a Promising Strategy for Mass Oral Vaccination of Carps against KHV Infection. J Virol 2021; 95:JVI.00415-21. [PMID: 33827944 DOI: 10.1128/jvi.00415-21] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 03/26/2021] [Indexed: 01/21/2023] Open
Abstract
Koi herpesvirus (KHV) is highly contagious and lethal to cyprinid fish, causing significant economic losses to the carp aquaculture industry, particularly to koi carp breeders. Vaccines delivered through intramuscular needle injection or gene gun are not suitable for mass vaccination of carp. So, the development of cost-effective oral vaccines that are easily applicable at a farm level is highly desirable. In this study, we utilized chitosan-alginate capsules as an oral delivery system for a live probiotic (Lactobacillus rhamnosus) vaccine, pYG-KHV-ORF81/LR CIQ249, expressing KHV ORF81 protein. The tolerance of the encapsulated recombinant Lactobacillus to various digestive environments and the ability of the probiotic strain to colonize the intestine of carp was tested. The immunogenicity and the protective efficacy of the encapsulated probiotic vaccine was evaluated by determining IgM levels, lymphocyte proliferation, expression of immune-related genes, and viral challenge to vaccinated fish. It was clear that the chitosan-alginate capsules protected the probiotic vaccine effectively against extreme digestive environments, and a significant level (P < 0.01) of antigen-specific IgM with KHV-neutralizing activity was detected, which provided a protection rate of ca. 85% for koi carp against KHV challenge. The strategy of using chitosan-alginate capsules to deliver probiotic vaccines is easily applicable for mass oral vaccination of fish.IMPORTANCE An oral probiotic vaccine, pYG-KHV-ORF81/LR CIQ249, encapsulated by chitosan-alginate capsules as an oral delivery system was developed for koi carp against koi herpesvirus (KHV) infection. This encapsulated probiotic vaccine can be protected from various digestive environments and maintain effectively high viability, showing a good tolerance to digestive environments. This encapsulated probiotic vaccine has a good immunogenicity in koi carp via oral vaccination, and a significant level of antigen-specific IgM was effectively induced after oral vaccination, displaying effective KHV-neutralizing activity. This encapsulated probiotic vaccine can provide effective protection for koi carp against KHV challenge, which is handling-stress free for the fish, cost effective, and suitable for the mass oral vaccination of koi carp at a farm level, suggesting a promising vaccine strategy for fish.
Collapse
|
32
|
Shonyela SM, Shi C, Yang W, Cao X, Yang G, Wang C. Recombinant Lactobacillus plantarum NC8 strain expressing porcine rotavirus VP7 induces specific antibodies in BALB/c mice. Acta Biochim Biophys Sin (Shanghai) 2021; 53:707-718. [PMID: 33963824 DOI: 10.1093/abbs/gmab050] [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: 09/18/2020] [Indexed: 12/23/2022] Open
Abstract
The major etiologic agent that causes acute gastroenteritis worldwide in young animals and children is Group A rotavirus. Currently, commercially available vaccines do not often prevent porcine rotavirus (PRV) infection. In this study, we evaluated the efficacy of oral recombinant Lactobacillus vaccine against PRV in a mouse model. Lactobacillus plantarum NC8 was used as the host strain, and bacterial vectors were constructed, because the NC8 isolated has shown the capability to survive gastric transit and to colonize the intestinal tract of humans and other mammals. To explore the immunological mechanisms, lactic acid bacterial vectors were used to express VP7 antigen from PRV. We constructed an L. plantarum strain with surface-displayed VP7, named NC8-pSIP409-pgsA-VP7-DCpep. The expressed recombinant protein had a molecular weight of ∼37 kDa. The strain was used to immunize BALB/c mice to evaluate their immunomodulatory characteristics. Mice were orally immunized with recombinant L. plantarum NC8-pSIP409-pgsA-VP7-DCpep at a dose of 2 × 109 colony forming units/200 µl. The results showed that NC8-pSIP409-pgsA-VP7-DCpep significantly stimulated the differentiation of dendritic cells (DCs) in Peyer's patches (PPs) and increased the serum levels of IL-4 and IFN-γ, as measured by enzyme-linked immunosorbent assay in mice treated with NC8-pSIP409-pgsA-VP7-DCpep. Compared to the empty vector group, NC8-pSIP409-pgsA-VP7-DCpep significantly increased the production of B220+ B cells in mesenteric lymph nodes (MLNs) and PPs and also increased the titer levels of the VP7-specific antibodies, including IgG and sIgA. The administration of NC8-pSIP409-pgsA-VP7-DCpep mediated relatively broad cellular responses. This study reveals that clear alternatives exist for PRV control strategies and provides information on PRV infection.
Collapse
Affiliation(s)
- Seria Masole Shonyela
- 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
- Ministry of Livestock and Fisheries, P.O. Box 2870, Tanzania
| | - 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
| | - 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
| | | | - 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
| | - 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
|
33
|
Reens AL, Cabral DJ, Liang X, Norton JE, Therien AG, Hazuda DJ, Swaminathan G. Immunomodulation by the Commensal Microbiome During Immune-Targeted Interventions: Focus on Cancer Immune Checkpoint Inhibitor Therapy and Vaccination. Front Immunol 2021; 12:643255. [PMID: 34054810 PMCID: PMC8155485 DOI: 10.3389/fimmu.2021.643255] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/22/2021] [Indexed: 12/11/2022] Open
Abstract
Emerging evidence in clinical and preclinical studies indicates that success of immunotherapies can be impacted by the state of the microbiome. Understanding the role of the microbiome during immune-targeted interventions could help us understand heterogeneity of treatment success, predict outcomes, and develop additional strategies to improve efficacy. In this review, we discuss key studies that reveal reciprocal interactions between the microbiome, the immune system, and the outcome of immune interventions. We focus on cancer immune checkpoint inhibitor treatment and vaccination as two crucial therapeutic areas with strong potential for immunomodulation by the microbiota. By juxtaposing studies across both therapeutic areas, we highlight three factors prominently involved in microbial immunomodulation: short-chain fatty acids, microbe-associate molecular patterns (MAMPs), and inflammatory cytokines. Continued interrogation of these models and pathways may reveal critical mechanistic synergies between the microbiome and the immune system, resulting in novel approaches designed to influence the efficacy of immune-targeted interventions.
