1
|
Wang T, Zhu B, Zhao J, Li S. Research progress in methods of acquisition, structure elucidation, and quality control of Chinese herbal polysaccharides. Chin J Nat Med 2025; 23:143-157. [PMID: 39986691 DOI: 10.1016/s1875-5364(25)60819-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/09/2024] [Accepted: 08/04/2024] [Indexed: 02/24/2025]
Abstract
The therapeutic efficacy of traditional Chinese medicine has been widely acknowledged due to its extensive history of clinical effectiveness. However, the precise active components underlying each prescription remain incompletely understood. Polysaccharides, as a major constituent of water decoctions-the most common preparation method for Chinese medicinals-may provide a crucial avenue for deepening our understanding of the efficacy principles of Chinese medicine and establishing a framework for its modern development. The structural complexity and diversity of Chinese herbal polysaccharides present significant challenges in their separation and analysis compared to small molecules. This paper aims to explore the potential of Chinese herbal polysaccharides efficiently by briefly summarizing recent advancements in polysaccharide chemical research, focusing on methods of acquisition, structure elucidation, and quality control.
Collapse
Affiliation(s)
- Tingting Wang
- Joint Laboratory of Chinese Herbal Glycoengineering and Testing Technology, University of Macau & National Glycoengineering Research Center, Macao SAR 999078, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Baojie Zhu
- Joint Laboratory of Chinese Herbal Glycoengineering and Testing Technology, University of Macau & National Glycoengineering Research Center, Macao SAR 999078, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Jing Zhao
- Joint Laboratory of Chinese Herbal Glycoengineering and Testing Technology, University of Macau & National Glycoengineering Research Center, Macao SAR 999078, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China.
| | - Shaoping Li
- Joint Laboratory of Chinese Herbal Glycoengineering and Testing Technology, University of Macau & National Glycoengineering Research Center, Macao SAR 999078, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China; Macao Centre for Testing of Chinese Medicine, University of Macau, Macao SAR 999078, China.
| |
Collapse
|
2
|
Chagas DB, Santos FDS, de Oliveira NR, Bohn TLO, Dellagostin OA. Recombinant Live-Attenuated Salmonella Vaccine for Veterinary Use. Vaccines (Basel) 2024; 12:1319. [PMID: 39771981 PMCID: PMC11680399 DOI: 10.3390/vaccines12121319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/14/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Vaccination is essential for maintaining animal health, with priority placed on safety and cost effectiveness in veterinary use. The development of recombinant live-attenuated Salmonella vaccines (RASVs) has enabled the construction of balanced lethal systems, ensuring the stability of plasmid vectors encoding protective antigens post-immunization. These vaccines are particularly suitable for production animals, providing long-term immunity against a range of bacterial, viral, and parasitic pathogens. This review summarizes the progress made in this field, with a focus on clinical trials demonstrating the efficacy and commercial potential of RASVs in veterinary medicine.
Collapse
Affiliation(s)
- Domitila Brzoskowski Chagas
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-610, Rio Grande do Sul, Brazil (T.L.O.B.)
| | - Francisco Denis Souza Santos
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-610, Rio Grande do Sul, Brazil (T.L.O.B.)
- Faculdade de Medicina, Universidade Federal do Rio Grande, Rio Grande 96200-400, Rio Grande do Sul, Brazil
| | - Natasha Rodrigues de Oliveira
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-610, Rio Grande do Sul, Brazil (T.L.O.B.)
| | - Thaís Larré Oliveira Bohn
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-610, Rio Grande do Sul, Brazil (T.L.O.B.)
| | - Odir Antônio Dellagostin
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-610, Rio Grande do Sul, Brazil (T.L.O.B.)
| |
Collapse
|
3
|
Huang X, Yang S, Zhao J, Yang J, Jiang H, Li S, Wang C, Liu G. Generation and evaluation of Salmonella entericaserovar Choleraesuis mutant strains as a potential live-attenuated vaccine. Vaccine 2024; 42:126262. [PMID: 39197218 DOI: 10.1016/j.vaccine.2024.126262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/02/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND Salmonella entericaserovar Choleraesuis (S.C) is a swine enteric pathogen causing paratyphoid fever, enterocolitis, and septicemia in piglets. S. C is mainly transmitted through the fecal-oral route. Vaccination is an effective strategy for preventing and controlling Salmonella infection. RESULTS Herein, we used CRISPR-Cas9 technology to knockout the virulence regulatory genes, rpoS, and slyA of S. C and constructed the ∆rpoS, ∆slyA, and ∆rpoS ∆slyA strains. The phenotypic characteristics of the mutant strains remained unchanged compared with the parental wild-type strain. In vivo study, unlike the wild-type strain, the ∆slyA and ∆rpoS ∆slyA strains alleviated splenomegaly, colon atrophy, and lower bacterial loads in the spleen, liver, ileum, and colon. These mutant strains survived in Peyer's patches (PPs) and mesenteric lymph nodes (MLN) for up to 15 days post-infection. Furthermore, the immunization of the ∆rpoS ∆slyA strain induced robust humoral and cellular immune responses. CONCLUSIONS Consequently, vaccination with ∆rpoS ∆slyA conferred a high percentage of protection against lethal invasive Salmonella, specifically S. C, in mice. This study provided novel insights into the development of live-attenuated vaccines against the infection of S. C.
Collapse
Affiliation(s)
- Xin Huang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Shanshan Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.; Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, Jiangsu, China
| | - Jing Zhao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Jing Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.; Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liege, Belgium
| | - Huazheng Jiang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Shuxian Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Caiying Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.; Cell Biology and Immunology Group, Wageningen University and Research, Wageningen, the Netherlands
| | - Guangliang Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China.
| |
Collapse
|
4
|
Danielson SM, Lefferts AR, Norman E, Regner EH, Schulz HM, Sansone-Poe D, Orlicky DJ, Kuhn KA. Myeloid Cells and Sphingosine-1-Phosphate Are Required for TCRαβ Intraepithelial Lymphocyte Recruitment to the Colon Epithelium. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1843-1854. [PMID: 38568091 PMCID: PMC11105980 DOI: 10.4049/jimmunol.2200556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 03/16/2024] [Indexed: 04/07/2024]
Abstract
Intraepithelial lymphocytes (IELs) are T cells important for the maintenance of barrier integrity in the intestine. Colon IELs are significantly reduced in both MyD88-deficient mice and those lacking an intact microbiota, suggesting that MyD88-mediated detection of bacterial products is important for the recruitment and/or retention of these cells. Here, using conditionally deficient MyD88 mice, we show that myeloid cells are the key mediators of TCRαβ+ IEL recruitment to the colon. Upon exposure to luminal bacteria, myeloid cells produce sphingosine-1-phosphate (S1P) in a MyD88-dependent fashion. TCRαβ+ IEL recruitment may be blocked using the S1P receptor antagonist FTY720, confirming the importance of S1P in the recruitment of TCRαβ+ IELs to the colon epithelium. Finally, using the TNFΔARE/+ model of Crohn's-like bowel inflammation, we show that disruption of colon IEL recruitment through myeloid-specific MyD88 deficiency results in reduced pathology. Our results illustrate one mechanism for recruitment of a subset of IELs to the colon.
Collapse
Affiliation(s)
- Sarah Mann Danielson
- Division of Rheumatology, Department of Medicine University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Adam R. Lefferts
- Division of Rheumatology, Department of Medicine University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Eric Norman
- Division of Rheumatology, Department of Medicine University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Emilie H. Regner
- Division of Rheumatology, Department of Medicine University of Colorado Anschutz Medical Campus, Aurora, CO
- Division of Gastroenterology and Hepatology, Department of Medicine University of Colorado Anschutz Medical Campus, Aurora, CO
- Current affiliation: Division of Gastroenterology, Department of Medicine, Oregon Health Sciences University, Portland, OR
| | - Hanna M. Schulz
- Division of Rheumatology, Department of Medicine University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Danielle Sansone-Poe
- Division of Rheumatology, Department of Medicine University of Colorado Anschutz Medical Campus, Aurora, CO
| | - David J. Orlicky
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO
| | - Kristine A. Kuhn
- Division of Rheumatology, Department of Medicine University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
5
|
Mkangara M. Prevention and Control of Human Salmonella enterica Infections: An Implication in Food Safety. INTERNATIONAL JOURNAL OF FOOD SCIENCE 2023; 2023:8899596. [PMID: 37727836 PMCID: PMC10506869 DOI: 10.1155/2023/8899596] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 08/19/2023] [Accepted: 08/26/2023] [Indexed: 09/21/2023]
Abstract
Salmonella is a foodborne zoonotic pathogen causing diarrhoeal disease to humans after consuming contaminated water, animal, and plant products. The bacterium is the third leading cause of human death among diarrhoeal diseases worldwide. Therefore, human salmonellosis is of public health concern demanding integrated interventions against the causative agent, Salmonella enterica. The prevention of salmonellosis in humans is intricate due to several factors, including an immune-stable individual infected with S. enterica continuing to shed live bacteria without showing any clinical signs. Similarly, the asymptomatic Salmonella animals are the source of salmonellosis in humans after consuming contaminated food products. Furthermore, the contaminated products of plant and animal origin are a menace in food industries due to Salmonella biofilms, which enhance colonization, persistence, and survival of bacteria on equipment. The contaminated food products resulting from bacteria on equipment offset the economic competition of food industries and partner institutions in international business. The most worldwide prevalent broad-range Salmonella serovars affecting humans are Salmonella Typhimurium and Salmonella Enteritidis, and poultry products, among others, are the primary source of infection. The broader range of Salmonella serovars creates concern over multiple strategies for preventing and controlling Salmonella contamination in foods to enhance food safety for humans. Among the strategies for preventing and controlling Salmonella spread in animal and plant products include biosecurity measures, isolation and quarantine, epidemiological surveillance, farming systems, herbs and spices, and vaccination. Other measures are the application of phages, probiotics, prebiotics, and nanoparticles reduced and capped with antimicrobial agents. Therefore, Salmonella-free products, such as beef, pork, poultry meat, eggs, milk, and plant foods, such as vegetables and fruits, will prevent humans from Salmonella infection. This review explains Salmonella infection in humans caused by consuming contaminated foods and the interventions against Salmonella contamination in foods to enhance food safety and quality for humans.
