1
|
Salerno-Goncalves R, Chen H, Bafford AC, Sztein MB. Epigenetic regulation in epithelial cells and innate lymphocyte responses to S. Typhi infection: insights into IFN-γ production and intestinal immunity. Front Immunol 2024; 15:1448717. [PMID: 39372404 PMCID: PMC11450450 DOI: 10.3389/fimmu.2024.1448717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/08/2024] [Indexed: 10/08/2024] Open
Abstract
Infection by Salmonella enterica serovar Typhi (S. Typhi), the cause of enteric fevers, is low in high-income countries but persistent in low- and middle-income countries, resulting in 65,400-187,700 deaths yearly. Drug resistance, including in the United States, exacerbates this issue. Evidence indicates that innate lymphocytes (INLs), such as natural killer (NK) cells, and unconventional T lymphocytes (e.g., Mucosal-associated invariant T (MAIT) cells and T-cell receptor gamma delta (TCR-γδ) cells) can impact the intestinal epithelial barrier, the primary site of exposure to S. Typhi. Moreover, INL production of IFN-γ is central in controlling S. Typhi infection. However, the impact of epithelial cells (EC) on the secretion of IFN-γ by INLs and the relationship between these events and epigenetic changes remains unknown. Epigenetic modifications in host cells are fundamental for their differentiation and function, including IFN-γ production. Herein, using a human organoid-derived polarized intestinal epithelial cell monolayer, we investigated the role of H3K4me3 and H3K27me3 epigenetic marks in intestinal immunity, focusing on the function of EC, NK, MAIT, and TCR-γδ cells in response to S. Typhi. This study builds on our previous findings that MAIT subsets exhibiting specific IFN-γ pattern signatures were associated with protection against typhoid fever and that S. Typhi infection regulates changes in chromatin marks that depend on individual cell subsets. Here, we show that cultures exposed to S. Typhi without EC exhibit a significant increase in NK and MAIT cells, and, to a lesser extent, TCR-γδ cells, expressing IFN-γ and H3K4me3 but not H3K27me3 marks, contrasting with cultures where EC is present. The influence of EC on INL H3K4me3 marks might be indirectly mediated through the modulation of IL-18 secretion via the Histone Deacetylase 6 gene during S. Typhi infection.
Collapse
Affiliation(s)
- Rosângela Salerno-Goncalves
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Haiyan Chen
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Andrea C. Bafford
- Division of General and Oncologic Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Marcelo B. Sztein
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Program in Oncology, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| |
Collapse
|
2
|
Shaw C, Weimer BC, Gann R, Desai PT, Shah JD. The Yin and Yang of pathogens and probiotics: interplay between Salmonella enterica sv. Typhimurium and Bifidobacterium infantis during co-infection. Front Microbiol 2024; 15:1387498. [PMID: 38812689 PMCID: PMC11133690 DOI: 10.3389/fmicb.2024.1387498] [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: 02/17/2024] [Accepted: 04/12/2024] [Indexed: 05/31/2024] Open
Abstract
Probiotic bacteria have been proposed as an alternative to antibiotics for the control of antimicrobial resistant enteric pathogens. The mechanistic details of this approach remain unclear, in part because pathogen reduction appears to be both strain and ecology dependent. Here we tested the ability of five probiotic strains, including some from common probiotic genera Lactobacillus and Bifidobacterium, to reduce binding of Salmonella enterica sv. Typhimurium to epithelial cells in vitro. Bifidobacterium longum subsp. infantis emerged as a promising strain; however, S. Typhimurium infection outcome in epithelial cells was dependent on inoculation order, with B. infantis unable to rescue host cells from preceding or concurrent infection. We further investigated the complex mechanisms underlying this interaction between B. infantis, S. Typhimurium, and epithelial cells using a multi-omics approach that included gene expression and altered metabolism via metabolomics. Incubation with B. infantis repressed apoptotic pathways and induced anti-inflammatory cascades in epithelial cells. In contrast, co-incubation with B. infantis increased in S. Typhimurium the expression of virulence factors, induced anaerobic metabolism, and repressed components of arginine metabolism as well as altering the metabolic profile. Concurrent application of the probiotic and pathogen notably generated metabolic profiles more similar to that of the probiotic alone than to the pathogen, indicating a central role for metabolism in modulating probiotic-pathogen-host interactions. Together these data imply crosstalk via small molecules between the epithelial cells, pathogen and probiotic that consistently demonstrated unique molecular mechanisms specific probiotic/pathogen the individual associations.