Collapse
Affiliation(s)
- Abigail L. Reens
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, United States
| | - Damien J. Cabral
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, United States
| | - Xue Liang
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, United States
| | - James E. Norton
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, United States
| | - Alex G. Therien
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, United States
| | - Daria J. Hazuda
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, United States
- Infectious Disease and Vaccine Research, Merck & Co., Inc., West Point, PA, United States
| | - Gokul Swaminathan
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, United States
| |
Collapse
|
34
|
Lin-Zhao Z, Tong-Yang B, Yi-Xuan Y, Ning-Guo S, Xing-Zhang D, Nan-Ji S, Lv B, Huan-Kang Y, Feng-Shan X, Mei-Shi Q, Wen-Sun W, Dong-Qian A. Construction and immune efficacy of recombinant Lactobacillus casei expressing OmpAI of Aeromonas veronii C5-I as molecular adjuvant. Microb Pathog 2021; 156:104827. [PMID: 33892129 DOI: 10.1016/j.micpath.2021.104827] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 12/30/2020] [Accepted: 02/16/2021] [Indexed: 12/19/2022]
Abstract
Despite advancements in diagnosis and control, Aeromonas infections are considered the leading cause of economic aquaculture loss. In this study, to enhance DNA vaccine efficacy against Aeromonas infections, a fused DNA fragment (1504 bp) of the OmpAI gene from Aeromonas veronii (A. veronii) combined with the C5-I gene from the common carp was generated with splicing by overlapping PCR (SOE-PCR) and expressed in Lactobacillus casei strain CC16. Protein C5-I served as a molecular adjuvant for the antigen OmpAI. Two types of fusion antigens were developed (anchored and secretory). Generally, anchored-type antigens are more effective in inducing immune responses in fish than secretory antigens. Western blot analysis showed that the bands of both antigens were present at 58 kDa. After oral immunization, both DNA vaccines enhanced the serum levels of AKP, ACP, SOD and LZM in immunized carp; the genes IL-10, IL-1β, TNF-α, and IFN-γ in the heart, liver, spleen, head kidney, and intestinal tract were upregulated; and a stronger phagocytic response was triggered in immunized fish. In addition, common carp administered the fused antigens were more protected from Aeromonas challenge (60-73.3% protection). Recombinant Lactobacillus bacteria expressing the fused protein showed a greater propensity for colonization in the intestinal tract in immunized fish than in controls. Here, we provide a promising approach to improve DNA vaccine immunogenicity for protecting common carp from A. veronii infections.
Collapse
Affiliation(s)
- Ze Lin-Zhao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Bin Tong-Yang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China; College of Life Science, Changchun Sci-Tech University Shuangyang District, Changchun, Jilin, 130118, China
| | - Yang Yi-Xuan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Song Ning-Guo
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Dong Xing-Zhang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Sheng Nan-Ji
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Bing Lv
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Yuan Huan-Kang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Xiao Feng-Shan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Qiu Mei-Shi
- Provincial Key Laboratory of Preventive Veterinary Medicine, Hebei Normal University of Science and Technology, Qinhuangdao, 066004, China
| | - Wu Wen-Sun
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China.
| | - Ai Dong-Qian
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
35
|
Antilisterial Potential of Lactic Acid Bacteria in Eliminating Listeria monocytogenes in Host and Ready-to-Eat Food Application. MICROBIOLOGY RESEARCH 2021. [DOI: 10.3390/microbiolres12010017] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Listeriosis is a severe food borne disease with a mortality rate of up to 30% caused by pathogenic Listeria monocytogenes via the production of several virulence factors including listeriolysin O (LLO), transcriptional activator (PrfA), actin (Act), internalin (Int), etc. It is a foodborne disease predominantly causing infections through consumption of contaminated food and is often associated with ready-to-eat food (RTE) and dairy products. Common medication for listeriosis such as antibiotics might cause an eagle effect and antibiotic resistance if it is overused. Therefore, exploration of the use of lactic acid bacteria (LAB) with probiotic characteristics and multiple antimicrobial properties is increasingly getting attention for their capability to treat listeriosis, vaccine development, and hurdle technologies. The antilisterial gene, a gene coding to produce antimicrobial peptide (AMP), one of the inhibitory substances found in LAB, is one of the potential key factors in listeriosis treatment, coupled with the vast array of functions and strategies; this review summarizes the various strategies by LAB against L. monocytogenes and the prospect in development of a ‘generally regarded as safe’ LAB for treatment of listeriosis.
Collapse
|
36
|
Afchangi A, Latifi T, Jalilvand S, Marashi SM, Shoja Z. Combined use of lactic-acid-producing bacteria as probiotics and rotavirus vaccine candidates expressing virus-specific proteins. Arch Virol 2021; 166:995-1006. [PMID: 33533975 DOI: 10.1007/s00705-021-04964-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/03/2020] [Indexed: 12/24/2022]
Abstract
Due to the lower efficacy of currently approved live attenuated rotavirus (RV) vaccines in developing countries, a new approach to the development of safe mucosally administered live bacterial vectors is being considered, using probiotic bacteria as an efficient delivery platform for heterologous RV antigens. Lactic acid bacteria (LAB), which are considered food-grade bacteria and normal microbiota, have been utilized throughout history as probiotics and developed since the 1990s as a delivery system for recombinant heterologous proteins. Over the last decade, LAB have frequently been used as a platform for the delivery of various RV antigens to the mucosa. Given the appropriate safety profile for neonates and providing the benefits of probiotics, recombinant LAB-based vaccines could potentially address the need for a subunit RV vaccine. The present review focuses mainly on different recombinant LAB vaccine constructs for RV and their potential as an alternative recombinant vaccine against RV disease.
