1
|
Archer EW, Chisnall T, Tano-Debrah K, Card RM, Duodu S, Kunadu APH. Prevalence and genomic characterization of Salmonella isolates from commercial chicken eggs retailed in traditional markets in Ghana. Front Microbiol 2023; 14:1283835. [PMID: 38029182 PMCID: PMC10646427 DOI: 10.3389/fmicb.2023.1283835] [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: 08/27/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
Salmonella enterica are important foodborne bacterial pathogens globally associated with poultry. Exposure to Salmonella-contaminated eggs and egg-related products is a major risk for human salmonellosis. Presently, there is a huge data gap regarding the prevalence and circulating serovars of Salmonella in chicken eggs sold in Ghana. In this study, 2,304 eggs (pools of six per sample unit) collected from informal markets in Accra, Kumasi and Tamale, representing the three ecological belts across Ghana, were tested for Salmonella. Antimicrobial susceptibility testing and Whole Genome Sequencing (WGS) of the isolates were performed using standard microdilution protocols and the Illumina NextSeq platform, respectively. The total prevalence of Salmonella was 5.5% with a higher rate of contamination in eggshell (4.9%) over egg content (1.8%). The serovars identified were S. Ajiobo (n = 1), S. Chester (n = 6), S. Hader (n = 7), S. enteritidis (n = 2); and S. I 4:b:- (n = 8). WGS analysis revealed varied sequence types (STs) that were serovar specific. The S. I 4:b:- isolates had a novel ST (ST8938), suggesting a local origin. The two S. enteritidis isolates belonged to ST11 and were identified with an invasive lineage of a global epidemic clade. All isolates were susceptible to ampicillin, azithromycin, cefotaxime, ceftazidime, gentamicin, meropenem, and tigecycline. The phenotypic resistance profiles to seven antimicrobials: chloramphenicol (13%), ciprofloxacin (94%), and nalidixic acid (94%), colistin (13%), trimethoprim (50%) sulfamethoxazole (50%) and tetracycline (50%) corresponded with the presence of antimicrobial resistance (AMR) determinants including quinolones (gyrA (D87N), qnrB81), aminoglycosides (aadA1), (aph(3")-Ib aph(6)-Id), tetracyclines (tet(A)), phenicols (catA1), trimethoprim (dfrA14 and dfrA1). The S. enteritidis and S. Chester isolates were multidrug resistant (MDR). Several virulence factors were identified, notably cytolethal distending toxin (cdtB gene), rck, pef and spv that may promote host invasion and disease progression in humans. The findings from this study indicate the presence of multidrug resistant and virulent strains of Salmonella serovars in Ghanaian chicken eggs, with the potential to cause human infections. This is a critical baseline information that could be used for Salmonella risk assessment in the egg food chain to mitigate potential future outbreaks.
Collapse
Affiliation(s)
- Edward W. Archer
- Nutrition and Food Science Department, University of Ghana, Accra, Ghana
- Food and Drug Authority, Food Safety Management Department, Accra, Ghana
| | - Tom Chisnall
- Animal and Plant Health Agency, Addlestone, United Kingdom
| | - Kwaku Tano-Debrah
- Nutrition and Food Science Department, University of Ghana, Accra, Ghana
| | | | - Samuel Duodu
- Biochemistry Cell and Molecular Biology Department, University of Ghana, Accra, Ghana
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
| | | |
Collapse
|
2
|
Siggins MK, MacLennan CA. An adsorption method to prepare specific antibody-depleted normal human serum as a source of complement for human serum bactericidal assays for Salmonella. Vaccine 2021; 39:7503-7509. [PMID: 34794820 DOI: 10.1016/j.vaccine.2021.10.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 10/19/2022]
Abstract
Serum bactericidal assays (SBA) are valuable for assessing the functional activity of natural and vaccine-induced antibodies against many Gram-negative bacteria, such as meningococcus and Salmonella. However, SBA often require an exogenous source of complement and the presence of pre-existing naturally acquired antibodies limits the use of human complement for this purpose. To remove pre-existing Salmonella-specific antibodies, in the context of SBA for Salmonella vaccine research, we incubated human sera with preparations of Salmonella. By incubating at 4 °C, pre-existing antibodies were adsorbed onto the Salmonella bacteria with only minimal complement deposition. We assessed the effects of adsorption on specific antibody levels, complement activity and the bactericidal activity of sera using flow cytometry, SBA and haemolytic assays. Adsorption removed Salmonella-specific antibodies and bactericidal activity against Salmonella from whole serum but was not detrimental to serum complement activity, even after five adsorption cycles. Bactericidal activity could be reconstituted in the adsorbed serum by the addition of exogenous specific antibodies. Sera preadsorbed with Salmonella are suitable as a source of human complement to measure the bactericidal activity of Salmonella antibodies. The adsorption method can be used to deplete, simply and rapidly, specific antibodies from serum to prepare a source of human complement for use in SBA for vaccine research and assessment.
Collapse
Affiliation(s)
- Matthew K Siggins
- The Institute of Immunology and Immunotherapy, University of Birmingham, B15 2TT, United Kingdom; National Heart and Lung Institute, St Mary's Hospital, Faculty of Medicine, Imperial College London, W2 1PG, United Kingdom.
| | - Calman A MacLennan
- The Institute of Immunology and Immunotherapy, University of Birmingham, B15 2TT, United Kingdom; The Jenner Institute, Nuffield Department of Medicine, University of Oxford, OX3 7DQ, United Kingdom; Bill & Melinda Gates Foundation, 62 Buckingham Gate, London SW1E 6AJ, United Kingdom.
| |
Collapse
|
3
|
Di Benedetto R, Alfini R, Carducci M, Aruta MG, Lanzilao L, Acquaviva A, Palmieri E, Giannelli C, Necchi F, Saul A, Micoli F. Novel Simple Conjugation Chemistries for Decoration of GMMA with Heterologous Antigens. Int J Mol Sci 2021; 22:10180. [PMID: 34638530 PMCID: PMC8508390 DOI: 10.3390/ijms221910180] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/17/2021] [Indexed: 11/30/2022] Open
Abstract
Outer Membrane Vesicles (OMV) constitute a promising platform for the development of efficient vaccines. OMV can be decorated with heterologous antigens (proteins or polysaccharides), becoming attractive novel carriers for the development of multicomponent vaccines. Chemical conjugation represents a tool for linking antigens, also from phylogenetically distant pathogens, to OMV. Here we develop two simple and widely applicable conjugation chemistries targeting proteins or lipopolysaccharides on the surface of Generalized Modules for Membrane Antigens (GMMA), OMV spontaneously released from Gram-negative bacteria mutated to increase vesicle yield and reduce potential reactogenicity. A Design of Experiment approach was used to identify optimal conditions for GMMA activation before conjugation, resulting in consistent processes and ensuring conjugation efficiency. Conjugates produced by both chemistries induced strong humoral response against the heterologous antigen and GMMA. Additionally, the use of the two orthogonal chemistries allowed to control the linkage of two different antigens on the same GMMA particle. This work supports the further advancement of this novel platform with great potential for the design of effective vaccines.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena, Italy; (R.D.B.); (R.A.); (M.C.); (M.G.A.); (L.L.); (A.A.); (E.P.); (C.G.); (F.N.); (A.S.)
| |
Collapse
|
4
|
Appiah GD, Mpimbaza A, Lamorde M, Freeman M, Kajumbula H, Salah Z, Kugeler K, Mikoleit M, White PB, Kapisi J, Borchert J, Sserwanga A, Van Dyne S, Mead P, Kim S, Lauer AC, Winstead A, Manabe YC, Flick RJ, Mintz E. Salmonella Bloodstream Infections in Hospitalized Children with Acute Febrile Illness-Uganda, 2016-2019. Am J Trop Med Hyg 2021; 105:37-46. [PMID: 33999850 DOI: 10.4269/ajtmh.20-1453] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/19/2021] [Indexed: 12/23/2022] Open
Abstract
Invasive Salmonella infection is a common cause of acute febrile illness (AFI) among children in sub-Saharan Africa; however, diagnosing Salmonella bacteremia is challenging in settings without blood culture. The Uganda AFI surveillance system includes blood culture-based surveillance for etiologies of bloodstream infection (BSIs) in hospitalized febrile children in Uganda. We analyzed demographic, clinical, blood culture, and antimicrobial resistance data from hospitalized children at six sentinel AFI sites from July 2016 to January 2019. A total of 47,261 children were hospitalized. Median age was 2 years (interquartile range, 1-4) and 26,695 (57%) were male. Of 7,203 blood cultures, 242 (3%) yielded bacterial pathogens including Salmonella (N = 67, 28%), Staphylococcus aureus (N = 40, 17%), Escherichia spp. (N = 25, 10%), Enterococcus spp. (N = 18, 7%), and Klebsiella pneumoniae (N = 17, 7%). Children with BSIs had longer median length of hospitalization (5 days versus 4 days), and a higher case-fatality ratio (13% versus 2%) than children without BSI (all P < 0.001). Children with Salmonella BSIs did not differ significantly in length of hospitalization or mortality from children with BSI resulting from other organisms. Serotype and antimicrobial susceptibility results were available for 49 Salmonella isolates, including 35 (71%) non-typhoidal serotypes and 14 Salmonella serotype Typhi (Typhi). Among Typhi isolates, 10 (71%) were multi-drug resistant and 13 (93%) had decreased ciprofloxacin susceptibility. Salmonella strains, particularly non-typhoidal serotypes and drug-resistant Typhi, were the most common cause of BSI. These data can inform regional Salmonella surveillance in East Africa and guide empiric therapy and prevention in Uganda.
Collapse
Affiliation(s)
- Grace D Appiah
- 1Division of Foodborne, Waterborne, and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Arthur Mpimbaza
- 2Infectious Disease Research Collaboration, Kampala, Uganda.,3Child Health and Development Center, Makerere University, Kampala, Uganda
| | | | - Molly Freeman
- 1Division of Foodborne, Waterborne, and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Henry Kajumbula
- 5Department of Microbiology, Makerere University, Kampala, Uganda
| | - Zainab Salah
- 1Division of Foodborne, Waterborne, and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Kiersten Kugeler
- 6Division of Vector-Borne Disease, Centers for Disease Control and Prevention, Fort Collins, Colorado
| | - Matthew Mikoleit
- 7Division of Global Health Protection, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Porscha Bumpus White
- 1Division of Foodborne, Waterborne, and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - James Kapisi
- 2Infectious Disease Research Collaboration, Kampala, Uganda
| | - Jeff Borchert
- 6Division of Vector-Borne Disease, Centers for Disease Control and Prevention, Fort Collins, Colorado
| | | | - Susan Van Dyne
- 1Division of Foodborne, Waterborne, and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Paul Mead
- 6Division of Vector-Borne Disease, Centers for Disease Control and Prevention, Fort Collins, Colorado
| | - Sunkyung Kim
- 1Division of Foodborne, Waterborne, and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Ana C Lauer
- 1Division of Foodborne, Waterborne, and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Alison Winstead
- 8Division of Parasitic Disease and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Yukari C Manabe
- 9Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert J Flick
- 9Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Eric Mintz
- 1Division of Foodborne, Waterborne, and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
5
|
Fiorino F, Pettini E, Koeberling O, Ciabattini A, Pozzi G, Martin LB, Medaglini D. Long-Term Anti-Bacterial Immunity against Systemic Infection by Salmonella enterica Serovar Typhimurium Elicited by a GMMA-Based Vaccine. Vaccines (Basel) 2021; 9:495. [PMID: 34065899 PMCID: PMC8150838 DOI: 10.3390/vaccines9050495] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/27/2022] Open
Abstract
Salmonella Typhimurium (STm) represents the most prevalent cause of invasive non-typhoidal Salmonella (iNTS) disease, and currently no licensed vaccine is available. In this work we characterized the long-term anti-bacterial immunity elicited by a STm vaccine based on Generalized Modules of Membrane Antigens (GMMA) delivering O:4,5 antigen, using a murine model of systemic infection. Subcutaneous immunization of mice with STmGMMA/Alhydrogel elicited rapid, high, and persistent antigen-specific serum IgG and IgM responses. The serum was bactericidal in vitro. O:4,5-specific IgG were also detected in fecal samples after immunization and positively correlated with IgG observed in intestinal washes. Long-lived plasma cells and O:4,5-specific memory B cells were detected in spleen and bone marrow. After systemic STm challenge, a significant reduction of bacterial load in blood, spleen, and liver, as well as a reduction of circulating neutrophils and G-CSF glycoprotein was observed in STmGMMA/Alhydrogel immunized mice compared to untreated animals. Taken together, these data support the development of a GMMA-based vaccine for prevention of iNTS disease.
Collapse
Affiliation(s)
- Fabio Fiorino
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (F.F.); (E.P.); (A.C.); (G.P.)
| | - Elena Pettini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (F.F.); (E.P.); (A.C.); (G.P.)
| | - Oliver Koeberling
- GSK Vaccines Institute for Global Health S.r.l., 53100 Siena, Italy;
| | - Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (F.F.); (E.P.); (A.C.); (G.P.)
| | - Gianni Pozzi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (F.F.); (E.P.); (A.C.); (G.P.)
| | - Laura B. Martin
- GSK Vaccines Institute for Global Health S.r.l., 53100 Siena, Italy;
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (F.F.); (E.P.); (A.C.); (G.P.)
| |
Collapse
|
6
|
Abstract
Enteric viral and bacterial infections continue to be a leading cause of mortality and morbidity in young children in low-income and middle-income countries, the elderly, and immunocompromised individuals. Vaccines are considered an effective and practical preventive approach against the predominantly fecal-to-oral transmitted gastroenteritis particularly in the resource-limited countries or regions where implementation of sanitation systems and supply of safe drinking water are not quickly achievable. While vaccines are available for a few enteric pathogens including rotavirus and cholera, there are no vaccines licensed for many other enteric viral and bacterial pathogens. Challenges in enteric vaccine development include immunological heterogeneity among pathogen strains or isolates, a lack of animal challenge models to evaluate vaccine candidacy, undefined host immune correlates to protection, and a low protective efficacy among young children in endemic regions. In this article, we briefly updated the progress and challenges in vaccines and vaccine development for the leading enteric viral and bacterial pathogens including rotavirus, human calicivirus, Shigella, enterotoxigenic Escherichia coli (ETEC), cholera, nontyphoidal Salmonella, and Campylobacter, and introduced a novel epitope- and structure-based vaccinology platform known as MEFA (multiepitope fusion antigen) and the application of MEFA for developing broadly protective multivalent vaccines against heterogenous pathogens.
Collapse
Affiliation(s)
- Hyesuk Seo
- University of Illinois at Urbana-Champaign, Department of Pathobiology, Urbana, Illinois, USA
| | - Qiangde Duan
- University of Yangzhou, Institute of Comparative Medicine, Yangzhou, PR China
| | - Weiping Zhang
- University of Illinois at Urbana-Champaign, Department of Pathobiology, Urbana, Illinois, USA,CONTACT Weiping Zhang, University of Illinois at Urbana-Champaign, Department of Pathobiology, Urbana, Illinois, USA
| |
Collapse
|
7
|
Micoli F, Alfini R, Di Benedetto R, Necchi F, Schiavo F, Mancini F, Carducci M, Palmieri E, Balocchi C, Gasperini G, Brunelli B, Costantino P, Adamo R, Piccioli D, Saul A. GMMA Is a Versatile Platform to Design Effective Multivalent Combination Vaccines. Vaccines (Basel) 2020; 8:E540. [PMID: 32957610 PMCID: PMC7564227 DOI: 10.3390/vaccines8030540] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/12/2020] [Accepted: 09/14/2020] [Indexed: 01/21/2023] Open
Abstract
Technology platforms are an important strategy to facilitate the design, development and implementation of vaccines to combat high-burden diseases that are still a threat for human populations, especially in low- and middle-income countries, and to address the increasing number and global distribution of pathogens resistant to antimicrobial drugs. Generalized Modules for Membrane Antigens (GMMA), outer membrane vesicles derived from engineered Gram-negative bacteria, represent an attractive technology to design affordable vaccines. Here, we show that GMMA, decorated with heterologous polysaccharide or protein antigens, leads to a strong and effective antigen-specific humoral immune response in mice. Importantly, GMMA promote enhanced immunogenicity compared to traditional formulations (e.g., recombinant proteins and glycoconjugate vaccines), without negative impact to the anti-GMMA immune response. Our findings support the use of GMMA as a "plug and play" technology for the development of effective combination vaccines targeting different bugs at the same time.