Collapse
Affiliation(s)
- Mwanaisha Mkangara
- Department of Science and Laboratory Technology, Dar es Salaam Institute of Technology, P.O. Box 2958, Dar es Salaam, Tanzania
| |
Collapse
|
6
|
Zhou G, Tian J, Tian Y, Ma Q, Li Q, Wang S, Shi H. Recombinant-attenuated Salmonella enterica serovar Choleraesuis vector expressing the PlpE protein of Pasteurella multocida protects mice from lethal challenge. BMC Vet Res 2023; 19:128. [PMID: 37598169 PMCID: PMC10439597 DOI: 10.1186/s12917-023-03679-0] [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: 08/31/2022] [Accepted: 07/27/2023] [Indexed: 08/21/2023] Open
Abstract
BACKGROUND Bacterial surface proteins play key roles in pathogenicity and often contribute to microbial adhesion and invasion. Pasteurella lipoprotein E (PlpE), a Pasteurella multocida (P. multocida) surface protein, has recently been identified as a potential vaccine candidate. Live attenuated Salmonella strains have a number of potential advantages as vaccine vectors, including immunization with live vector can mimic natural infections by organisms, lead to the induction of mucosal, humoral, and cellular immune responses. In this study, a previously constructed recombinant attenuated Salmonella Choleraesuis (S. Choleraesuis) vector rSC0016 was used to synthesize and secrete the surface protein PlpE of P. multocida to form the vaccine candidate rSC0016(pS-PlpE). Subsequently, the immunogenicity of S. Choleraesuis rSC0016(pS-PlpE) as an oral vaccine to induce protective immunity against P. multocida in mice was evaluated. RESULTS After immunization, the recombinant attenuated S. Choleraesuis vector can efficiently delivered P. multocida PlpE protein in vivo and induced a specific immune response against this heterologous antigen in mice. In addition, compared with the inactivated vaccine, empty vector (rSC0016(pYA3493)) and PBS immunized groups, the rSC0016(pS-PlpE) vaccine candidate group induced higher antigen-specific mucosal, humoral and mixed Th1/Th2 cellular immune responses. After intraperitoneal challenge, the rSC0016(pS-PlpE) immunized group had a markedly enhanced survival rate (80%), a better protection efficiency than 60% of the inactivated vaccine group, and significantly reduced tissue damage. CONCLUSIONS In conclusion, our study found that the rSC0016(pS-PlpE) vaccine candidate provided good protection against challenge with wild-type P. multocida serotype A in a mouse infection model, and may potentially be considered for use as a universal vaccine against multiple serotypes of P. multocida in livestock, including pigs.
Collapse
Affiliation(s)
- Guodong Zhou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Jiashuo Tian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Yichen Tian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Qifeng Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Quan Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Shifeng Wang
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32611-0880, USA
| | - Huoying Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China.
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University (JIRLAAPS), Yangzhou, China.
| |
Collapse
|
7
|
Teklemariam AD, Al-Hindi RR, Albiheyri RS, Alharbi MG, Alghamdi MA, Filimban AAR, Al Mutiri AS, Al-Alyani AM, Alseghayer MS, Almaneea AM, Albar AH, Khormi MA, Bhunia AK. Human Salmonellosis: A Continuous Global Threat in the Farm-to-Fork Food Safety Continuum. Foods 2023; 12:foods12091756. [PMID: 37174295 PMCID: PMC10178548 DOI: 10.3390/foods12091756] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Salmonella is one of the most common zoonotic foodborne pathogens and a worldwide public health threat. Salmonella enterica is the most pathogenic among Salmonella species, comprising over 2500 serovars. It causes typhoid fever and gastroenteritis, and the serovars responsible for the later disease are known as non-typhoidal Salmonella (NTS). Salmonella transmission to humans happens along the farm-to-fork continuum via contaminated animal- and plant-derived foods, including poultry, eggs, fish, pork, beef, vegetables, fruits, nuts, and flour. Several virulence factors have been recognized to play a vital role in attaching, invading, and evading the host defense system. These factors include capsule, adhesion proteins, flagella, plasmids, and type III secretion systems that are encoded on the Salmonella pathogenicity islands. The increased global prevalence of NTS serovars in recent years indicates that the control approaches centered on alleviating the food animals' contamination along the food chain have been unsuccessful. Moreover, the emergence of antibiotic-resistant Salmonella variants suggests a potential food safety crisis. This review summarizes the current state of the knowledge on the nomenclature, microbiological features, virulence factors, and the mechanism of antimicrobial resistance of Salmonella. Furthermore, it provides insights into the pathogenesis and epidemiology of Salmonella infections. The recent outbreaks of salmonellosis reported in different clinical settings and geographical regions, including Africa, the Middle East and North Africa, Latin America, Europe, and the USA in the farm-to-fork continuum, are also highlighted.
Collapse
Affiliation(s)
- Addisu D Teklemariam
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Rashad R Al-Hindi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Raed S Albiheyri
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mona G Alharbi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mashail A Alghamdi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Amani A R Filimban
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Abdullah S Al Mutiri
- Laboratory Department, Saudi Food and Drug Authority, Riyadh 12843, Saudi Arabia
| | - Abdullah M Al-Alyani
- Laboratory Department, Saudi Food and Drug Authority, Jeddah 22311, Saudi Arabia
| | - Mazen S Alseghayer
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Monitoring and Risk Assessment Department, Saudi Food and Drug Authority, Riyadh 13513, Saudi Arabia
| | - Abdulaziz M Almaneea
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Monitoring and Risk Assessment Department, Saudi Food and Drug Authority, Riyadh 13513, Saudi Arabia
| | - Abdulgader H Albar
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Microbiology and Medical Parasitology, Faculty of Medicine, Jeddah University, Jeddah 23218, Saudi Arabia
| | - Mohsen A Khormi
- Department of Biological Sciences, Faculty of Sciences, Jazan University, Jazan 82817, Saudi Arabia
| | - Arun K Bhunia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA
- Purdue University Interdisciplinary Life Science Program (PULSe), West Lafayette, IN 47907, USA
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
8
|
Zhou G, Zhao Y, Ma Q, Li Q, Wang S, Shi H. Manipulation of host immune defenses by effector proteins delivered from multiple secretion systems of Salmonella and its application in vaccine research. Front Immunol 2023; 14:1152017. [PMID: 37081875 PMCID: PMC10112668 DOI: 10.3389/fimmu.2023.1152017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/23/2023] [Indexed: 04/07/2023] Open
Abstract
Salmonella is an important zoonotic bacterial species and hazardous for the health of human beings and livestock globally. Depending on the host, Salmonella can cause diseases ranging from gastroenteritis to life-threatening systemic infection. In this review, we discuss the effector proteins used by Salmonella to evade or manipulate four different levels of host immune defenses: commensal flora, intestinal epithelial-mucosal barrier, innate and adaptive immunity. At present, Salmonella has evolved a variety of strategies against host defense mechanisms, among which various effector proteins delivered by the secretory systems play a key role. During its passage through the digestive system, Salmonella has to face the intact intestinal epithelial barrier as well as competition with commensal flora. After invasion of host cells, Salmonella manipulates inflammatory pathways, ubiquitination and autophagy processes with the help of effector proteins. Finally, Salmonella evades the adaptive immune system by interfering the migration of dendritic cells and interacting with T and B lymphocytes. In conclusion, Salmonella can manipulate multiple aspects of host defense to promote its replication in the host.
Collapse
Affiliation(s)
- Guodong Zhou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yuying Zhao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Qifeng Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Quan Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Shifeng Wang
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Huoying Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University (JIRLAAPS), Yangzhou, China
| |
Collapse
|
9
|
Salmonella in Poultry and Other Birds. Infect Dis (Lond) 2023. [DOI: 10.1007/978-1-0716-2463-0_1092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
10
|
Cao X, Du X, Jiao H, An Q, Chen R, Fang P, Wang J, Yu B. Carbohydrate-based drugs launched during 2000 -2021. Acta Pharm Sin B 2022; 12:3783-3821. [PMID: 36213536 PMCID: PMC9532563 DOI: 10.1016/j.apsb.2022.05.020] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/18/2022] [Accepted: 05/12/2022] [Indexed: 01/09/2023] Open
Abstract
Carbohydrates are fundamental molecules involved in nearly all aspects of lives, such as being involved in formating the genetic and energy materials, supporting the structure of organisms, constituting invasion and host defense systems, and forming antibiotics secondary metabolites. The naturally occurring carbohydrates and their derivatives have been extensively studied as therapeutic agents for the treatment of various diseases. During 2000 to 2021, totally 54 carbohydrate-based drugs which contain carbohydrate moities as the major structural units have been approved as drugs or diagnostic agents. Here we provide a comprehensive review on the chemical structures, activities, and clinical trial results of these carbohydrate-based drugs, which are categorized by their indications into antiviral drugs, antibacterial/antiparasitic drugs, anticancer drugs, antidiabetics drugs, cardiovascular drugs, nervous system drugs, and other agents.