Collapse
Affiliation(s)
| | - Bart C. Weimer
- Department of Population Health and Reproduction, School of Veterinary Medicine, 100K Pathogen Genome Project, University of California, Davis, Davis, CA, United States
| | | | | | | |
Collapse
|
3
|
Salerno-Goncalves R, Chen H, Bafford AC, Izquierdo M, Hormazábal JC, Lagos R, Tettelin H, D’Mello A, Booth JS, Fasano A, Levine MM, Sztein MB. Early host immune responses in a human organoid-derived gallbladder monolayer to Salmonella Typhi strains from patients with acute and chronic infections: a comparative analysis. Front Immunol 2024; 15:1334762. [PMID: 38533492 PMCID: PMC10963533 DOI: 10.3389/fimmu.2024.1334762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/26/2024] [Indexed: 03/28/2024] Open
Abstract
Salmonella enterica serovar Typhi (S. Typhi), a human-restricted pathogen, invades the host through the gut to cause typhoid fever. Recent calculations of the typhoid fever burden estimated that more than 10 million new typhoid fever cases occur in low and middle-income countries, resulting in 65,400-187,700 deaths yearly. Interestingly, if not antibiotic-treated, upon the resolution of acute disease, 1%-5% of patients become asymptomatic chronic carriers. Chronically infected hosts are not only critical reservoirs of infection that transmit the disease to naive individuals but are also predisposed to developing gallbladder carcinoma. Nevertheless, the molecular mechanisms involved in the early interactions between gallbladder epithelial cells and S. Typhi remain largely unknown. Based on our previous studies showing that closely related S. Typhi strains elicit distinct innate immune responses, we hypothesized that host molecular pathways activated by S. Typhi strains derived from acutely and chronically infected patients would differ. To test this hypothesis, we used a novel human organoid-derived polarized gallbladder monolayer model, and S. Typhi strains derived from acutely and chronically infected patients. We found that S. Typhi strains derived from acutely and chronically infected patients differentially regulate host mitogen-activated protein kinase (MAPK) and S6 transcription factors. These variations might be attributed to differential cytokine signaling, predominantly via TNF-α and IL-6 production and appear to be influenced by the duration the isolate was subjected to selective pressures in the gallbladder. These findings represent a significant leap in understanding the complexities behind chronic S. Typhi infections in the gallbladder and may uncover potential intervention targets.
Collapse
Affiliation(s)
- Rosângela Salerno-Goncalves
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Haiyan Chen
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Andrea C. Bafford
- Division of General and Oncologic Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Mariana Izquierdo
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Juan Carlos Hormazábal
- Seccion Bacteriologia, Subdepartamento de Enfermedades Infecciosas, Departamento de Laboratorio Biomédico, Instituto de Salud Pública de Chile (ISP), Santiago, Chile
| | - Rosanna Lagos
- Seccion Bacteriologia, Subdepartamento de Enfermedades Infecciosas, Departamento de Laboratorio Biomédico, Instituto de Salud Pública de Chile (ISP), Santiago, Chile
| | - Hervé Tettelin
- Department of Microbiology and Immunology and Institute for Genome Sciences (IGS), University of Maryland School of Medicine, Baltimore, MD, United States
| | - Adonis D’Mello
- Department of Microbiology and Immunology and Institute for Genome Sciences (IGS), University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jayaum S. Booth
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Alessio Fasano
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA, United States
| | - Myron M. Levine
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Marcelo B. Sztein
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Program in Oncology, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| |
Collapse
|
4
|
Barton A, Hill J, O'Connor D, Jones C, Jones E, Camara S, Shrestha S, Jin C, Gibani MM, Dobinson HC, Waddington C, Darton TC, Blohmke CJ, Pollard AJ. Early transcriptional responses to human enteric fever challenge. Infect Immun 2023; 91:e0010823. [PMID: 37725060 PMCID: PMC10581002 DOI: 10.1128/iai.00108-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/29/2023] [Indexed: 09/21/2023] Open
Abstract
Enteric fever, caused by oral infection with typhoidal Salmonella serovars, presents as a non-specific febrile illness preceded by an incubation period of 5 days or more. The enteric fever human challenge model provides a unique opportunity to investigate the innate immune response during this incubation period, and how this response is altered by vaccination with the Vi polysaccharide or conjugate vaccine. We find that on the same day as ingestion of typhoidal Salmonella, there is already evidence of an immune response, with 199 genes upregulated in the peripheral blood transcriptome 12 hours post-challenge (false discovery rate <0.05). Gene sets relating to neutrophils, monocytes, and innate immunity were over-represented (false discovery rate <0.05). Estimating cell proportions from gene expression data suggested a possible increase in activated monocytes 12 hours post-challenge (P = 0.036, paired Wilcoxon signed-rank test). Furthermore, plasma TNF-α rose following exposure (P = 0.011, paired Wilcoxon signed-rank test). There were no significant differences in gene expression (false discovery rate <0.05) in the 12 hours response between those who did and did not subsequently develop clinical or blood culture confirmed enteric fever or between vaccination groups. Together, these results demonstrate early perturbation of the peripheral blood transcriptome after enteric fever challenge and provide initial insight into early mechanisms of protection.