Collapse
Affiliation(s)
- Atefeh Afchangi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Tayebeh Latifi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Jalilvand
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sayed Mahdi Marashi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Zabihollah Shoja
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
37
|
Protective Immunity Against Enterotoxigenic Escherichia coli by Oral Vaccination of Engineered Lactococcus lactis. Curr Microbiol 2021; 78:3464-3473. [PMID: 34264362 PMCID: PMC8280578 DOI: 10.1007/s00284-021-02601-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 04/13/2021] [Indexed: 02/07/2023]
Abstract
Enterotoxigenic Escherichia coli (ETEC) is one of the leading causes of diarrhea in children globally, and thus suitable vaccines are desired. Antigen display on lactic acid bacteria is a reliable approach for efficient oral vaccination and preventing bowel diseases. To develop an oral vaccine against ETEC, the gene of the binding domain from heat-labile toxin (LTB), a key ETEC virulence factor, was codon-optimized and cloned into a construct containing a signal peptide and an anchor for display on L. lactis. Bioinformatics analysis showed a codon adaptation index of 0.95 for the codon-optimized gene. Cell surface expression of LTB was confirmed by transmission electron microscopy and blotting. White New Zealand rabbits were immunized per os (PO) with the recombinant L. lactis, and the antibody titers were assayed with ELISA. In vitro neutralization assay was performed using mouse adrenal tumor cells and rabbit ileal loop test was performed as the in vivo assay. ELISA results indicated that oral administration of the engineered L. lactis elicited a significant production of IgA in the intestine. In vitro neutralization assay showed that the effect of the toxin could be neutralized with 500 µg/ml of IgG isolated from the oral vaccine group. Furthermore, the dose of ETEC causing fluid accumulation in the ileal loop test showed a tenfold increase in rabbits immunized with either recombinant L. lactis or LTB protein compared to other groups. Our results imply that recombinant L. lactis could potentially be an effective live oral vaccine against ETEC toxicity.
Collapse
|
38
|
The Role of Mucosal Immunity and Recombinant Probiotics in SARS-CoV2 Vaccine Development. Probiotics Antimicrob Proteins 2021; 13:1239-1253. [PMID: 33770348 PMCID: PMC7996120 DOI: 10.1007/s12602-021-09773-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2021] [Indexed: 01/07/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV2), causing the 2019 novel coronavirus disease (COVID-19), was introduced by WHO (World Health Organization) as "pandemic" in March 2020. According to WHO, thus far (23 November 2020) 58,425,681 infected cases including 1,385,218 deaths have been reported worldwide. In order to reduce transmission and spread of this lethal virus, attempts are globally being made to develop an appropriate vaccine. Intending to neutralize pathogens at their initial entrance site, protective mucosal immunity is inevitably required. In SARS-CoV2 infection and transmission, respiratory mucosa plays a key role; hence, apparently mucosal vaccination could be a superior approach to elicit mucosal and systemic immune responses simultaneously. In this review, the advantages of mucosal vaccination to control COVID-19 infection, limitations, and outcomes of mucosal vaccines have been highlighted. Considering the gut microbiota dysregulation in COVID-19, we further provide evidences on utilization of recombinant probiotics, particularly lactic acid bacteria (LAB) as vaccine carrier. Their intrinsic immunomodulatory features, natural adjuvanticity, and feasible expression of relevant antigen in the mucosal surface make them more appealing as live cell factory. Among all available platforms, bioengineered probiotics are considered as the most affordable, most practical, and safest vaccination approach to halt this emerging virus.
Collapse
|
39
|
S S, S R. Cyclic peptide production from lactic acid bacteria (LAB) and their diverse applications. Crit Rev Food Sci Nutr 2020; 62:2909-2927. [PMID: 33356473 DOI: 10.1080/10408398.2020.1860900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent years, cyclic peptides gave gained increasing attention owing to their pH tolerance, heat stability and resistance to enzymatic actions. The increasing outbreaks of antibiotic resistant pathogens and food spoilage have prompted researchers to search for new approaches to combat them. The increasing number of reports on novel cyclic peptides from lactic acid bacteria (LAB) is considered as a breakthrough due to their potential applications. Although an extensive investigation is required to understand the mechanism of action and range of applications, LAB cyclic peptides can be considered as potential substitutes for commercially available antibiotics and bio preservatives. This review summarizes the current updates of LAB cyclic peptides with emphasis on their structure, mode of action and applications. Recent trends in cyclic peptide applications are also discussed.