Collapse
Affiliation(s)
- Xin Cao
- Zhongshan Hospital Institute of Clinical Science, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Xiaojing Du
- Zhongshan Hospital Institute of Clinical Science, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Heng Jiao
- Zhongshan Hospital Institute of Clinical Science, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Quanlin An
- Zhongshan Hospital Institute of Clinical Science, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Ruoxue Chen
- Zhongshan Hospital Institute of Clinical Science, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Pengfei Fang
- State Key Laboratory of Bio-organic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Jing Wang
- State Key Laboratory of Bio-organic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Biao Yu
- State Key Laboratory of Bio-organic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| |
Collapse
|
11
|
Mishra R, Chiang Tan Y, Adel Ahmed Abd El-Aal A, Lahiri C. Computational Identification of the Plausible Molecular Vaccine Candidates of Multidrug-Resistant Salmonella enterica. SALMONELLA SPP. - A GLOBAL CHALLENGE 2021. [DOI: 10.5772/intechopen.95856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Salmonella enterica serovars are responsible for the life-threatening, fatal, invasive diseases that are common in children and young adults. According to the most recent estimates, globally, there are approximately 11–20 million cases of morbidity and between 128,000 and 161,000 mortality per year. The high incidence rates of diseases like typhoid, caused by the serovars Typhi and Paratyphi, and gastroenteritis, caused by the non-typhoidal Salmonellae, have become worse, with the ever-increasing pathogenic strains being resistant to fluoroquinolones or almost even the third generation cephalosporins, such as ciprofloxacin and ceftriaxone. With vaccination still being one of the chosen methods of eradicating this disease, identification of candidate proteins, to be utilized for effective molecular vaccines, has probably remained a challenging issue. In our study here, we portray the usage of computational tools to analyze and predict potential vaccine candidate(s) for the multi-drug resistant serovars of S. enterica.
Collapse
|
12
|
Epidemiological Investigation of Salmonella enterica Isolates in Children with Diarrhea in Chengdu, China. Jundishapur J Microbiol 2021. [DOI: 10.5812/jjm.119034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: Children with the immature intestinal immune system are prone to Salmonella infection through the fecal-oral route causing diarrhea. Non-typhoid Salmonella (NTS) is difficult to treat and eliminate due to its zoonosis. Salmonella typhi, including typhoid and paratyphoid A, B, and C, only infect humans and cause invasive infectious diseases. Salmonella typhi infection is serious and requires antibiotic treatment. The bacterial resistance caused by conventional antibacterial drugs brings great difficulties to treatment. Objectives: This study aimed to investigate the epidemiology of S. enterica in children with diarrhea in Chengdu, China. Methods: Fresh stool specimens or rectal swabs from 6656 children aged 1 day to 13 years with diarrhea were collected, cultured, identified, and tested for antimicrobial susceptibility. Analytical Profile index 20E was used for biochemical identification, and the Kirby-Bauer method was used for the bacterial sensitivity test. The whole process was conducted in accordance with the fourth edition of the National Clinical Examination procedures, and the drug sensitivity test was conducted in accordance with the Clinical and Laboratory Standards Institute 2020 guidelines. Results: A total of 649 Salmonella strains were isolated from 6656 children with suspected Salmonella infection, among which the isolation rates of NTS and S. typhi were 8.92% and 0.83%, respectively. The infection rate of S. typhimurium was the highest every year (74.88%). Salmonella infections are on the rise, especially typhimurium, Dublin, Typhi, and London. Paratyphi is unstable, presenting a phenomenon of transition and replacement (the male to female ratio:1.12:1). The infection rate was the lowest within 1 day and 6 months (P < 0.05). Salmonella mainly infected children under 3 years of age, and the positive rate was reported as 88.29%. Within June-September, the infection rate of Salmonella was the highest, with a positive rate of 72.73%. The isolated 649 Salmonella strains had good susceptibility to cefotaxime and ciprofloxacin (87.67% and 79.20%, respectively), almost no susceptibility to ampicillin, and a drug resistance rate of 92.91%. Conclusions: The typhoid and paratyphoid vaccines should be considered together, and vaccines should focus on children under 3 years of age. Antibiotics should be rationally selected according to the drug sensitivity test and disease condition.
Collapse
|
13
|
Bearson SMD. Salmonella in Swine: Prevalence, Multidrug Resistance, and Vaccination Strategies. Annu Rev Anim Biosci 2021; 10:373-393. [PMID: 34699256 DOI: 10.1146/annurev-animal-013120-043304] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
An estimated 1.3 million Salmonella infections and 420 deaths occur annually in the United States, with an estimated economic burden of $3.7 billion. More than 50% of US swine operations test positive for Salmonella according to the National Animal Health Monitoring System, and 20% of Salmonella from swine are multidrug resistant (resistant to ≥3 antimicrobial classes) as reported by the National Antimicrobial Resistance Monitoring System. This review on Salmonella in swine addresses the current status of these topics by discussing antimicrobial resistance and metal tolerance in Salmonella and the contribution of horizontal gene transfer. A major challenge in controlling Salmonella is that Salmonella is a foodborne pathogen in humans but is often a commensal in food animals and thereby establishes an asymptomatic reservoir state in such animals, including swine. As food animal production systems continue to expand and antimicrobial usage becomes more limited, the need for Salmonella interventions has intensified. A promising mitigation strategy is vaccination against Salmonella in swine to limit animal, environmental, and food contamination. Expected final online publication date for the Annual Review of Animal Biosciences, Volume 10 is February 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Shawn M D Bearson
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Center, US Department of Agriculture, Ames, Iowa, USA;
| |
Collapse
|
14
|
Avendaño C, Vidal S, Villamizar-Sarmiento MG, Guzmán M, Hidalgo H, Lapierre L, Valenzuela C, Sáenz L. Encapsulation of Cochleates Derived from Salmonella Infantis with Biopolymers to Develop a Potential Oral Poultry Vaccine. Polymers (Basel) 2021; 13:3426. [PMID: 34641241 PMCID: PMC8512329 DOI: 10.3390/polym13193426] [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: 09/13/2021] [Accepted: 09/28/2021] [Indexed: 12/31/2022] Open
Abstract
The aim of this study was to develop and characterize Salmonellaenterica serovar Infantis (S. Infantis) cochleates protected by encapsulation technology as a potential vaccine and to determine its safety in pullets. Cochleates were encapsulated by two technologies, spray drying and ionotropic gelation at different concentrations (0-15% v/v), and were characterized by physicochemical properties, protein content and Fourier Transform Infrared Spectroscopy (FTIR). The cochleates were white liquid suspensions with tubular shapes and a protein content of 1.0-2.1 mg/mL. After encapsulation by spray drying, microparticles ranged in size from 10.4-16.9 µm, were spherical in shape, and the protein content was 0.7-1.8 mg/g. After encapsulation by ionotropic gelation, beads ranged in size from 1620-1950 µm and were spherical in shape with a protein content of 1.0-2.5 mg/g. FTIR analysis indicated that both encapsulation processes were efficient. The cochleates encapsulated by ionotropic gelation were then tested for safety in pullets. No ill effect on the health of animals was observed upon physical or postmortem examination. In conclusion, this study was the first step in developing a potential oral S. Infantis vaccine safe for poultry using a novel cochleate encapsulation technology. Future studies are needed to determine the effectiveness of the vaccine.
Collapse
Affiliation(s)
- Constanza Avendaño
- Faculty of Veterinary Sciences, University of Chile, Santiago 8820808, Chile; (C.A.); (S.V.); (M.G.V.-S.); (M.G.); (H.H.); (L.L.)
| | - Sonia Vidal
- Faculty of Veterinary Sciences, University of Chile, Santiago 8820808, Chile; (C.A.); (S.V.); (M.G.V.-S.); (M.G.); (H.H.); (L.L.)
| | - María Gabriela Villamizar-Sarmiento
- Faculty of Veterinary Sciences, University of Chile, Santiago 8820808, Chile; (C.A.); (S.V.); (M.G.V.-S.); (M.G.); (H.H.); (L.L.)
- Department of Sciences and Pharmaceutical Technology, University of Chile, Santiago 8380494, Chile
| | - Miguel Guzmán
- Faculty of Veterinary Sciences, University of Chile, Santiago 8820808, Chile; (C.A.); (S.V.); (M.G.V.-S.); (M.G.); (H.H.); (L.L.)
