1
|
Lentsch V, Woller A, Rocker A, Aslani S, Moresi C, Ruoho N, Larsson L, Fattinger SA, Wenner N, Barazzone EC, Hardt WD, Loverdo C, Diard M, Slack E. Vaccine-enhanced competition permits rational bacterial strain replacement in the gut. Science 2025; 388:74-81. [PMID: 40179176 DOI: 10.1126/science.adp5011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 02/06/2025] [Indexed: 04/05/2025]
Abstract
Colonization of the intestinal lumen precedes invasive infection for a wide range of enteropathogenic and opportunistic pathogenic bacteria. We show that combining oral vaccination with engineered or selected niche-competitor strains permits pathogen exclusion and strain replacement in the mouse gut lumen. This approach can be applied either prophylactically to prevent invasion of nontyphoidal Salmonella strains, or therapeutically to displace an established Escherichia coli. Both intact adaptive immunity and metabolic niche competition are necessary for efficient vaccine-enhanced competition. Our findings imply that mucosal antibodies have evolved to work in the context of gut microbial ecology by influencing the outcome of competition. This has broad implications for the elimination of pathogenic and antibiotic-resistant bacterial reservoirs and for rational microbiota engineering.
Collapse
Affiliation(s)
- Verena Lentsch
- Institute for Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
- Medical Research Council (MRC) Translational Immunology Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Aurore Woller
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine (IBPS), Laboratoire Jean Perrin (LJP), Paris, France
- Unité de Chronobiologie théorique, Faculté des Sciences, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - Selma Aslani
- Institute for Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Claudia Moresi
- Institute for Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Niina Ruoho
- Institute for Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Louise Larsson
- Institute for Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Stefan A Fattinger
- Institute for Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | | | | | - Wolf-Dietrich Hardt
- Institute for Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Claude Loverdo
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine (IBPS), Laboratoire Jean Perrin (LJP), Paris, France
| | - Médéric Diard
- Biozentrum, University of Basel, Basel, Switzerland
- Basel Research Centre for Child Health, Basel, Switzerland
| | - Emma Slack
- Institute for Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
- Basel Research Centre for Child Health, Basel, Switzerland
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| |
Collapse
|
2
|
Śliwa-Dominiak J, Czechowska K, Blanco A, Sielatycka K, Radaczyńska M, Skonieczna-Żydecka K, Marlicz W, Łoniewski I. Flow Cytometry in Microbiology: A Review of the Current State in Microbiome Research, Probiotics, and Industrial Manufacturing. Cytometry A 2025; 107:145-164. [PMID: 40028773 DOI: 10.1002/cyto.a.24920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/22/2024] [Accepted: 01/25/2025] [Indexed: 03/05/2025]
Abstract
Flow cytometry (FC) is a versatile and powerful tool in microbiology, enabling precise analysis of single cells for a variety of applications, including the detection and quantification of bacteria, viruses, fungi, as well as algae, phytoplankton, and parasites. Its utility in assessing cell viability, metabolic activity, immune responses, and pathogen-host interactions makes it indispensable in both research and diagnostics. The analysis of microbiota (community of microorganisms) and microbiome (collective genomes of the microorganisms) has become essential for understanding the intricate role of microbial communities in health, disease, and physiological functions. FC offers a promising complement, providing rapid, cost-effective, and dynamic profiling of microbial communities, with the added ability to isolate and sort bacterial populations for further analysis. In the probiotic industry, FC facilitates fast, affordable, and versatile analyses, helping assess both probiotics and postbiotics. It also supports the study of bacterial viability under stress conditions, including gastric acid and bile, improving insight into probiotic survival and adhesion to the intestinal mucosa. Additionally, the integration of Machine Learning in microbiology research has transformative potential, improving data analysis and supporting advances in personalized medicine and probiotic formulations. Despite the need for further standardization, FC continues to evolve as a key tool in modern microbiology and clinical diagnostics.
Collapse
Affiliation(s)
- Joanna Śliwa-Dominiak
- Research and Development Centre, Sanprobi, Szczecin, Poland
- Department of Biochemical Science, Faculty of Health Sciences, Pomeranian Medical University, Szczecin, Poland
| | | | - Alfonso Blanco
- Flow Cytometry Core Technology, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Katarzyna Sielatycka
- Research and Development Centre, Sanprobi, Szczecin, Poland
- Institute of Biology, Faculty of Exact and Natural Sciences, University of Szczecin, Szczecin, Poland
| | - Martyna Radaczyńska
- Research and Development Centre, Sanprobi, Szczecin, Poland
- Department of Biochemical Science, Faculty of Health Sciences, Pomeranian Medical University, Szczecin, Poland
| | - Karolina Skonieczna-Żydecka
- Research and Development Centre, Sanprobi, Szczecin, Poland
- Department of Biochemical Science, Faculty of Health Sciences, Pomeranian Medical University, Szczecin, Poland
| | - Wojciech Marlicz
- Research and Development Centre, Sanprobi, Szczecin, Poland
- Department of Gastroenterology, Faculty of Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Igor Łoniewski
- Research and Development Centre, Sanprobi, Szczecin, Poland
- Department of Biochemical Science, Faculty of Health Sciences, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
3
|
Thomas DE, Kinskie KS, Brown KM, Flanagan LA, Davalos RV, Hyler AR. Dielectrophoretic Microfluidic Designs for Precision Cell Enrichments and Highly Viable Label-Free Bacteria Recovery from Blood. MICROMACHINES 2025; 16:236. [PMID: 40047707 PMCID: PMC11857104 DOI: 10.3390/mi16020236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 03/09/2025]
Abstract
Conducting detailed cellular analysis of complex biological samples poses challenges in cell sorting and recovery for downstream analysis. Label-free microfluidics provide a promising solution for these complex applications. In this work, we investigate particle manipulation on two label-free microdevice designs using cDEP to enrich E. coli from whole human blood to mimic infection workflows. E. coli is still a growing source of bacteremia, sepsis, and other infections in modern countries, affecting millions of patients globally. The two microfluidic designs were evaluated for throughput, scaling, precision targeting, and high-viability recovery. While CytoChip D had the potential for higher throughput, given its continuous method of DEP-based sorting to accommodate larger clinical samples like a 10 mL blood draw, it could not effectively recover the bacteria. CytoChip B achieved a high-purity recovery of over 98% of bacteria from whole human blood, even in concentrations on the order of <100 CFU/mL, demonstrating the feasibility of processing and recovering ultra-low concentrations of bacteria for downstream analysis, culture, and drug testing. Future work will aim to scale CytoChip B for larger volume throughput while still achieving high bacteria recovery.
Collapse
Affiliation(s)
- Dean E. Thomas
- CytoRecovery, Inc., Blacksburg, VA 24060, USA; (D.E.T.); (K.S.K.); (K.M.B.)
| | - Kyle S. Kinskie
- CytoRecovery, Inc., Blacksburg, VA 24060, USA; (D.E.T.); (K.S.K.); (K.M.B.)
| | - Kyle M. Brown
- CytoRecovery, Inc., Blacksburg, VA 24060, USA; (D.E.T.); (K.S.K.); (K.M.B.)
| | - Lisa A. Flanagan
- Departments of Neurology, Biomedical Engineering, and Anatomy & Neurobiology, University of California Irvine, Irvine, CA 92697, USA;
| | - Rafael V. Davalos
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech-Emory University, Atlanta, GA 30318, USA;
| | - Alexandra R. Hyler
- CytoRecovery, Inc., Blacksburg, VA 24060, USA; (D.E.T.); (K.S.K.); (K.M.B.)