Collapse
Affiliation(s)
- Silpa S
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankalathur, Tamilnadu, India
| | - Rupachandra S
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankalathur, Tamilnadu, India
| |
Collapse
|
40
|
Uriza PJ, Trautman C, Palomino MM, Fina Martin J, Ruzal SM, Roset MS, Briones G. Development of an Antigen Delivery Platform Using Lactobacillus acidophilus Decorated With Heterologous Proteins: A Sheep in Wolf's Clothing Story. Front Microbiol 2020; 11:509380. [PMID: 33193117 PMCID: PMC7652789 DOI: 10.3389/fmicb.2020.509380] [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: 11/01/2019] [Accepted: 10/01/2020] [Indexed: 01/18/2023] Open
Abstract
S-layers are bacterial structures present on the surface of several Gram-positive and Gram-negative bacteria that play a role in bacterial protection. In Lactobacillus acidophilus (L. acidophilus ATCC 4356), the S-layer is mainly composed of the protein SlpA. A tandem of two copies of the protein domain SLP-A (pfam: 03217) was identified at the C-terminal of SlpA, being this double SLP-A protein domain (in short dSLP-A) necessary and sufficient for the association of the protein to the L. acidophilus cell wall. A variety of proteins fused to the dSLP-A domain were able to spontaneously associate with high affinity to the cell wall of L. acidophilus and Bacillus subtilis var. natto, in a process that we termed decoration. Binding of dSLP-A-containing-proteins to L. acidophilus was stable at conditions that mimic the gastrointestinal transit in terms of pH, proteases, and bile salts. To evaluate if protein decoration of L. acidophilus can be adapted to generate an oral vaccine platform, a chimeric antigen derived from the bacterial pathogen Shiga-toxin-producing Escherichia coli (STEC) was constructed by fusing the sequences encoding the polypeptides EspA36–192, Intimin653–953, Tir240–378, and H7 flagellin352–374 (EITH7) to the dSLP-A domain (EITH7-dSLP-A). Recombinantly expressed EITH7-dSLP-A protein was affinity purified and combined with L. acidophilus cultures to allow the association of the chimeric antigen to the bacterial surface. EITH7-decorated L. acidophilus was orally administered to BALB/c mice and the induction of anti-EITH7 specific antibodies in sera and feces determined by ELISA. Mice presenting significantly higher anti-EITH7 antibodies titers were able to control more efficiently an experimental STEC infection than mice that received the non-decorated L. acidophilus carrier, indicating that antigen-decorated L. acidophilus can be adapted as a mucosal immunization delivery platform to elicit a protective immune response for vaccine purposes.
Collapse
Affiliation(s)
- Paula J Uriza
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, IIB-UNSAM (IIBIO-CONICET), Buenos Aires, Argentina
| | - Cynthia Trautman
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, IIB-UNSAM (IIBIO-CONICET), Buenos Aires, Argentina
| | - María M Palomino
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Buenos Aires, Argentina.,CONICET - Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Joaquina Fina Martin
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Buenos Aires, Argentina.,CONICET - Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Sandra M Ruzal
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Buenos Aires, Argentina.,CONICET - Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Mara S Roset
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, IIB-UNSAM (IIBIO-CONICET), Buenos Aires, Argentina
| | - Gabriel Briones
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, IIB-UNSAM (IIBIO-CONICET), Buenos Aires, Argentina
| |
Collapse
|
41
|
Construction and evaluation of recombinant Lactobacillus plantarum NC8 delivering one single or two copies of G protein fused with a DC-targeting peptide (DCpep) as novel oral rabies vaccine. Vet Microbiol 2020; 251:108906. [PMID: 33160196 DOI: 10.1016/j.vetmic.2020.108906] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 10/20/2020] [Indexed: 11/24/2022]
Abstract
Rabies remains an important public health threat in most developing countries. To develop a more effective and safe oral vaccine against rabies, we constructed recombinant Lactobacillus plantarum NC8 carrying one or two copies of the G gene with a dendritic cell-targeting peptide (DCpep) fused at the C-terminal designated NC8-pSIP409-sRVG or NC8-pSIP409-dRVG, respectively. The immunogenicity and protective efficacy of these recombinant Lactobacillus plantarum against RABV were evaluated by oral administration in a mouse model. The results showed that recombinant NC8-pSIP409-dRVG possessed more G protein, resulting in more functional maturation of DCs. After three cycle of oral immunization, NC8-pSIP409-dRVG induced significantly higher levels of specific IgG antibody and mixed Th1/Th2 with a strong Th1-biasd immune response in mice. Most importantly, although the titers of RABV neutralizing antibody (VNA) were below the threshold of 0.5 IU/mL, the NC8-pSIP409-dRVG could protect 60 % of inoculated mice against lethal RABV challenge. These data reveal that recombinant NC8-pSIP409-dRVG may be a novel and promising oral vaccine candidate to prevent and control of animal rabies.
Collapse
|
42
|
Jia S, Huang X, Li H, Zheng D, Wang L, Qiao X, Jiang Y, Cui W, Tang L, Li Y, Xu Y. Immunogenicity evaluation of recombinant Lactobacillus casei W56 expressing bovine viral diarrhea virus E2 protein in conjunction with cholera toxin B subunit as an adjuvant. Microb Cell Fact 2020; 19:186. [PMID: 33004035 PMCID: PMC7527787 DOI: 10.1186/s12934-020-01449-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 09/25/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Bovine viral diarrhea virus (BVDV) is one of the main causes of infectious diseases in cattle and causes large financial losses to the cattle industry worldwide. In this study, Lactobacillus casei strain W56 (Lc W56) was used as antigen deliver carrier to construct a recombinant Lactobacillus vaccine pPG-E2-ctxB/Lc W56 constitutively expressing BVDV E2 protein fused with cholera toxin B subunit (ctxB) as an adjuvant, and its immunogenicity against BVDV infection in mice model by oral route was explored. RESULTS Our results suggested that pPG-E2-ctxB/Lc W56 can effectively activate dendritic cells (DCs) in the Peyer's patches, up-regulate the expression of Bcl-6, and promote T-follicular helper (Tfh) cells differentiation, as well as enhance B lymphocyte proliferation and promote them differentiate into specific IgA-secreting plasma cells, secreting anti-E2 mucosal sIgA antibody with BVDV-neutralizing activity. Moreover, significant levels (p < 0.01) of BVDV-neutralizing antigen-specific serum antibodies were induced in the pPG-E2-ctxB/LC W56 group post-vaccination. The recombinant Lactobacillus vaccine can induce cellular immune responses, and significant levels (p < 0.01) of Th1-associated cytokines (IL-2, IL-12, and IFN-γ), Th2-associated cytokines (IL-4, IL-10) and Th17-associated cytokine (IL-17) were determined in the serum of vaccinated mice. Significantly, the recombinant Lactobacillus vaccine provides immune protection against BVDV infection, which can be cleared effectively by the vaccine post-challenge in orally vaccinated animals. CONCLUSIONS The genetically engineered Lactobacillus vaccine constructed in this study is immunogenic in mice and can induce mucosal, humoral, and cellular immune responses, providing effective anti-BVDV immune protection. It thus represents a promising strategy for vaccine development against BVDV.