- Nucleus of Applied Research in Veterinary and Agronomic Sciences, NIAVA, Faculty of Veterinary Medicine and Agronomy, Universidad de las Américas, Santiago 9250000, Chile
| | - Héctor Hidalgo
- Faculty of Veterinary Sciences, University of Chile, Santiago 8820808, Chile; (C.A.); (S.V.); (M.G.V.-S.); (M.G.); (H.H.); (L.L.)
| | - Lisette Lapierre
- Faculty of Veterinary Sciences, University of Chile, Santiago 8820808, Chile; (C.A.); (S.V.); (M.G.V.-S.); (M.G.); (H.H.); (L.L.)
| | - Carolina Valenzuela
- Faculty of Veterinary Sciences, University of Chile, Santiago 8820808, Chile; (C.A.); (S.V.); (M.G.V.-S.); (M.G.); (H.H.); (L.L.)
| | - Leonardo Sáenz
- Faculty of Veterinary Sciences, University of Chile, Santiago 8820808, Chile; (C.A.); (S.V.); (M.G.V.-S.); (M.G.); (H.H.); (L.L.)
| |
Collapse
|
15
|
Kuźmińska-Bajor M, Śliwka P, Ugorski M, Korzeniowski P, Skaradzińska A, Kuczkowski M, Narajaczyk M, Wieliczko A, Kolenda R. Genomic and functional characterization of five novel Salmonella-targeting bacteriophages. Virol J 2021; 18:183. [PMID: 34496915 PMCID: PMC8425127 DOI: 10.1186/s12985-021-01655-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/29/2021] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND The host-unrestricted, non-typhoidal Salmonella enterica serovar Enteritidis (S. Enteritidis) and the serovar Typhimurium (S. Typhimurium) are major causative agents of food-borne gastroenteritis, and the host-restricted Salmonella enterica serovar Gallinarum (S. Gallinarum) is responsible for fowl typhoid. Increasing drug resistance in Salmonella contributes to the reduction of effective therapeutic and/or preventive options. Bacteriophages appear to be promising antibacterial tools, able to combat infectious diseases caused by a wide range of Salmonella strains belonging to both host-unrestricted and host-restricted Salmonella serovars. METHODS In this study, five novel lytic Salmonella phages, named UPWr_S1-5, were isolated and characterized, including host range determination by plaque formation, morphology visualization with transmission electron microscopy, and establishment of physiological parameters. Moreover, phage genomes were sequenced, annotated and analyzed, and their genomes were compared with reference Salmonella phages by use of average nucleotide identity, phylogeny, dot plot, single nucleotide variation and protein function analysis. RESULTS It was found that UPWr_S1-5 phages belong to the genus Jerseyvirus within the Siphoviridae family. All UPWr_S phages were found to efficiently infect various Salmonella serovars. Host range determination revealed differences in host infection profiles and exhibited ability to infect Salmonella enterica serovars such as Enteritidis, Gallinarum, Senftenberg, Stanley and Chester. The lytic life cycle of UPWr_S phages was confirmed using the mitomycin C test assay. Genomic analysis revealed that genomes of UPWr_S phages are composed of 51 core and 19 accessory genes, with 33 of all predicted genes having assigned functions. UPWr_S genome organization comparison revealed 3 kinds of genomes and mosaic structure. UPWr_S phages showed very high sequence similarity to each other, with more than 95% average nucleotide identity. CONCLUSIONS Five novel UPWr_S1-5 bacteriophages were isolated and characterized. They exhibit host lysis range within 5 different serovars and are efficient in lysis of both host-unrestricted and host-restricted Salmonella serovars. Therefore, because of their ability to infect various Salmonella serovars and lytic life cycle, UPWr_S1-5 phages can be considered as useful tools in biological control of salmonellosis.
Collapse
Affiliation(s)
- Marta Kuźmińska-Bajor
- Department of Biotechnology and Food Microbiology, Faculty of Biotechnology and Food Sciences, Wrocław University of Environmental and Life Sciences, Wrocław, Poland.
| | - Paulina Śliwka
- Department of Biotechnology and Food Microbiology, Faculty of Biotechnology and Food Sciences, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Maciej Ugorski
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Paweł Korzeniowski
- Department of Biotechnology and Food Microbiology, Faculty of Biotechnology and Food Sciences, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Aneta Skaradzińska
- Department of Biotechnology and Food Microbiology, Faculty of Biotechnology and Food Sciences, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Maciej Kuczkowski
- Department of Epizootiology and Clinic of Birds and Exotic Animals, Faculty of Veterinary Medicine, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Magdalena Narajaczyk
- Department of Electron Microscopy, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | - Alina Wieliczko
- Department of Epizootiology and Clinic of Birds and Exotic Animals, Faculty of Veterinary Medicine, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Rafał Kolenda
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| |
Collapse
|
16
|
Ji HJ, Byun EB, Chen F, Ahn KB, Jung HK, Han SH, Lim JH, Won Y, Moon JY, Hur J, Seo HS. Radiation-Inactivated S. gallinarum Vaccine Provides a High Protective Immune Response by Activating Both Humoral and Cellular Immunity. Front Immunol 2021; 12:717556. [PMID: 34484221 PMCID: PMC8415480 DOI: 10.3389/fimmu.2021.717556] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/22/2021] [Indexed: 11/19/2022] Open
Abstract
Salmonella enterica subsp. enterica serovar Gallinarum (SG) is a common pathogen in chickens, and causes an acute systemic disease that leads to high mortality. The live attenuated vaccine 9R is able to successfully protect chickens older than six weeks by activating a robust cell-mediated immune response, but its safety and efficacy in young chickens remains controversial. An inactivated SG vaccine is being used as an alternative, but because of its low cellular immune response, it cannot be used as a replacement for live attenuated 9R vaccine. In this study, we employed gamma irradiation instead of formalin as an inactivation method to increase the efficacy of the inactivated SG vaccine. Humoral, cellular, and protective immune responses were compared in both mouse and chicken models. The radiation-inactivated SG vaccine (r-SG) induced production of significantly higher levels of IgG2b and IgG3 antibodies than the formalin-inactivated vaccine (f-SG), and provided a homogeneous functional antibody response against group D, but not group B Salmonella. Moreover, we found that r-SG vaccination could provide a higher protective immune response than f-SG by inducing higher Th17 activation. These results indicate that r-SG can provide a protective immune response similar to the live attenuated 9R vaccine by activating a higher humoral immunity and a lower, but still protective, cellular immune response. Therefore, we expect that the radiation inactivation method might substitute for the 9R vaccine with little or no side effects in chickens younger than six weeks.
Collapse
Affiliation(s)
- Hyun Jung Ji
- Research Division for Radiation Science, Korea Atomic Energy Research Institute, Jeongeup, South Korea.,Department of Oral Microbiology and Immunology, and DRI, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Eui-Baek Byun
- Research Division for Radiation Science, Korea Atomic Energy Research Institute, Jeongeup, South Korea
| | - Fengjia Chen
- Research Division for Radiation Science, Korea Atomic Energy Research Institute, Jeongeup, South Korea
| | - Ki Bum Ahn
- Research Division for Radiation Science, Korea Atomic Energy Research Institute, Jeongeup, South Korea
| | - Ho Kyoung Jung
- Research and Development Center, HONGCHEON CTCVAC Co., Ltd., Hongcheon, South Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, and DRI, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Jae Hyang Lim
- Department of Microbiology, Ewha Womans University College of Medicine, Seoul, South Korea.,Ewha Education & Research Center for Infection, Ewha Womans University Medical Center, Seoul, South Korea
| | - Yongkwan Won
- Research and Development Center, HONGCHEON CTCVAC Co., Ltd., Hongcheon, South Korea
| | - Ja Young Moon
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - Jin Hur
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - Ho Seong Seo
- Research Division for Radiation Science, Korea Atomic Energy Research Institute, Jeongeup, South Korea.,Department of Radiation Science, University of Science and Technology, Daejeon, South Korea
| |
Collapse
|
17
|
An Outer Membrane Vesicle-Adjuvanted Oral Vaccine Protects Against Lethal, Oral Salmonella Infection. Pathogens 2021; 10:pathogens10050616. [PMID: 34069796 PMCID: PMC8157261 DOI: 10.3390/pathogens10050616] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/14/2021] [Accepted: 05/16/2021] [Indexed: 11/23/2022] Open
Abstract
Non-typhoidal salmonellosis, caused by Salmonella enterica serovar Typhimurium is a common fecal-oral disease characterized by mild gastrointestinal distress resulting in diarrhea, chills, fever, abdominal cramps, head and body aches, nausea, and vomiting. Increasing incidences of antibiotic resistant invasive non-typhoidal Salmonella infections makes this a global threat requiring novel treatment strategies including next-generation vaccines. The goal of the current study was to formulate a novel vaccine platform against Salmonella infection that could be delivered orally. To accomplish this, we created a Salmonella-specific vaccine adjuvanted with Burkholderia pseudomallei outer membrane vesicles (OMVs). We show that adding OMVs to a heat-killed oral Salmonella vaccine (HKST + OMVs) protects against a lethal, oral challenge with Salmonella. Further, we show that opsonizing anti-Salmonella antibodies are induced in response to immunization and that CD4 T cells and B cells can be induced when OMVs are used as the oral adjuvant. This study represents a novel oral vaccine approach to combatting the increasing problem of invasive Salmonella infections.