Collapse
Affiliation(s)
- Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | - Renzo Alfini
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | - Roberta Di Benedetto
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | - Francesca Necchi
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | - Fabiola Schiavo
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | - Francesca Mancini
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | - Martina Carducci
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | - Elena Palmieri
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | | | - Gianmarco Gasperini
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | | | | | - Roberto Adamo
- GSK, 53100 Siena, Italy; (C.B.); (B.B.); (P.C.); (R.A.); (D.P.)
| | - Diego Piccioli
- GSK, 53100 Siena, Italy; (C.B.); (B.B.); (P.C.); (R.A.); (D.P.)
| | - Allan Saul
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| |
Collapse
|
8
|
Ahmed A, Akhade AS, Qadri A. Accessibility of O Antigens Shared between Salmonella Serovars Determines Antibody-Mediated Cross-Protection. THE JOURNAL OF IMMUNOLOGY 2020; 205:438-446. [PMID: 32540995 DOI: 10.4049/jimmunol.1900624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 05/05/2020] [Indexed: 11/19/2022]
Abstract
Pathogenic Salmonella serovars produce clinical manifestations ranging from systemic infection typhoid to invasive nontyphoidal Salmonella disease in humans. These serovars share a high degree of homology at the genome and the proteome level. However, whether infection or immunization with one serovar provides protection against other serovars has not been well studied. We show in this study that immunization of mice with live typhoidal serovar, Salmonella Typhi, generates cross-reactive immune responses, which provide far greater resistance against challenge with nontyphoidal serovar Salmonella Enteritidis than with another nontyphoidal serovar, Salmonella Typhimurium. Splenic T cells from these immunized mice produced similar levels of IL-2 and IFN-γ upon ex vivo stimulation with Ags prepared from S Enteritidis and S Typhimurium. In contrast, Abs against S Typhi interacted with live intact S Enteritidis but did not bind intact S Typhimurium. These pathogen-reactive Abs were largely directed against oligosaccharide (O)-antigenic determinant of LPS that S Typhi shares with S Enteritidis. Abs against the O determinant, which S Typhi shares with S Typhimurium, were present in the sera of immunized mice but did not bind live intact Salmonella because of surface inaccessibility of this determinant. Similar accessibility-regulated interaction was seen with Abs generated against S Typhimurium and S Enteritidis. Our results suggest that the ability of protective Abs elicited with one Salmonella serovar to engage with and consequently provide protection against another Salmonella serovar is determined by the accessibility of shared O Ags. These findings have significant and broader implications for immunity and vaccine development against pathogenic Salmonellae.
Collapse
Affiliation(s)
- Anees Ahmed
- Hybridoma Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Ajay Suresh Akhade
- Hybridoma Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Ayub Qadri
- Hybridoma Laboratory, National Institute of Immunology, New Delhi 110067, India
| |
Collapse
|
9
|
Albert MJ, Bulach D, Alfouzan W, Izumiya H, Carter G, Alobaid K, Alatar F, Sheikh AR, Poirel L. Non-typhoidal Salmonella blood stream infection in Kuwait: Clinical and microbiological characteristics. PLoS Negl Trop Dis 2019; 13:e0007293. [PMID: 30986214 PMCID: PMC6483562 DOI: 10.1371/journal.pntd.0007293] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/25/2019] [Accepted: 03/12/2019] [Indexed: 12/12/2022] Open
Abstract
Non-typhoidal Salmonella (NTS) bacteremia is a significant cause of morbidity and mortality worldwide. It is considered to be an emerging and neglected tropical disease in Africa. We studied this in two tertiary hospitals–Al Farwaniya and Al Amiri–in Kuwait, a subtropical country, from April 2013-May 2016. NTS bacteremia was present in 30 of 53,860 (0.75%) and 31 of 290,36 (1.33%) blood cultures in the two hospitals respectively. In Al Farwaniya hospital, one-third of the patients were from some tropical developing countries of Asia. About 66% of all patients (40/61) had diarrhea, and of these, 65% had the corresponding blood serovar isolated from stool culture. A few patients had Salmonella cultured from urine. Patients were either young or old. Most of the patients had co-morbidities affecting the immune system. Two patients each died in both hospitals. The number of different serovars cultured in each hospital was 13, and most infections were due to S. Enteritidis (all sequence type [ST]) 11) and S. Typhimurium (all ST19) except in a subgroup of expatriate patients from tropical developing countries in Al Farwaniya hospital. About a quarter of the isolates were multidrug-resistant. Most patients were treated with a cephalosporin with or without other antibiotics. S. Enteritidis and S. Typhimurium isolates were typed by pulsed field-gel electrophoresis (PFGE) and a selected number of isolates were whole-genome sequenced. Up to four different clades were present by PFGE in either species. Whole-genome sequenced isolates showed antibiotic-resistance genes that showed phenotypic correlation, and in some cases, phenotypes showed absence of specific genes. Whole-genome sequenced isolates showed presence of genes that contributed to blood-stream infection. Phylogeny by core genome analysis showed a close relationship with S. Typhimurium and S. Enteritidis from other parts of the world. The uniqueness of our study included the finding of a low prevalence of infection, mortality and multidrug-resistance, a relatively high prevalence of gastrointestinal infection in patients, and the characterization of selected isolates of S. Typhimurium and S. Enteritidis serovars by whole-genome sequencing that shed light on phylogeny, virulence and resistance. Similarities with studies from developing countries especially Africa included infection in patients with co-morbidities affecting the immune system, predominance of S. Typhimurium and S. Enteritidis serovars and presence of drug-resistance in isolates. Salmonella organisms are classified into typhoidal Salmonella (causing enteric fever) and non-typhoidal Salmonella (NTS) (causing infections other than enteric fever). Apart from causing other infections, NTS causes blood-stream infection (bacteremia and septicemia). NTS blood stream infection (NTS-BI) is considered to be an emerging and neglected tropical disease in Africa. It causes a very high morbidity and mortality in Africa. The individuals affected in Africa are children, malnourished people, patients with malaria or HIV etc. These conditions affect the immune system and make them vulnerable to infection with NTS. In these patients, diarrheal disease due to NTS is rare. The majority of infections are due to two types of NTS: Typhimurium and Enteritidis. There is a very high prevalence of multidrug-resistance in NTS making the infection difficult to treat. NTS-BI is also present in other parts of the world including developed countries albeit at a lower prevalence. Kuwait is a high-income, subtropical country in transition (from a developing to developed country), located in the Middle East. We studied NTS-BI in Al Farwaniya and Al Amiri hospitals in Kuwait during April 2013 to May 2016. Out of nearly 30,000 to more than 50,000 blood cultures done in these hospitals, NTS was present in 0. 75 to 1.33% of blood cultures, representing a very small proportion of blood cultures, unlike in Africa. This showed that 31 patients in Al Farwaniya hospital and 30 patients in Al Amari hospital had NTS-BI. Most of these patients had underlying illnesses such as diabetes, lung infection, cancer etc. that affect the immune system, as in Africa. Many patients also had diarrheal disease caused by the same NTS that caused blood stream infection, unlike in Africa. Only two patients in each hospital died, a low mortality, unlike in Africa. The majority of the isolates belonged to Typhimurium and Enteritidis as in Africa. Even though resistance to drugs was a problem, about quarter of the isolates only were multidrug-resistant, a lower prevalence compared to in Africa. In Kuwait, we performed a detailed genetic study of a selected number of Typhimurium and Enteritidis isolates by a modern technique called whole genome sequencing. This revealed genetic determinants encoding drug-resistance and virulence causing blood-stream infection. This type of study was not performed in African isolates. Thus, our study revealed similarities and differences with studies of NTS-BI in Africa.
Collapse
Affiliation(s)
- M. John Albert
- Department of Microbiology, Faculty of Medicine, Kuwait University, Jabriya, Kuwait
- * E-mail:
| | - Dieter Bulach
- Microbiological Diagnostic Unit, Public Health Laboratory, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Victoria, Australia
| | - Wadha Alfouzan
- Department of Microbiology, Faculty of Medicine, Kuwait University, Jabriya, Kuwait
- Microbiology Unit, Department of Laboratories, Al Farwaniya Hospital, Sabah Al-Nasser, Kuwait
| | | | - Glen Carter
- Microbiological Diagnostic Unit, Public Health Laboratory, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Victoria, Australia
| | - Khaled Alobaid
- Department of Microbiology, Al Amiri Hospital, Sharq, Kuwait
| | - Fatemah Alatar
- Microbiology Unit, Muabarak Al Kabeer Hospital, Jabriya, Kuwait, and
| | | | - Laurent Poirel
- Department of Medicine, University of Fribourg, Switzerland
| |
Collapse
|
10
|
Mooney JP, Galloway LJ, Riley EM. Malaria, anemia, and invasive bacterial disease: A neutrophil problem? J Leukoc Biol 2018; 105:645-655. [PMID: 30570786 PMCID: PMC6487965 DOI: 10.1002/jlb.3ri1018-400r] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/29/2018] [Accepted: 12/01/2018] [Indexed: 12/16/2022] Open
Abstract
Invasive bacterial disease is well described in immunocompromised hosts, including those with malaria infection. One bacterial infection frequently observed in children with Plasmodium falciparum infection is nontyphoidal salmonella (NTS) infection, in which a typically intestinal infection becomes systemic with serious, often fatal, consequences. In this review, we consider the role of malaria‐induced immunoregulatory responses in tipping the balance from tissue homeostasis during malaria infection to risk of invasive NTS. Also, neutrophils are crucial in the clearance of NTS but their ability to mount an oxidative burst and kill intracellular Salmonella is severely compromised during, and for some time after, an acute malaria infection. Here, we summarize the evidence linking malaria and invasive NTS infections; describe the role of neutrophils in clearing NTS infections; review evidence for neutrophil dysfunction in malaria infections; and explore roles of heme oxygenase‐1, IL‐10, and complement in mediating this dysfunction. Finally, given the epidemiological evidence that low density, subclinical malaria infections pose a risk for invasive NTS infections, we consider whether the high prevalence of such infections might underlie the very high incidence of invasive bacterial disease across much of sub‐Saharan Africa.
Collapse
Affiliation(s)
- Jason P Mooney
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, United Kingdom
| | - Lauren J Galloway
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, United Kingdom
| | - Eleanor M Riley
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, United Kingdom
| |
Collapse
|
11
|
Comparative immunogenicity and efficacy of equivalent outer membrane vesicle and glycoconjugate vaccines against nontyphoidal Salmonella. Proc Natl Acad Sci U S A 2018; 115:10428-10433. [PMID: 30262653 PMCID: PMC6187145 DOI: 10.1073/pnas.1807655115] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Nontyphoidal Salmonellae cause a devastating burden of invasive disease in sub-Saharan Africa with high levels of antimicrobial resistance. Vaccination has potential for a major global health impact, but no licensed vaccine is available. The lack of commercial incentive makes simple, affordable technologies the preferred route for vaccine development. Here we compare equivalent Generalized Modules for Membrane Antigens (GMMA) outer membrane vesicles and O-antigen-CRM197 glycoconjugates to deliver lipopolysaccharide O-antigen in bivalent Salmonella Typhimurium and Enteritidis vaccines. Salmonella strains were chosen and tolR deleted to induce GMMA production. O-antigens were extracted from wild-type bacteria and conjugated to CRM197 Purified GMMA and glycoconjugates were characterized and tested in mice for immunogenicity and ability to reduce Salmonella infection. GMMA and glycoconjugate O-antigen had similar structural characteristics, O-acetylation, and glucosylation levels. Immunization with GMMA induced higher anti-O-antigen IgG than glycoconjugate administered without Alhydrogel adjuvant. With Alhydrogel, antibody levels were similar. GMMA induced a diverse antibody isotype profile with greater serum bactericidal activity than glycoconjugate, which induced almost exclusively IgG1. Immunization reduced bacterial colonization of mice subsequently infected with SalmonellaS Typhimurium numbers were lower in tissues of mice vaccinated with GMMA compared with glycoconjugate. S. Enteritidis burden in the tissues was similar in mice immunized with either vaccine. With favorable immunogenicity, low cost, and ability to induce functional antibodies and reduce bacterial burden, GMMA offer a promising strategy for the development of a nontyphoidal Salmonella vaccine compared with established glycoconjugates. GMMA technology is potentially attractive for development of vaccines against other bacteria of global health significance.
Collapse
|
12
|
Milanez GP, Werle CH, Amorim MR, Ribeiro RA, Tibo LHS, Roque-Barreira MC, Oliveira AF, Brocchi M. HU-Lacking Mutants of Salmonella enterica Enteritidis Are Highly Attenuated and Can Induce Protection in Murine Model of Infection. Front Microbiol 2018; 9:1780. [PMID: 30186241 PMCID: PMC6113365 DOI: 10.3389/fmicb.2018.01780] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 07/16/2018] [Indexed: 01/31/2023] Open
Abstract
Salmonella enterica infection is a major public health concern worldwide, particularly when associated with other medical conditions. The serovars Typhimurium and Enteritidis are frequently associated with an invasive illness that primarily affects immunocompromised adults and children with HIV, malaria, or malnutrition. These serovars can also cause infections in a variety of animal hosts, and they are the most common isolates in poultry materials. Here, we described S. Enteritidis mutants, where hupA and hupB genes were deleted, and evaluated their potential use as live-attenuated vaccine candidates. In vitro, the mutants behaved like S. Typhimurium described previously, but there were some particularities in macrophage invasion and survival experiments. The virulence and immunogenicity of the mutant lacking both hupA and hupB (PT4ΔhupAB) were evaluated in a BALB/c mice model. This mutant was highly attenuated and could, therefore, be administrated at doses higher than 109 CFU/treatment, which was sufficient to protect all treated mice challenged with the wild-type parental strain with a single dose. Additionally, the PT4ΔhupAB strain induced production of specific IgG and IgA antibodies against Salmonella and TH1-related cytokines (IFN-γ and TNF-α), indicating that this strain can induce systemic and mucosal protection in the murine model. Additional studies are needed to better understand the mechanisms that lead to attenuation of the double-mutant PT4ΔhupAB and to elucidate the immune response induced by immunization using this strain. However, our data allow us to state that hupAB mutants could be potential candidates to be explore as live-attenuated vaccines.