Collapse
Affiliation(s)
- Shuo Jia
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Xinning Huang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Hua Li
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Dianzhong Zheng
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Li Wang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Xinyuan Qiao
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Yanping Jiang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Wen Cui
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Lijie Tang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
- Northeast Science Inspection Station, Key Laboratory of Animal Pathogen Biology of Ministry of Agriculture of China, Harbin, P. R. China
| | - Yijing Li
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
- Northeast Science Inspection Station, Key Laboratory of Animal Pathogen Biology of Ministry of Agriculture of China, Harbin, P. R. China
| | - Yigang Xu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
- Northeast Science Inspection Station, Key Laboratory of Animal Pathogen Biology of Ministry of Agriculture of China, Harbin, P. R. China
| |
Collapse
|
43
|
Chen Y, Hua X, Ren X, Duan K, Gao S, Sun J, Feng Y, Zhou Y, Guan X, Li D, Wang N, Li J, Yang J, Xia D, Shi W, Liu M. Oral immunization with recombinant Lactobacillus casei displayed AHA1-CK6 and VP2 induces protection against infectious pancreatic necrosis in rainbow trout (Oncorhynchus mykiss). FISH & SHELLFISH IMMUNOLOGY 2020; 100:18-26. [PMID: 32142871 DOI: 10.1016/j.fsi.2020.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/16/2020] [Accepted: 03/02/2020] [Indexed: 06/10/2023]
Abstract
Infectious pancreatic necrosis virus (IPNV) primarily infects larvae and young salmonid with serious economic losses, which causes haemorrhage and putrescence of hepatopancreas. To develop a more effective oral vaccine against IPNV infection, the aeromonas hydrophila adhesion (AHA1) gene was used as a targeting molecule for intestinal epithelial cells. A genetically engineered Lactobacillus casei (pPG-612-AHA1-CK6-VP2/L. casei 393) was constructed to express the AHA1-CK6-VP2 fusion protein. The expression of interest protein was confirmed by western blotting and the immunogenicity of pPG-612-AHA1-CK6-VP2/L. casei 393 was evaluated. And the results showed that more pPG-612-AHA1-CK6-VP2/L. casei 393 were found in the intestinal mucosal surface of the immunized group. The Lactobacillus-derived AHA1-CK6-VP2 fusion protein could induce the production of serum IgM and skin mucus IgT specific for IPNV with neutralizing activity in rainbow trouts. The levels of IL-1β, IL-8 and TNF-α isolated from the lymphocytes stimulated by AHA1-CK6-EGFP produced were significantly higher than EGFP group. For transcription levels of IL-1β, IL-8, CK6, MHC-II, Mx and TNF-1α in the spleen, the result indicated that the adhesion and target chemokine recruit more immune cells to induce cellular immunity. The level of IPNV in the immunized group of pPG-612-AHA1-CK6-VP2/L. casei 393 was significantly lower than that in the control groups. These data indicated that the adhesion and target chemokine could enhance antigen delivery efficiency, which provides a valuable strategy for the development of IPNV recombination Lactobacillus casei oral vaccine in the future.
Collapse
Affiliation(s)
- Yaping Chen
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Xiaojing Hua
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Xuanyu Ren
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Kexin Duan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Shuai Gao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Jinhui Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Ying Feng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Ying Zhou
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Xin Guan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Dechuan Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Na Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Jiahui Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Jiawei Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Dong Xia
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Wen Shi
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China.
| | - Min Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
44
|
Raya Tonetti F, Arce L, Salva S, Alvarez S, Takahashi H, Kitazawa H, Vizoso-Pinto MG, Villena J. Immunomodulatory Properties of Bacterium-Like Particles Obtained From Immunobiotic Lactobacilli: Prospects for Their Use as Mucosal Adjuvants. Front Immunol 2020; 11:15. [PMID: 32038659 PMCID: PMC6989447 DOI: 10.3389/fimmu.2020.00015] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 01/06/2020] [Indexed: 01/26/2023] Open
Abstract
Non-viable lactic acid bacteria (LAB) have been proposed as antigen delivery platforms called bacterium-like particles (BLPs). Most studies have been performed with Lactococcus lactis-derived BLPs where multiple antigens were attached to the peptidoglycan surface and used to successfully induce specific immune responses. It is well-established that the immunomodulatory properties of LAB are strain dependent and therefore, the BLPs derived from each individual strain could have different adjuvant capacities. In this work, we obtained BLPs from immunomodulatory (immunobiotics) and non-immunomodulatory Lactobacillus rhamnosus and Lactobacillus plantarum strains and comparatively evaluated their ability to improve the intestinal and systemic immune responses elicited by an attenuated rotavirus vaccine. Results demonstrated that orally administered BLPs from non-immunomodulatory strains did not induce significant changes in the immune response triggered by rotavirus vaccine in mice. On the contrary, BLPs derived from immunobiotic lactobacilli were able to improve the levels of anti-rotavirus intestinal IgA and serum IgG, the numbers of CD24+B220+ B and CD4+ T cells in Peyer's patches and spleen as well as the production of IFN-γ by immune cells. Interestingly, among immunobiotics-derived BLPs, those obtained from L. rhamnosus CRL1505 and L. rhamnosus IBL027 enhanced more efficiently the intestinal and systemic humoral immune responses when compared to BLPs from other immunobiotic bacteria. The findings of this work indicate that it is necessary to perform an appropriate selection of BLPs in order to find those with the most efficient adjuvant properties. We propose the term Immunobiotic-like particles (IBLPs) for the BLPs derived from CRL1505 and IBL027 strains that are an excellent alternative for the development of mucosal vaccines.