Collapse
|
18
|
Erazo AB, Wang N, Standke L, Semeniuk AD, Fülle L, Cengiz SC, Thiem M, Weighardt H, Strugnell RA, Förster I. CCL17-expressing dendritic cells in the intestine are preferentially infected by Salmonella but CCL17 plays a redundant role in systemic dissemination. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:891-904. [PMID: 33945673 PMCID: PMC8342217 DOI: 10.1002/iid3.445] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/10/2021] [Accepted: 04/13/2021] [Indexed: 12/14/2022]
Abstract
Introduction Salmonella spp. are a recognized and global cause of serious health issues from gastroenteritis to invasive disease. The mouse model of human typhoid fever, which uses Salmonella enterica serovar Typhimurium (STM) in susceptible mouse strains, has revealed that the bacteria gain access to extraintestinal tissues from the gastrointestinal tract to cause severe systemic disease. Previous analysis of the immune responses against Salmonella spp. revealed the crucial role played by dendritic cells (DCs) in carrying STM from the intestinal mucosa to the mesenteric lymph nodes (mLNs), a key site for antigen presentation and T cell activation. In this study, we investigated the influence of chemokine CCL17 on the dissemination of STM. Methods WT, CCL17/EGFP reporter, or CCL17‐deficient mice were infected orally with STM (SL1344) or mCherry‐expressing STM for 1–3 days. Colocalization of STM with CCL17‐expressing DCs in Peyer's patches (PP) and mLN was analyzed by fluorescence microscopy. In addition, DCs and myeloid cell populations from naïve and Salmonella‐infected mice were analyzed by flow cytometry. Bacterial load was determined in PP, mLN, spleen, and liver 1 and 3 days after infection. Results Histological analysis revealed that CCL17‐expressing cells are located in close proximity to STM in the dome area of PP. We show that, in mLN, STM were preferentially located within CCL17+ rather than CCL17− DCs, besides other mononuclear phagocytes, and identified the CD103+ CD11b− DC subset as the main STM‐carrying DC population in the intestine. STM infection triggered upregulation of CCL17 expression in specific intestinal DC subsets in a tissue‐specific manner. The dissemination of STM from the gut to the mLN, however, was only moderately influenced by the presence of CCL17. Conclusion CCL17‐expressing DCs were preferentially infected by Salmonella in the intestine in comparison to other DC. Nevertheless, the production of CCL17 was not essential for the early dissemination of Salmonella from the gut to systemic organs.
Collapse
Affiliation(s)
- Anna B Erazo
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.,Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Nancy Wang
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Lena Standke
- Department for Innate Immunity and Metaflammation, Institute of Innate Immunity, University Hospital Bonn, Medical Faculty, Bonn, Germany
| | - Adrian D Semeniuk
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.,Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Lorenz Fülle
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Sevgi C Cengiz
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Manja Thiem
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Heike Weighardt
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Richard A Strugnell
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Irmgard Förster
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
19
|
Ontiveros-Padilla L, García-Lozano A, Tepale-Segura A, Rivera-Hernández T, Pastelin-Palacios R, Isibasi A, Arriaga-Pizano LA, Bonifaz LC, López-Macías C. CD4+ and CD8+ Circulating Memory T Cells Are Crucial in the Protection Induced by Vaccination with Salmonella Typhi Porins. Microorganisms 2021; 9:microorganisms9040770. [PMID: 33916894 PMCID: PMC8067540 DOI: 10.3390/microorganisms9040770] [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: 03/09/2021] [Revised: 03/23/2021] [Accepted: 03/29/2021] [Indexed: 11/16/2022] Open
Abstract
Salmonella enterica serovar Typhi (S. Typhi) porins, OmpC and OmpF, are potent inducers of the immune response against S. Typhi in mice and humans. Vaccination with porins induces the protection against 500 LD50 of S. Typhi, life-lasting bactericidal antibodies and effector T cell responses in mice; however, the nature of the memory T cell compartment and its contribution to protection remains unknown. In this work, we firstly observed that vaccination with porins induces in situ (skin) CD4+ and CD8+ T cell responses. Analysis of the porin-specific functional responses of skin CD4+ and CD8+ T cells showed IFN-gamma- and IL-17-producing cells in both T cell populations. The memory phenotype of porin-specific T cells indicated the presence of resident and effector memory phenotypes in the skin, and a central memory phenotype in the skin-draining lymph node. In addition, we demonstrated that vaccination with porins via skin reduces the bacterial burden following challenge. Finally, evaluating the role of the circulating T cell memory population in protection, we showed that circulating memory CD4+ and CD8+ T cells are crucial in porin-mediated protection against S. Typhi. Overall, this study highlights the importance of inducing circulating memory T cell responses in order to achieve the optimal protection provided by porins, showing a mechanism that could be sought in the rational development of vaccines.
Collapse
Affiliation(s)
- Luis Ontiveros-Padilla
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades del Centro Médico Nacional “Siglo XXI”, Instituto Mexicano del Seguro Social (IMSS), Cuauhtemoc, Ciudad de Mexico 06720, Mexico; (L.O.-P.); (A.G.-L.); (A.T.-S.); (T.R.-H.); (A.I.); (L.A.A.-P.)
- Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Coyoacan, Ciudad de Mexico 04510, Mexico;
| | - Alberto García-Lozano
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades del Centro Médico Nacional “Siglo XXI”, Instituto Mexicano del Seguro Social (IMSS), Cuauhtemoc, Ciudad de Mexico 06720, Mexico; (L.O.-P.); (A.G.-L.); (A.T.-S.); (T.R.-H.); (A.I.); (L.A.A.-P.)
| | - Araceli Tepale-Segura
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades del Centro Médico Nacional “Siglo XXI”, Instituto Mexicano del Seguro Social (IMSS), Cuauhtemoc, Ciudad de Mexico 06720, Mexico; (L.O.-P.); (A.G.-L.); (A.T.-S.); (T.R.-H.); (A.I.); (L.A.A.-P.)
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Miguel Hidalgo, Ciudad de Mexico 11340, Mexico
| | - Tania Rivera-Hernández
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades del Centro Médico Nacional “Siglo XXI”, Instituto Mexicano del Seguro Social (IMSS), Cuauhtemoc, Ciudad de Mexico 06720, Mexico; (L.O.-P.); (A.G.-L.); (A.T.-S.); (T.R.-H.); (A.I.); (L.A.A.-P.)
- Consejo Nacional de Ciencia y Tecnología (CONACYT), Benito Juárez, Ciudad de Mexico 03940, Mexico
| | - Rodolfo Pastelin-Palacios
- Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Coyoacan, Ciudad de Mexico 04510, Mexico;
| | - Armando Isibasi
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades del Centro Médico Nacional “Siglo XXI”, Instituto Mexicano del Seguro Social (IMSS), Cuauhtemoc, Ciudad de Mexico 06720, Mexico; (L.O.-P.); (A.G.-L.); (A.T.-S.); (T.R.-H.); (A.I.); (L.A.A.-P.)
| | - Lourdes A. Arriaga-Pizano
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades del Centro Médico Nacional “Siglo XXI”, Instituto Mexicano del Seguro Social (IMSS), Cuauhtemoc, Ciudad de Mexico 06720, Mexico; (L.O.-P.); (A.G.-L.); (A.T.-S.); (T.R.-H.); (A.I.); (L.A.A.-P.)
| | - Laura C. Bonifaz
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades del Centro Médico Nacional “Siglo XXI”, Instituto Mexicano del Seguro Social (IMSS), Cuauhtemoc, Ciudad de Mexico 06720, Mexico; (L.O.-P.); (A.G.-L.); (A.T.-S.); (T.R.-H.); (A.I.); (L.A.A.-P.)
- Correspondence: (L.C.B.); or (C.L.-M.)
| | - Constantino López-Macías
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades del Centro Médico Nacional “Siglo XXI”, Instituto Mexicano del Seguro Social (IMSS), Cuauhtemoc, Ciudad de Mexico 06720, Mexico; (L.O.-P.); (A.G.-L.); (A.T.-S.); (T.R.-H.); (A.I.); (L.A.A.-P.)
- Correspondence: (L.C.B.); or (C.L.-M.)
| |
Collapse
|
20
|
Sears KT, Galen JE, Tennant SM. Advances in the development of Salmonella-based vaccine strategies for protection against Salmonellosis in humans. J Appl Microbiol 2021; 131:2640-2658. [PMID: 33665941 PMCID: PMC9292744 DOI: 10.1111/jam.15055] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/18/2021] [Accepted: 03/02/2021] [Indexed: 11/29/2022]
Abstract
Salmonella spp. are important human pathogens globally causing millions of cases of typhoid fever and non‐typhoidal salmonellosis annually. There are only a few vaccines licensed for use in humans which all target Salmonella enterica serovar Typhi. Vaccine development is hampered by antigenic diversity between the thousands of serovars capable of causing infection in humans. However, a number of attenuated candidate vaccine strains are currently being developed. As facultative intracellular pathogens with multiple systems for transporting effector proteins to host cells, attenuated Salmonella strains can also serve as ideal tools for the delivery of foreign antigens to create multivalent live carrier vaccines for simultaneous immunization against several unrelated pathogens. Further, the ease with which Salmonella can be genetically modified and the extensive knowledge of the virulence mechanisms of this pathogen means that this bacterium has often served as a model organism to test new approaches. In this review we focus on (1) recent advances in live attenuated Salmonella vaccine development, (2) improvements in expression of foreign antigens in carrier vaccines and (3) adaptation of attenuated strains as sources of purified antigens and vesicles that can be used for subunit and conjugate vaccines or together with attenuated vaccine strains in heterologous prime‐boosting immunization strategies. These advances have led to the development of new vaccines against Salmonella which have or will soon be tested in clinical trials.