Collapse
Affiliation(s)
- Guilherme P Milanez
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Catierine H Werle
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Mariene R Amorim
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Rafael A Ribeiro
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Luiz H S Tibo
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Maria Cristina Roque-Barreira
- Department of Cellular and Molecular Biology, Faculdade de Medicina de Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Aline F Oliveira
- Department of Cellular and Molecular Biology, Faculdade de Medicina de Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Marcelo Brocchi
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
| |
Collapse
|
13
|
Rycroft JA, Gollan B, Grabe GJ, Hall A, Cheverton AM, Larrouy-Maumus G, Hare SA, Helaine S. Activity of acetyltransferase toxins involved in Salmonella persister formation during macrophage infection. Nat Commun 2018; 9:1993. [PMID: 29777131 PMCID: PMC5959882 DOI: 10.1038/s41467-018-04472-6] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 04/27/2018] [Indexed: 12/31/2022] Open
Abstract
Non-typhoidal Salmonella strains are responsible for invasive infections associated with high mortality and recurrence in sub-Saharan Africa, and there is strong evidence for clonal relapse following antibiotic treatment. Persisters are non-growing bacteria that are thought to be responsible for the recalcitrance of many infections to antibiotics. Toxin-antitoxin systems are stress-responsive elements that are important for Salmonella persister formation, specifically during infection. Here, we report the analysis of persister formation of clinical invasive strains of Salmonella Typhimurium and Enteritidis in human primary macrophages. We show that all the invasive clinical isolates of both serovars that we tested produce high levels of persisters following internalization by human macrophages. Our genome comparison reveals that S. Enteritidis and S. Typhimurium strains contain three acetyltransferase toxins that we characterize structurally and functionally. We show that all induce the persister state by inhibiting translation through acetylation of aminoacyl-tRNAs. However, they differ in their potency and target partially different subsets of aminoacyl-tRNAs, potentially accounting for their non-redundant effect.
Collapse
Affiliation(s)
- Julian A Rycroft
- Section of Microbiology, Medical Research Council Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London, SW7 2AZ, UK
| | - Bridget Gollan
- Section of Microbiology, Medical Research Council Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London, SW7 2AZ, UK
| | - Grzegorz J Grabe
- Section of Microbiology, Medical Research Council Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London, SW7 2AZ, UK
| | - Alexander Hall
- Section of Microbiology, Medical Research Council Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London, SW7 2AZ, UK
| | - Angela M Cheverton
- Section of Microbiology, Medical Research Council Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London, SW7 2AZ, UK
| | - Gerald Larrouy-Maumus
- Department of Life Sciences, Medical Research Council Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London, SW7 2AZ, UK
| | - Stephen A Hare
- Department of Life Sciences, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
- School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Sophie Helaine
- Section of Microbiology, Medical Research Council Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London, SW7 2AZ, UK.
| |
Collapse
|
14
|
Gilchrist JJ, Rautanen A, Fairfax BP, Mills TC, Naranbhai V, Trochet H, Pirinen M, Muthumbi E, Mwarumba S, Njuguna P, Mturi N, Msefula CL, Gondwe EN, MacLennan JM, Chapman SJ, Molyneux ME, Knight JC, Spencer CCA, Williams TN, MacLennan CA, Scott JAG, Hill AVS. Risk of nontyphoidal Salmonella bacteraemia in African children is modified by STAT4. Nat Commun 2018. [PMID: 29523850 PMCID: PMC5844948 DOI: 10.1038/s41467-017-02398-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Nontyphoidal Salmonella (NTS) is a major cause of bacteraemia in Africa. The disease typically affects HIV-infected individuals and young children, causing substantial morbidity and mortality. Here we present a genome-wide association study (180 cases, 2677 controls) and replication analysis of NTS bacteraemia in Kenyan and Malawian children. We identify a locus in STAT4, rs13390936, associated with NTS bacteraemia. rs13390936 is a context-specific expression quantitative trait locus for STAT4 RNA expression, and individuals carrying the NTS-risk genotype demonstrate decreased interferon-γ (IFNγ) production in stimulated natural killer cells, and decreased circulating IFNγ concentrations during acute NTS bacteraemia. The NTS-risk allele at rs13390936 is associated with protection against a range of autoimmune diseases. These data implicate interleukin-12-dependent IFNγ-mediated immunity as a determinant of invasive NTS disease in African children, and highlight the shared genetic architecture of infectious and autoimmune disease.
Collapse
Affiliation(s)
- James J Gilchrist
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK. .,Department of Paediatrics, University of Oxford, Oxford, OX3 9DU, UK.
| | - Anna Rautanen
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Benjamin P Fairfax
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Tara C Mills
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Vivek Naranbhai
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Holly Trochet
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Matti Pirinen
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK.,Institute for Molecular Medicine, Finland (FIMM) University of Helsinki, FI-00014, Helsinki, Finland
| | - Esther Muthumbi
- KEMRI-Wellcome Trust Research Programme, Kilifi, 80108, Kenya
| | - Salim Mwarumba
- KEMRI-Wellcome Trust Research Programme, Kilifi, 80108, Kenya
| | | | - Neema Mturi
- KEMRI-Wellcome Trust Research Programme, Kilifi, 80108, Kenya
| | - Chisomo L Msefula
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, P.O. Box 30096, Chichiri, Blantyre, Malawi.,Pathology Department, College of Medicine, P.O. Box 360, Chichiri, Blantyre, Malawi
| | - Esther N Gondwe
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, P.O. Box 30096, Chichiri, Blantyre, Malawi
| | - Jenny M MacLennan
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, P.O. Box 30096, Chichiri, Blantyre, Malawi.,Department of Zoology, University of Oxford, South Parks Road, Oxford, OX1 3PS, UK
| | - Stephen J Chapman
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK.,Oxford Centre for Respiratory Medicine, Churchill Hospital Site, Oxford University Hospitals, Oxford, OX3 7LE, UK
| | - Malcolm E Molyneux
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, P.O. Box 30096, Chichiri, Blantyre, Malawi
| | - Julian C Knight
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Chris C A Spencer
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Thomas N Williams
- KEMRI-Wellcome Trust Research Programme, Kilifi, 80108, Kenya.,Department of Medicine, Imperial College, Norfolk Place, London, W2 1PG, UK
| | - Calman A MacLennan
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, P.O. Box 30096, Chichiri, Blantyre, Malawi.,The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - J Anthony G Scott
- KEMRI-Wellcome Trust Research Programme, Kilifi, 80108, Kenya.,Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Adrian V S Hill
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK. .,The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK.
| |
Collapse
|
15
|
IgG Responses to Porins and Lipopolysaccharide within an Outer Membrane-Based Vaccine against Nontyphoidal Salmonella Develop at Discordant Rates. mBio 2018; 9:mBio.02379-17. [PMID: 29511082 PMCID: PMC5844998 DOI: 10.1128/mbio.02379-17] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Antibodies acquired after vaccination or natural infection with Gram-negative bacteria, such as invasive Salmonella enterica serovar Typhimurium, can protect against disease. Immunization with naturally shed outer membrane vesicles from Gram-negative bacteria is being studied for its potential to protect against many infections, since antigens within vesicles maintain their natural conformation and orientation. Shedding can be enhanced through genetic modification, and the resulting particles, generalized modules for membrane antigens (GMMA), not only offer potential as vaccines but also can facilitate the study of B-cell responses to bacterial antigens. Here we show that the response to immunization with GMMA from S. Typhimurium (STmGMMA) provides B-cell-dependent protection and induces antibodies to two immunodominant antigens, lipopolysaccharide (LPS) and porins. Antibodies to LPS O antigen (O-Ag) markedly enhance protection in the spleen, but this effect is less marked in the liver. Strikingly, IgG responses to LPS and porins develop with distinct kinetics. In the first week after immunization, there is a dramatic T-cell-independent B1b-cell-associated induction of all IgG isotypes, except IgG1, to porins but not to LPS. In contrast, production of IgG1 to either antigen was delayed and T cell dependent. Nevertheless, after 1 month, cells in the bone marrow secreting IgG against porins or LPS were present at a similar frequency. Unexpectedly, immunization with O-Ag-deficient STmGMMA did not substantially enhance the anti-porin response. Therefore, IgG switching to all antigens does not develop synchronously within the same complex and so the rate of IgG switching to a single component does not necessarily reflect its frequency within the antigenic complex. Vaccines save millions of lives, yet for some infections there are none. This includes some types of Salmonella infections, killing hundreds of thousands of people annually. We show how a new type of vaccine, called GMMA, that is made from blebs shed from the Salmonella cell wall, works to protect against infection in mice by inducing host proteins (antibodies) specifically recognizing bacterial components (antigens). The rate of development of IgG antibody to antigens within GMMA occurred with different kinetics. However, the antibody response to GMMA persists and is likely to provide prolonged protection for those who need it. These results help show how antibody responses to bacterial antigens develop and how vaccines like GMMA can work and help prevent infection.
Collapse
|
16
|
Characterization and Protective Efficacy of Type III Secretion Proteins as a Broadly Protective Subunit Vaccine against Salmonella enterica Serotypes. Infect Immun 2018; 86:IAI.00473-17. [PMID: 29311233 DOI: 10.1128/iai.00473-17] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 12/17/2017] [Indexed: 12/20/2022] Open
Abstract
Nontyphoidal Salmonella enterica serotypes (NTS) are the leading cause of hospitalization and death due to foodborne illnesses. NTS are the costliest of the foodborne pathogens and cause ∼$4 billion annually in health care costs. In Africa, new invasive NTS are the leading cause of bacteremia, especially in HIV-positive children and adults. Current vaccines against S. enterica are not broadly protective and most are directed at the typhoid-causing serotypes, not the NTS. All S. enterica strains require two type III secretion systems (T3SS) for virulence. The T3SS needle tip protein and the first translocator are localized to the T3SS needle tip and are required for pathogenesis of S. enterica Collectively they are 95 to 98% conserved at the amino acid sequence level among all S. enterica strains. The Salmonella pathogenicity island 1 or 2 tip and first translocator proteins were genetically fused to produce the S1 and S2 fusion proteins, respectively, as potential vaccine candidates. S1 and S2 were then characterized using spectroscopic techniques to understand their structural and biophysical properties. Formulated at the proper pH, S1, S2, or S1 plus S2 (S1S2), admixed with adjuvant, was used to immunize mice followed by a lethal challenge with S. enterica serotype Typhimurium or S. enterica serotype Enteritidis. The S1S2 formulation provided the highest protective efficacy, thus demonstrating that an S1S2 subunit vaccine can provide broad, serotype-independent protection, possibly against all S. enterica serotypes. Such a finding would be transformative in improving human health.
Collapse
|
17
|
Immunological bases of increased susceptibility to invasive nontyphoidal Salmonella infection in children with malaria and anaemia. Microbes Infect 2017; 20:589-598. [PMID: 29248635 PMCID: PMC6250906 DOI: 10.1016/j.micinf.2017.11.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 11/17/2017] [Accepted: 11/20/2017] [Indexed: 01/01/2023]
Abstract
Malaria and anaemia are key underlying factors for iNTS disease in African children. Knowledge of clinical and epidemiological risk-factors for iNTS disease has not been paralleled by an in-depth knowledge of the immunobiology of the disease. Herein, we review human and animal studies on mechanisms of increased susceptibility to iNTS in children.
Collapse
|
18
|
MacLennan CA, Msefula CL, Gondwe EN, Gilchrist JJ, Pensulo P, Mandala WL, Mwimaniwa G, Banda M, Kenny J, Wilson LK, Phiri A, MacLennan JM, Molyneux EM, Molyneux ME, Graham SM. Presentation of life-threatening invasive nontyphoidal Salmonella disease in Malawian children: A prospective observational study. PLoS Negl Trop Dis 2017; 11:e0006027. [PMID: 29216183 PMCID: PMC5745124 DOI: 10.1371/journal.pntd.0006027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 12/27/2017] [Accepted: 10/10/2017] [Indexed: 12/17/2022] Open
Abstract
Nontyphoidal Salmonellae commonly cause invasive disease in African children that is often fatal. The clinical diagnosis of these infections is hampered by the absence of a clear clinical syndrome. Drug resistance means that empirical antibiotic therapy is often ineffective and currently no vaccine is available. The study objective was to identify risk factors for mortality among children presenting to hospital with invasive Salmonella disease in Africa. We conducted a prospective study enrolling consecutive children with microbiologically-confirmed invasive Salmonella disease admitted to Queen Elizabeth Central Hospital, Blantyre, in 2006. Data on clinical presentation, co-morbidities and outcome were used to identify children at risk of inpatient mortality through logistic-regression modeling. Over one calendar year, 263 consecutive children presented with invasive Salmonella disease. Median age was 16 months (range 0-15 years) and 52/256 children (20%; 95%CI 15-25%) died. Nontyphoidal serovars caused 248/263 (94%) of cases. 211/259 (81%) of isolates were multi-drug resistant. 251/263 children presented with bacteremia, 6 with meningitis and 6 with both. Respiratory symptoms were present in 184/240 (77%; 95%CI 71-82%), 123/240 (51%; 95%CI 45-58%) had gastrointestinal symptoms and 101/240 (42%; 95%CI 36-49%) had an overlapping clinical syndrome. Presentation at <7 months (OR 10.0; 95%CI 2.8-35.1), dyspnea (OR 4.2; 95%CI 1.5-12.0) and HIV infection (OR 3.3; 95%CI 1.1-10.2) were independent risk factors for inpatient mortality. Invasive Salmonella disease in Malawi is characterized by high mortality and prevalence of multi-drug resistant isolates, along with non-specific presentation. Young infants, children with dyspnea and HIV-infected children bear a disproportionate burden of the Salmonella-associated mortality in Malawi. Strategies to improve prevention, diagnosis and management of invasive Salmonella disease should be targeted at these children.
Collapse
Affiliation(s)
- Calman A. MacLennan
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- School of Immunity and Infection, College of Medicine and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Malawi
| | - Chisomo L. Msefula
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Malawi
- Department of Microbiology, College of Medicine, University of Malawi, Malawi
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| | - Esther N. Gondwe
- School of Immunity and Infection, College of Medicine and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Malawi
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
- Department of Biochemistry, College of Medicine, University of Malawi, Malawi
| | - James J. Gilchrist
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Malawi
- Department of Paediatrics, University of Oxford, United Kingdom
- Wellcome Trust Centre for Human Genetics, University of Oxford, United Kingdom
| | - Paul Pensulo
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Malawi
| | - Wilson L. Mandala
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Malawi
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
- Department of Basic Medical Sciences, College of Medicine, University of Malawi, Blantyre, Malawi
- Academy of Medical Sciences, Malawi University of Science and Technology, Thyolo, Malawi
| | - Grace Mwimaniwa
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Malawi
| | - Meraby Banda
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Malawi
| | - Julia Kenny
- Department of Paediatrics, College of Medicine, University of Malawi, Malawi
- Infectious Diseases and Microbiology Unit, Institute of Child Health, University College London, London, United Kingdom
| | - Lorna K. Wilson
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Malawi
| | - Amos Phiri
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Malawi
| | - Jenny M. MacLennan
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Malawi
- Department of Zoology, University of Oxford, Oxford, United Kingdom
| | | | - Malcolm E. Molyneux
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Malawi
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
- Department of Medicine, College of Medicine, University of Malawi, Malawi
| | - Stephen M. Graham
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Malawi
- Department of Paediatrics, College of Medicine, University of Malawi, Malawi
- Centre for International Child Health, University of Melbourne and Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Australia
| |
Collapse
|
19
|
Role of sapA and yfgA in Susceptibility to Antibody-Mediated Complement-Dependent Killing and Virulence of Salmonella enterica Serovar Typhimurium. Infect Immun 2017; 85:IAI.00419-17. [PMID: 28674031 PMCID: PMC5563563 DOI: 10.1128/iai.00419-17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 06/16/2017] [Indexed: 01/18/2023] Open
Abstract
The ST313 pathovar of Salmonella enterica serovar Typhimurium contributes to a high burden of invasive disease among African infants and HIV-infected adults. It is characterized by genome degradation (loss of coding capacity) and has increased resistance to antibody-dependent complement-mediated killing compared with enterocolitis-causing strains of S. Typhimurium. Vaccination is an attractive disease-prevention strategy, and leading candidates focus on the induction of bactericidal antibodies. Antibody-resistant strains arising through further gene deletion could compromise such a strategy. Exposing a saturating transposon insertion mutant library of S. Typhimurium to immune serum identified a repertoire of S. Typhimurium genes that, when interrupted, result in increased resistance to serum killing. These genes included several involved in bacterial envelope biogenesis, protein translocation, and metabolism. We generated defined mutant derivatives using S. Typhimurium SL1344 as the host. Based on their initial levels of enhanced resistance to killing, yfgA and sapA mutants were selected for further characterization. The S. Typhimurium yfgA mutant lost the characteristic Salmonella rod-shaped appearance, exhibited increased sensitivity to osmotic and detergent stress, lacked very long lipopolysaccharide, was unable to invade enterocytes, and demonstrated decreased ability to infect mice. In contrast, the S. Typhimurium sapA mutants had similar sensitivity to osmotic and detergent stress and lipopolysaccharide profile and an increased ability to infect enterocytes compared with the wild type, but it had no increased ability to cause in vivo infection. These findings indicate that increased resistance to antibody-dependent complement-mediated killing secondary to genetic deletion is not necessarily accompanied by increased virulence and suggest the presence of different mechanisms of antibody resistance.