| |
Collapse
|
4
|
Liu R, Galiwango RM, Park D, Huibner S, Aziz M, Anok A, Nnamutete J, Isbirye Y, Wasswa JB, Male D, Kigozi G, Tobian AAR, Prodger JL, Liu C, Coburn B, Kaul R. Gardnerella vaginalis-binding IgA in the urethra of sexually experienced males. MICROBIOME 2025; 13:29. [PMID: 39881368 PMCID: PMC11776119 DOI: 10.1186/s40168-024-02007-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 12/16/2024] [Indexed: 01/31/2025]
Abstract
BACKGROUND Genital inflammation increases HIV susceptibility and is associated with the density of pro-inflammatory anaerobes in the vagina and coronal sulcus. The penile urethra is a critical site of HIV acquisition, although correlates of urethral HIV acquisition are largely unknown. While Streptococcus mitis is a consistent component of the urethral flora, the presence of Gardnerella vaginalis has been linked with prior penile-vaginal sex and urethral inflammation. Here, we use a flow cytometry-based bacterial assay to quantify urethral IgA and IgG that bind G. vaginalis and S. mitis in a cross-sectional cohort of 45 uncircumcised Ugandan men and to evaluate their association with the urethral microbiome and local soluble immune factors. RESULTS Urethral antibodies binding both bacterial species were readily detectable, with G. vaginalis predominantly bound by IgA, and S. mitis equivalently by IgA and IgG. Gardnerella vaginalis-binding IgA was elevated in participants with detectable urethral Gardnerella, with the latter only present in participants who reported prior penile-vaginal sex. In contrast, detectable urethral S. mitis was not associated with sexual history or levels of S. mitis-binding IgA/IgG. The time from the last penile-vaginal sex was inversely correlated with the urethral concentrations of total IgA, G. vaginalis-binding IgA, and chemokines IL-8 and MIP-1β; these inflammatory chemokines were independently associated with higher total IgA concentration, but not with G. vaginalis-binding IgA. CONCLUSIONS This first description of microbe-binding antibodies in the penile urethra suggests that urethral colonization by Gardnerella after penile-vaginal sex specifically induces a G. vaginalis-binding IgA response. Prospective studies of the host-microbe relationship in the urethra may have implications for the development of vaccines against sexually-transmitted bacteria. Video Abstract.
Collapse
Affiliation(s)
- Rachel Liu
- Department of Medicine, University of Toronto, Toronto, Canada.
| | - R M Galiwango
- Department of Immunology, University of Toronto, Toronto, Canada
- Rakai Health Science Program, Kalisizo, Uganda
| | - Daniel Park
- Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington, DC, USA
| | - Sanja Huibner
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Maliha Aziz
- Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington, DC, USA
| | - Aggrey Anok
- Rakai Health Science Program, Kalisizo, Uganda
| | | | | | | | - Deo Male
- Rakai Health Science Program, Kalisizo, Uganda
| | | | - Aaron A R Tobian
- Department of Pathology, John Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jessica L Prodger
- Department of Microbiology and Immunology, Western University, London, ON, Canada
| | - Cindy Liu
- Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington, DC, USA
| | - Bryan Coburn
- Department of Medicine, University of Toronto, Toronto, Canada
- Department of Immunology, University of Toronto, Toronto, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Rupert Kaul
- Department of Medicine, University of Toronto, Toronto, Canada
- Department of Immunology, University of Toronto, Toronto, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| |
Collapse
|
5
|
Shen D, Seco BMS, Teixeira Alves LG, Yao L, Bräutigam M, Opitz B, Witzenrath M, Fries BC, Seeberger PH. Semisynthetic Glycoconjugate Vaccine Lead against Klebsiella pneumoniae Serotype O2afg Induces Functional Antibodies and Reduces the Burden of Acute Pneumonia. J Am Chem Soc 2024; 146:35356-35366. [PMID: 39666976 DOI: 10.1021/jacs.4c13972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Carbapenem-resistant Klebsiella pneumoniae (CR-Kp) bacteria are a serious global health concern due to their drug-resistance to nearly all available antibiotics, fast spread, and high mortality rate. O2afg is a major CR-Kp serotype in the sequence type 258 group (KPST258) that is weakly immunogenic in humans. Here, we describe the creation and evaluation of semisynthetic O2afg glycoconjugate vaccine leads containing one and two repeating units of the polysaccharide epitope that covers the surface of the bacteria conjugated to the carrier protein CRM197. The semisynthetic glycoconjugate containing two repeating units induced functional IgG antibodies in rabbits with opsonophagocytic killing activity and enhanced complement activation and complement-mediated killing of CR-Kp. Passive immunization reduced the burden of acute pneumonia in mice and may represent an alternative to antimicrobial therapy. The semisynthetic glycoconjugate vaccine lead against CR-Kp expressing O2afg antigen is awaiting preclinical development.
Collapse
Affiliation(s)
- Dacheng Shen
- Department of Bimolecular System, Max Planck Institute of Colloids and Interfaces; 14476 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Bruna M S Seco
- Department of Bimolecular System, Max Planck Institute of Colloids and Interfaces; 14476 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Luiz Gustavo Teixeira Alves
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charite-Universitätsmedizin Berlin; 10117 Berlin, Germany
| | - Ling Yao
- Department of Bimolecular System, Max Planck Institute of Colloids and Interfaces; 14476 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Maria Bräutigam
- Department of Bimolecular System, Max Planck Institute of Colloids and Interfaces; 14476 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Bastian Opitz
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charite-Universitätsmedizin Berlin; 10117 Berlin, Germany
- German Center for Lung Research (DZL), 12203 Berlin, Germany
| | - Martin Witzenrath
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charite-Universitätsmedizin Berlin; 10117 Berlin, Germany
- German Center for Lung Research (DZL), 12203 Berlin, Germany
| | - Bettina C Fries
- Department of Medicine, Infectious Disease Division, Stony Brook University; Stony Brook, New York 11794, United States
- Veteran's Administration Medical Center, Northport, New York 11768, United States
| | - Peter H Seeberger
- Department of Bimolecular System, Max Planck Institute of Colloids and Interfaces; 14476 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| |
Collapse
|
6
|
Peralta Alvarez MP, Downward K, White A, Harris SA, Satti I, Li S, Morrison A, Sibley L, Sarfas C, Dennis M, Azema HR, Sharpe S, McShane H, Tanner R. Intravenous BCG vaccination in non-human primates induces superior serum antibody titers with enhanced avidity and opsonizing capacity compared to the intradermal route. Vaccine 2024; 42:126444. [PMID: 39522337 PMCID: PMC11906387 DOI: 10.1016/j.vaccine.2024.126444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/07/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024]
Abstract
A new and more effective tuberculosis (TB) vaccine is urgently needed, but development is hampered by the lack of validated immune correlates of protection. Bacillus Calmette Guérin (BCG) vaccination by the aerosol (AE) and intravenous (IV) routes has been shown to confer superior levels of protection from challenge with Mycobacterium tuberculosis (M.tb) in non-human primates (NHP) compared with standard intradermal (ID) administration. This finding offers a valuable opportunity to investigate which aspects of immunity are associated with improved control of M.tb and may represent biomarkers or correlates of protection. As TB vaccine research to date has focused largely on cellular immunity, we aimed to better characterize the poorly-understood serum antibody response to BCG administered by different routes of vaccination in NHP. We demonstrate superior M.tb-specific IgG, IgA, and IgM titers in serum following IV BCG vaccination compared to the ID or AE routes. We also observe improved capacity of IgG induced by IV BCG to opsonize the surface of mycobacteria, and report for the first time that M.tb-specific IgG from IV BCG vaccinated animals is of higher avidity compared with IgG from ID or AE BCG vaccinated animals. Notably, we identified a significant correlation between IgG avidity and measures of protection from aerosol M.tb challenge. Our findings highlight a potential role for antibodies as markers and/or mediators of the superior vaccine-induced protection IV BCG confers against TB and suggest that quality, as well as quantity, of antibodies should be considered when developing and evaluating TB vaccine candidates.