Collapse
Affiliation(s)
- K T Sears
- Center for Vaccine Development and Global Health, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - J E Galen
- Center for Vaccine Development and Global Health, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - S M Tennant
- Center for Vaccine Development and Global Health, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
21
|
Protection Conferred by Drinking Water Administration of a Nanoparticle-Based Vaccine against Salmonella Enteritidis in Hens. Vaccines (Basel) 2021; 9:vaccines9030216. [PMID: 33802556 PMCID: PMC8001700 DOI: 10.3390/vaccines9030216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/23/2021] [Accepted: 02/23/2021] [Indexed: 12/22/2022] Open
Abstract
Salmonellosis remains a major medical and an unmet socioeconomic challenge. Worldwide, more than three million deaths per year are associated with Salmonella enterica serovar Enteritidis infections. Although commercially available vaccines for use in poultry exist, their efficacy is limited. We previously described a method for isolating a heat extract (HE) fraction of the cell surface of S. Enteritidis that contained major antigenic complexes immunogenic in hens naturally infected with the bacterium. One single dose of S. Enteritidis’ HE induced protection against lethal salmonellosis in mice. Furthermore, HE encapsulation in nanoparticles of the copolymer of methyl vinyl ether and maleic anhydride (PVM/MA), Gantrez AN, improved and prolonged the protection against the disease in mice. We formulated new preparations of Gantrez AN nanoparticles with HE S. Enteritidis and assessed their stability in drinking water and their efficacy in hens after experimental infection. The oral treatment of six-week-old hens with two doses of HE nanoparticles significantly reduced the Salmonella excretion in hens. Due to the effectiveness of the treatment in reducing bacterial excretion, we conclude that HE nanoencapsulation obtained from S. Enteritidis is a viable novel vaccination approach against salmonellosis in farms.
Collapse
|
22
|
Zhao X, Zeng X, Dai Q, Hou Y, Zhu D, Wang M, Jia R, Chen S, Liu M, Yang Q, Wu Y, Zhang S, Huang J, Ou X, Mao S, Gao Q, Zhang L, Liu Y, Yu Y, Cheng A. Immunogenicity and protection efficacy of a Salmonella enterica serovar Typhimurium fnr, arcA and fliC mutant. Vaccine 2020; 39:588-595. [PMID: 33341307 DOI: 10.1016/j.vaccine.2020.12.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 11/05/2020] [Accepted: 12/01/2020] [Indexed: 01/17/2023]
Abstract
Salmonella enterica serovar Typhimurium is a major food-borne pathogen that can cause self-limited gastroenteritis or life-threatening invasive diseases in humans. There is no licensed S. Typhimurium vaccine for humans to date. In this study, we attempted to construct a live attenuated vaccine strain of S. Typhimurium based on three genes, namely, the two global regulator genes fnr and arcA and the flagellin subunit gene fliC. The S. Typhimurium three-gene mutant, named SLT39 (ΔfnrΔarcAΔfliC), exhibited a high level of attenuation with a colonization defect in mouse tissues and approximately 104-fold decreased virulence compared with that of the wild-type strain. To evaluate the immunogenicity and protection efficacy of STL39, mice were inoculated twice with a dose of 107 CFU or 108 CFU at a 28-day interval, and the immunized mice were challenged with a lethal dose of the wild-type S. Typhimurium strain one month post second immunization. Compared with mock immunization, SLT39 immunization with either dose elicited significant serum total IgG, IgG1 and IgG2a and faecal IgA responses against inactivated S. Typhimurium antigens at a comparable level post second immunization, whereas the 108 CFU group induced higher levels of duodenal and caecal IgA than the 107 CFU group. Furthermore, the bacterial loads in mouse tissues, including Peyer's patches, spleen and liver, significantly decreased in the two SLT39 immunization groups compared to those in the control group post challenge. Additionally, all mice in the SLT39 (108 CFU) group and 80% of the mice in the SLT39 (107 CFU) group survived the lethal challenge, suggesting full protection and 80% protection efficacy, respectively. Thus, the S. Typhimurium fnr, arcA and fliC mutant proved to be a potential attenuated live vaccine candidate for prevention of homologous infection.
Collapse
Affiliation(s)
- Xinxin Zhao
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China; Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xiaoli Zeng
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Qinlong Dai
- Liziping National Nature Reserve, Shimian, Sichuan, China
| | - Yulong Hou
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Dekang Zhu
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China; Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Mingshu Wang
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China; Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Renyong Jia
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China; Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Shun Chen
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China; Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Mafeng Liu
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China; Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Qiao Yang
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China; Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Ying Wu
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China; Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Shaqiu Zhang
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China; Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Juan Huang
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China; Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xumin Ou
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China; Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Sai Mao
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China; Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Qun Gao
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China; Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Ling Zhang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China; Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yunya Liu
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China; Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yanling Yu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China; Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Anchun Cheng
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China; Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China.
| |
Collapse
|
23
|
Masuet-Aumatell C, Atouguia J. Typhoid fever infection - Antibiotic resistance and vaccination strategies: A narrative review. Travel Med Infect Dis 2020; 40:101946. [PMID: 33301931 DOI: 10.1016/j.tmaid.2020.101946] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023]
Abstract
Typhoid fever is a bacterial infection caused by the Gram-negative bacterium Salmonella enterica subspecies enterica serovar Typhi (S. Typhi), prevalent in many low- and middle-income countries. In high-income territories, typhoid fever is predominantly travel-related, consequent to travel in typhoid-endemic regions; however, data show that the level of typhoid vaccination in travellers is low. Successful management of typhoid fever using antibiotics is becoming increasingly difficult due to drug resistance; emerging resistance has spread geographically due to factors such as increasing travel connectivity, affecting those in endemic regions and travellers alike. This review provides an overview of: the epidemiology and diagnosis of typhoid fever; the emergence of drug-resistant typhoid strains in the endemic setting; drug resistance observed in travellers; vaccines currently available to prevent typhoid fever; vaccine recommendations for people living in typhoid-endemic regions; strategies for the introduction of typhoid vaccines and stakeholders in vaccination programmes; and travel recommendations for a selection of destinations with a medium or high incidence of typhoid fever.
Collapse
Affiliation(s)
- Cristina Masuet-Aumatell
- Preventive Medicine Department, Bellvitge Biomedical Research Institute (IDIBELL), University Hospital of Bellvitge, Faculty of Medicine, University of Barcelona, Feixa Llarga s/n, L'Hospitalet de Llobregat, 08907, Barcelona, Catalonia, Spain.
| | - Jorge Atouguia
- Instituto Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junquiera, 100, Lisbon, Portugal.
| |
Collapse
|
24
|
Inhibition of invasive salmonella by orally administered IgA and IgG monoclonal antibodies. PLoS Negl Trop Dis 2020; 14:e0007803. [PMID: 32203503 PMCID: PMC7117778 DOI: 10.1371/journal.pntd.0007803] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 04/02/2020] [Accepted: 02/26/2020] [Indexed: 12/15/2022] Open
Abstract
Non-typhoidal Salmonella enterica strains, including serovar Typhimurium (STm), are an emerging cause of invasive disease among children and the immunocompromised, especially in regions of sub-Saharan Africa. STm invades the intestinal mucosa through Peyer's patch tissues before disseminating systemically. While vaccine development efforts are ongoing, the emergence of multidrug resistant strains of STm affirms the need to seek alternative strategies to protect high-risk individuals from infection. In this report, we investigated the potential of an orally administered O5 serotype-specific IgA monoclonal antibody (mAb), called Sal4, to prevent infection of invasive Salmonella enterica serovar Typhimurium (STm) in mice. Sal4 IgA was delivered to mice prior to or concurrently with STm challenge. Infectivity was measured as bacterial burden in Peyer's patch tissues one day after challenge. Using this model, we defined the minimal amount of Sal4 IgA required to significantly reduce STm uptake into Peyer's patches. The relative efficacy of Sal4 in dimeric and secretory IgA (SIgA) forms was compared. To assess the role of isotype in oral passive immunization, we engineered a recombinant IgG1 mAb carrying the Sal4 variable regions and evaluated its ability to block invasion of STm into epithelial cells in vitro and Peyer's patch tissues. Our results demonstrate the potential of orally administered monoclonal IgA and SIgA, but not IgG, to passively immunize against invasive Salmonella. Nonetheless, the prophylactic window of IgA/SIgA in the mouse was on the order of minutes, underscoring the need to develop formulations to protect mAbs in the gastric environment and to permit sustained release in the small intestine.
Collapse
|
25
|
Wang M, Qazi IH, Wang L, Zhou G, Han H. Salmonella Virulence and Immune Escape. Microorganisms 2020; 8:microorganisms8030407. [PMID: 32183199 PMCID: PMC7143636 DOI: 10.3390/microorganisms8030407] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/02/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023] Open
Abstract
Salmonella genus represents the most common foodborne pathogens causing morbidity, mortality, and burden of disease in all regions of the world. The introduction of antimicrobial agents and Salmonella-specific phages has been considered as an effective intervention strategy to reduce Salmonella contamination. However, data from the United States, European countries, and low- and middle-income countries indicate that Salmonella cases are still a commonly encountered cause of bacterial foodborne diseases globally. The control programs have not been successful and even led to the emergence of some multidrug-resistant Salmonella strains. It is known that the host immune system is able to effectively prevent microbial invasion and eliminate microorganisms. However, Salmonella has evolved mechanisms of resisting host physical barriers and inhibiting subsequent activation of immune response through their virulence factors. There has been a high interest in understanding how Salmonella interacts with the host. Therefore, in the present review, we characterize the functions of Salmonella virulence genes and particularly focus on the mechanisms of immune escape in light of evidence from the emerging mainstream literature.