Collapse
|
20
|
Lee SJ, Benoun J, Sheridan BS, Fogassy Z, Pham O, Pham QM, Puddington L, McSorley SJ. Dual Immunization with SseB/Flagellin Provides Enhanced Protection against Salmonella Infection Mediated by Circulating Memory Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:1353-1361. [PMID: 28710253 PMCID: PMC5548602 DOI: 10.4049/jimmunol.1601357] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 06/19/2017] [Indexed: 01/09/2023]
Abstract
The development of a subunit Salmonella vaccine has been hindered by the absence of detailed information about antigenic targets of protective Salmonella-specific T and B cells. Recent studies have identified SseB as a modestly protective Ag in susceptible C57BL/6 mice, but the mechanism of protective immunity remains undefined. In this article, we report that simply combining Salmonella SseB with flagellin substantially enhances protective immunity, allowing immunized C57BL/6 mice to survive for up to 30 d following challenge with virulent bacteria. Surprisingly, the enhancing effect of flagellin did not require flagellin Ag targeting during secondary responses or recognition of flagellin by TLR5. Although coimmunization with flagellin did not affect SseB-specific Ab responses, it modestly boosted CD4 responses. In addition, protective immunity was effectively transferred in circulation to parabionts of immunized mice, demonstrating that tissue-resident memory is not required for vaccine-induced protection. Finally, protective immunity required host expression of IFN-γR but was independent of induced NO synthase expression. Taken together, these data indicate that Salmonella flagellin has unique adjuvant properties that improve SseB-mediated protective immunity provided by circulating memory.
Collapse
Affiliation(s)
- Seung-Joo Lee
- Center for Comparative Medicine, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616; and
| | - Joseph Benoun
- Center for Comparative Medicine, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616; and
| | - Brian S Sheridan
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Zachary Fogassy
- Center for Comparative Medicine, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616; and
| | - Oanh Pham
- Center for Comparative Medicine, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616; and
| | - Quynh-Mai Pham
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Lynn Puddington
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Stephen J McSorley
- Center for Comparative Medicine, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616;
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616; and
| |
Collapse
|
21
|
Zhao X, Dai Q, Zhu D, Liu M, Chen S, Sun K, Yang Q, Wu Y, Kong Q, Jia R. Recombinant attenuated Salmonella Typhimurium with heterologous expression of the Salmonella Choleraesuis O-polysaccharide: high immunogenicity and protection. Sci Rep 2017; 7:7127. [PMID: 28754982 PMCID: PMC5533773 DOI: 10.1038/s41598-017-07689-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 07/03/2017] [Indexed: 11/29/2022] Open
Abstract
Non-typhoidal Salmonella are associated with gastrointestinal disease worldwide and invasive disease in Africa. We constructed novel bivalent vaccines through the recombinant expression of heterologous O-antigens from Salmonella Choleraesuis in Salmonella Typhimurium. A recombinant Asd+ plasmid pCZ1 with the cloned Salmonella Choleraesuis O-antigen gene cluster was introduced into three constructed Salmonella Typhimurium Δasd mutants: SLT11 (ΔrfbP), SLT12 (ΔrmlB-rfbP) and SLT16 (ΔrfbP ∆pagL::TT araCPBADrfbP). Immunoblotting demonstrated that SLT11 (pCZ1) and SLT12 (pCZ1) efficiently expressed the heterologous O-antigen. In the presence of arabinose, SLT16 (pCZ1) expressed both the homologous and heterologous O-antigens, whereas in the absence of arabinose, SLT16 (pCZ1) mainly expressed the heterologous O-antigen. We deleted the crp/cya genes in SLT12 (pCZ1) and SLT16 (pCZ1) for attenuation purposes, generating the recombinant vaccine strains SLT17 (pCZ1) and SLT18 (pCZ1). Immunization with either SLT17 (pCZ1) or SLT18 (pCZ1) induced specific IgG against the heterologous O-antigen, which mediated significant killing of Salmonella Choleraesuis and provided full protection against a lethal homologous challenge in mice. Furthermore, SLT17 (pCZ1) or SLT18 (pCZ1) immunization resulted in 83% or 50% heterologous protection against Salmonella Choleraesuis challenge, respectively. Our study demonstrates that heterologous O-antigen expression is a promising strategy for the development of multivalent Salmonella vaccines.
Collapse
Affiliation(s)
- Xinxin Zhao
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan, 611130, P.R. China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang, Chengdu, Sichuan, 611130, P.R. China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan, 611130, P.R. China
| | - Qinlong Dai
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan, 611130, P.R. China
| | - Dekang Zhu
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan, 611130, P.R. China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang, Chengdu, Sichuan, 611130, P.R. China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan, 611130, P.R. China
| | - Mafeng Liu
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan, 611130, P.R. China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang, Chengdu, Sichuan, 611130, P.R. China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan, 611130, P.R. China
| | - Shun Chen
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan, 611130, P.R. China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang, Chengdu, Sichuan, 611130, P.R. China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan, 611130, P.R. China
| | - Kunfeng Sun
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan, 611130, P.R. China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang, Chengdu, Sichuan, 611130, P.R. China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan, 611130, P.R. China
| | - Qiao Yang
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan, 611130, P.R. China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang, Chengdu, Sichuan, 611130, P.R. China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan, 611130, P.R. China
| | - Ying Wu
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan, 611130, P.R. China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang, Chengdu, Sichuan, 611130, P.R. China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan, 611130, P.R. China
| | - Qingke Kong
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan, 611130, P.R. China. .,Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287-5401, USA.
| | - Renyong Jia
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan, 611130, P.R. China. .,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang, Chengdu, Sichuan, 611130, P.R. China. .,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, Sichuan, 611130, P.R. China.
| |
Collapse
|
22
|
Fiorino F, Rondini S, Micoli F, Lanzilao L, Alfini R, Mancini F, MacLennan CA, Medaglini D. Immunogenicity of a Bivalent Adjuvanted Glycoconjugate Vaccine against Salmonella Typhimurium and Salmonella Enteritidis. Front Immunol 2017; 8:168. [PMID: 28289411 PMCID: PMC5326758 DOI: 10.3389/fimmu.2017.00168] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 02/02/2017] [Indexed: 12/12/2022] Open
Abstract
Salmonella enterica serovars Typhimurium and Enteritidis are the predominant causes of invasive non-typhoidal Salmonella (iNTS) disease. Considering the co-endemicity of S. Typhimurium and S. Enteritidis, a bivalent vaccine formulation against both pathogens is necessary for protection against iNTS disease, thus investigation of glycoconjugate combination is required. In the present work, we investigated the immune responses induced by S. Typhimurium and S. Enteritidis monovalent and bivalent glycoconjugate vaccines adjuvanted with aluminum hydroxide (alum) only or in combination with cytosine-phosphorothioate-guanine oligodeoxynucleotide (CpG). Humoral and cellular, systemic and local, immune responses were characterized in two different mouse strains. All conjugate vaccines elicited high levels of serum IgG against the respective O-antigens (OAg) with bactericidal activity. The bivalent conjugate vaccine induced systemic production of antibodies against both S. Typhimurium and S. Enteritidis OAg. The presence of alum or alum + CpG adjuvants in vaccine formulations significantly increased the serum antigen-specific antibody production. The alum + CpG bivalent vaccine formulation triggered the highest systemic anti-OAg antibodies and also a significant increase of anti-OAg IgG in intestinal washes and fecal samples, with a positive correlation with serum levels. These data demonstrate the ability of monovalent and bivalent conjugate vaccines against S. Typhimurium and S. Enteritidis to induce systemic and local immune responses in different mouse strains, and highlight the suitability of a bivalent glycoconjugate formulation, especially when adjuvanted with alum + CpG, as a promising candidate vaccine against iNTS disease.
Collapse
Affiliation(s)
- Fabio Fiorino
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena , Siena , Italy
| | - Simona Rondini
- GSK Vaccines Institute for Global Health S.r.l. (formerly Novartis Vaccines Institute for Global Health S.r.l.) , Siena , Italy
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health S.r.l. (formerly Novartis Vaccines Institute for Global Health S.r.l.) , Siena , Italy
| | - Luisa Lanzilao
- GSK Vaccines Institute for Global Health S.r.l. (formerly Novartis Vaccines Institute for Global Health S.r.l.) , Siena , Italy
| | - Renzo Alfini
- GSK Vaccines Institute for Global Health S.r.l. (formerly Novartis Vaccines Institute for Global Health S.r.l.) , Siena , Italy
| | - Francesca Mancini
- GSK Vaccines Institute for Global Health S.r.l. (formerly Novartis Vaccines Institute for Global Health S.r.l.) , Siena , Italy
| | - Calman A MacLennan
- Jenner Institute, Nuffield Department of Medicine, University of Oxford , Oxford , UK
| | - Donata Medaglini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena , Siena , Italy
| |
Collapse
|
23
|
Mandala WL, Msefula CL, Gondwe EN, Drayson MT, Molyneux ME, MacLennan CA. Monocyte activation and cytokine production in Malawian children presenting with P. falciparum malaria. Parasite Immunol 2017; 38:317-25. [PMID: 27027867 PMCID: PMC4850749 DOI: 10.1111/pim.12319] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/24/2016] [Indexed: 12/20/2022]
Abstract
Malaria in malaria‐naïve adults is associated with an inflammatory response characterized by expression of specific activation markers on innate immune cells. Here, we investigate activation and adhesion marker expression, and cytokine production in monocytes from children presenting with cerebral malaria (CM, n = 36), severe malarial anaemia (SMA, n = 42) or uncomplicated malaria (UM, n = 66), and healthy aparasitemic children (n = 52) in Blantyre, Malawi. In all malaria groups, but particularly in the two severe malaria groups, monocyte expression of CD11b, CD11c, CD18, HLA‐DR and CD86, and percentages of TNF‐α‐ and IL‐6‐producing monocytes were lower than in healthy controls, while expression of CD11a, TLR2 and TLR4 was lower in children with severe malaria compared with controls. These levels mostly normalized during convalescence, but percentages of cytokine‐producing monocytes remained suppressed in children with SMA. In all malaria groups, especially the SMA group, a greater proportion of monocytes were loaded with haemozoin than among controls. In a P. falciparum hyperendemic area, monocytes in children with acute symptomatic malaria have reduced expression of adhesion molecules and activation markers and reduced inflammatory cytokine production. This immune suppression could be due to accumulation of haemozoin and/or previous exposure to P. falciparum.
Collapse
Affiliation(s)
- W L Mandala
- Malawi-Liverpool Wellcome Trust Clinical Research Programme, Blantyre, Malawi.,Department of Biomedical Sciences, College of Medicine, University of Malawi, Blantyre, Malawi.,Liverpool School of Tropical Medicine, Pembroke Place, University of Liverpool, Liverpool, UK
| | - C L Msefula
- Malawi-Liverpool Wellcome Trust Clinical Research Programme, Blantyre, Malawi.,Liverpool School of Tropical Medicine, Pembroke Place, University of Liverpool, Liverpool, UK
| | - E N Gondwe
- Malawi-Liverpool Wellcome Trust Clinical Research Programme, Blantyre, Malawi.,Liverpool School of Tropical Medicine, Pembroke Place, University of Liverpool, Liverpool, UK
| | - M T Drayson
- Medical Research Council Centre for Immune Regulation and Clinical Immunology Service, Institute of Biomedical Research, School of Immunity and Infection, College of Medicine and Dental Sciences, University of Birmingham, Birmingham, UK
| | - M E Molyneux
- Malawi-Liverpool Wellcome Trust Clinical Research Programme, Blantyre, Malawi.,Liverpool School of Tropical Medicine, Pembroke Place, University of Liverpool, Liverpool, UK.,Department of Medicine, College of Medicine, University of Malawi, Blantyre, Malawi
| | - C A MacLennan
- Malawi-Liverpool Wellcome Trust Clinical Research Programme, Blantyre, Malawi.,Medical Research Council Centre for Immune Regulation and Clinical Immunology Service, Institute of Biomedical Research, School of Immunity and Infection, College of Medicine and Dental Sciences, University of Birmingham, Birmingham, UK.,The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
24
|
Uche IV, MacLennan CA, Saul A. A Systematic Review of the Incidence, Risk Factors and Case Fatality Rates of Invasive Nontyphoidal Salmonella (iNTS) Disease in Africa (1966 to 2014). PLoS Negl Trop Dis 2017; 11:e0005118. [PMID: 28056035 PMCID: PMC5215826 DOI: 10.1371/journal.pntd.0005118] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 10/19/2016] [Indexed: 11/19/2022] Open
Abstract
This study systematically reviews the literature on the occurrence, incidence and case fatality rate (CFR) of invasive nontyphoidal Salmonella (iNTS) disease in Africa from 1966 to 2014. Data on the burden of iNTS disease in Africa are sparse and generally have not been aggregated, making it difficult to describe the epidemiology that is needed to inform the development and implementation of effective prevention and control policies. This study involved a comprehensive search of PubMed and Embase databases. It documents the geographical spread of iNTS disease over time in Africa, and describes its reported incidence, risk factors and CFR. We found that Nontyphoidal Salmonella (NTS) have been reported as a cause of bacteraemia in 33 out of 54 African countries, spanning the five geographical regions of Africa, and especially in sub-Saharan Africa since 1966. Our review indicates that NTS have been responsible for up to 39% of community acquired blood stream infections in sub-Saharan Africa with an average CFR of 19%. Salmonella Typhimurium and Enteritidis are the major serovars implicated and together have been responsible for 91%% of the cases of iNTS disease, (where serotype was determined), reported in Africa. The study confirms that iNTS disease is more prevalent amongst Human Immunodeficiency Virus (HIV)-infected individuals, infants, and young children with malaria, anaemia and malnutrition. In conclusion, iNTS disease is a substantial cause of community-acquired bacteraemia in Africa. Given the high morbidity and mortality of iNTS disease in Africa, it is important to develop effective prevention and control strategies including vaccination.