Collapse
Affiliation(s)
- Amber Barton
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Jennifer Hill
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Daniel O'Connor
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Claire Jones
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Elizabeth Jones
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Susana Camara
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Sonu Shrestha
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Celina Jin
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
- Department of Pathology, Royal Melbourne Hospital, Melbourne, Australia
- Infectious Diseases and Immune Defence Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Malick M. Gibani
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
- Department of Infectious Disease, Imperial College, London, United Kingdom
| | - Hazel C. Dobinson
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Claire Waddington
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
- Department of Infectious Disease, Imperial College, London, United Kingdom
| | - Thomas C. Darton
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease and The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, United Kingdom
| | - Christoph J. Blohmke
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Andrew J. Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| |
Collapse
|
5
|
Mahdally SM, Izquierdo M, Viscardi RM, Magder LS, Crowley HM, Bafford AC, Drachenberg CB, Farfan MJ, Fasano A, Sztein MB, Salerno-Goncalves R. Secretory-IgA binding to intestinal microbiota attenuates inflammatory reactions as the intestinal barrier of preterm infants matures. Clin Exp Immunol 2023; 213:339-356. [PMID: 37070830 PMCID: PMC10570995 DOI: 10.1093/cei/uxad042] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/09/2023] [Accepted: 04/11/2023] [Indexed: 04/19/2023] Open
Abstract
Previous work has shown that Secretory-IgA (SIgA) binding to the intestinal microbiota is variable and may regulate host inflammatory bowel responses. Nevertheless, the impact of the SIgA functional binding to the microbiota remains largely unknown in preterm infants whose immature epithelial barriers make them particularly susceptible to inflammation. Here, we investigated SIgA binding to intestinal microbiota isolated from stools of preterm infants <33 weeks gestation with various levels of intestinal permeability. We found that SIgA binding to intestinal microbiota attenuates inflammatory reactions in preterm infants. We also observed a significant correlation between SIgA affinity to the microbiota and the infant's intestinal barrier maturation. Still, SIgA affinity was not associated with developing host defenses, such as the production of mucus and inflammatory calprotectin protein, but it depended on the microbiota shifts as the intestinal barrier matures. In conclusion, we reported an association between the SIgA functional binding to the microbiota and the maturity of the preterm infant's intestinal barrier, indicating that the pattern of SIgA coating is altered as the intestinal barrier matures.
Collapse
Affiliation(s)
- Sarah M Mahdally
- Division of Neonatology, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mariana Izquierdo
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rose M Viscardi
- Division of Neonatology, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Laurence S Magder
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Helena M Crowley
- Division of Pediatric Surgery and Urology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Andrea C Bafford
- Division of General and Oncologic Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Cinthia B Drachenberg
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mauricio J Farfan
- Departamento de Pediatría y Cirugía Infantil, Facultad de Medicina, Hospital Dr. Luis Calvo Mackenna, Universidad de Chile, Santiago, Chile
| | - Alessio Fasano
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA, USA
| | - Marcelo B Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rosangela Salerno-Goncalves
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
6
|
Salerno-Gonçalves R, Fresnay S, Magder L, Darton TC, Waddington CS, Blohmke CJ, Angus B, Levine MM, Pollard AJ, Sztein MB. Mucosal-Associated Invariant T cells exhibit distinct functional signatures associated with protection against typhoid fever. Cell Immunol 2022; 378:104572. [PMID: 35772315 PMCID: PMC9377420 DOI: 10.1016/j.cellimm.2022.104572] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 11/26/2022]
Abstract
First demonstration of cytokine-secreting MAIT cell kinetics after human challenge with Salmonella enterica serovar Typhi. MAIT cell's functional signatures and association with typhoid fever protection. Predictive value of MAIT cell cytokine pattern. Lack of association between the number of cytokines expressed by MAIT cells and prevention against typhoid fever.
We have previously demonstrated that Mucosal-Associated Invariant T (MAIT) cells secrete multiple cytokines after exposure to Salmonella enterica serovar Typhi (S. Typhi), the causative agent of typhoid fever in humans. However, whether cytokine secreting MAIT cells can enhance or attenuate the clinical severity of bacterial infections remain debatable. This study characterizes human MAIT cell functions in subjects participating in a wild-type S. Typhi human challenge model. Here, we found that MAIT cells exhibit distinct functional signatures associated with protection against typhoid fever. We also observed that the cytokine patterns of MAIT cell responses, rather than the average number of cytokines expressed, are more predictive of typhoid fever outcomes. These results might enable us to objectively, based on functional parameters, identify cytokine patterns that may serve as predictive biomarkers during natural infection and vaccination.