Collapse
Affiliation(s)
- Mengyao Wang
- Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (M.W.); (L.W.)
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Izhar Hyder Qazi
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China;
- Department of Veterinary Anatomy and Histology, Shaheed Benazir Bhutto University of Veterinary and Animal Sciences, Sakrand 67210, Pakistan
| | - Linli Wang
- Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (M.W.); (L.W.)
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Guangbin Zhou
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China;
- Correspondence: (H.H.); (G.Z.)
| | - Hongbing Han
- Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (M.W.); (L.W.)
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
- Correspondence: (H.H.); (G.Z.)
| |
Collapse
|
26
|
Li M, Wang Y, Sun Y, Cui H, Zhu SJ, Qiu HJ. Mucosal vaccines: Strategies and challenges. Immunol Lett 2019; 217:116-125. [PMID: 31669546 DOI: 10.1016/j.imlet.2019.10.013] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/08/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023]
Abstract
Mucosal immunization has potential benefits over conventional parenteral immunization, eliciting immune defense in both mucosal and systemic tissue for protecting from pathogen invasion at mucosal surfaces. To provide a first line of protection at these entry ports, mucosal vaccines have been developed and hold a significant promise for reducing the burden of infectious diseases. However, until very recently, only limited mucosal vaccines are available. This review summarizes recent advances in selected aspects regarding mucosal vaccination, including appropriate administration routes, reasonable formulations, antigen-sampling and immune responses of mucosal immunity, and the strategies used to improve mucosal vaccine efficacy. Finally, the challenges of developing successful mucosal vaccines and the potential solutions are discussed.
Collapse
Affiliation(s)
- Miao Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yi Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yuan Sun
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hongyu Cui
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Shu J Zhu
- College of Animal Science, Zhejiang University, Hangzhou, China.
| | - Hua-Ji Qiu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| |
Collapse
|
27
|
Zafar H, Rahman SU, Ali S, Javed MT. Evaluation of a Salmonella Strain Isolated from Honeybee Gut as a Potential Live Oral Vaccine Against Lethal Infection of Salmonella Typhimurium. Pol J Microbiol 2019; 68:173-183. [PMID: 31257790 DOI: 10.21307/pjm-2019-017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 01/16/2019] [Accepted: 01/19/2019] [Indexed: 11/11/2022] Open
Abstract
In this research, Salmonella species were isolated from the animal, insect and human enteric sources in Faisalabad, Punjab, Pakistan. These species were characterized by different microbiological and molecular techniques including polymerase chain reaction (PCR) by amplification of the 16S rRNA gene. Furthermore, sequencing of the amplicons confirmed all ten isolates as Salmonella strains. The antigenic cross-reactivity was found maximum between the HB1 (strain isolated from honeybee) antiserum and its antigen with an antibody titer of 1:128, while the HB1 antiserum showed a cross-reactive titer range of 1:8 to 1:64. On the basis of the highest geometric mean titer (GMT) shown by the antiserum of the HB1 antigen, it was selected as the best candidate for a cross-reactive live Salmonella oral antigen. Moreover, the HB1 antigen was used a live oral antigen (1 × 1010 CFU/ml) in a safety test in rabbits and proved to be avirulent. During the animal trial, three different oral doses of the HB1 live oral antigen were evaluated in four different rabbits' groups (R1, R2, R3, and R4). The dose number 2 of 0.5 ml (two drops orally and repeated after one week) gave the best GMT measured by indirect hemagglutination (IHA) as compared to the other two doses, while R4 group was kept as control. Results of the challenge protection test also validated the efficacy of the double dose of the HB1 live vaccine, which gave the highest survival percentage. Results of this study lay the foundation for a potential cross-reactive live oral Salmonella vaccine that has proved to be immunogenic in rabbits. In this research, Salmonella species were isolated from the animal, insect and human enteric sources in Faisalabad, Punjab, Pakistan. These species were characterized by different microbiological and molecular techniques including polymerase chain reaction (PCR) by amplification of the 16S rRNA gene. Furthermore, sequencing of the amplicons confirmed all ten isolates as Salmonella strains. The antigenic cross-reactivity was found maximum between the HB1 (strain isolated from honeybee) antiserum and its antigen with an antibody titer of 1:128, while the HB1 antiserum showed a cross-reactive titer range of 1:8 to 1:64. On the basis of the highest geometric mean titer (GMT) shown by the antiserum of the HB1 antigen, it was selected as the best candidate for a cross-reactive live Salmonella oral antigen. Moreover, the HB1 antigen was used a live oral antigen (1 × 1010 CFU/ml) in a safety test in rabbits and proved to be avirulent. During the animal trial, three different oral doses of the HB1 live oral antigen were evaluated in four different rabbits’ groups (R1, R2, R3, and R4). The dose number 2 of 0.5 ml (two drops orally and repeated after one week) gave the best GMT measured by indirect hemagglutination (IHA) as compared to the other two doses, while R4 group was kept as control. Results of the challenge protection test also validated the efficacy of the double dose of the HB1 live vaccine, which gave the highest survival percentage. Results of this study lay the foundation for a potential cross-reactive live oral Salmonella vaccine that has proved to be immunogenic in rabbits.
Collapse
Affiliation(s)
- Hassan Zafar
- Institute of Microbiology, University of Agriculture , Faisalabad, Punjab , Pakistan
| | - Sajjad Ur Rahman
- Institute of Microbiology, University of Agriculture , Faisalabad, Punjab , Pakistan
| | - Sultan Ali
- Institute of Microbiology, University of Agriculture , Faisalabad, Punjab , Pakistan
| | - Muhammad Tariq Javed
- Department of Veterinary Pathology, University of Agriculture , Faisalabad, Punjab , Pakistan
| |
Collapse
|
28
|
Zafar H, Rahman SU, Ali S, Javed MT. Evaluation of a Salmonella Strain Isolated from Honeybee Gut as a Potential Live Oral Vaccine Against Lethal Infection of Salmonella Typhimurium. Pol J Microbiol 2019. [PMID: 31257790 PMCID: PMC7260634 DOI: 10.33073/pjm-2019-017] [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] [Indexed: 11/06/2022] Open
Abstract
In this research, Salmonella species were isolated from the animal, insect and human enteric sources in Faisalabad, Punjab, Pakistan. These species were characterized by different microbiological and molecular techniques including polymerase chain reaction (PCR) by amplification of the 16S rRNA gene. Furthermore, sequencing of the amplicons confirmed all ten isolates as Salmonella strains. The antigenic cross-reactivity was found maximum between the HB1 (strain isolated from honeybee) antiserum and its antigen with an antibody titer of 1:128, while the HB1 antiserum showed a cross-reactive titer range of 1:8 to 1:64. On the basis of the highest geometric mean titer (GMT) shown by the antiserum of the HB1 antigen, it was selected as the best candidate for a cross-reactive live Salmonella oral antigen. Moreover, the HB1 antigen was used a live oral antigen (1 × 1010 CFU/ml) in a safety test in rabbits and proved to be avirulent. During the animal trial, three different oral doses of the HB1 live oral antigen were evaluated in four different rabbits’ groups (R1, R2, R3, and R4). The dose number 2 of 0.5 ml (two drops orally and repeated after one week) gave the best GMT measured by indirect hemagglutination (IHA) as compared to the other two doses, while R4 group was kept as control. Results of the challenge protection test also validated the efficacy of the double dose of the HB1 live vaccine, which gave the highest survival percentage. Results of this study lay the foundation for a potential cross-reactive live oral Salmonella vaccine that has proved to be immunogenic in rabbits.
Collapse
Affiliation(s)
- Hassan Zafar
- Institute of Microbiology, University of Agriculture , Faisalabad, Punjab , Pakistan
| | - Sajjad Ur Rahman
- Institute of Microbiology, University of Agriculture , Faisalabad, Punjab , Pakistan
| | - Sultan Ali
- Institute of Microbiology, University of Agriculture , Faisalabad, Punjab , Pakistan
| | - Muhammad Tariq Javed
- Department of Veterinary Pathology, University of Agriculture , Faisalabad, Punjab , Pakistan
| |
Collapse
|
29
|
Feodorova VA, Lyapina AM, Zaitsev SS, Khizhnyakova MA, Sayapina LV, Ulianova OV, Ulyanov SS, Motin VL. New Promising Targets for Synthetic Omptin-Based Peptide Vaccine against Gram-Negative Pathogens. Vaccines (Basel) 2019; 7:vaccines7020036. [PMID: 30974891 PMCID: PMC6630670 DOI: 10.3390/vaccines7020036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 03/24/2019] [Accepted: 04/04/2019] [Indexed: 12/18/2022] Open
Abstract
Omptins represent a family of proteases commonly found in various Gram-negative pathogens. These proteins play an important role in host-pathogen interaction and have been recognized as key virulence factors, highlighting the possibility of developing an omptin-based broad-spectrum vaccine. The prototypical omptin, His-tagged recombinant Pla, was used as a model target antigen. In total, 46 linear and 24 conformational epitopes for the omptin family were predicted by the use of ElliPro service. Among these we selected highly conserved, antigenic, non-allergenic, and immunogenic B-cell epitopes. Five epitopes (2, 6, 8, 10, and 11 corresponding to Pla regions 52-60, 146-150, 231-234, 286-295, and 306-311, respectively) could be the first choice for the development of the new generation of target-peptide-based vaccine against plague. The partial residues of omptin epitopes 6, 8, and 10 (regions 136-145, 227-230, and 274-285) could be promising targets for the multi-pathogen vaccine against a group of enterobacterial infections. The comparative analysis and 3D modeling of amino acid sequences of several omptin family proteases, such as Pla (Yersinia pestis), PgtE (Salmonella enterica), SopA (Shigella flexneri), OmpT, and OmpP (Escherichia coli), confirmed their high cross-homology with respect to the identified epitope clusters and possible involvement of individual epitopes in host-pathogen interaction.