Collapse
Affiliation(s)
| | | | - Allan Saul
- Novartis Vaccines Institute for Global Health, Siena, Italy
| |
Collapse
|
25
|
De Benedetto G, Alfini R, Cescutti P, Caboni M, Lanzilao L, Necchi F, Saul A, MacLennan CA, Rondini S, Micoli F. Characterization of O-antigen delivered by Generalized Modules for Membrane Antigens (GMMA) vaccine candidates against nontyphoidal Salmonella. Vaccine 2016; 35:419-426. [PMID: 27998639 DOI: 10.1016/j.vaccine.2016.11.089] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/21/2016] [Accepted: 11/27/2016] [Indexed: 12/20/2022]
Abstract
Invasive nontyphoidal Salmonella disease (iNTS) is a leading cause of death and morbidity in Africa. The most common pathogens are Salmonella enterica serovars Typhimurium and Enteritidis. The O-antigen portion of their lipopolysaccharide is a target of protective immunity and vaccines targeting O-antigen are currently in development. Here we investigate the use of Generalized Modules for Membrane Antigens (GMMA) as delivery system for S. Typhimurium and S. Enteritidis O-antigen. Gram-negative bacteria naturally shed outer membrane in a blebbing process. By deletion of the tolR gene, the level of shedding was greatly enhanced. Further genetic modifications were introduced into the GMMA-producing strains in order to reduce reactogenicity, by detoxifying the lipid A moiety of lipopolysaccharide. We found that genetic mutations can impact on expression of O-antigen chains. All S. Enteritidis GMMA characterized had an O-antigen to protein w/w ratio higher than 0.6, while the ratio was 0.7 for S. Typhimurium ΔtolR GMMA, but decreased to less than 0.1 when further mutations for lipid A detoxification were introduced. Changes were also observed in O-antigen chain length and level and/or position of O-acetylation. When tested in mice, the GMMA induced high levels of anti-O-antigen-specific IgG functional antibodies, despite variation in density and O-antigen structural modifications. In conclusion, simplicity of manufacturing process and low costs of production, coupled with encouraging immunogenicity data, make GMMA an attractive strategy to further investigate for the development of a vaccine against iNTS.
Collapse
Affiliation(s)
- G De Benedetto
- GSK Vaccines Institute for Global Health (GVGH) S.r.l. (former Novartis Vaccines Institute for Global Health, NVGH), Via Fiorentina 1, 53100 Siena, Italy; Dipartimento di Scienze della Vita, Ed. C11, Università degli Studi di Trieste, via L. Giorgieri 1, 34127 Trieste, Italy
| | - R Alfini
- GSK Vaccines Institute for Global Health (GVGH) S.r.l. (former Novartis Vaccines Institute for Global Health, NVGH), Via Fiorentina 1, 53100 Siena, Italy
| | - P Cescutti
- Dipartimento di Scienze della Vita, Ed. C11, Università degli Studi di Trieste, via L. Giorgieri 1, 34127 Trieste, Italy
| | - M Caboni
- Antimicrobial Discovery Center, Department of Biology, 360 Huntington Ave., Boston, MA 02115, United States
| | - L Lanzilao
- GSK Vaccines Institute for Global Health (GVGH) S.r.l. (former Novartis Vaccines Institute for Global Health, NVGH), Via Fiorentina 1, 53100 Siena, Italy
| | - F Necchi
- GSK Vaccines Institute for Global Health (GVGH) S.r.l. (former Novartis Vaccines Institute for Global Health, NVGH), Via Fiorentina 1, 53100 Siena, Italy
| | - A Saul
- GSK Vaccines Institute for Global Health (GVGH) S.r.l. (former Novartis Vaccines Institute for Global Health, NVGH), Via Fiorentina 1, 53100 Siena, Italy
| | - C A MacLennan
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - S Rondini
- GSK Vaccines Institute for Global Health (GVGH) S.r.l. (former Novartis Vaccines Institute for Global Health, NVGH), Via Fiorentina 1, 53100 Siena, Italy
| | - F Micoli
- GSK Vaccines Institute for Global Health (GVGH) S.r.l. (former Novartis Vaccines Institute for Global Health, NVGH), Via Fiorentina 1, 53100 Siena, Italy.
| |
Collapse
|
26
|
Park SE, Pak GD, Aaby P, Adu-Sarkodie Y, Ali M, Aseffa A, Biggs HM, Bjerregaard-Andersen M, Breiman RF, Crump JA, Cruz Espinoza LM, Eltayeb MA, Gasmelseed N, Hertz JT, Im J, Jaeger A, Parfait Kabore L, von Kalckreuth V, Keddy KH, Konings F, Krumkamp R, MacLennan CA, Meyer CG, Montgomery JM, Ahmet Niang A, Nichols C, Olack B, Panzner U, Park JK, Rabezanahary H, Rakotozandrindrainy R, Sampo E, Sarpong N, Schütt-Gerowitt H, Sooka A, Soura AB, Sow AG, Tall A, Teferi M, Yeshitela B, May J, Wierzba TF, Clemens JD, Baker S, Marks F. The Relationship Between Invasive Nontyphoidal Salmonella Disease, Other Bacterial Bloodstream Infections, and Malaria in Sub-Saharan Africa. Clin Infect Dis 2016; 62 Suppl 1:S23-31. [PMID: 26933016 DOI: 10.1093/cid/civ893] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Country-specific studies in Africa have indicated that Plasmodium falciparum is associated with invasive nontyphoidal Salmonella (iNTS) disease. We conducted a multicenter study in 13 sites in Burkina Faso, Ethiopia, Ghana, Guinea-Bissau, Kenya, Madagascar, Senegal, South Africa, Sudan, and Tanzania to investigate the relationship between the occurrence of iNTS disease, other systemic bacterial infections, and malaria. METHODS Febrile patients received a blood culture and a malaria test. Isolated bacteria underwent antimicrobial susceptibility testing, and the association between iNTS disease and malaria was assessed. RESULTS A positive correlation between frequency proportions of malaria and iNTS was observed (P = .01; r = 0.70). Areas with higher burden of malaria exhibited higher odds of iNTS disease compared to other bacterial infections (odds ratio [OR], 4.89; 95% CI, 1.61-14.90; P = .005) than areas with lower malaria burden. Malaria parasite positivity was associated with iNTS disease (OR, 2.44; P = .031) and gram-positive bacteremias, particularly Staphylococcus aureus, exhibited a high proportion of coinfection with Plasmodium malaria. Salmonella Typhimurium and Salmonella Enteritidis were the predominant NTS serovars (53/73; 73%). Both moderate (OR, 6.05; P = .0001) and severe (OR, 14.62; P < .0001) anemia were associated with iNTS disease. CONCLUSIONS A positive correlation between iNTS disease and malaria endemicity, and the association between Plasmodium parasite positivity and iNTS disease across sub-Saharan Africa, indicates the necessity to consider iNTS as a major cause of febrile illness in malaria-holoendemic areas. Prevention of iNTS disease through iNTS vaccines for areas of high malaria endemicity, targeting high-risk groups for Plasmodium parasitic infection, should be considered.
Collapse
Affiliation(s)
- Se Eun Park
- International Vaccine Institute, Seoul, Republic of Korea
| | - Gi Deok Pak
- International Vaccine Institute, Seoul, Republic of Korea
| | - Peter Aaby
- Bandim Health Project, Bissau, Guinea-Bissau Research Center for Vitamins and Vaccines, Copenhagen, Denmark
| | - Yaw Adu-Sarkodie
- Kumasi Centre for Collaborative Research in Tropical Medicine School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Mohammad Ali
- International Vaccine Institute, Seoul, Republic of Korea Johns Hopkins University, Baltimore, Maryland
| | - Abraham Aseffa
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Holly M Biggs
- Division of Infectious Diseases and International Health, Duke University Medical Center, Durham, North Carolina Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | | | - Robert F Breiman
- Centers for Disease Control and Prevention, Nairobi, Kenya Emory Global Health Institute, Emory University, Atlanta, Georgia
| | - John A Crump
- Division of Infectious Diseases and International Health, Duke University Medical Center, Durham, North Carolina Kilimanjaro Christian Medical Centre, Moshi, Tanzania Duke Global Health Institute, Duke University, Durham, North Carolina Centre for International Health, University of Otago, Dunedin, New Zealand
| | | | | | | | - Julian T Hertz
- Division of Infectious Diseases and International Health, Duke University Medical Center, Durham, North Carolina Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Justin Im
- International Vaccine Institute, Seoul, Republic of Korea
| | - Anna Jaeger
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | | | - Karen H Keddy
- National Institute for Communicable Diseases Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Frank Konings
- International Vaccine Institute, Seoul, Republic of Korea
| | - Ralf Krumkamp
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Calman A MacLennan
- Jenner Institute, Nuffield Department of Medicine, University of Oxford Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Christian G Meyer
- Institute of Tropical Medicine, Eberhard-Karls University Tübingen, Germany
| | | | | | | | | | - Ursula Panzner
- International Vaccine Institute, Seoul, Republic of Korea
| | - Jin Kyung Park
- International Vaccine Institute, Seoul, Republic of Korea
| | | | | | - Emmanuel Sampo
- Schiphra Hospital, Ouagadougou, Burkina Faso Institut Supérieur des Sciences de la Population, University of Ouagadougou, Burkina Faso
| | - Nimako Sarpong
- Kumasi Centre for Collaborative Research in Tropical Medicine
| | - Heidi Schütt-Gerowitt
- International Vaccine Institute, Seoul, Republic of Korea Institute of Medical Microbiology, University of Cologne, Germany
| | | | | | - Amy Gassama Sow
- Institute Pasteur Senegal, Dakar Université Cheikh Anta Diop de Dakar, Senegal
| | | | | | | | - Jürgen May
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | - John D Clemens
- International Vaccine Institute, Seoul, Republic of Korea International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka Fielding School of Public Health, University of California, Los Angeles
| | - Stephen Baker
- Hospital for Tropical Diseases, Wellcome Trust Major Overseas Programme, Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Florian Marks
- International Vaccine Institute, Seoul, Republic of Korea
| |
Collapse
|
27
|
Yang M, Simon R, MacKerell AD. Conformational Preference of Serogroup B Salmonella O Polysaccharide in Presence and Absence of the Monoclonal Antibody Se155-4. J Phys Chem B 2016; 121:3412-3423. [PMID: 28423910 DOI: 10.1021/acs.jpcb.6b08955] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Salmonella infection is a major public health problem worldwide. Antibodies directed toward the O polysaccharide (OPS) of S. Typhimurium, a serogroup B nontyphoidal Salmonella serovar, have protected against fatal infection in animal models. The OPS is known to undergo O-acetylation, though the impact of these modifications on antibody binding is poorly understood. Using molecular simulations, we assessed the conformational properties and antigen-antibody interactions of deacetylated and O-acetylated S. Typhimurium OPS when bound by monoclonal anti-OPS IgG Se155-4. Our findings indicate that (i) the α-d-abequose (8) monosaccharide makes important interactions with Se155-4, (ii) the deacetylated form binds to the antibody in two conformations, (iii) the acetyl group at α-l-rhamnose (5) traps the acetylated O-antigenic saccharide in one of those two conformations when bound to the antibody; (iv) the dominant conformation sampled by both unbound saccharides only occurs in the deacetylated-antibody complex; and (v) both unbound saccharides sample the second bound conformation to a small extent (2-4%). These observations provide insights into the conformational preference of an antigenic saccharide when bound to a well-characterized specific monoclonal antibody, and suggest possible important properties of vaccine induced antibodies following immunization with live attenuated and OPS-based subunit vaccines.
Collapse
Affiliation(s)
- Mingjun Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland , Baltimore, Maryland 21201, United States
| | - Raphael Simon
- Center for Vaccine Development, Institute for Global Health, School of Medicine, University of Maryland , Baltimore, Maryland 21201, United States
| | - Alexander D MacKerell
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland , Baltimore, Maryland 21201, United States
| |
Collapse
|
28
|
Cytokine Profiles during Invasive Nontyphoidal Salmonella Disease Predict Outcome in African Children. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 23:601-9. [PMID: 27170644 PMCID: PMC4933780 DOI: 10.1128/cvi.00128-16] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 05/07/2016] [Indexed: 01/01/2023]
Abstract
Nontyphoidal Salmonella is a leading cause of sepsis in African children. Cytokine responses are central to the pathophysiology of sepsis and predict sepsis outcome in other settings. In this study, we investigated cytokine responses to invasive nontyphoidal Salmonella (iNTS) disease in Malawian children. We determined serum concentrations of 48 cytokines with multiplexed immunoassays in Malawian children during acute iNTS disease (n = 111) and in convalescence (n = 77). Principal component analysis and logistic regression were used to identify cytokine signatures of acute iNTS disease. We further investigated whether these responses are altered by HIV coinfection or severe malnutrition and whether cytokine responses predict inpatient mortality. Cytokine changes in acute iNTS disease were associated with two distinct cytokine signatures. The first is characterized by increased concentrations of mediators known to be associated with macrophage function, and the second is characterized by raised pro- and anti-inflammatory cytokines typical of responses reported in sepsis secondary to diverse pathogens. These cytokine responses were largely unaltered by either severe malnutrition or HIV coinfection. Children with fatal disease had a distinctive cytokine profile, characterized by raised mediators known to be associated with neutrophil function. In conclusion, cytokine responses to acute iNTS infection in Malawian children are reflective of both the cytokine storm typical of sepsis secondary to diverse pathogens and the intramacrophage replicative niche of NTS. The cytokine profile predictive of fatal disease supports a key role of neutrophils in the pathogenesis of NTS sepsis.
Collapse
|
29
|
Goh YS, Necchi F, O’Shaughnessy CM, Micoli F, Gavini M, Young SP, Msefula CL, Gondwe EN, Mandala WL, Gordon MA, Saul AJ, MacLennan CA. Bactericidal Immunity to Salmonella in Africans and Mechanisms Causing Its Failure in HIV Infection. PLoS Negl Trop Dis 2016; 10:e0004604. [PMID: 27057743 PMCID: PMC4825999 DOI: 10.1371/journal.pntd.0004604] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 03/12/2016] [Indexed: 12/17/2022] Open
Abstract
Background Nontyphoidal strains of Salmonella are a leading cause of death among HIV-infected Africans. Antibody-induced complement-mediated killing protects healthy Africans against Salmonella, but increased levels of anti-lipopolysaccharide (LPS) antibodies in some HIV-infected African adults block this killing. The objective was to understand how these high levels of anti-LPS antibodies interfere with the killing of Salmonella. Methodology/Principal Findings Sera and affinity-purified antibodies from African HIV-infected adults that failed to kill invasive S. Typhimurium D23580 were compared to sera from HIV-uninfected and HIV-infected subjects with bactericidal activity. The failure of sera from certain HIV-infected subjects to kill Salmonella was found to be due to an inherent inhibitory effect of anti-LPS antibodies. This inhibition was concentration-dependent and strongly associated with IgA and IgG2 anti-LPS antibodies (p<0.0001 for both). IgG anti-LPS antibodies, from sera of HIV-infected individuals that inhibit killing at high concentration, induced killing when diluted. Conversely, IgG, from sera of HIV-uninfected adults that induce killing, inhibited killing when concentrated. IgM anti-LPS antibodies from all subjects also induced Salmonella killing. Finally, the inhibitory effect of high concentrations of anti-LPS antibodies is seen with IgM as well as IgG and IgA. No correlation was found between affinity or avidity, or complement deposition or consumption, and inhibition of killing. Conclusion/Significance IgG and IgM classes of anti-S. Typhimurium LPS antibodies from HIV-infected and HIV-uninfected individuals are bactericidal, while at very high concentrations, anti-LPS antibodies of all classes inhibit in vitro killing of Salmonella. This could be due to a variety of mechanisms relating to the poor ability of IgA and IgG2 to activate complement, and deposition of complement at sites where it cannot insert in the bacterial membrane. Vaccine trials are required to understand the significance of lack of in vitro killing by anti-LPS antibodies from a minority of HIV-infected individuals with impaired immune homeostasis. Bacteremia caused by nontyphoidal Salmonellae are a major health burden in Africa. While antibody-induced complement-mediated killing protects healthy Africans against Salmonella, increased levels of anti-LPS antibodies in some HIV-infected Africans block this killing. Little is known about the mechanism of the interference of killing by these antibodies. Here, we compared sera and affinity-purified antibodies from African HIV-infected adults that are unable to kill invasive S. Typhimurium D23580, with sera from HIV-uninfected and HIV-infected subjects with bactericidal activity. We found that the blocking effect of anti-LPS antibodies is a factor of antibody concentration, rather than antibody structure or specificity. While all three isotypes (IgG, IgA and IgM) can inhibit killing of Salmonella at grossly high concentrations, the IgG and IgM isotypes of the anti-LPS antibodies have in vitro bactericidal activity against invasive African S. Typhimurium. Inhibition of killing did not associate with antibody affinity or avidity, or complement deposition or consumption. It is possible that a LPS-based vaccine would induce antibodies at bactericidal rather than inhibitory concentrations in HIV-uninfected individuals. In HIV-infected individuals, it is uncertain whether vaccination will induce a protective response or a dysregulated excess of anti-LPS antibodies that impairs serum killing of Salmonella.