Collapse
|
7
|
Wang H, Xie Z, Yang F, Wang Y, Jiang H, Huang X, Zhang Y. Salmonella enterica serovar Typhi influences inflammation and autophagy in macrophages. Braz J Microbiol 2022; 53:525-534. [PMID: 35274232 PMCID: PMC9151981 DOI: 10.1007/s42770-022-00719-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 02/01/2022] [Indexed: 02/01/2023] Open
Abstract
Salmonella enterica serovar Typhi (S. Typhi) is a human enteropathogen that can survive in macrophages and cause systemic infection. Autophagy and inflammation are two important immune responses of macrophages that contribute to the elimination of pathogens. However, Salmonella has derived many strategies to evade inflammation and autophagy. This study investigated inflammation-related NF-κB signaling pathways and autophagy in S. Typhi-infected macrophages. RNA-seq and quantitative real-time PCR indicated that mRNA levels of NF-κB signaling pathway and autophagy-related genes were dynamically influenced in S. Typhi-infected macrophages. Western blots revealed that S. Typhi activated the NF-κB signaling pathway and induced the expression of inhibitor protein IκBζ. In addition, S. Typhi enhanced autophagy during early stages of infection and may inhibit autophagy during late stages of infection. Thus, we propose that S. Typhi can influence the NF-κB signaling pathway and autophagy in macrophages.
Collapse
Affiliation(s)
- Huiyun Wang
- JiangYin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, 214400, Jiangsu, China
| | - Zhongyi Xie
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Fanfan Yang
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Yurou Wang
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Haiqiang Jiang
- JiangYin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, 214400, Jiangsu, China
| | - Xinxiang Huang
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Ying Zhang
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
8
|
Salerno-Gonçalves R, Rezwan T, Luo D, Tettelin H, Sztein MB. B Cells Control Mucosal-Associated Invariant T Cell Responses to Salmonella enterica Serovar Typhi Infection Through the CD85j HLA-G Receptor. Front Immunol 2021; 12:728685. [PMID: 34659215 PMCID: PMC8517411 DOI: 10.3389/fimmu.2021.728685] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/16/2021] [Indexed: 11/13/2022] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are an innate-like population of T cells that display a TCR Vα7.2+ CD161+ phenotype and are restricted by the nonclassical MHC-related molecule 1 (MR1). Although B cells control MAIT cell development and function, little is known about the mechanisms underlying their interaction(s). Here, we report, for the first time, that during Salmonella enterica serovar Typhi (S. Typhi) infection, HLA-G expression on B cells downregulates IFN-γ production by MAIT cells. In contrast, blocking HLA-G expression on S. Typhi-infected B cells increases IFN-γ production by MAIT cells. After interacting with MAIT cells, kinetic studies show that B cells upregulate HLA-G expression and downregulate the inhibitory HLA-G receptor CD85j on MAIT cells resulting in their loss. These results provide a new role for HLA-G as a negative feedback loop by which B cells control MAIT cell responses to antigens.
Collapse
Affiliation(s)
- Rosângela Salerno-Gonçalves
- Center for Vaccine Development and Global Health (CVD), Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Tasmia Rezwan
- Center for Vaccine Development and Global Health (CVD), Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - David Luo
- Center for Vaccine Development and Global Health (CVD), Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Hervé Tettelin
- Department of Microbiology and Immunology and Institute for Genome Sciences (IGS), University of Maryland School of Medicine, Baltimore, MD, United States
| | - Marcelo B. Sztein
- Center for Vaccine Development and Global Health (CVD), Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Program in Oncology, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| |
Collapse
|
9
|
Schultz BM, Melo-Gonzalez F, Salazar GA, Porto BN, Riedel CA, Kalergis AM, Bueno SM. New Insights on the Early Interaction Between Typhoid and Non-typhoid Salmonella Serovars and the Host Cells. Front Microbiol 2021; 12:647044. [PMID: 34276584 PMCID: PMC8282409 DOI: 10.3389/fmicb.2021.647044] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 06/08/2021] [Indexed: 11/13/2022] Open
Abstract
Salmonella enterica is a common source of food and water-borne infections, causing a wide range of clinical ailments in both human and animal hosts. Immunity to Salmonella involves an interplay between different immune responses, which are rapidly initiated to control bacterial burden. However, Salmonella has developed several strategies to evade and modulate the host immune responses. In this sense, the main knowledge about the pathogenicity of this bacterium has been obtained by the study of mouse models with non-typhoidal serovars. However, this knowledge is not representative of all the pathologies caused by non-typhoidal serovars in the human. Here we review the most important features of typhoidal and non-typhoidal serovars and the diseases they cause in the human host, describing the virulence mechanisms used by these pathogens that have been identified in different models of infection.