Collapse
Affiliation(s)
- Fernanda Raya Tonetti
- Infection Biology Lab, Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, Tucumán, Argentina.,Facultad de Medicina, Universidad Nacional de Tucumán (UNT), Tucumán, Argentina.,Laboratorio de Ciencias Básicas & Or. Genética, Facultad de Medicina, Universidad Nacional de Tucumán, Tucumán, Argentina
| | - Lorena Arce
- Infection Biology Lab, Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, Tucumán, Argentina.,Facultad de Medicina, Universidad Nacional de Tucumán (UNT), Tucumán, Argentina.,Laboratorio de Ciencias Básicas & Or. Genética, Facultad de Medicina, Universidad Nacional de Tucumán, Tucumán, Argentina
| | - Susana Salva
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucumán, Argentina
| | - Susana Alvarez
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucumán, Argentina
| | - Hideki Takahashi
- Laboratory of Plant Pathology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Plant Immunology Unit, International Education and Research Center for Food Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Maria Guadalupe Vizoso-Pinto
- Infection Biology Lab, Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, Tucumán, Argentina.,Facultad de Medicina, Universidad Nacional de Tucumán (UNT), Tucumán, Argentina.,Laboratorio de Ciencias Básicas & Or. Genética, Facultad de Medicina, Universidad Nacional de Tucumán, Tucumán, Argentina
| | - Julio Villena
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucumán, Argentina.,Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| |
Collapse
|
45
|
Todorov SD, Kang HJ, Ivanova IV, Holzapfel WH. Bacteriocins From LAB and Other Alternative Approaches for the Control of Clostridium and Clostridiodes Related Gastrointestinal Colitis. Front Bioeng Biotechnol 2020; 8:581778. [PMID: 33042979 PMCID: PMC7517946 DOI: 10.3389/fbioe.2020.581778] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/25/2020] [Indexed: 12/14/2022] Open
Abstract
The gut microbiome is considered as a promising target for future non-conventional therapeutic treatment of inflammatory and infectious diseases. The search for appropriate safe and beneficial (lactic acid bacterial and other) putative probiotic strains and/or their antimicrobial metabolites represents a challenging approach for combating several problematic and emerging infections. The process of selecting suitable strains, especially of lactic acid bacteria (LAB) with superior properties, has been accelerated and intensified during the past two decades, also thanks to recent developments in lab techniques. Currently, special focus is on the potential of antimicrobial metabolites produced by some LAB strains and their application as active therapeutic agents. The vision is to develop a scientific basis for 'biotherapeutics' as alternative to conventional approaches in both human and veterinary medicine. Consequently, innovative and promising applications of LAB to the therapeutic practice are presently emerging. An overview of the existing literature indicates that some antimicrobial metabolites such as bacteriocins, widely produced by different bacterial species including LAB, are promising biotherapeutic agents for controlling infections caused by potential pathogens, such as Clostridium and Clostridiodes. Non-conventional, safe and well designed therapeutic treatments may contribute to the improvement of gut dysbiotic conditions. Thereby gut homeostasis can be restored and inflammatory conditions such as gastrointestinal colitis ameliorated. Combining the knowledge on the production, characterization and application of bacteriocins from probiotic LAB, together with their antibacterial properties, appears to be a promising and novel approach in biotherapy. In this overview, different scenarios for the control of Clostridium spp. by application of bacteriocins as therapeutic agents, also in synergistic combination with antibiotics, will be discussed.
Collapse
Affiliation(s)
- Svetoslav D. Todorov
- Advanced Green Energy and Environment Institute (AGEE), Handong Global University, Pohang, South Korea
| | - Hye-Ji Kang
- Advanced Green Energy and Environment Institute (AGEE), Handong Global University, Pohang, South Korea
- HEM Inc., Handong Global University, Pohang, South Korea
| | - Iskra V. Ivanova
- Department of General and Applied Microbiology, Faculty of Biology, Sofia University “St. Kliment Ohridski”, Sofia, Bulgaria
| | - Wilhelm H. Holzapfel
- Advanced Green Energy and Environment Institute (AGEE), Handong Global University, Pohang, South Korea
- HEM Inc., Handong Global University, Pohang, South Korea
- *Correspondence: Wilhelm H. Holzapfel,
| |
Collapse
|
46
|
Wiese-Szadkowska M, Helmin-Basa A, Eljaszewicz A, Gackowska L, Januszewska M, Motyl I, Andryszczyk M, Wieczynska J, Michalkiewicz J. Selected commensal bacteria change profiles of Helicobacter pylori-induced T cells via dendritic cell modulation. Helicobacter 2019; 24:e12614. [PMID: 31328382 DOI: 10.1111/hel.12614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 05/07/2019] [Accepted: 05/17/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND The mechanisms of downregulation of protective immunity against Helicobacter pylori (Hp) infection strongly depend on dendritic cell (DC)-induced T-lymphocyte differentiation pattern. Lactic acid bacteria (LAB) strains can modulate Hp-induced immunoresponse by changes in DC activation profiles. Here, we want to find out if the LAB-pulsed DCs will change Hp-induced T-cell responsiveness patterns. MATERIALS AND METHODS The naive peripheral CD4+ T cells were co-cultured with Hp CagA + pulsed monocyte-derived DCs (DC/CD4+ T cell) in the presence/absence of the feces-derived probiotics: antagonistic or non-antagonistic to Hp (Lactobacillus rhamnosus 900, Lr, Lactobacillus paracasei 915, Lp, respectively), as assessed by the agar slab method. The regulatory T-cell (Treg) population was assessed by flow cytometry, and IFN-γ, IL-12p70, IL-10, and IL-17A levels were evaluated by ELISA method. RESULTS The Hp-pulsed DC/CD4+ T-cell co-cultures were characterized by high IL-10, decreased IL-12p70 and IFN-γ levels, and elevated Treg population. In contrast, Lr-pulsed DC/CD4+ T-cell co-cultures expressed low IL-10, high IL-12p70 and IFN-γ levels and declined Treg population; this responsiveness pattern was not changed by Hp. The responsiveness pattern of the Lp/Hp-pulsed DC/CD4+ T-cell co-cultures did not differ from those pulsed with Hp alone. CONCLUSION In contrast to Lp, Lr probiotic strain overcomes Hp-mediated immune profile in the DC/T-cell co-cultures toward Th1 pattern and limited generation of Tregs in vitro. Lr may therefore be used as a component of anti-Hp treatment.