Collapse
Affiliation(s)
- Valentina A Feodorova
- Laboratory for Molecular Biology and NanoBiotechnology, Federal Research Center for Virology and Microbiology, Branch in Saratov, 410028 Saratov, Russia.
| | - Anna M Lyapina
- Laboratory for Molecular Biology and NanoBiotechnology, Federal Research Center for Virology and Microbiology, Branch in Saratov, 410028 Saratov, Russia.
| | - Sergey S Zaitsev
- Laboratory for Molecular Biology and NanoBiotechnology, Federal Research Center for Virology and Microbiology, Branch in Saratov, 410028 Saratov, Russia.
| | - Maria A Khizhnyakova
- Laboratory for Molecular Biology and NanoBiotechnology, Federal Research Center for Virology and Microbiology, Branch in Saratov, 410028 Saratov, Russia.
| | - Lidiya V Sayapina
- Department of Vaccine Control, Scientific Center on Expertise of Medical Application Products, 127051, Moscow, Russia.
| | - Onega V Ulianova
- Laboratory for Molecular Biology and NanoBiotechnology, Federal Research Center for Virology and Microbiology, Branch in Saratov, 410028 Saratov, Russia.
| | - Sergey S Ulyanov
- Department for Medical Optics, Saratov State University, 410012, Saratov, Russia.
| | - Vladimir L Motin
- Department of Pathology, Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
30
|
Muruganandah V, Sathkumara HD, Navarro S, Kupz A. A Systematic Review: The Role of Resident Memory T Cells in Infectious Diseases and Their Relevance for Vaccine Development. Front Immunol 2018; 9:1574. [PMID: 30038624 PMCID: PMC6046459 DOI: 10.3389/fimmu.2018.01574] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 06/25/2018] [Indexed: 12/12/2022] Open
Abstract
Background Resident memory T cells have emerged as key players in the immune response generated against a number of pathogens. Their ability to take residence in non-lymphoid peripheral tissues allows for the rapid deployment of secondary effector responses at the site of pathogen entry. This ability to provide enhanced regional immunity has gathered much attention, with the generation of resident memory T cells being the goal of many novel vaccines. Objectives This review aimed to systematically analyze published literature investigating the role of resident memory T cells in human infectious diseases. Known effector responses mounted by these cells are summarized and key strategies that are potentially influential in the rational design of resident memory T cell inducing vaccines have also been highlighted. Methods A Boolean search was applied to Medline, SCOPUS, and Web of Science. Studies that investigated the effector response generated by resident memory T cells and/or evaluated strategies for inducing these cells were included irrespective of published date. Studies must have utilized an established technique for identifying resident memory T cells such as T cell phenotyping. Results While over 600 publications were revealed by the search, 147 articles were eligible for inclusion. The reference lists of included articles were also screened for other eligible publications. This resulted in the inclusion of publications that studied resident memory T cells in the context of over 25 human pathogens. The vast majority of studies were conducted in mouse models and demonstrated that resident memory T cells mount protective immune responses. Conclusion Although the role resident memory T cells play in providing immunity varies depending on the pathogen and anatomical location they resided in, the evidence overall suggests that these cells are vital for the timely and optimal protection against a number of infectious diseases. The induction of resident memory T cells should be further investigated and seriously considered when designing new vaccines.
Collapse
Affiliation(s)
- Visai Muruganandah
- Centre for Biosecurity and Tropical Infectious Diseases, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Harindra D Sathkumara
- Centre for Biosecurity and Tropical Infectious Diseases, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Severine Navarro
- Centre for Biosecurity and Tropical Infectious Diseases, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia.,QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Andreas Kupz
- Centre for Biosecurity and Tropical Infectious Diseases, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
31
|
McWhorter AR, Chousalkar KK. A Long-Term Efficacy Trial of a Live, Attenuated Salmonella Typhimurium Vaccine in Layer Hens. Front Microbiol 2018; 9:1380. [PMID: 29997596 PMCID: PMC6028619 DOI: 10.3389/fmicb.2018.01380] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 06/06/2018] [Indexed: 01/13/2023] Open
Abstract
Salmonella remains one of the most common causes of bacterial foodborne gastrointestinal disease in humans. Raw eggs or food items containing undercooked eggs are frequently identified as the source of Salmonella. Salmonella Typhimurium contamination of table eggs most commonly occurs when they are laid in a contaminated environment. Several control strategies, including vaccination, are widely used to mitigate the total Salmonella load. It is unclear, however, whether live attenuated Salmonella vaccines are efficacious over the life span of a layer hen. Live attenuated Salmonella vaccines have been favored due to their ability to illicit a strong humoral immune response. The lifespan of a layer hen ranges between 60 and 80 weeks and the long term efficacy of attenuated vaccine strains has not been investigated. In this study, commercial brown layer chicks were vaccinated at day old, 6 weeks of age, and again at 10 weeks of age with the Bioproperties VaxsafeTM STM1 aroA mutant vaccine. Birds were challenged at 18 weeks of age with Salmonella Typhimurium DT9 (MLVA 03 15 08 11 550). Feces and eggs were monitored for S. Typhimurium for 40 weeks post-infection. Birds produced a strong immune response following the final dose which was administered intramuscularly. The serum antibody response to S. Typhimurium DT9 infection did not differ between challenged groups. Fecal shedding and egg contamination was highly variable and did not differ significantly between vaccinated and unvaccinated birds that had been challenged with S. Typhimurium DT9. Total bacterial load in feces was quantified using qPCR. No significant difference was detected between unvaccinated and vaccinated birds after challenge.
Collapse
Affiliation(s)
- Andrea R McWhorter
- School of Animal and Veterinary Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Kapil K Chousalkar
- School of Animal and Veterinary Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
32
|
Abstract
The number of human salmonellosis within the European Union tended to increase since 2013. One of the reasons might be Salmonella Enteritidis rising in laying hens flocks by around 17% in 2015 vs 2014 and by 57% in 2016 vs 2015. The most important sources of food-borne Salmonella outbreaks are still eggs and egg products as well as ready-to-eat foods having a long shelf life. Specific actions are suggested to restart decreasing the number of human salmonellosis: (1) revision of sampling schemes to solve pathogen under detection in both animals and foods; (2) integration of microbiological criteria with fit for purpose performance objectives and food safety objectives; and (3) improvement of epidemiological investigations of human, food, and animal isolates by using whole-genome sequencing in order to effectively track salmonellosis and verify which prevention measures are most effective.
Collapse
|
33
|
Amarachintha S, Harmel-Laws E, Steinbrecher KA. Guanylate cyclase C reduces invasion of intestinal epithelial cells by bacterial pathogens. Sci Rep 2018; 8:1521. [PMID: 29367634 PMCID: PMC5784150 DOI: 10.1038/s41598-018-19868-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 12/20/2017] [Indexed: 12/24/2022] Open
Abstract
The guanylate cyclase C (GC-C) receptor regulates electrolyte and water secretion into the gut following activation by the E. coli enterotoxin STa, or by weaker endogenous agonists guanylin and uroguanylin. Our previous work has demonstrated that GC-C plays an important role in controlling initial infection as well as carrying load of non-invasive bacterial pathogens in the gut. Here, we use Salmonella enterica serovar Typhimurium to determine whether GC-C signaling is important in host defense against pathogens that actively invade enterocytes. In vitro studies indicated that GC-C signaling significantly reduces Salmonella invasion into Caco2-BBE monolayers. Relative to controls, GC-C knockout mice develop severe systemic illness following oral Salmonella infection, characterized by disrupted intestinal mucus layer, elevated cytokines and organ CFUs, and reduced animal survival. In Salmonella-infected wildtype mice, oral gavage of GC-C agonist peptide reduced host/pathogen physical interaction and diminished bacterial translocation to mesenteric lymph nodes. These studies suggest that early life susceptibility to STa-secreting enterotoxigenic E. coli may be counter-balanced by a critical role of GC-C in protecting the mucosa from non-STa producing, invasive bacterial pathogens.
Collapse
Affiliation(s)
- Surya Amarachintha
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Eleana Harmel-Laws
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Kris A Steinbrecher
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA. .,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45229, USA.
| |
Collapse
|