Collapse
Affiliation(s)
- Bárbara M Schultz
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe Melo-Gonzalez
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Geraldyne A Salazar
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bárbara N Porto
- Laboratory of Clinical and Experimental Immunology, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil.,Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Claudia A Riedel
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Millennium Institute on Immunology and Immunotherapy, Universidad Andrés Bello, Santiago, Chile
| | - Alexis M Kalergis
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
10
|
Maharjan A, Dhungel B, Bastola A, Thapa Shrestha U, Adhikari N, Banjara MR, Lekhak B, Ghimire P, Rijal KR. Antimicrobial Susceptibility Pattern of Salmonella spp. Isolated from Enteric Fever Patients in Nepal. Infect Dis Rep 2021; 13:388-400. [PMID: 33919283 PMCID: PMC8167555 DOI: 10.3390/idr13020037] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/13/2021] [Accepted: 04/19/2021] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION Enteric fever, a systemic infection caused by Salmonella enterica Typhi and S. enterica Paratyphi is one of the most common infections in developing countries such as Nepal. Aside from irrational practices of antibiotic use, mutations in chromosomal genes encoding DNA gyrase and Topoisomerase IV and by plasmid mediated quinolone resistant (PMQR) genes are suggested mechanisms for the development of resistance to nalidixic acid and reduced susceptibility to ciprofloxacin. Regardless of high endemicity of enteric fever in Nepal, there is paucity of studies on prevalence and drug-resistance of the pathogen. Therefore, this study aimed to assess the antibiotic susceptibility pattern of Salmonella isolates and determine the minimum inhibitory concentration of ciprofloxacin. METHODS A total of 1298 blood samples were obtained from patients with suspected enteric fever, attending Sukraraj Tropical and Infectious Disease Hospital (STIDH) during March-August, 2019. Blood samples were inoculated immediately into BACTEC culture bottles and further processed for isolation and identification of Salmonella Typhi and S. Paratyphi. Axenic cultures of the isolates were further subjected to antimicrobial susceptibility testing (AST) by using the modified Kirby-Bauer disc diffusion method based on the guidelines by CLSI. The minimum inhibitory concentration (MIC) of ciprofloxacin was determined by agar-dilution method. RESULTS Out of 1298 blood cultures, 40 (3.1%) were positive for Salmonella spp. among which 29 (72.5%) isolates were S. Typhi and 11 (27.5%) isolates were S. Paratyphi A. In AST, 12.5% (5/40), 15% (6/40) and 20% (8/40) of the Salmonella isolates were susceptible to nalidixic acid, ofloxacin and levofloxacin, respectively, whereas none of the isolates were susceptible to ciprofloxacin. The MIC value for ciprofloxacin ranged from 0.06-16 µg/mL in which, respectively, 5% (2/40) and 52.5% (21/40) of the isolates were susceptible and resistant to ciprofloxacin. None of the isolates showed multidrug-resistance (MDR) in this study. CONCLUSION This study showed high prevalence of quinolone-resistant Salmonella spp., while there was marked re-emergence of susceptibilities to traditional first option drugs. Hence, conventional first-line-drugs and third-generation cephalosporins may find potential usage as the empirical drugs for enteric fever. Although our reporting was free of MDR strains, extensive surveillance, augmentation of diagnostic facilities and treatment protocol aided by AST report are recommended for addressing the escalating drug-resistance in the country.
Collapse
Affiliation(s)
- Anu Maharjan
- Central Department of Microbiology, Tribhuvan University, Kirtipur, Kathmandu 44618, Nepal; (A.M.); (B.D.); (U.T.S.); (N.A.); (M.R.B.); (B.L.); (P.G.)
| | - Binod Dhungel
- Central Department of Microbiology, Tribhuvan University, Kirtipur, Kathmandu 44618, Nepal; (A.M.); (B.D.); (U.T.S.); (N.A.); (M.R.B.); (B.L.); (P.G.)
| | - Anup Bastola
- Sukraraj Tropical and Infectious Disease Hospital, Teku, Kathmandu 44600, Nepal;
| | - Upendra Thapa Shrestha
- Central Department of Microbiology, Tribhuvan University, Kirtipur, Kathmandu 44618, Nepal; (A.M.); (B.D.); (U.T.S.); (N.A.); (M.R.B.); (B.L.); (P.G.)
| | - Nabaraj Adhikari
- Central Department of Microbiology, Tribhuvan University, Kirtipur, Kathmandu 44618, Nepal; (A.M.); (B.D.); (U.T.S.); (N.A.); (M.R.B.); (B.L.); (P.G.)