Collapse
Affiliation(s)
- Marco Polo Peralta Alvarez
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK; Laboratorio Nacional de Vacunologia y Virus Tropicales, Departamento de Microbiologia, Escuela Nacional de Ciencias Biologicas, Instituto Politecnico Nacional, Ciudad de Mexico 11350, Mexico
| | - Keya Downward
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Andrew White
- UK Health Security Agency, Porton Down, Salisbury SP4 0JG, UK
| | - Stephanie A Harris
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Iman Satti
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Shuailin Li
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Laura Sibley
- UK Health Security Agency, Porton Down, Salisbury SP4 0JG, UK
| | | | - Mike Dennis
- UK Health Security Agency, Porton Down, Salisbury SP4 0JG, UK
| | - Hugo Redondo Azema
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Sally Sharpe
- UK Health Security Agency, Porton Down, Salisbury SP4 0JG, UK
| | - Helen McShane
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Rachel Tanner
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK; Department of Biology, University of Oxford, Oxford OX1 3RB, UK.
| |
Collapse
|
7
|
Xu S, Lee I, Kwon SJ, Kim E, Nevo L, Straight L, Murata H, Matyjaszewski K, Dordick JS. Split fluorescent protein-mediated multimerization of cell wall binding domain for highly sensitive and selective bacterial detection. N Biotechnol 2024; 82:54-64. [PMID: 38750815 DOI: 10.1016/j.nbt.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/22/2024] [Accepted: 05/08/2024] [Indexed: 05/18/2024]
Abstract
Cell wall peptidoglycan binding domains (CBDs) of cell lytic enzymes, including bacteriocins, autolysins and bacteriophage endolysins, enable highly selective bacterial binding, and thus, have potential as biorecognition molecules for nondestructive bacterial detection. Here, a novel design for a self-complementing split fluorescent protein (FP) complex is proposed, where a multimeric FP chain fused with specific CBDs ((FP-CBD)n) is assembled inside the cell, to improve sensitivity by enhancing the signal generated upon Staphylococcus aureus or Bacillus anthracis binding. Flow cytometry shows enhanced fluorescence on the cell surface with increasing FP stoichiometry and surface plasmon resonance reveals nanomolar binding affinity to isolated peptidoglycan. The breadth of function of these complexes is demonstrated through the use of CBD modularity and the ability to attach enzymatic detection modalities. Horseradish peroxidase-coupled (FP-CBD)n complexes generate a catalytic amplification, with the degree of amplification increasing as a function of FP length, reaching a limit of detection (LOD) of 103 cells/droplet (approximately 0.1 ng S. aureus or B. anthracis) within 15 min on a polystyrene surface. These fusion proteins can be multiplexed for simultaneous detection. Multimeric split FP-CBD fusions enable use as a biorecognition molecule with enhanced signal for use in bacterial biosensing platforms.
Collapse
Affiliation(s)
- Shirley Xu
- Department of Chemical and Biological Engineering, and Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY, USA
| | - Inseon Lee
- Department of Chemical and Biological Engineering, and Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY, USA
| | - Seok-Joon Kwon
- Department of Chemical and Biological Engineering, and Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY, USA
| | - Eunsol Kim
- Department of Chemical and Biological Engineering, and Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY, USA
| | - Liv Nevo
- Department of Chemical and Biological Engineering, and Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY, USA
| | - Lorelli Straight
- Department of Biology, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY, USA
| | - Hironobu Murata
- Department of Chemistry, Carnegie Mellon University, 5000 Forbes Ave, Pittsburgh, PA, USA
| | | | - Jonathan S Dordick
- Department of Chemical and Biological Engineering, and Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY, USA; Department of Biology, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY, USA.
| |
Collapse
|
8
|
Jordan C, Siebold K, Priegue P, Seeberger PH, Gilmour R. A Fluorinated Sialic Acid Vaccine Lead Against Meningitis B and C. J Am Chem Soc 2024; 146:15366-15375. [PMID: 38768956 PMCID: PMC11157539 DOI: 10.1021/jacs.4c03179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/22/2024]
Abstract
Inspired by the specificity of α-(2,9)-sialyl epitopes in bacterial capsular polysaccharides (CPS), a doubly fluorinated disaccharide has been validated as a vaccine lead against Neisseria meningitidis serogroups C and/or B. Emulating the importance of fluorine in drug discovery, this molecular editing approach serves a multitude of purposes, which range from controlling α-selective chemical sialylation to mitigating competing elimination. Conjugation of the disialoside with two carrier proteins (CRM197 and PorA) enabled a semisynthetic vaccine to be generated; this was then investigated in six groups of six mice. The individual levels of antibodies formed were compared and classified as highly glycan-specific and protective. All glycoconjugates induced a stable and long-term IgG response and binding to the native CPS epitope was achieved. The generated antibodies were protective against MenC and/or MenB; this was validated in vitro by SBA and OPKA assays. By merging the fluorinated glycan epitope of MenC with an outer cell membrane protein of MenB, a bivalent vaccine against both serogroups was created. It is envisaged that validation of this synthetic, fluorinated disialoside bioisostere as a potent antigen will open new therapeutic avenues.
Collapse
Affiliation(s)
- Christina Jordan
- Institute
for Organic Chemistry, University of Münster, Corrensstraße 36, Münster 48149, Germany
| | - Kathrin Siebold
- Institute
for Organic Chemistry, University of Münster, Corrensstraße 36, Münster 48149, Germany
| | - Patricia Priegue
- Department
of Biomolecular Systems, Max Planck Institute
for Colloids and Interfaces, Am Mühlenberg 1, Potsdam 14476, Germany
- Freie
Universität Berlin, Institute of
Chemistry and Biochemistry, Arnimallee 22, Berlin 14195, Germany
| | - Peter H. Seeberger
- Department
of Biomolecular Systems, Max Planck Institute
for Colloids and Interfaces, Am Mühlenberg 1, Potsdam 14476, Germany
- Freie
Universität Berlin, Institute of
Chemistry and Biochemistry, Arnimallee 22, Berlin 14195, Germany
| | - Ryan Gilmour
- Institute
for Organic Chemistry, University of Münster, Corrensstraße 36, Münster 48149, Germany
| |
Collapse
|
9
|
Filimonova I, Innocenti G, Vogl T. Phage Immunoprecipitation Sequencing (PhIP-Seq) for Analyzing Antibody Epitope Repertoires Against Food Antigens. Methods Mol Biol 2024; 2717:101-122. [PMID: 37737980 DOI: 10.1007/978-1-0716-3453-0_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
While thousands of food and environmental allergens have been reported, conventional methods for allergy testing typically rely on measuring immunoglobulin E (IgE) binding against panels of dozens to hundreds of antigens. Beyond IgE, also the specificity of other Ig (sub-)classes such as IgG4, has gained interest because of a potential protective role toward allergy.Phage immunoprecipitation sequencing (PhIP-Seq) allows to study hundreds of thousands of rationally selected peptide antigens and to resolve binding specificities of different Ig classes. This technology combines synthetic DNA libraries encoding antigens, with the display on the surface of T7 bacteriophages and next-generation sequencing (NGS) for quantitative readouts. Thereby binding of entire Ig repertoires can be measured to detect the exact epitopes of food allergens and to study potential cross-reactivity.In this chapter, we provide a summary of both the key experimental steps and various strategies for analyzing PhIP-Seq datasets, as well as comparing the advantages and disadvantages of this methodology for measuring antibody responses against food antigens.