Collapse
Affiliation(s)
- Yun Shan Goh
- Singapore Immunology Network, Agency for Science, Technology and Research, Biopolis, Singapore
| | - Francesca Necchi
- Sclavo Behring Vaccines Institute for Global Health, a GlaxoSmith Kline Company, Siena, Italy
| | - Colette M. O’Shaughnessy
- School of Immunity and Infection, College of Medicine and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Francesca Micoli
- Sclavo Behring Vaccines Institute for Global Health, a GlaxoSmith Kline Company, Siena, Italy
| | | | - Stephen P. Young
- Centre for Translational Inflammation Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Chisomo L. Msefula
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre, Malawi
- Department of Pathology, Division of Microbiology, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Esther N. Gondwe
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre, Malawi
- Department of Basic Medical Sciences, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Wilson L. Mandala
- Centre for Translational Inflammation Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Melita A. Gordon
- Department of Gastroenterology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Allan J. Saul
- Sclavo Behring Vaccines Institute for Global Health, a GlaxoSmith Kline Company, Siena, Italy
| | - Calman A. MacLennan
- School of Immunity and Infection, College of Medicine and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
30
|
Tennant SM, MacLennan CA, Simon R, Martin LB, Khan MI. Nontyphoidal salmonella disease: Current status of vaccine research and development. Vaccine 2016; 34:2907-2910. [PMID: 27032517 DOI: 10.1016/j.vaccine.2016.03.072] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 03/09/2016] [Indexed: 12/30/2022]
Abstract
Among more than 2500 nontyphoidal Salmonella enterica (NTS) serovars, S. enterica serovar Typhimurium and S. enterica serovar Enteritidis account for approximately fifty percent of all human isolates of NTS reported globally. The global incidence of NTS gastroenteritis in 2010 was estimated to be 93 million cases, approximately 80 million of which were contracted via food-borne transmission. It is estimated that 155,000 deaths resulted from NTS in 2010. NTS also causes severe, extra-intestinal, invasive bacteremia, referred to as invasive nontyphoidal Salmonella (iNTS) disease. iNTS disease usually presents as a febrile illness, frequently without gastrointestinal symptoms, in both adults and children. Symptoms of iNTS are similar to malaria, often including fever (>90%) and splenomegaly (>40%). The underlying reasons for the high rates of iNTS disease in Africa are still being elucidated. Evidence from animal and human studies supports the feasibility of developing a safe and effective vaccine against iNTS. Both antibodies and complement can kill Salmonella species in vitro. Proof-of-principle studies in animal models have demonstrated efficacy for live attenuated and subunit vaccines that target the O-antigens, flagellin proteins, and other outer membrane proteins of serovars Typhimurium and Enteritidis. More recently, a novel delivery strategy for NTS vaccines has been developed: the Generalized Modules for Membrane Antigens (GMMA) technology which presents surface polysaccharides and outer membrane proteins in their native conformation. GMMA technology is self-adjuvanting, as it delivers multiple pathogen-associated molecular pattern molecules. GMMA may be particularly relevant for low- and middle-income countries as it has the potential for high immunologic potency at a low cost and involves a relatively simple production process without the need for complex conjugation. Several vaccines for the predominant NTS serovars Typhimurium and Enteritidis, are currently under development.
Collapse
Affiliation(s)
- Sharon M Tennant
- Center for Vaccine Development and Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Calman A MacLennan
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom; Wellcome Trust Sanger Institute, Wellcome Trust Genomes Campus, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Raphael Simon
- Center for Vaccine Development and Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Laura B Martin
- GSK Vaccines Institute for Global Health, S.r.l. Via Fiorentina 1, 53100 Siena, Italy
| | - M Imran Khan
- Center of Excellence in Woman and Child Health, The Aga Khan University, Stadium Road Karachi 74800, Pakistan.
| |
Collapse
|
31
|
Hart PJ, O’Shaughnessy CM, Siggins MK, Bobat S, Kingsley RA, Goulding DA, Crump JA, Reyburn H, Micoli F, Dougan G, Cunningham AF, MacLennan CA. Differential Killing of Salmonella enterica Serovar Typhi by Antibodies Targeting Vi and Lipopolysaccharide O:9 Antigen. PLoS One 2016; 11:e0145945. [PMID: 26741681 PMCID: PMC4712142 DOI: 10.1371/journal.pone.0145945] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 12/10/2015] [Indexed: 01/08/2023] Open
Abstract
Salmonella enterica serovar Typhi expresses a capsule of Vi polysaccharide, while most Salmonella serovars, including S. Enteritidis and S. Typhimurium, do not. Both S. Typhi and S. Enteritidis express the lipopolysaccharide O:9 antigen, yet there is little evidence of cross-protection from anti-O:9 antibodies. Vaccines based on Vi polysaccharide have efficacy against typhoid fever, indicating that antibodies against Vi confer protection. Here we investigate the role of Vi capsule and antibodies against Vi and O:9 in antibody-dependent complement- and phagocyte-mediated killing of Salmonella. Using isogenic Vi-expressing and non-Vi-expressing derivatives of S. Typhi and S. Typhimurium, we show that S. Typhi is inherently more sensitive to serum and blood than S. Typhimurium. Vi expression confers increased resistance to both complement- and phagocyte-mediated modalities of antibody-dependent killing in human blood. The Vi capsule is associated with reduced C3 and C5b-9 deposition, and decreased overall antibody binding to S. Typhi. However, purified human anti-Vi antibodies in the presence of complement are able to kill Vi-expressing Salmonella, while killing by anti-O:9 antibodies is inversely related to Vi expression. Human serum depleted of antibodies to antigens other than Vi retains the ability to kill Vi-expressing bacteria. Our findings support a protective role for Vi capsule in preventing complement and phagocyte killing of Salmonella that can be overcome by specific anti-Vi antibodies, but only to a limited extent by anti-O:9 antibodies.
Collapse
Affiliation(s)
- Peter J. Hart
- School of Immunity and Infection, College of Medicine and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Colette M. O’Shaughnessy
- School of Immunity and Infection, College of Medicine and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Matthew K. Siggins
- School of Immunity and Infection, College of Medicine and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Saeeda Bobat
- School of Immunity and Infection, College of Medicine and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Robert A. Kingsley
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - David A. Goulding
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - John A. Crump
- Centre for International Health, University of Otago, Dunedin, New Zealand
- Kilimanjaro Christian Medical Centre and Kilimanjaro Christian Medical University College, Tumaini University, Moshi, Tanzania
- Division of Infectious Diseases and International Health, Duke University Medical Center, Durham, United States of America
- Duke Global Health Institute, Duke University, Durham, United States of America
| | - Hugh Reyburn
- London School of Hygiene and Tropical Medicine, Keppel Street, London, United Kingdom
| | - Francesca Micoli
- Sclavo-Behring Vaccines Institute for Global Health, a GlaxoSmithKline Company, Siena, Italy
| | - Gordon Dougan
- Centre for International Health, University of Otago, Dunedin, New Zealand
| | - Adam F. Cunningham
- School of Immunity and Infection, College of Medicine and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Calman A. MacLennan
- School of Immunity and Infection, College of Medicine and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
32
|
Goh YS, Armour KL, Clark MR, Grant AJ, Mastroeni P. Igg Subclasses Targeting the Flagella of Salmonella enterica Serovar Typhimurium Can Mediate Phagocytosis and Bacterial Killing. ACTA ACUST UNITED AC 2016; 7. [PMID: 27366588 DOI: 10.4172/2157-7560.1000322] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Invasive non-typhoidal Salmonella are a common cause of invasive disease in immuno-compromised individuals and in children. Multi-drug resistance poses challenges to disease control, with a critical need for effective vaccines. Flagellin is an attractive vaccine candidate due to surface exposure and high epitope copy number, but its potential as a target for opsonophacytic antibodies is unclear. We examined the effect of targeting flagella with different classes of IgG on the interaction between Salmonella Typhimurium and a human phagocyte-like cell line, THP-1. We tagged the FliC flagellar protein with a foreign CD52 mimotope (TSSPSAD) and bacteria were opsonized with a panel of humanised CD52 antibodies with the same antigen-binding V-region, but different constant regions. We found that IgG binding to flagella increases bacterial phagocytosis and reduces viable intracellular bacterial numbers. Opsonisation with IgG3, followed by IgG1, IgG4, and IgG2, resulted in the highest level of bacterial uptake and in the highest reduction in the intracellular load of viable bacteria. Taken together, our data provide proof-of-principle evidence that targeting flagella with antibodies can increase the antibacterial function of host cells, with IgG3 being the most potent subclass. These data will assist the rational design of urgently needed, optimised vaccines against iNTS disease.
Collapse
Affiliation(s)
- Yun Shan Goh
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - Kathryn L Armour
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| | - Michael R Clark
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| | - Andrew J Grant
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - Pietro Mastroeni
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| |
Collapse
|
33
|
Strain Selection for Generation of O-Antigen-Based Glycoconjugate Vaccines against Invasive Nontyphoidal Salmonella Disease. PLoS One 2015; 10:e0139847. [PMID: 26445460 PMCID: PMC4596569 DOI: 10.1371/journal.pone.0139847] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 09/17/2015] [Indexed: 01/09/2023] Open
Abstract
Nontyphoidal Salmonellae, principally S. Typhimurium and S. Enteritidis, are a major cause of invasive bloodstream infections in sub-Saharan Africa with no vaccine currently available. Conjugation of lipopolysaccharide O-antigen to a carrier protein constitutes a promising vaccination strategy. Here we describe a rational process to select the most appropriate isolates of Salmonella as source of O-antigen for developing a bivalent glycoconjugate vaccine. We screened a library of 30 S. Typhimurium and 21 S. Enteritidis in order to identify the most suitable strains for large scale O-antigen production and generation of conjugate vaccines. Initial screening was based on growth characteristics, safety profile of the isolates, O-antigen production, and O-antigen characteristics in terms of molecular size, O-acetylation and glucosylation level and position, as determined by phenol sulfuric assay, NMR, HPLC-SEC and HPAEC-PAD. Three animal isolates for each serovar were identified and used to synthesize candidate glycoconjugate vaccines, using CRM197 as carrier protein. The immunogenicity of these conjugates and the functional activity of the induced antibodies was investigated by ELISA, serum bactericidal assay and flow cytometry. S. Typhimurium O-antigen showed high structural diversity, including O-acetylation of rhamnose in a Malawian invasive strain generating a specific immunodominant epitope. S. Typhimurium conjugates provoked an anti-O-antigen response primarily against the O:5 determinant. O-antigen from S. Enteritidis was structurally more homogeneous than from S. Typhimurium, and no idiosyncratic antibody responses were detected for the S. Enteritidis conjugates. Of the three initially selected isolates, two S. Typhimurium (1418 and 2189) and two S. Enteritidis (502 and 618) strains generated glycoconjugates able to induce high specific antibody levels with high breadth of serovar-specific strain coverage, and were selected for use in vaccine production. The strain selection approach described is potentially applicable to the development of glycoconjugate vaccines against other bacterial pathogens.
Collapse
|
34
|
Marshall JM, Gunn JS. The O-Antigen Capsule of Salmonella enterica Serovar Typhimurium Facilitates Serum Resistance and Surface Expression of FliC. Infect Immun 2015; 83:3946-59. [PMID: 26195553 PMCID: PMC4567616 DOI: 10.1128/iai.00634-15] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 07/16/2015] [Indexed: 11/20/2022] Open
Abstract
Group IV polysaccharide capsules are common in enteric bacteria and have more recently been described in nontyphoidal Salmonella species. Such capsules are known as O-antigen (O-Ag) capsules, due to their high degree of similarity to the O-Ag of the lipopolysaccharide (LPSO-Ag). Capsular polysaccharides are known virulence factors of many bacterial pathogens, facilitating evasion of immune recognition and systemic dissemination within the host. Previous studies on the O-Ag capsule of salmonellae have focused primarily on its role in bacterial surface attachment and chronic infection; however, the potential effects of the O-Ag capsule on acute pathogenesis have yet to be investigated. While much of the in vivo innate immune resistance of Salmonella enterica serovar Typhimurium is attributed to the high-molecular-weight LPS, we hypothesized that the O-Ag capsule may enhance this resistance by diminishing surface expression of pathogen-associated molecular patterns, such as flagella, and increasing resistance to host immune molecules. To test this hypothesis, O-Ag capsule-deficient mutants were constructed, and the loss of O-Ag capsular surface expression was confirmed through microscopy and immunoblotting. Loss of O-Ag capsule production did not alter bacterial growth or production of LPS. Western blot analysis and confocal microscopy revealed that O-Ag capsule-deficient mutants demonstrate reduced resistance to killing by human serum. Furthermore, O-Ag capsule-deficient mutants produced exclusively phase I flagellin (FliC). Although O-Ag capsule-deficient mutants did not exhibit reduced virulence in a murine model of acute infection, in vitro results indicate that the O-Ag capsule may function to modify the antigenic nature of the bacterial surface, warranting additional investigation of a potential role of the structure in pathogenesis.
Collapse
Affiliation(s)
- Joanna M Marshall
- Department of Microbial Infection and Immunity, Center for Microbial Interface Biology, The Ohio State University, Columbus, Ohio, USA
| | - John S Gunn
- Department of Microbial Infection and Immunity, Center for Microbial Interface Biology, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
35
|
Mooney JP, Lee SJ, Lokken KL, Nanton MR, Nuccio SP, McSorley SJ, Tsolis RM. Transient Loss of Protection Afforded by a Live Attenuated Non-typhoidal Salmonella Vaccine in Mice Co-infected with Malaria. PLoS Negl Trop Dis 2015; 9:e0004027. [PMID: 26366739 PMCID: PMC4569369 DOI: 10.1371/journal.pntd.0004027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/03/2015] [Indexed: 11/19/2022] Open
Abstract
In immunocompetent individuals, non-typhoidal Salmonella serovars (NTS) are associated with gastroenteritis, however, there is currently an epidemic of NTS bloodstream infections in sub-Saharan Africa. Plasmodium falciparum malaria is an important risk factor for invasive NTS bloodstream in African children. Here we investigated whether a live, attenuated Salmonella vaccine could be protective in mice, in the setting of concurrent malaria. Surprisingly, mice acutely infected with the nonlethal malaria parasite Plasmodium yoelii 17XNL exhibited a profound loss of protective immunity to NTS, but vaccine-mediated protection was restored after resolution of malaria. Absence of protective immunity during acute malaria correlated with maintenance of antibodies to NTS, but a marked reduction in effector capability of Salmonella-specific CD4 and CD8 T cells. Further, increased expression of the inhibitory molecule PD1 was identified on memory CD4 T cells induced by vaccination. Blockade of IL-10 restored protection against S. Typhimurium, without restoring CD4 T cell effector function. Simultaneous blockade of CTLA-4, LAG3, and PDL1 restored IFN-γ production by vaccine-induced memory CD4 T cells but was not sufficient to restore protection. Together, these data demonstrate that malaria parasite infection induces a temporary loss of an established adaptive immune response via multiple mechanisms, and suggest that in the setting of acute malaria, protection against NTS mediated by live vaccines may be interrupted. In children, malaria is a predisposing factor for invasive bacterial infections with non-typhoidal Salmonella (NTS) serovars, a frequent cause of morbidity and mortality in sub-Saharan Africa. Since development of vaccines against NTS has been proposed as a strategy to protect African children against disseminated NTS infection, we interrogated the effect of malaria on vaccine-induced memory responses to NTS. Our results from a mouse infection model show that infection with malaria parasites temporarily suspends protective immunity conferred by a live, attenuated vaccine and suppresses adaptive immune responses to NTS that are mediated by T cells. These results suggest that in the setting of acute malaria, live attenuated NTS vaccines may lose their effectiveness.