| | - Megha Raj Banjara
- Central Department of Microbiology, Tribhuvan University, Kirtipur, Kathmandu 44618, Nepal; (A.M.); (B.D.); (U.T.S.); (N.A.); (M.R.B.); (B.L.); (P.G.)
| | - Binod Lekhak
- Central Department of Microbiology, Tribhuvan University, Kirtipur, Kathmandu 44618, Nepal; (A.M.); (B.D.); (U.T.S.); (N.A.); (M.R.B.); (B.L.); (P.G.)
| | - Prakash Ghimire
- Central Department of Microbiology, Tribhuvan University, Kirtipur, Kathmandu 44618, Nepal; (A.M.); (B.D.); (U.T.S.); (N.A.); (M.R.B.); (B.L.); (P.G.)
| | - Komal Raj Rijal
- Central Department of Microbiology, Tribhuvan University, Kirtipur, Kathmandu 44618, Nepal; (A.M.); (B.D.); (U.T.S.); (N.A.); (M.R.B.); (B.L.); (P.G.)
| |
Collapse
|
11
|
J Barton A, Hill J, J Blohmke C, J Pollard A. Host restriction, pathogenesis and chronic carriage of typhoidal Salmonella. FEMS Microbiol Rev 2021; 45:6159486. [PMID: 33733659 PMCID: PMC8498562 DOI: 10.1093/femsre/fuab014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 03/03/2021] [Indexed: 12/16/2022] Open
Abstract
While conjugate vaccines against typhoid fever have recently been recommended by the World Health Organization for deployment, the lack of a vaccine against paratyphoid, multidrug resistance and chronic carriage all present challenges for the elimination of enteric fever. In the past decade, the development of in vitro and human challenge models has resulted in major advances in our understanding of enteric fever pathogenesis. In this review, we summarise these advances, outlining mechanisms of host restriction, intestinal invasion, interactions with innate immunity and chronic carriage, and discuss how this knowledge may progress future vaccines and antimicrobials.
Collapse
Affiliation(s)
- Amber J Barton
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford OX3 7LE, UK.,National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford OX4 2PG, UK.,Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Jennifer Hill
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford OX3 7LE, UK.,National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford OX4 2PG, UK
| | - Christoph J Blohmke
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford OX3 7LE, UK.,National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford OX4 2PG, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford OX3 7LE, UK.,National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford OX4 2PG, UK
| |
Collapse
|
12
|
Salmonella enterica serovar Typhi exposure elicits ex vivo cell-type-specific epigenetic changes in human gut cells. Sci Rep 2020; 10:13581. [PMID: 32788681 PMCID: PMC7423951 DOI: 10.1038/s41598-020-70492-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 06/22/2020] [Indexed: 01/25/2023] Open
Abstract
Salmonella enterica serovar Typhi (S. Typhi) causes substantial morbidity and mortality worldwide, particularly among young children. Humans develop an array of mucosal immune responses following S. Typhi infection. Whereas the cellular mechanisms involved in S. Typhi infection have been intensively studied, very little is known about the early chromatin modifications occurring in the human gut microenvironment that influence downstream immune responses. To address this gap in knowledge, cells isolated from human terminal ileum exposed ex vivo to the wild-type S. Typhi strain were stained with a 33-metal-labeled antibody panel for mass cytometry analyses of the early chromatin modifications modulated by S. Typhi. We measured the cellular levels of 6 classes of histone modifications, and 1 histone variant in 11 major cell subsets (i.e., B, CD3 + T, CD4 + T, CD8 + T, NK, TCR-γδ, Mucosal associated invariant (MAIT), and NKT cells as well as monocytes, macrophages, and epithelial cells). We found that arginine methylation might regulate the early-differentiation of effector-memory CD4+ T-cells following exposure to S. Typhi. We also found S. Typhi-induced post-translational modifications in histone methylation and acetylation associated with epithelial cells, NKT, MAIT, TCR-γδ, Monocytes, and CD8 + T-cells that are related to both gene activation and silencing.