Collapse
Affiliation(s)
- Ioanna Filimonova
- Medical University of Vienna, Center for Cancer Research, Vienna, Austria
| | - Gabriel Innocenti
- Medical University of Vienna, Center for Cancer Research, Vienna, Austria
| | - Thomas Vogl
- Medical University of Vienna, Center for Cancer Research, Vienna, Austria.
| |
Collapse
|
10
|
Hunault L, Auria E, England P, Deschamps J, Briandet R, Kremer V, Iannascoli B, Vidal-Maison L, Guo C, Macdonald L, Péchiné S, Denève-Larrazet C, Dupuy B, Gorochov G, Bruhns P, Sterlin D. Anti-S-layer monoclonal antibodies impact Clostridioides difficile physiology. Gut Microbes 2024; 16:2301147. [PMID: 38289292 PMCID: PMC10829821 DOI: 10.1080/19490976.2023.2301147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/28/2023] [Indexed: 02/01/2024] Open
Abstract
Clostridioides difficile (C. difficile), a gram-positive anaerobic and spore-forming bacterium, is the leading cause of nosocomial antibiotic-associated diarrhea in adults which is characterized by high levels of recurrence and mortality. Surface (S)-layer Protein A (SlpA), the most abundantly expressed protein on the bacterial surface, plays a crucial role in the early stages of infection although the nature of its involvement in C. difficile physiology is yet to be fully understood. Anti-S-layer antibodies have been identified in the sera of convalescent patients and have been correlated with improved outcomes of C. difficile infection (CDI). However, the precise mechanisms by which anti-S-layer antibodies confer protection to the host remain unknown. In this study, we report the first monoclonal antibodies (mAbs) targeting the S-layer of reference strain 630. Characterization of these mAbs unraveled important roles for the S-layer protein in growth, toxin secretion, and biofilm formation by C. difficile, with differential and even opposite effects of various anti-SlpA mAbs on these functions. Moreover, one anti-SlpA mAb impaired C. difficile growth and conferred sensitivity to lysozyme-induced lysis. The results of this study show that anti-S-layer antibody responses can be beneficial or harmful for the course of CDI and provide important insights for the development of adequate S-layer-targeting therapeutics.
Collapse
Affiliation(s)
- Lise Hunault
- Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, Inserm, CNRS, Paris, France
- Antibodies in Therapy and Pathology, Institut Pasteur, Université Paris-Cité, Inserm UMR1222, Paris, France
- Collège doctoral, Sorbonne Université, Paris, France
| | - Emile Auria
- Laboratoire Pathogenèse des Bactéries Anaérobies, Institut Pasteur, Université Paris-Cité, UMR-CNRS 6047, Paris, France
| | - Patrick England
- Department of Structural Biology and Chemistry, Institut Pasteur, Université Paris Cité, CNRS UMR3528, Plateforme de Biophysique Moléculaire, Paris, France
| | - Julien Deschamps
- Institut Micalis, Université Paris-Saclay, INRAE, AgroParisTech, Jouy-en-Josas, France
| | - Romain Briandet
- Institut Micalis, Université Paris-Saclay, INRAE, AgroParisTech, Jouy-en-Josas, France
| | - Vanessa Kremer
- Antibodies in Therapy and Pathology, Institut Pasteur, Université Paris-Cité, Inserm UMR1222, Paris, France
- Inflammation, Microbiome and Immunosurveillance, Université Paris-Saclay, Inserm, Châtenay-Malabry, France
| | - Bruno Iannascoli
- Antibodies in Therapy and Pathology, Institut Pasteur, Université Paris-Cité, Inserm UMR1222, Paris, France
| | - Léo Vidal-Maison
- Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, Inserm, CNRS, Paris, France
| | | | | | - Séverine Péchiné
- Equipe Bactéries Pathogènes et Santé, Faculté de Pharmacie, Institut MICALIS (UMR 1319 Université Paris-Saclay, INRAE, AgroParisTech), Orsay, France
| | - Cécile Denève-Larrazet
- Equipe Bactéries Pathogènes et Santé, Faculté de Pharmacie, Institut MICALIS (UMR 1319 Université Paris-Saclay, INRAE, AgroParisTech), Orsay, France
| | - Bruno Dupuy
- Laboratoire Pathogenèse des Bactéries Anaérobies, Institut Pasteur, Université Paris-Cité, UMR-CNRS 6047, Paris, France
| | - Guy Gorochov
- Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, Inserm, CNRS, Paris, France
| | - Pierre Bruhns
- Antibodies in Therapy and Pathology, Institut Pasteur, Université Paris-Cité, Inserm UMR1222, Paris, France
| | - Delphine Sterlin
- Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, Inserm, CNRS, Paris, France
| |
Collapse
|
11
|
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: 7] [Impact Index Per Article: 3.5] [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
|
12
|
Thomas AM, Fidelle M, Routy B, Kroemer G, Wargo JA, Segata N, Zitvogel L. Gut OncoMicrobiome Signatures (GOMS) as next-generation biomarkers for cancer immunotherapy. Nat Rev Clin Oncol 2023; 20:583-603. [PMID: 37365438 PMCID: PMC11258874 DOI: 10.1038/s41571-023-00785-8] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2023] [Indexed: 06/28/2023]
Abstract
Oncogenesis is associated with intestinal dysbiosis, and stool shotgun metagenomic sequencing in individuals with this condition might constitute a non-invasive approach for the early diagnosis of several cancer types. The prognostic relevance of antibiotic intake and gut microbiota composition urged investigators to develop tools for the detection of intestinal dysbiosis to enable patient stratification and microbiota-centred clinical interventions. Moreover, since the advent of immune-checkpoint inhibitors (ICIs) in oncology, the identification of biomarkers for predicting their efficacy before starting treatment has been an unmet medical need. Many previous studies addressing this question, including a meta-analysis described herein, have led to the description of Gut OncoMicrobiome Signatures (GOMS). In this Review, we discuss how patients with cancer across various subtypes share several GOMS with individuals with seemingly unrelated chronic inflammatory disorders who, in turn, tend to have GOMS different from those of healthy individuals. We discuss findings from the aforementioned meta-analysis of GOMS patterns associated with clinical benefit from or resistance to ICIs across different cancer types (in 808 patients), with a focus on metabolic and immunological surrogate markers of intestinal dysbiosis, and propose practical guidelines to incorporate GOMS in decision-making for prospective clinical trials in immuno-oncology.
Collapse
Affiliation(s)
| | - Marine Fidelle
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) UMR 1015, ClinicObiome, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Pharmacology Department, Gustave Roussy, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS) 1428, Villejuif, France
| | - Bertrand Routy
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec, Canada
- Hematology-Oncology Division, Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Quebec, Canada
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, INSERM U1138, Equipe labellisée - Ligue Nationale contre le cancer, Université de Paris, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jennifer A Wargo
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Platform for Innovative Microbiome and Translational Research (PRIME-TR), MD Anderson Cancer Center, Houston, TX, USA
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Villejuif, France.
- Institut National de la Santé Et de la Recherche Médicale (INSERM) UMR 1015, ClinicObiome, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France.
- Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS) 1428, Villejuif, France.