Collapse
Affiliation(s)
- Jason P. Mooney
- Department of Microbiology & Immunology, School of Medicine, University of California Davis, Davis, California, United States of America
| | - Seung-Joo Lee
- Center for Comparative Medicine, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Kristen L. Lokken
- Department of Microbiology & Immunology, School of Medicine, University of California Davis, Davis, California, United States of America
| | - Minelva R. Nanton
- Center for Comparative Medicine, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Sean-Paul Nuccio
- Department of Microbiology & Immunology, School of Medicine, University of California Davis, Davis, California, United States of America
| | - Stephen J. McSorley
- Center for Comparative Medicine, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Renée M. Tsolis
- Department of Microbiology & Immunology, School of Medicine, University of California Davis, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
36
|
Gilchrist JJ, MacLennan CA, Hill AVS. Genetic susceptibility to invasive Salmonella disease. Nat Rev Immunol 2015; 15:452-63. [PMID: 26109132 DOI: 10.1038/nri3858] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Invasive Salmonella disease, in the form of enteric fever and invasive non-typhoidal Salmonella (iNTS) disease, causes substantial morbidity and mortality in children and adults in the developing world. The study of genetic variations in humans and mice that influence susceptibility of the host to Salmonella infection provides important insights into immunity to Salmonella. In this Review, we discuss data that have helped to elucidate the host genetic determinants of human enteric fever and iNTS disease, alongside data from the mouse model of Salmonella infection. Considered together, these studies provide a detailed picture of the immunobiology of human invasive Salmonella disease.
Collapse
Affiliation(s)
- James J Gilchrist
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, University of Oxford, Oxford OX3 7BN, UK
| | - Calman A MacLennan
- 1] Jenner Institute, Nuffield Department of Medicine, Old Road Campus Research Building, Roosevelt Drive, University of Oxford, Oxford, OX3 7DQ, UK. [2] Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Adrian V S Hill
- 1] Wellcome Trust Centre for Human Genetics, Roosevelt Drive, University of Oxford, Oxford OX3 7BN, UK. [2] Jenner Institute, Nuffield Department of Medicine, Old Road Campus Research Building, Roosevelt Drive, University of Oxford, Oxford, OX3 7DQ, UK
| |
Collapse
|
37
|
Monoclonal Antibodies of a Diverse Isotype Induced by an O-Antigen Glycoconjugate Vaccine Mediate In Vitro and In Vivo Killing of African Invasive Nontyphoidal Salmonella. Infect Immun 2015; 83:3722-31. [PMID: 26169269 PMCID: PMC4534659 DOI: 10.1128/iai.00547-15] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 06/30/2015] [Indexed: 12/15/2022] Open
Abstract
Nontyphoidal Salmonella (NTS), particularly Salmonella enterica serovars Typhimurium and Enteritidis, is responsible for a major global burden of invasive disease with high associated case-fatality rates. We recently reported the development of a candidate O-antigen–CRM197 glycoconjugate vaccine against S. Typhimurium. Here, using a panel of mouse monoclonal antibodies generated by the vaccine, we examined the relative efficiency of different antibody isotypes specific for the O:4 antigen of S. Typhimurium to effect in vitro and in vivo killing of the invasive African S. Typhimurium strain D23580. All O:4-specific antibody isotypes could mediate cell-free killing and phagocytosis of S. Typhimurium by mouse blood cells. Opsonization of Salmonella with O:4-specific IgA, IgG1, IgG2a, and IgG2b, but not IgM, resulted in cell-dependent bacterial killing. At high concentrations, O:4-specific antibodies inhibited both cell-free complement-mediated and cell-dependent opsonophagocytic killing of S. Typhimurium in vitro. Using passive immunization in mice, the O:4-specific antibodies provided in vivo functional activity by decreasing the bacterial load in the blood and tissues, with IgG2a and IgG2b being the most effective isotypes. In conclusion, an O-antigen–CRM197 glycoconjugate vaccine can induce O-antigen-specific antibodies of different isotypes that exert in vitro and in vivo killing of S. Typhimurium.
Collapse
|
38
|
Nhampossa T, Mandomando I, Acacio S, Quintó L, Vubil D, Ruiz J, Nhalungo D, Sacoor C, Nhabanga A, Nhacolo A, Aide P, Machevo S, Sigaúque B, Nhama A, Kotloff K, Farag T, Nasrin D, Bassat Q, Macete E, Levine MM, Alonso P. Diarrheal Disease in Rural Mozambique: Burden, Risk Factors and Etiology of Diarrheal Disease among Children Aged 0-59 Months Seeking Care at Health Facilities. PLoS One 2015; 10:e0119824. [PMID: 25973880 PMCID: PMC4431848 DOI: 10.1371/journal.pone.0119824] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 02/03/2015] [Indexed: 11/26/2022] Open
Abstract
Background Diarrheal disease remains a leading cause of illness and death, particularly in low-income countries. Its burden, microbiological causes and risk factors were examined in children aged 0–59 months living in Manhiça, rural southern Mozambique. Methods Trends of diarrhea-related burden of disease were estimated during the period 2001–2012. A prospective, age-stratified and matched (by age, gender and geographical origin), case-control study was conducted during 2007–2011. Clinical, epidemiology, anthropometric measurement and fecal samples obtained from recruited children were used to estimate moderate-to-severe diarrhea (MSD) weighted attributable fractions. Results Over the last decade the incidence of acute diarrhea has dropped by about 80%. Incidence of MSD per 100 child years at risk for the period 2007–2011 was 9.85, 7.73 and 2.10 for children aged 0–11, 12–23 and 24–59 months respectively. By adjusted population attributable fractions, most cases of MSD were due to rotavirus, Cryptosporidium, ETEC ST (ST only or ST/LT), Shigella and Adenovirus 40/41. Washing hands and having facilities to dispose child’s stools were associated with a reduced risk of MSD, while giving stored water to the child was associated with an increased risk of MSD. Conclusions Despite the predominantly decreasing trends observed throughout the last decade, diarrheal diseases remain today a major cause of morbidity among children aged 0–59 months living in this rural Mozambican area. Rotavirus, cryptosporidium, Shigella, ETEC ST and Adenovirus 40/41 were the most important aetiologies of MSD. Thus, well-known preventive strategies such as washing hands, improving the treatment of stored water, having facilities to dispose children stools, and accelerating the introduction of the rotavirus vaccine should be promoted on a wider scale to reduce the current burden of diarrheal diseases.
Collapse
Affiliation(s)
- Tacilta Nhampossa
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Instituto Nacional de Saúde, Ministério de Saúde, Maputo, Mozambique
- * E-mail:
| | - Inacio Mandomando
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Instituto Nacional de Saúde, Ministério de Saúde, Maputo, Mozambique
| | - Sozinho Acacio
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Instituto Nacional de Saúde, Ministério de Saúde, Maputo, Mozambique
| | - Llorenç Quintó
- Barcelona Center for International Health Research (CRESIB, Hospital Clínic-Universitat de Barcelona), Barcelona, Spain
| | - Delfino Vubil
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Joaquin Ruiz
- Barcelona Center for International Health Research (CRESIB, Hospital Clínic-Universitat de Barcelona), Barcelona, Spain
| | - Delino Nhalungo
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Charfudin Sacoor
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Arnaldo Nhabanga
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Ariel Nhacolo
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Instituto Nacional de Saúde, Ministério de Saúde, Maputo, Mozambique
| | - Pedro Aide
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Instituto Nacional de Saúde, Ministério de Saúde, Maputo, Mozambique
| | - Sónia Machevo
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Faculdade de Medicina, Universidade Eduardo Mondlane, Maputo, Mozambique
| | - Betuel Sigaúque
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Instituto Nacional de Saúde, Ministério de Saúde, Maputo, Mozambique
| | - Abel Nhama
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Instituto Nacional de Saúde, Ministério de Saúde, Maputo, Mozambique
| | - Karen Kotloff
- Center for Vaccine Development (CVD), University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Tamer Farag
- Center for Vaccine Development (CVD), University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Dilruba Nasrin
- Center for Vaccine Development (CVD), University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Quique Bassat
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Barcelona Center for International Health Research (CRESIB, Hospital Clínic-Universitat de Barcelona), Barcelona, Spain
| | - Eusebio Macete
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Instituto Nacional de Saúde, Ministério de Saúde, Maputo, Mozambique
| | - Myron M. Levine
- Center for Vaccine Development (CVD), University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Pedro Alonso
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Barcelona Center for International Health Research (CRESIB, Hospital Clínic-Universitat de Barcelona), Barcelona, Spain
| |
Collapse
|
39
|
Boyd MA, Tennant SM, Melendez JH, Toema D, Galen JE, Geddes CD, Levine MM. Adaptation of red blood cell lysis represents a fundamental breakthrough that improves the sensitivity of Salmonella detection in blood. J Appl Microbiol 2015; 118:1199-209. [PMID: 25630831 PMCID: PMC4418380 DOI: 10.1111/jam.12769] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 01/20/2015] [Accepted: 01/22/2015] [Indexed: 11/29/2022]
Abstract
AIMS Isolation of Salmonella Typhi from blood culture is the standard diagnostic for confirming typhoid fever but it is unavailable in many developing countries. We previously described a Microwave Accelerated Metal Enhanced Fluorescence (MAMEF)-based assay to detect Salmonella in medium. Attempts to detect Salmonella in blood were unsuccessful, presumably due to the interference of erythrocytes. The objective of this study was to evaluate various blood treatment methods that could be used prior to PCR, real-time PCR or MAMEF to increase sensitivity of detection of Salmonella. METHODS AND RESULTS We tested ammonium chloride and erythrocyte lysis buffer, water, Lymphocyte Separation Medium, BD Vacutainer(®) CPT(™) Tubes and dextran. Erythrocyte lysis buffer was the best isolation method as it is fast, inexpensive and works with either fresh or stored blood. The sensitivity of PCR- and real-time PCR detection of Salmonella in spiked blood was improved when whole blood was first lysed using erythrocyte lysis buffer prior to DNA extraction. Removal of erythrocytes and clotting factors also enabled reproducible lysis of Salmonella and fragmentation of DNA, which are necessary for MAMEF sensing. CONCLUSIONS Use of the erythrocyte lysis procedure prior to DNA extraction has enabled improved sensitivity of Salmonella detection by PCR and real-time PCR and has allowed lysis and fragmentation of Salmonella using microwave radiation (for future detection by MAMEF). SIGNIFICANCE AND IMPACT OF THE STUDY Adaptation of the blood lysis method represents a fundamental breakthrough that improves the sensitivity of DNA-based detection of Salmonella in blood.
Collapse
Affiliation(s)
- M A Boyd
- Center for Vaccine Development, University of Maryland Baltimore, Baltimore, MD, USA; Department of Pediatrics, University of Maryland Baltimore, Baltimore, MD, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Onsare RS, Micoli F, Lanzilao L, Alfini R, Okoro CK, Muigai AW, Revathi G, Saul A, Kariuki S, MacLennan CA, Rondini S. Relationship between antibody susceptibility and lipopolysaccharide O-antigen characteristics of invasive and gastrointestinal nontyphoidal Salmonellae isolates from Kenya. PLoS Negl Trop Dis 2015; 9:e0003573. [PMID: 25739091 PMCID: PMC4352093 DOI: 10.1371/journal.pntd.0003573] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 01/28/2015] [Indexed: 01/01/2023] Open
Abstract
Background Nontyphoidal Salmonellae (NTS) cause a large burden of invasive and gastrointestinal disease among young children in sub-Saharan Africa. No vaccine is currently available. Previous reports indicate the importance of the O-antigen of Salmonella lipopolysaccharide for virulence and resistance to antibody-mediated killing. We hypothesised that isolates with more O-antigen have increased resistance to antibody-mediated killing and are more likely to be invasive than gastrointestinal. Methodology/Principal Findings We studied 192 NTS isolates (114 Typhimurium, 78 Enteritidis) from blood and stools, mostly from paediatric admissions in Kenya 2000–2011. Isolates were tested for susceptibility to antibody-mediated killing, using whole adult serum. O-antigen structural characteristics, including O-acetylation and glucosylation, were investigated. Overall, isolates were susceptible to antibody-mediated killing, but S. Enteritidis were less susceptible and expressed more O-antigen than Typhimurium (p<0.0001 for both comparisons). For S. Typhimurium, but not Enteritidis, O-antigen expression correlated with reduced sensitivity to killing (r = 0.29, 95% CI = 0.10-0.45, p = 0.002). Both serovars expressed O-antigen populations ranging 21–33 kDa average molecular weight. O-antigen from most Typhimurium were O-acetylated on rhamnose and abequose residues, while Enteritidis O-antigen had low or no O-acetylation. Both Typhimurium and Enteritidis O-antigen were approximately 20%–50% glucosylated. Amount of S. Typhimurium O-antigen and O-antigen glucosylation level were inversely related. There was no clear association between clinical presentation and antibody susceptibility, O-antigen level or other O-antigen features. Conclusion/Significance Kenyan S. Typhimurium and Enteritidis clinical isolates are susceptible to antibody-mediated killing, with degree of susceptibility varying with level of O-antigen for S. Typhimurium. This supports the development of an antibody-inducing vaccine against NTS for Africa. No clear differences were found in the phenotype of isolates from blood and stool, suggesting that the same isolates can cause invasive disease and gastroenteritis. Genome studies are required to understand whether invasive and gastrointestinal isolates differ at the genotypic level. Nontyphoidal Salmonellae (NTS) are an emerging major cause of invasive bacterial disease in African children aged less than 5 years and immunocompromised adults, with an estimated case fatality rate of 20–25%. NTS also cause diarrhoea, a killer of about 1.5 million young children annually, mainly in low- and middle-income countries. No vaccine against NTS is available, but improved understanding of the Salmonella bacteria that cause disease in Africa would help the development of new vaccines. The authors characterized a collection of 192 Kenyan NTS strains (114 S. Typhimurium and 78 S. Enteritidis) from blood and stool specimens. All strains could be killed to differing extents by antibodies present in the blood of healthy HIV-uninfected African adults, supporting the development of a vaccine that will induce protective antibodies when given to African children. Differences in killing by antibody were partly related to the amount of O-antigen on the bacterial surface. There were no clear distinction between stains causing invasive disease and diarrhoea, suggesting that the same strains may be capable of causing both forms of disease. Clarification of this will require genomic analysis.