Collapse
|
13
|
Qian H, Cheng S, Liu G, Tan Z, Dong C, Bao J, Hong J, Jin D, Bao C, Gu B. Discovery of seven novel mutations of gyrB, parC and parE in Salmonella Typhi and Paratyphi strains from Jiangsu Province of China. Sci Rep 2020; 10:7359. [PMID: 32355184 PMCID: PMC7193621 DOI: 10.1038/s41598-020-64346-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 04/15/2020] [Indexed: 01/25/2023] Open
Abstract
Objective: To investigate the prevalence of Salmonella Typhi and Paratyphi resistance to quinolones and characterize the underlying mechanism in Jiangsu Province of China. Methods: Antimicrobial susceptibility testing was performed using Kirby-Bauer disc diffusion system. Quinolone resistance-determining region (QRDR), plasmid-mediated quinolone resistance (PMQR) determinant genes were detected by PCR and sequencing. Results: Out of 239 Salmonella isolates, 164 were S. Typhi and 75 were S. Paratyphi. 128 (53.6%) Salmonella isolates were resistant to nalidixic acid; 11 (4.6%) isolates to ciprofloxacin and 66 (27.6%) isolates were intermediate to ciprofloxacin. QRDR were present in 69 S. Typhi isolates, among which mutation at codon 83 (n = 45) and 133 (n = 61) predominated. In S. Paratyphi, the most common mutations were detected in gyrA at codon 83(n = 24) and parC: T57S (n = 8). Seven mutations were first reported in Salmonella isolates including gyrB: S426G, parC: D79G and parE: [S498T, E543K, V560G, I444S, Y434S]. PMQR genes including qnrD1, qnrA1, qnrB4, aac (6′)-Ib-cr4 and qnrS1 were detected in 1, 2, 3, 7 and 9 isolates, relatively. Conclusions: High resistance to quinolones in Salmonella remains a serious problem in Jiangsu, China. The presence of the novel mutations increases the complexity of quinolone-resistant genotypes and poses a threat to public health. Subject terms: Salmonella Typhi, Salmonella Paratyphi, antimicrobial resistance, QRDR, PMQR.
Collapse
Affiliation(s)
- Huimin Qian
- Department of Acute Infectious Disease Prevention and Control, Jiangsu Provincial Center for Disease Prevention and Control, Nanjing, 210029, China
| | - Siyun Cheng
- Xuzhou Medical University School of Medical Technology, Xuzhou, 221004, China
| | - Guoye Liu
- Xuzhou Medical University School of Medical Technology, Xuzhou, 221004, China
| | - Zhongming Tan
- Department of Acute Infectious Disease Prevention and Control, Jiangsu Provincial Center for Disease Prevention and Control, Nanjing, 210029, China
| | - Chen Dong
- Department of Acute Infectious Disease Prevention and Control, Jiangsu Provincial Center for Disease Prevention and Control, Nanjing, 210029, China
| | - Jinfeng Bao
- Xuzhou Medical University School of Medical Technology, Xuzhou, 221004, China
| | - Jie Hong
- Department of Acute Infectious Disease Prevention and Control, Jiangsu Provincial Center for Disease Prevention and Control, Nanjing, 210029, China
| | - Dazhi Jin
- Centre of Laboratory Medicine, Zhejiang Provincial People Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.,School of Laboratory Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, 310053, China
| | - Changjun Bao
- Department of Acute Infectious Disease Prevention and Control, Jiangsu Provincial Center for Disease Prevention and Control, Nanjing, 210029, China.
| | - Bing Gu
- Xuzhou Medical University School of Medical Technology, Xuzhou, 221004, China. .,Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China.
| |
Collapse
|
14
|
Verma S, Senger S, Cherayil BJ, Faherty CS. Spheres of Influence: Insights into Salmonella Pathogenesis from Intestinal Organoids. Microorganisms 2020; 8:microorganisms8040504. [PMID: 32244707 PMCID: PMC7232497 DOI: 10.3390/microorganisms8040504] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/27/2020] [Accepted: 03/28/2020] [Indexed: 12/19/2022] Open
Abstract
The molecular complexity of host-pathogen interactions remains poorly understood in many infectious diseases, particularly in humans due to the limited availability of reliable and specific experimental models. To bridge the gap between classical two-dimensional culture systems, which often involve transformed cell lines that may not have all the physiologic properties of primary cells, and in vivo animal studies, researchers have developed the organoid model system. Organoids are complex three-dimensional structures that are generated in vitro from primary cells and can recapitulate key in vivo properties of an organ such as structural organization, multicellularity, and function. In this review, we discuss how organoids have been deployed in exploring Salmonella infection in mice and humans. In addition, we summarize the recent advancements that hold promise to elevate our understanding of the interactions and crosstalk between multiple cell types and the microbiota with Salmonella. These models have the potential for improving clinical outcomes and future prophylactic and therapeutic intervention strategies.
Collapse
Affiliation(s)
- Smriti Verma
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Charlestown Navy Yard, Boston, 02129 MA, USA; (S.S.); (B.J.C.); (C.S.F.)
- Harvard Medical School, Boston, 02115 MA, USA
- Correspondence: ; Tel.: +1-617-726-7991
| | - Stefania Senger
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Charlestown Navy Yard, Boston, 02129 MA, USA; (S.S.); (B.J.C.); (C.S.F.)
- Harvard Medical School, Boston, 02115 MA, USA
| | - Bobby J. Cherayil
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Charlestown Navy Yard, Boston, 02129 MA, USA; (S.S.); (B.J.C.); (C.S.F.)