- Université Paris-Saclay, Gif-sur-Yvette, France.
| |
Collapse
|
13
|
Lingasamy P, Modhukur V, Mändar R, Salumets A. Exploring Immunome and Microbiome Interplay in Reproductive Health: Current Knowledge, Challenges, and Novel Diagnostic Tools. Semin Reprod Med 2023; 41:172-189. [PMID: 38262441 PMCID: PMC10846929 DOI: 10.1055/s-0043-1778017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
The dynamic interplay between the immunome and microbiome in reproductive health is a complex and rapidly advancing research field, holding tremendously vast possibilities for the development of reproductive medicine. This immunome-microbiome relationship influences the innate and adaptive immune responses, thereby affecting the onset and progression of reproductive disorders. However, the mechanisms governing these interactions remain elusive and require innovative approaches to gather more understanding. This comprehensive review examines the current knowledge on reproductive microbiomes across various parts of female reproductive tract, with special consideration of bidirectional interactions between microbiomes and the immune system. Additionally, it explores innate and adaptive immunity, focusing on immunoglobulin (Ig) A and IgM antibodies, their regulation, self-antigen tolerance mechanisms, and their roles in immune homeostasis. This review also highlights ongoing technological innovations in microbiota research, emphasizing the need for standardized detection and analysis methods. For instance, we evaluate the clinical utility of innovative technologies such as Phage ImmunoPrecipitation Sequencing (PhIP-Seq) and Microbial Flow Cytometry coupled to Next-Generation Sequencing (mFLOW-Seq). Despite ongoing advancements, we emphasize the need for further exploration in this field, as a deeper understanding of immunome-microbiome interactions holds promise for innovative diagnostic and therapeutic strategies for reproductive health, like infertility treatment and management of pregnancy.
Collapse
Affiliation(s)
| | - Vijayachitra Modhukur
- Competence Centre on Health Technologies, Tartu, Estonia
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Reet Mändar
- Competence Centre on Health Technologies, Tartu, Estonia
- Department of Microbiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Andres Salumets
- Competence Centre on Health Technologies, Tartu, Estonia
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
14
|
Lentsch V, Aslani S, Echtermann T, Preet S, Cappio Barazzone E, Hoces D, Moresi C, Kümmerlen D, Slack E. "EvoVax" - A rationally designed inactivated Salmonella Typhimurium vaccine induces strong and long-lasting immune responses in pigs. Vaccine 2023; 41:5545-5552. [PMID: 37517910 DOI: 10.1016/j.vaccine.2023.07.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/25/2023] [Accepted: 07/25/2023] [Indexed: 08/01/2023]
Abstract
Salmonella enterica subspecies enterica serovar Typhimurium (S.Tm) poses a considerable threat to public health due to its zoonotic potential. Human infections are mostly foodborne, and pork and pork products are ranked among the top culprits for transmission. In addition, the high percentage of antibiotic resistance, especially in monophasic S.Tm, limits treatment options when needed. Better S.Tm control would therefore be of benefit both for farm animals and for safety of the human food chain. A promising pre-harvest intervention is vaccination. In this study we tested safety and immunogenicity of an oral inactivated S.Tm vaccine, which has been recently shown to generate an "evolutionary trap" and to massively reduce S.Tm colonization and transmission in mice. We show that this vaccine is highly immunogenic and safe in post-weaning pigs and that administration of a single oral dose results in a strong and long-lasting serum IgG response. This has several advantages over existing - mainly live - vaccines against S.Tm, both in improved seroconversion and reduced risk of vaccine-strain persistence and reversion to virulence.
Collapse
Affiliation(s)
- Verena Lentsch
- Institute for Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Selma Aslani
- Institute for Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Thomas Echtermann
- Division of Swine Medicine, Department for Farm Animals, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Swapan Preet
- Institute for Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | | | - Daniel Hoces
- Institute for Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Claudia Moresi
- Institute for Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Dolf Kümmerlen
- Division of Swine Medicine, Department for Farm Animals, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Emma Slack
- Institute for Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland; Botnar Research Centre for Child Health, Basel, Switzerland.
| |
Collapse
|
15
|
Mermans F, Mattelin V, Van den Eeckhoudt R, García-Timermans C, Van Landuyt J, Guo Y, Taurino I, Tavernier F, Kraft M, Khan H, Boon N. Opportunities in optical and electrical single-cell technologies to study microbial ecosystems. Front Microbiol 2023; 14:1233705. [PMID: 37692384 PMCID: PMC10486927 DOI: 10.3389/fmicb.2023.1233705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/03/2023] [Indexed: 09/12/2023] Open
Abstract
New techniques are revolutionizing single-cell research, allowing us to study microbes at unprecedented scales and in unparalleled depth. This review highlights the state-of-the-art technologies in single-cell analysis in microbial ecology applications, with particular attention to both optical tools, i.e., specialized use of flow cytometry and Raman spectroscopy and emerging electrical techniques. The objectives of this review include showcasing the diversity of single-cell optical approaches for studying microbiological phenomena, highlighting successful applications in understanding microbial systems, discussing emerging techniques, and encouraging the combination of established and novel approaches to address research questions. The review aims to answer key questions such as how single-cell approaches have advanced our understanding of individual and interacting cells, how they have been used to study uncultured microbes, which new analysis tools will become widespread, and how they contribute to our knowledge of ecological interactions.
Collapse
Affiliation(s)
- Fabian Mermans
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Ghent University, Ghent, Belgium
- Department of Oral Health Sciences, KU Leuven, Leuven, Belgium
| | - Valérie Mattelin
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Ghent University, Ghent, Belgium
| | - Ruben Van den Eeckhoudt
- Micro- and Nanosystems (MNS), Department of Electrical Engineering (ESAT), KU Leuven, Leuven, Belgium
| | - Cristina García-Timermans
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Ghent University, Ghent, Belgium
| | - Josefien Van Landuyt
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Ghent University, Ghent, Belgium
| | - Yuting Guo
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Ghent University, Ghent, Belgium
| | - Irene Taurino
- Micro- and Nanosystems (MNS), Department of Electrical Engineering (ESAT), KU Leuven, Leuven, Belgium
- Semiconductor Physics, Department of Physics and Astronomy, KU Leuven, Leuven, Belgium
| | - Filip Tavernier
- MICAS, Department of Electrical Engineering (ESAT), KU Leuven, Leuven, Belgium
| | - Michael Kraft
- Micro- and Nanosystems (MNS), Department of Electrical Engineering (ESAT), KU Leuven, Leuven, Belgium
- Leuven Institute of Micro- and Nanoscale Integration (LIMNI), KU Leuven, Leuven, Belgium
| | - Hira Khan
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Ghent University, Ghent, Belgium
| | - Nico Boon
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Ghent University, Ghent, Belgium
| |
Collapse
|
16
|
Johnson-Hence CB, Gopalakrishna KP, Bodkin D, Coffey KE, Burr AH, Rahman S, Rai AT, Abbott DA, Sosa YA, Tometich JT, Das J, Hand TW. Stability and heterogeneity in the antimicrobiota reactivity of human milk-derived immunoglobulin A. J Exp Med 2023; 220:e20220839. [PMID: 37462916 PMCID: PMC10354535 DOI: 10.1084/jem.20220839] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 04/11/2023] [Accepted: 06/15/2023] [Indexed: 07/21/2023] Open
Abstract
Immunoglobulin A (IgA) is secreted into breast milk and is critical for both protecting against enteric pathogens and shaping the infant intestinal microbiota. The efficacy of breast milk-derived maternal IgA (BrmIgA) is dependent upon its specificity; however, heterogeneity in BrmIgA binding ability to the infant microbiota is not known. Using a flow cytometric array, we analyzed the reactivity of BrmIgA against bacteria common to the infant microbiota and discovered substantial heterogeneity between all donors, independent of preterm or term delivery. Surprisingly, we also observed intradonor variability in the BrmIgA response to closely related bacterial isolates. Conversely, longitudinal analysis showed that the antibacterial BrmIgA reactivity was relatively stable through time, even between sequential infants, indicating that mammary gland IgA responses are durable. Together, our study demonstrates that the antibacterial BrmIgA reactivity displays interindividual heterogeneity but intraindividual stability. These findings have important implications for how breast milk shapes the development of the preterm infant microbiota and protects against necrotizing enterocolitis.