Collapse
Affiliation(s)
- Robert S. Onsare
- Centre for Microbiology Research (CMR), Kenya Medical Research Institute (KEMRI), Nairobi, Kenya
- Jomo Kenyatta University of Agriculture and Technology (JKUAT), Nairobi, Kenya
| | - Francesca Micoli
- Novartis Vaccines Institute for Global Health (NVGH), Siena, Italy
| | - Luisa Lanzilao
- Novartis Vaccines Institute for Global Health (NVGH), Siena, Italy
| | - Renzo Alfini
- Novartis Vaccines Institute for Global Health (NVGH), Siena, Italy
| | - Chinyere K. Okoro
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Anne W. Muigai
- Jomo Kenyatta University of Agriculture and Technology (JKUAT), Nairobi, Kenya
| | - Gunturu Revathi
- Division of Microbiology, Department of Pathology, Aga Khan University Hospital, Nairobi, Kenya
| | - Allan Saul
- Novartis Vaccines Institute for Global Health (NVGH), Siena, Italy
| | - Samuel Kariuki
- Centre for Microbiology Research (CMR), Kenya Medical Research Institute (KEMRI), Nairobi, Kenya
| | | | - Simona Rondini
- Novartis Vaccines Institute for Global Health (NVGH), Siena, Italy
- * E-mail:
| |
Collapse
|
41
|
Matsui H, Fukiya S, Kodama-Akaboshi C, Eguchi M, Yamamoto T. Mouse models for assessing the cross-protective efficacy of oral non-typhoidal Salmonella vaccine candidates harbouring in-frame deletions of the ATP-dependent protease lon and other genes. J Med Microbiol 2015; 64:295-302. [PMID: 25589672 DOI: 10.1099/jmm.0.000014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In BALB/c mouse models of Salmonella enterica serovar Typhimurium infection, a single oral immunization with a mutant strain with an insertion of the chloramphenicol resistance gene into the ATP-dependent protease clpP or lon gene decreased the number of salmonellae in each tissue sample 5 days after oral challenge with virulent S. Typhimurium at weeks 26 and 54 post-immunization. These data suggested that an oral immunization with the ClpP- or Lon-disrupted S. Typhimurium strain could provide long-term protection against oral challenge with virulent S. Typhimurium. Accordingly, recombinant oral non-typhoidal Salmonella (NTS) vaccines were constructed by incorporating mutants of both S. Typhimurium and S. enterica serovar Enteritidis harbouring stable in-frame markerless deletions of the clpP-lon-sulA (suppressor of lon), lon-sulA or lon-msbB (acyltransferase) genes. Amongst these orally administered vaccine candidates, those with the lon-sulA gene deletion mutants of S. Typhimurium and S. Enteritidis protected BALB/c and C57BL/6J mice against oral challenge with both virulent S. Typhimurium and virulent S. Enteritidis. Therefore, the in-frame markerless lon-sulA gene deletion mutant of S. Typhimurium or S. Enteritidis could be a promising cross-protective NTS live vaccine candidate for practical use in humans.
Collapse
Affiliation(s)
- Hidenori Matsui
- Kitasato Institute for Life Sciences and Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Satoru Fukiya
- Kitasato Institute for Life Sciences and Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Chie Kodama-Akaboshi
- Kitasato Institute for Life Sciences and Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Masahiro Eguchi
- National Institute of Animal Health, National Agriculture and Food Research Organization, 3-1-5 Kannondai, Tsukuba, Ibaraki 305-0856, Japan
| | - Tomoko Yamamoto
- Graduate School of Pharmaceutical Sciences, Chiba University, Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| |
Collapse
|
42
|
Pakkanen SH, Kantele JM, Savolainen LE, Rombo L, Kantele A. Specific and cross-reactive immune response to oral Salmonella Typhi Ty21a and parenteral Vi capsular polysaccharide typhoid vaccines administered concomitantly. Vaccine 2015; 33:451-8. [DOI: 10.1016/j.vaccine.2014.11.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 11/05/2014] [Accepted: 11/17/2014] [Indexed: 01/02/2023]
|
43
|
MacLennan CA. Antibodies and protection against invasive salmonella disease. Front Immunol 2014; 5:635. [PMID: 25566248 PMCID: PMC4273658 DOI: 10.3389/fimmu.2014.00635] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 11/27/2014] [Indexed: 11/29/2022] Open
Affiliation(s)
- Calman A MacLennan
- Novartis Vaccines Institute for Global Health , Siena , Italy ; School of Immunity and Infection, College of Medicine and Dental Sciences, University of Birmingham , Birmingham , UK
| |
Collapse
|
44
|
Stefanetti G, Rondini S, Lanzilao L, Saul A, MacLennan CA, Micoli F. Impact of conjugation chemistry on the immunogenicity of S. Typhimurium conjugate vaccines. Vaccine 2014; 32:6122-9. [PMID: 25192974 DOI: 10.1016/j.vaccine.2014.08.056] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 08/12/2014] [Accepted: 08/27/2014] [Indexed: 10/24/2022]
Abstract
Salmonella Typhimurium is major cause of invasive nontyphoidal Salmonella disease in Africa. Conjugation of S. Typhimurium O-antigen to an appropriate carrier protein constitutes a possible strategy for the development of a vaccine against this disease, for which no vaccines are currently available. The conjugation chemistry used is one of the parameters that can affect the immunogenicity of glycoconjugate vaccines. Herein different glycoconjugates were synthesized to investigate the impact of this variable on the immunogenicity of S. Typhimurium conjugate vaccines in mice, all with CRM₁₉₇ as carrier protein. Random derivatization along the O-antigen chain was compared with site-directed activation of the terminal KDO sugar residue of the core oligosaccharide. In particular, two different random approaches were used, based on the oxidation of the polysaccharide, which differently impact the structure and conformation of the O-antigen chain. For the selective conjugation methods, linkers of two different lengths were compared. When tested in mice, all conjugates induced anti-O-antigen IgG antibodies with serum bactericidal activity. Similar anti-O-antigen antibody levels were elicited independent of the chemistry used and a higher degree of saccharide derivatization did not impact negatively on the anti-O-antigen IgG response. Bactericidal activity of serum antibodies induced by selective conjugates was similar independent of the length of the spacer used. Random conjugates elicited antibodies with greater bactericidal activity than selective ones, and an inverse correlation was found between degree of O-antigen modification and antibody functional activity.
Collapse
Affiliation(s)
- G Stefanetti
- Novartis Vaccines Institute for Global Health, Via Fiorentina 1, 53100 Siena, Italy
| | - S Rondini
- Novartis Vaccines Institute for Global Health, Via Fiorentina 1, 53100 Siena, Italy
| | - L Lanzilao
- Novartis Vaccines Institute for Global Health, Via Fiorentina 1, 53100 Siena, Italy
| | - A Saul
- Novartis Vaccines Institute for Global Health, Via Fiorentina 1, 53100 Siena, Italy
| | - C A MacLennan
- Novartis Vaccines Institute for Global Health, Via Fiorentina 1, 53100 Siena, Italy
| | - F Micoli
- Novartis Vaccines Institute for Global Health, Via Fiorentina 1, 53100 Siena, Italy.
| |
Collapse
|
45
|
Abstract
With the 2010s declared the Decade of Vaccines, and Millennium Development Goals 4 and 5 focused on reducing diseases that are potentially vaccine preventable, now is an exciting time for vaccines against poverty, that is, vaccines against diseases that disproportionately affect low- and middle-income countries (LMICs). The Global Burden of Disease Study 2010 has helped better understand which vaccines are most needed. In 2012, US$1.3 billion was spent on research and development for new vaccines for neglected infectious diseases. However, the majority of this went to three diseases: HIV/AIDS, malaria, and tuberculosis, and not neglected diseases. Much of it went to basic research rather than development, with an ongoing decline in funding for product development partnerships. Further investment in vaccines against diarrheal diseases, hepatitis C, and group A Streptococcus could lead to a major health impact in LMICs, along with vaccines to prevent sepsis, particularly among mothers and neonates. The Advanced Market Commitment strategy of the Global Alliance for Vaccines and Immunisation (GAVI) Alliance is helping to implement vaccines against rotavirus and pneumococcus in LMICs, and the roll out of the MenAfriVac meningococcal A vaccine in the African Meningitis Belt represents a paradigm shift in vaccines against poverty: the development of a vaccine primarily targeted at LMICs. Global health vaccine institutes and increasing capacity of vaccine manufacturers in emerging economies are helping drive forward new vaccines for LMICs. Above all, partnership is needed between those developing and manufacturing LMIC vaccines and the scientists, health care professionals, and policy makers in LMICs where such vaccines will be implemented.
Collapse
|
46
|
Bumann D. Identification of Protective Antigens for Vaccination against Systemic Salmonellosis. Front Immunol 2014; 5:381. [PMID: 25157252 PMCID: PMC4127814 DOI: 10.3389/fimmu.2014.00381] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 07/25/2014] [Indexed: 12/21/2022] Open
Abstract
There is an urgent medical need for improved vaccines with broad serovar coverage and high efficacy against systemic salmonellosis. Subunit vaccines offer excellent safety profiles but require identification of protective antigens, which remains a challenging task. Here, I review crucial properties of Salmonella antigens that might help to narrow down the number of potential candidates from more than 4000 proteins encoded in Salmonella genomes, to a more manageable number of 50–200 most promising antigens. I also discuss complementary approaches for antigen identification and potential limitations of current pre-clinical vaccine testing.
Collapse
Affiliation(s)
- Dirk Bumann
- Focal Area Infection Biology, Biozentrum, University of Basel , Basel , Switzerland
| |
Collapse
|
47
|
Gal-Mor O, Boyle EC, Grassl GA. Same species, different diseases: how and why typhoidal and non-typhoidal Salmonella enterica serovars differ. Front Microbiol 2014; 5:391. [PMID: 25136336 PMCID: PMC4120697 DOI: 10.3389/fmicb.2014.00391] [Citation(s) in RCA: 272] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 07/12/2014] [Indexed: 12/22/2022] Open
Abstract
Human infections by the bacterial pathogen Salmonella enterica represent major disease burdens worldwide. This highly ubiquitous species consists of more than 2600 different serovars that can be divided into typhoidal and non-typhoidal Salmonella (NTS) serovars. Despite their genetic similarity, these two groups elicit very different diseases and distinct immune responses in humans. Comparative analyses of the genomes of multiple Salmonella serovars have begun to explain the basis of the variation in disease manifestations. Recent advances in modeling both enteric fever and intestinal gastroenteritis in mice will facilitate investigation into both the bacterial- and host-mediated mechanisms involved in salmonelloses. Understanding the genetic and molecular mechanisms responsible for differences in disease outcome will augment our understanding of Salmonella pathogenesis, host immunity, and the molecular basis of host specificity. This review outlines the differences in epidemiology, clinical manifestations, and the human immune response to typhoidal and NTS infections and summarizes the current thinking on why these differences might exist.
Collapse
Affiliation(s)
- Ohad Gal-Mor
- The Infectious Diseases Research Laboratory, Sheba Medical Center Tel-Hashomer, Israel
| | - Erin C Boyle
- Bernhard Nocht Institute for Tropical Medicine Hamburg, Germany
| | - Guntram A Grassl
- Institute for Experimental Medicine, Christian Albrechts University Kiel Kiel, Germany ; Research Center Borstel Borstel, Germany
| |
Collapse
|
48
|
Workalemahu G, Wang H, Puan KJ, Nada MH, Kuzuyama T, Jones BD, Jin C, Morita CT. Metabolic engineering of Salmonella vaccine bacteria to boost human Vγ2Vδ2 T cell immunity. THE JOURNAL OF IMMUNOLOGY 2014; 193:708-21. [PMID: 24943221 DOI: 10.4049/jimmunol.1302746] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Human Vγ2Vδ2 T cells monitor isoprenoid metabolism by recognizing foreign (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (HMBPP), a metabolite in the 2-C-methyl-D-erythritol-4-phosphate pathway used by most eubacteria and apicomplexan parasites, and self isopentenyl pyrophosphate, a metabolite in the mevalonate pathway used by humans. Whereas microbial infections elicit prolonged expansion of memory Vγ2Vδ2 T cells, immunization with prenyl pyrophosphates or aminobisphosphonates elicit short-term Vγ2Vδ2 expansion with rapid anergy and deletion upon subsequent immunizations. We hypothesized that a live, attenuated bacterial vaccine that overproduces HMBPP would elicit long-lasting Vγ2Vδ2 T cell immunity by mimicking a natural infection. Therefore, we metabolically engineered the avirulent aroA(-) Salmonella enterica serovar Typhimurium SL7207 strain by deleting the gene for LytB (the downstream enzyme from HMBPP) and functionally complementing for this loss with genes encoding mevalonate pathway enzymes. LytB(-) Salmonella SL7207 had high HMBPP levels, infected human cells as efficiently as did the wild-type bacteria, and stimulated large ex vivo expansions of Vγ2Vδ2 T cells from human donors. Importantly, vaccination of a rhesus monkey with live lytB(-) Salmonella SL7207 stimulated a prolonged expansion of Vγ2Vδ2 T cells without significant side effects or anergy induction. These studies provide proof-of-principle that metabolic engineering can be used to derive live bacterial vaccines that boost Vγ2Vδ2 T cell immunity. Similar engineering of metabolic pathways to produce lipid Ags or B vitamin metabolite Ags could be used to derive live bacterial vaccine for other unconventional T cells that recognize nonpeptide Ags.
Collapse
Affiliation(s)
- Grefachew Workalemahu
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242; Department of Veterans Affairs, Iowa City Health Care System, Iowa City, IA 52246
| | - Hong Wang
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242; Department of Veterans Affairs, Iowa City Health Care System, Iowa City, IA 52246
| | - Kia-Joo Puan
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242; Singapore Immunology Network, Agency for Science, Technology and Research, Singapore 138648
| | - Mohanad H Nada
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242; Department of Veterans Affairs, Iowa City Health Care System, Iowa City, IA 52246; Interdisciplinary Graduate Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242
| | - Tomohisa Kuzuyama
- Biotechnology Research Center, The University of Tokyo, Tokyo 113-8657, Japan
| | - Bradley D Jones
- Department of Microbiology, University of Iowa Carver College of Medicine, Iowa City, IA 52242; Interdisciplinary Graduate Program in Genetics, University of Iowa Carver College of Medicine, Iowa City, IA 52242; and Inflammation Program, University of Iowa Carver College of Medicine, Iowa City, IA 52242
| | - Chenggang Jin
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242; Department of Veterans Affairs, Iowa City Health Care System, Iowa City, IA 52246
| | - Craig T Morita
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242; Department of Veterans Affairs, Iowa City Health Care System, Iowa City, IA 52246; Interdisciplinary Graduate Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242;
| |
Collapse
|
49
|
MacLennan CA, Martin LB, Micoli F. Vaccines against invasive Salmonella disease: current status and future directions. Hum Vaccin Immunother 2014; 10:1478-93. [PMID: 24804797 PMCID: PMC4185946 DOI: 10.4161/hv.29054] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Though primarily enteric pathogens, Salmonellae are responsible for a considerable yet under-appreciated global burden of invasive disease. In South and South-East Asia, this manifests as enteric fever caused by serovars Typhi and Paratyphi A. In sub-Saharan Africa, a similar disease burden results from invasive nontyphoidal Salmonellae, principally serovars Typhimurium and Enteritidis. The existing Ty21a live-attenuated and Vi capsular polysaccharide vaccines target S. Typhi and are not effective in young children where the burden of invasive Salmonella disease is highest. After years of lack of investment in new Salmonella vaccines, recent times have seen increased interest in the area led by emerging-market manufacturers, global health vaccine institutes and academic partners. New glycoconjugate vaccines against S. Typhi are becoming available with similar vaccines against other invasive serovars in development. With other new vaccines under investigation, including live-attenuated, protein-based and GMMA vaccines, now is an exciting time for the Salmonella vaccine field.
Collapse
Affiliation(s)
- Calman A MacLennan
- Novartis Vaccines Institute for Global Health; Siena, Italy; Medical Research Council Centre for Immune Regulation and Clinical Immunology Service; Institute of Biomedical Research, School of Immunity and Infection; College of Medicine and Dental Sciences; University of Birmingham; Birmingham, UK
| | - Laura B Martin
- Novartis Vaccines Institute for Global Health; Siena, Italy
| | | |
Collapse
|
50
|
Maclennan CA. Out of Africa: links between invasive nontyphoidal Salmonella disease, typhoid fever, and malaria. Clin Infect Dis 2013; 58:648-50. [PMID: 24336912 PMCID: PMC3922216 DOI: 10.1093/cid/cit803] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|