Collapse
Affiliation(s)
| | - Anna Helmin-Basa
- Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Andrzej Eljaszewicz
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Bialystok, Poland
| | - Lidia Gackowska
- Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | | | - Ilona Motyl
- Faculty of Biotechnology and Food Sciences, The Institute of Technology Fermentation and Microbiology, Technical University of Lodz, Łodz, Poland
| | - Marek Andryszczyk
- Faculty of Mechanical Engineering, University of Technology and Sciences in Bydgoszcz, Bydgoszcz, Poland
| | - Jolanta Wieczynska
- Department of Clinical Microbiology and Immunology, Children's Memorial Hospital, Warsaw, Poland
| | - Jacek Michalkiewicz
- Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland.,Department of Clinical Microbiology and Immunology, Children's Memorial Hospital, Warsaw, Poland
| |
Collapse
|
47
|
Kuczkowska K, Øverland L, Rocha SDC, Eijsink VGH, Mathiesen G. Comparison of eight Lactobacillus species for delivery of surface-displayed mycobacterial antigen. Vaccine 2019; 37:6371-6379. [PMID: 31526620 DOI: 10.1016/j.vaccine.2019.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 09/04/2019] [Indexed: 12/26/2022]
Abstract
Lactobacillus spp. comprise a large group of Gram-positive lactic acid bacteria with varying physiological, ecological and immunomodulatory properties that are widely exploited by mankind, primarily in food production and as health-promoting probiotics. Recent years have shown increased interest in using lactobacilli for delivery of vaccines, mainly due to their ability to skew the immune system towards pro-inflammatory responses. We have compared the potential of eight Lactobacillus species, L. plantarum, L. brevis, L. curvatus, L. rhamnosus, L. sakei, L. gasseri, L. acidophilus and L. reuteri, as immunogenic carriers of the Ag85B-ESAT-6 antigen from Mycobacterium tuberculosis. Surface-display of the antigen was achieved in L. plantarum, L. brevis, L. gasseri and L. reuteri and these strains were further analyzed in terms of their in vitro and in vivo immunogenicity. All strains activated human dendritic cells in vitro. Immunization of mice using a homologous prime-boost regimen comprising a primary subcutaneous immunization followed by three intranasal boosters, led to slightly elevated IgG levels in serum in most strains, and, importantly, to significantly increased levels of antigen-specific mucosal IgA. Cellular immunity was assessed by studying antigen-specific T cell responses in splenocytes, which did not reveal proliferation as assessed by the expression of Ki67, but which showed clear antigen-specific IFN-γ and IL-17 responses for some of the groups. Taken together, the present results indicate that L. plantarum and L. brevis are the most promising carriers of TB vaccines.
Collapse
Affiliation(s)
- Katarzyna Kuczkowska
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), P.O. Box 5003, 1432 Aas, Norway.
| | - Lise Øverland
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), P.O. Box 5003, 1432 Aas, Norway
| | - Sergio D C Rocha
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), P.O. Box 5003, 1432 Aas, Norway
| | - Vincent G H Eijsink
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), P.O. Box 5003, 1432 Aas, Norway
| | - Geir Mathiesen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), P.O. Box 5003, 1432 Aas, Norway
| |
Collapse
|
48
|
Zhao L, Tang X, Sheng X, Xing J, Zhan W. Surface display of hirame novirhabdovirus (HIRRV) G protein in Lactococcus lactis and its immune protection in flounder (Paralichthys olivaceus). Microb Cell Fact 2019; 18:142. [PMID: 31434565 PMCID: PMC6704618 DOI: 10.1186/s12934-019-1195-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 08/14/2019] [Indexed: 01/07/2023] Open
Abstract
Background Hirame novirhabdovirus (HIRRV) can infect a wide range of marine and freshwater fish, causing huge economic losses to aquaculture industry. Vaccine development, especially oral vaccine, has become an effective and convenient way to control aquatic infectious diseases. HIRRV glycoprotein (G), an immunogenic viral protein is a potential vaccine candidate for prevention of the disease. Here, we aimed to construct a recombinant Lactococcus lactis strain expressing HIRRV-G on the cell surface as an oral vaccine to prevent HIRRV. Results Glycoprotein gene of HIRRV was successfully cloned and expressed in L. lactis NZ9000 in a surface-displayed form, yielding Ll:pSLC-G. An approximately 81 kDa recombinant G protein (containing LysM anchoring motif) was confirmed by SDS-PAGE, western blotting and mass spectrometry analysis. The surface-displayed G protein was also verified by immunofluorescence and flow cytometry assays. Furthermore, to evaluate the potential of Ll:pSLC-G as oral vaccine candidate, flounders were continuously fed with commercial diet pellets coated with 1.0 × 109 cfu/g of induced Ll:pSLC-G for 1 week. Four weeks later, booster vaccination was performed with the same procedure. Compared with the controls, Ll:pSLC-G elicited significantly higher levels of specific IgM against HIRRV in flounder gut mucus at the second week and in serum at the fourth week (p < 0.05). Meanwhile, oral immunization with Ll:pSLC-G could provide 60.7% protection against HIRRV infection and a significantly lower virus load was detected than the controls on the third day post-challenge (p < 0.01). Moreover, on the first day post 1-week feeding, approximately 104–105 recombinant L. lactis cells were detected in every gram of foregut, midgut and hindgut of flounder, which were mainly localized at the bottom of gut mucus layer; and on day 21, 102–103L. lactis cells could still be recovered. Conclusions HIRRV-G protein was successfully expressed on the surface of L. lactis cells, which could trigger mucosal and humoral immune response of flounder and provide considerable immune protection against HIRRV. It suggests that genetically engineered L. lactis expressing G protein can be employed as a promising oral vaccine against HIRRV infection.