Collapse
Affiliation(s)
- Chelseá B. Johnson-Hence
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kathyayini P. Gopalakrishna
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Darren Bodkin
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kara E. Coffey
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pediatrics, Division of Allergy and Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ansen H.P. Burr
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Syed Rahman
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ali T. Rai
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Darryl A. Abbott
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yelissa A. Sosa
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Justin T. Tometich
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jishnu Das
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Timothy W. Hand
- Pediatrics Department, Infectious Disease Section, R.K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
17
|
Bourgonje AR, Andreu-Sánchez S, Vogl T, Hu S, Vich Vila A, Gacesa R, Leviatan S, Kurilshikov A, Klompus S, Kalka IN, van Dullemen HM, Weinberger A, Visschedijk MC, Festen EAM, Faber KN, Wijmenga C, Dijkstra G, Segal E, Fu J, Zhernakova A, Weersma RK. Phage-display immunoprecipitation sequencing of the antibody epitope repertoire in inflammatory bowel disease reveals distinct antibody signatures. Immunity 2023; 56:1393-1409.e6. [PMID: 37164015 DOI: 10.1016/j.immuni.2023.04.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/13/2022] [Accepted: 04/14/2023] [Indexed: 05/12/2023]
Abstract
Inflammatory bowel diseases (IBDs), e.g., Crohn's disease (CD) and ulcerative colitis (UC), are chronic immune-mediated inflammatory diseases. A comprehensive overview of an IBD-specific antibody epitope repertoire is, however, lacking. Using high-throughput phage-display immunoprecipitation sequencing (PhIP-Seq), we identified antibodies against 344,000 antimicrobial, immune, and food antigens in 497 individuals with IBD compared with 1,326 controls. IBD was characterized by 373 differentially abundant antibody responses (202 overrepresented and 171 underrepresented), with 17% shared by both IBDs, 55% unique to CD, and 28% unique to UC. Antibody reactivities against bacterial flagellins dominated in CD and were associated with ileal involvement, fibrostenotic disease, and anti-Saccharomyces cerevisiae antibody positivity, but not with fecal microbiome composition. Antibody epitope repertoires accurately discriminated CD from controls (area under the curve [AUC] = 0.89), and similar discrimination was achieved when using only ten antibodies (AUC = 0.87). Individuals with IBD thus show a distinct antibody repertoire against selected peptides, allowing clinical stratification and discovery of immunological targets.
Collapse
Affiliation(s)
- Arno R Bourgonje
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Sergio Andreu-Sánchez
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Thomas Vogl
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel; Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University Graz, Graz, Austria; Center for Cancer Research, Medical University of Vienna, Wien, Austria
| | - Shixian Hu
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Arnau Vich Vila
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Ranko Gacesa
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Sigal Leviatan
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Alexander Kurilshikov
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Shelley Klompus
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Iris N Kalka
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Hendrik M van Dullemen
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Adina Weinberger
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Marijn C Visschedijk
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Eleonora A M Festen
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Klaas Nico Faber
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Gerard Dijkstra
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Eran Segal
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Jingyuan Fu
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
18
|
Conrey PE, Denu L, O’Boyle KC, Rozich I, Green J, Maslanka J, Lubin JB, Duranova T, Haltzman BL, Gianchetti L, Oldridge DA, De Luna N, Vella LA, Allman D, Spergel J, Tanes C, Bittinger K, Henrickson SE, Silverman MA. IgA deficiency destabilizes homeostasis toward intestinal microbes and increases systemic immune dysregulation. Sci Immunol 2023; 8:eade2335. [PMID: 37235682 PMCID: PMC11623094 DOI: 10.1126/sciimmunol.ade2335] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 05/05/2023] [Indexed: 05/28/2023]
Abstract
The ability of most patients with selective immunoglobulin A (IgA) deficiency (SIgAD) to remain apparently healthy has been a persistent clinical conundrum. Compensatory mechanisms, including IgM, have been proposed, yet it remains unclear how secretory IgA and IgM work together in the mucosal system and, on a larger scale, whether the systemic and mucosal anti-commensal responses are redundant or have unique features. To address this gap in knowledge, we developed an integrated host-commensal approach combining microbial flow cytometry and metagenomic sequencing (mFLOW-Seq) to comprehensively define which microbes induce mucosal and systemic antibodies. We coupled this approach with high-dimensional immune profiling to study a cohort of pediatric patients with SIgAD and household control siblings. We found that mucosal and systemic antibody networks cooperate to maintain homeostasis by targeting a common subset of commensal microbes. In IgA-deficiency, we find increased translocation of specific bacterial taxa associated with elevated levels of systemic IgG targeting fecal microbiota. Associated features of immune system dysregulation in IgA-deficient mice and humans included elevated levels of inflammatory cytokines, enhanced follicular CD4 T helper cell frequency and activation, and an altered CD8 T cell activation state. Although SIgAD is clinically defined by the absence of serum IgA, the symptomatology and immune dysregulation were concentrated in the SIgAD participants who were also fecal IgA deficient. These findings reveal that mucosal IgA deficiency leads to aberrant systemic exposures and immune responses to commensal microbes, which increase the likelihood of humoral and cellular immune dysregulation and symptomatic disease in patients with IgA deficiency.
Collapse
Affiliation(s)
- Peyton E. Conrey
- Division of Allergy Immunology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Lidiya Denu
- Division of Infectious Disease, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Kaitlin C. O’Boyle
- Division of Allergy Immunology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Isaiah Rozich
- Division of Infectious Disease, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jamal Green
- Division of Infectious Disease, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jeffrey Maslanka
- Division of Infectious Disease, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jean-Bernard Lubin
- Division of Infectious Disease, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Tereza Duranova
- Division of Infectious Disease, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | - Lauren Gianchetti
- Division of Infectious Disease, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Derek A. Oldridge
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Institute for Immunology, IFI, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Nina De Luna
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Laura A. Vella
- Division of Infectious Disease, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Immunology, IFI, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - David Allman
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Institute for Immunology, IFI, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Jonathan Spergel
- Division of Allergy Immunology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Institute for Immunology, IFI, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Ceylan Tanes
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Kyle Bittinger
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Sarah E. Henrickson
- Division of Allergy Immunology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Immunology, IFI, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael A. Silverman
- Division of Infectious Disease, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Immunology, IFI, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
19
|
Burkhard R, Koegler M, Brown K, Wilson K, Mager LF, Zucoloto AZ, Thomson C, Hebbandi Nanjundappa R, Skalosky I, Ahmadi S, McDonald B, Geuking MB. Intestinal colonization regulates systemic anti-commensal immune sensitivity and hyperreactivity. Front Immunol 2023; 14:1030395. [PMID: 37283756 PMCID: PMC10239946 DOI: 10.3389/fimmu.2023.1030395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 05/04/2023] [Indexed: 06/08/2023] Open
Abstract
Healthy host-microbial mutualism with our intestinal microbiota relies to a large degree on compartmentalization and careful regulation of adaptive mucosal and systemic anti-microbial immune responses. However, commensal intestinal bacteria are never exclusively or permanently restricted to the intestinal lumen and regularly reach the systemic circulation. This results in various degrees of commensal bacteremia that needs to be appropriately dealt with by the systemic immune system. While most intestinal commensal bacteria, except for pathobionts or opportunistic pathogen, have evolved to be non-pathogenic, this does not mean that they are non-immunogenic. Mucosal immune adaptation is carefully controlled and regulated to avoid an inflammatory response, but the systemic immune system usually responds differently and more vigorously to systemic bacteremia. Here we show that germ-free mice have increased systemic immune sensitivity and display anti-commensal hyperreactivity in response to the addition of a single defined T helper cell epitope to the outer membrane porin C (OmpC) of a commensal Escherichia coli strain demonstrated by increased E. coli-specific T cell-dependent IgG responses following systemic priming. This increased systemic immune sensitivity was not observed in mice colonized with a defined microbiota at birth indicating that intestinal commensal colonization also regulates systemic, and not only mucosal, anti-commensal responses. The observed increased immunogenicity of the E. coli strain with the modified OmpC protein was not due to a loss of function and associated metabolic changes as a control E. coli strain without OmpC did not display increased immunogenicity.