- Harvard Medical School, Boston, 02115 MA, USA
| | - Christina S. Faherty
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Charlestown Navy Yard, Boston, 02129 MA, USA; (S.S.); (B.J.C.); (C.S.F.)
- Harvard Medical School, Boston, 02115 MA, USA
| |
Collapse
|
15
|
Salerno-Gonçalves R, Tettelin H, Luo D, Guo Q, Ardito MT, Martin WD, De Groot AS, Sztein MB. Differential functional patterns of memory CD4 + and CD8 + T-cells from volunteers immunized with Ty21a typhoid vaccine observed using a recombinant Escherichia coli system expressing S. Typhi proteins. Vaccine 2019; 38:258-270. [PMID: 31629569 DOI: 10.1016/j.vaccine.2019.10.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 10/03/2019] [Accepted: 10/07/2019] [Indexed: 02/01/2023]
Abstract
It is widely accepted that CD4+ and CD8+ T-cells play a significant role in protection against Salmonella enterica serovar Typhi (S. Typhi), the causative agent of the typhoid fever. However, the antigen specificity of these T-cells remains largely unknown. Previously, we demonstrated the feasibility of using a recombinant Escherichia coli (E. coli) expression system to uncover the antigen specificity of CD4+ and CD8+ T cells. Here, we expanded these studies to include the evaluation of 12 additional S. Typhi proteins: 4 outer membrane proteins (OmpH, OmpL, OmpR, OmpX), 3 Vi-polysaccharide biosynthesis proteins (TviA, TviB, TviE), 3 cold shock proteins (CspA, CspB, CspC), and 2 conserved hypothetical proteins (Chp 1 and Chp2), all selected based on the bioinformatic analyses of the content of putative T-cell epitopes. CD4+ and CD8+ T cells from 15 adult volunteers, obtained before and 42 days after immunization with oral live attenuated Ty21a vaccine, were assessed for their functionality (i.e., production of cytokines and cytotoxic expression markers in response to stimulation with selected antigens) as measured by flow cytometry. Although volunteers differed on their T-cell antigen specificity, we observed T-cell immune responses against all S. Typhi proteins evaluated. These responses included 9 proteins, OmpH, OmpR, TviA, TviE, CspA, CspB, CspC, Chp 1 and Chp 2, which have not been previously reported to elicit T-cell responses. Interestingly, we also observed that, regardless of the protein, the functional patterns of the memory T-cells were different between CD4+ and CD8+ T cells. In sum, these studies demonstrated the feasibility of using bioinformatic analysis and the E. coli expressing system described here to uncover novel immunogenic T-cell proteins that could serve as potential targets for the production of protein-based vaccines.
Collapse
Affiliation(s)
- Rosângela Salerno-Gonçalves
- Center for Vaccine Development and Global Health (CVD), Department of Pediatrics, University of Maryland School of Medicine, 685 West Baltimore Street, HSF1, Baltimore, MD 21201, USA.
| | - Hervé Tettelin
- Department of Microbiology and Immunology and Institute for Genome Sciences (IGS), University of Maryland School of Medicine, 670 West Baltimore Street, HSF3, Baltimore, MD 21201, USA
| | - David Luo
- Center for Vaccine Development and Global Health (CVD), Department of Pediatrics, University of Maryland School of Medicine, 685 West Baltimore Street, HSF1, Baltimore, MD 21201, USA
| | - Qin Guo
- Department of Microbiology and Immunology and Institute for Genome Sciences (IGS), University of Maryland School of Medicine, 670 West Baltimore Street, HSF3, Baltimore, MD 21201, USA
| | - Matthew T Ardito
- Institute for Immunology and Informatics (iCubed), Department of Cell and Molecular Biology, University of Rhode Island, 80 Washington Street, Providence, RI, USA; EpiVax, Inc., 188 Valley Street Suite 424, Providence, RI, USA
| | - William D Martin
- Institute for Immunology and Informatics (iCubed), Department of Cell and Molecular Biology, University of Rhode Island, 80 Washington Street, Providence, RI, USA; EpiVax, Inc., 188 Valley Street Suite 424, Providence, RI, USA
| | - Anne S De Groot
- Institute for Immunology and Informatics (iCubed), Department of Cell and Molecular Biology, University of Rhode Island, 80 Washington Street, Providence, RI, USA; EpiVax, Inc., 188 Valley Street Suite 424, Providence, RI, USA
| | - Marcelo B Sztein
- Center for Vaccine Development and Global Health (CVD), Department of Pediatrics, University of Maryland School of Medicine, 685 West Baltimore Street, HSF1, Baltimore, MD 21201, USA
| |
Collapse
|