Collapse
Affiliation(s)
- Lining Zhao
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China. .,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China.
| | - Xiuzhen Sheng
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Jing Xing
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China
| | - Wenbin Zhan
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China
| |
Collapse
|
49
|
Loh FK, Nathan S, Chow SC, Fang CM. Vaccination challenges and strategies against long-lived Toxoplasma gondii. Vaccine 2019; 37:3989-4000. [PMID: 31186188 DOI: 10.1016/j.vaccine.2019.05.083] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 04/05/2019] [Accepted: 05/21/2019] [Indexed: 01/03/2023]
Abstract
Since the discovery of Toxoplasma gondii in 1908, it is estimated that one-third of the global population has been exposed to this ubiquitous intracellular protozoan. The complex life cycle of T. gondii has enabled itself to overcome stress and transmit easily within a broad host range thus achieving a high seroprevalence worldwide. To date, toxoplasmosis remains one of the most prevalent HIV-associated opportunistic central nervous system infections. This review presents a comprehensive overview of different vaccination approaches ranging from traditional inactivated whole-T. gondii vaccines to the popular DNA vaccines. Extensive discussions are made to highlight the challenges in constructing these vaccines, selecting adjuvants as well as delivery methods, immunisation approaches and developing study models. Herein we also deliberate over the latest and promising enhancement strategies that can address the limitations in developing an effective T. gondii prophylactic vaccine.
Collapse
Affiliation(s)
- Fei-Kean Loh
- Division of Biomedical Sciences, School of Pharmacy, The University of Nottingham Malaysia Campus, 43500 Semenyih, Selangor, Malaysia
| | - Sheila Nathan
- School of Biosciences and Biotechnology, Faculty of Science & Technology, Universiti Kebangsaan Malaysia, 43600 Bangi, Selangor, Malaysia
| | - Sek-Chuen Chow
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| | - Chee-Mun Fang
- Division of Biomedical Sciences, School of Pharmacy, The University of Nottingham Malaysia Campus, 43500 Semenyih, Selangor, Malaysia.
| |
Collapse
|
50
|
Takahashi K, Orito N, Tokunoh N, Inoue N. Current issues regarding the application of recombinant lactic acid bacteria to mucosal vaccine carriers. Appl Microbiol Biotechnol 2019; 103:5947-5955. [PMID: 31175431 DOI: 10.1007/s00253-019-09912-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/21/2022]
Abstract
Over the past two decades, lactic acid bacteria (LAB) have been intensively studied as potential bacterial carriers for therapeutic materials, such as vaccine antigens, to the mucosal tissues. LAB have several attractive advantages as carriers of mucosal vaccines, and the effectiveness of LAB vaccines has been demonstrated in numerous studies. Research on LAB vaccines to date has focused on whether antigen-specific immunity, particularly antibody responses, can be induced. However, with recent developments in immunology, microbiology, and vaccinology, more detailed analyses of the underlying mechanisms, especially, of the induction of cell-mediated immunity and memory cells, have been required for vaccine development and licensure. In this mini-review, we will discuss the issues, including (i) immune responses other than antibody production, (ii) persistence of LAB vaccine immunity, (iii) comparative evaluation of LAB vaccines with any existing or reference vaccines, (iv) strategies for increasing the effectiveness of LAB vaccines, and (iv) effects of microbiota on the efficacy of LAB vaccines. Although these issues have been rarely studied or discussed to date in relation to LAB vaccine research, further understanding of them is critical for the practical application of LAB vaccine systems.
Collapse
Affiliation(s)
- Keita Takahashi
- Department of Microbiology and Immunology, Gifu Pharmaceutical University, 1-25-4 Daigaku Nishi, Gifu, 501-1196, Japan.
| | - Nozomi Orito
- Department of Microbiology and Immunology, Gifu Pharmaceutical University, 1-25-4 Daigaku Nishi, Gifu, 501-1196, Japan
| | - Nagisa Tokunoh
- Department of Microbiology and Immunology, Gifu Pharmaceutical University, 1-25-4 Daigaku Nishi, Gifu, 501-1196, Japan
| | - Naoki Inoue
- Department of Microbiology and Immunology, Gifu Pharmaceutical University, 1-25-4 Daigaku Nishi, Gifu, 501-1196, Japan.
| |
Collapse
|