Collapse
Affiliation(s)
- Regula Burkhard
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mia Koegler
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kirsty Brown
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kirsten Wilson
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Lukas F. Mager
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Amanda Z. Zucoloto
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Carolyn Thomson
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Roopa Hebbandi Nanjundappa
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Isla Skalosky
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Shokouh Ahmadi
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Braedon McDonald
- Snyder Institute of Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Immunology Research Group, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Markus B. Geuking
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute of Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Immunology Research Group, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
20
|
Johnson-Hence CB, Gopalakrishna KP, Bodkin D, Coffey KE, Burr AH, Rahman S, Rai AT, Abbott DA, Sosa YA, Tometich JT, Das J, Hand TW. Stability and heterogeneity in the anti-microbiota reactivity of human milk-derived Immunoglobulin A. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.16.532940. [PMID: 36993366 PMCID: PMC10055037 DOI: 10.1101/2023.03.16.532940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
UNLABELLED Immunoglobulin A (IgA) is secreted into breast milk and is critical to both protecting against enteric pathogens and shaping the infant intestinal microbiota. The efficacy of breast milk-derived maternal IgA (BrmIgA) is dependent upon its specificity, however heterogeneity in BrmIgA binding ability to the infant microbiota is not known. Using a flow cytometric array, we analyzed the reactivity of BrmIgA against bacteria common to the infant microbiota and discovered substantial heterogeneity between all donors, independent of preterm or term delivery. We also observed intra-donor variability in the BrmIgA response to closely related bacterial isolates. Conversely, longitudinal analysis showed that the anti-bacterial BrmIgA reactivity was relatively stable through time, even between sequential infants, indicating that mammary gland IgA responses are durable. Together, our study demonstrates that the anti-bacterial BrmIgA reactivity displays inter-individual heterogeneity but intra-individual stability. These findings have important implications for how breast milk shapes the development of the infant microbiota and protects against Necrotizing Enterocolitis. SUMMARY We analyze the ability of breast milk-derived Immunoglobulin A (IgA) antibodies to bind the infant intestinal microbiota. We discover that each mother secretes into their breast milk a distinct set of IgA antibodies that are stably maintained over time.
Collapse
Affiliation(s)
- Chelseá B. Johnson-Hence
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh PA, 15224
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Texas Southwestern Medical Center
| | - Kathyayini P. Gopalakrishna
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh PA, 15224
| | - Darren Bodkin
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh PA, 15224
| | - Kara E. Coffey
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh PA, 15224
- Department of Pediatrics, Division of Allergy and Immunology, University of Pittsburgh School of Medicine
| | - Ansen H.P. Burr
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh PA, 15224
- Department of Immunology, University of Pittsburgh School of Medicine
| | - Syed Rahman
- Department of Immunology, University of Pittsburgh School of Medicine
- Center for Systems Immunology, University of Pittsburgh School of Medicine
| | - Ali T. Rai
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh PA, 15224
| | - Darryl A. Abbott
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh PA, 15224
| | - Yelissa A. Sosa
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh PA, 15224
| | - Justin T. Tometich
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh PA, 15224
| | - Jishnu Das
- Department of Immunology, University of Pittsburgh School of Medicine
- Center for Systems Immunology, University of Pittsburgh School of Medicine
| | - Timothy W. Hand
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh PA, 15224
- Department of Immunology, University of Pittsburgh School of Medicine
| |
Collapse
|
21
|
Ceglia S, Berthelette A, Howley K, Li Y, Mortzfeld B, Bhattarai SK, Yiew NKH, Xu Y, Brink R, Cyster JG, Hooper LV, Randolph GJ, Bucci V, Reboldi A. An epithelial cell-derived metabolite tunes immunoglobulin A secretion by gut-resident plasma cells. Nat Immunol 2023; 24:531-544. [PMID: 36658240 PMCID: PMC10243503 DOI: 10.1038/s41590-022-01413-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 12/14/2022] [Indexed: 01/21/2023]
Abstract
Immunoglobulin A (IgA) secretion by plasma cells, terminally differentiated B cells residing in the intestinal lamina propria, assures microbiome homeostasis and protects the host against enteric infections. Exposure to diet-derived and commensal-derived signals provides immune cells with organizing cues that instruct their effector function and dynamically shape intestinal immune responses at the mucosal barrier. Recent data have described metabolic and microbial inputs controlling T cell and innate lymphoid cell activation in the gut; however, whether IgA-secreting lamina propria plasma cells are tuned by local stimuli is completely unknown. Although antibody secretion is considered to be imprinted during B cell differentiation and therefore largely unaffected by environmental changes, a rapid modulation of IgA levels in response to intestinal fluctuations might be beneficial to the host. In the present study, we showed that dietary cholesterol absorption and commensal recognition by duodenal intestinal epithelial cells lead to the production of oxysterols, evolutionarily conserved lipids with immunomodulatory functions. Using conditional cholesterol 25-hydroxylase deleter mouse line we demonstrated that 7α,25-dihydroxycholesterol from epithelial cells is critical to restrain IgA secretion against commensal- and pathogen-derived antigens in the gut. Intestinal plasma cells sense oxysterols via the chemoattractant receptor GPR183 and couple their tissue positioning with IgA secretion. Our findings revealed a new mechanism linking dietary cholesterol and humoral immune responses centered around plasma cell localization for efficient mucosal protection.
Collapse
Affiliation(s)
- Simona Ceglia
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Alyssa Berthelette
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Kelsey Howley
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Yun Li
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Benedikt Mortzfeld
- Department of Microbiology and Physiological systems, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Shakti K Bhattarai
- Department of Microbiology and Physiological systems, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Nicole K H Yiew
- Department of Pathology and Immunology, Washington University Medical School, St. Louis, MO, USA
| | - Ying Xu
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Robert Brink
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Lora V Hooper
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University Medical School, St. Louis, MO, USA
| | - Vanni Bucci
- Department of Microbiology and Physiological systems, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Andrea Reboldi
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
22
|
Weller S, Sterlin D, Fadeev T, Coignard E, de los Aires AV, Goetz C, Fritzen R, Bahuaud M, Batteux F, Gorochov G, Weill JC, Reynaud CA. T-independent responses to polysaccharides in humans mobilize marginal zone B cells prediversified against gut bacterial antigens. Sci Immunol 2023; 8:eade1413. [PMID: 36706172 PMCID: PMC7614366 DOI: 10.1126/sciimmunol.ade1413] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 01/04/2023] [Indexed: 01/29/2023]
Abstract
Marginal zone (MZ) B cells are one of the main actors of T-independent (TI) responses in mice. To identify the B cell subset(s) involved in such responses in humans, we vaccinated healthy individuals with Pneumovax, a model TI vaccine. By high-throughput repertoire sequencing of plasma cells (PCs) isolated 7 days after vaccination and of different B cell subpopulations before and after vaccination, we show that the PC response mobilizes large clones systematically, including an immunoglobulin M component, whose diversification and amplification predated the pneumococcal vaccination. These clones could be mainly traced back to MZ B cells, together with clonally related IgA+ and, to a lesser extent, IgG+CD27+ B cells. Recombinant monoclonal antibodies isolated from large PC clones recognized a wide array of bacterial species from the gut flora, indicating that TI responses in humans largely mobilize MZ and switched B cells that most likely prediversified during mucosal immune responses against bacterial antigens and acquired pneumococcal cross-reactivity through somatic hypermutation.
Collapse
Affiliation(s)
- Sandra Weller
- Université Paris Cité, INSERM U1151, CNRS UMR-8253, Institut Necker Enfants Malades (INEM), F-75015 Paris, France
| | - Delphine Sterlin
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), F-75013 Paris, France
- Département d’Immunologie, Assistance Publique Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, F-75013 Paris, France
| | - Tatiana Fadeev
- Université Paris Cité, INSERM U1151, CNRS UMR-8253, Institut Necker Enfants Malades (INEM), F-75015 Paris, France
| | - Eva Coignard
- Université Paris Cité, INSERM U1151, CNRS UMR-8253, Institut Necker Enfants Malades (INEM), F-75015 Paris, France
| | - Alba Verge de los Aires
- Université Paris Cité, INSERM U1151, CNRS UMR-8253, Institut Necker Enfants Malades (INEM), F-75015 Paris, France
| | - Clara Goetz
- Université Paris Cité, INSERM U1151, CNRS UMR-8253, Institut Necker Enfants Malades (INEM), F-75015 Paris, France
| | - Rémi Fritzen
- Université Paris Cité, INSERM U1151, CNRS UMR-8253, Institut Necker Enfants Malades (INEM), F-75015 Paris, France
- School of Medicine, University of St Andrews, St Andrews, UK
| | - Mathilde Bahuaud
- Université Paris Cité, INSERM U1016, Institut Cochin, F-75014 Paris, France
- Service d’Immunologie Biologique, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, F-75014 Paris, France
| | - Frederic Batteux
- Université Paris Cité, INSERM U1016, Institut Cochin, F-75014 Paris, France
- Service d’Immunologie Biologique, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, F-75014 Paris, France
| | - Guy Gorochov
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), F-75013 Paris, France
- Département d’Immunologie, Assistance Publique Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, F-75013 Paris, France
| | - Jean-Claude Weill
- Université Paris Cité, INSERM U1151, CNRS UMR-8253, Institut Necker Enfants Malades (INEM), F-75015 Paris, France
| | - Claude-Agnès Reynaud
- Université Paris Cité, INSERM U1151, CNRS UMR-8253, Institut Necker Enfants Malades (INEM), F-75015 Paris, France
| |
Collapse
|
23
|
Metabolic reconstitution of germ-free mice by a gnotobiotic microbiota varies over the circadian cycle. PLoS Biol 2022; 20:e3001743. [PMID: 36126044 PMCID: PMC9488797 DOI: 10.1371/journal.pbio.3001743] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 07/06/2022] [Indexed: 12/17/2022] Open
Abstract
The capacity of the intestinal microbiota to degrade otherwise indigestible diet components is known to greatly improve the recovery of energy from food. This has led to the hypothesis that increased digestive efficiency may underlie the contribution of the microbiota to obesity. OligoMM12-colonized gnotobiotic mice have a consistently higher fat mass than germ-free (GF) or fully colonized counterparts. We therefore investigated their food intake, digestion efficiency, energy expenditure, and respiratory quotient using a novel isolator-housed metabolic cage system, which allows long-term measurements without contamination risk. This demonstrated that microbiota-released calories are perfectly balanced by decreased food intake in fully colonized versus gnotobiotic OligoMM12 and GF mice fed a standard chow diet, i.e., microbiota-released calories can in fact be well integrated into appetite control. We also observed no significant difference in energy expenditure after normalization by lean mass between the different microbiota groups, suggesting that cumulative small differences in energy balance, or altered energy storage, must underlie fat accumulation in OligoMM12 mice. Consistent with altered energy storage, major differences were observed in the type of respiratory substrates used in metabolism over the circadian cycle: In GF mice, the respiratory exchange ratio (RER) was consistently lower than that of fully colonized mice at all times of day, indicative of more reliance on fat and less on glucose metabolism. Intriguingly, the RER of OligoMM12-colonized gnotobiotic mice phenocopied fully colonized mice during the dark (active/eating) phase but phenocopied GF mice during the light (fasting/resting) phase. Further, OligoMM12-colonized mice showed a GF-like drop in liver glycogen storage during the light phase and both liver and plasma metabolomes of OligoMM12 mice clustered closely with GF mice. This implies the existence of microbiota functions that are required to maintain normal host metabolism during the resting/fasting phase of circadian cycle and which are absent in the OligoMM12 consortium.
Collapse
|
24
|
Vujkovic-Cvijin I, Welles HC, Ha CWY, Huq L, Mistry S, Brenchley JM, Trinchieri G, Devkota S, Belkaid Y. The systemic anti-microbiota IgG repertoire can identify gut bacteria that translocate across gut barrier surfaces. Sci Transl Med 2022; 14:eabl3927. [PMID: 35976997 PMCID: PMC9741845 DOI: 10.1126/scitranslmed.abl3927] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Unique gut microbiota compositions have been associated with inflammatory diseases, but identifying gut bacterial functions linked to immune activation in humans remains challenging. Translocation of pathogens from mucosal surfaces into peripheral tissues can elicit immune activation, although whether and which gut commensal bacteria translocate in inflammatory diseases is difficult to assess. We report that a subset of commensal gut microbiota constituents that translocate across the gut barrier in mice and humans are associated with heightened systemic immunoglobulin G (IgG) responses. We present a modified high-throughput, culture-independent approach to quantify systemic IgG against gut commensal bacteria in human serum samples without the need for paired stool samples. Using this approach, we highlight several commensal bacterial species that elicit elevated IgG responses in patients with inflammatory bowel disease (IBD) including taxa within the clades Collinsella, Bifidobacterium, Lachnospiraceae, and Ruminococcaceae. These and other taxa identified as translocating bacteria or targets of systemic immunity in IBD concomitantly exhibited heightened transcriptional activity and growth rates in IBD patient gut microbiomes. Our approach represents a complementary tool to illuminate interactions between the host and its gut microbiota and may provide an additional method to identify microbes linked to inflammatory disease.
Collapse
Affiliation(s)
- Ivan Vujkovic-Cvijin
- Metaorganism Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Hugh C. Welles
- Metaorganism Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Connie W. Y. Ha
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Lutfi Huq
- Metaorganism Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA
| | - Shreni Mistry
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Jason M. Brenchley
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA
| | - Giorgio Trinchieri
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Suzanne Devkota
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| |
Collapse
|
25
|
Rice TA, Bielecka AA, Nguyen MT, Rosen CE, Song D, Sonnert ND, Yang Y, Cao Y, Khetrapal V, Catanzaro JR, Martin AL, Rashed SA, Leopold SR, Hao L, Yu X, van Dijk D, Ring AM, Flavell RA, de Zoete MR, Palm NW. Interspecies commensal interactions have nonlinear impacts on host immunity. Cell Host Microbe 2022; 30:988-1002.e6. [PMID: 35640610 PMCID: PMC9283318 DOI: 10.1016/j.chom.2022.05.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/22/2022] [Accepted: 05/04/2022] [Indexed: 02/03/2023]
Abstract
The impacts of individual commensal microbes on immunity and disease can differ dramatically depending on the surrounding microbial context; however, the specific bacterial combinations that dictate divergent immunological outcomes remain largely undefined. Here, we characterize an immunostimulatory Allobaculum species from an inflammatory bowel disease patient that exacerbates colitis in gnotobiotic mice. Allobaculum inversely associates with the taxonomically divergent immunostimulatory species Akkermansia muciniphila in human-microbiota-associated mice and human cohorts. Co-colonization with A. muciniphila ameliorates Allobaculum-induced intestinal epithelial cell activation and colitis in mice, whereas Allobaculum blunts the A.muciniphila-specific systemic antibody response and reprograms the immunological milieu in mesenteric lymph nodes by blocking A.muciniphila-induced dendritic cell activation and T cell expansion. These studies thus identify a pairwise reciprocal interaction between human gut bacteria that dictates divergent immunological outcomes. Furthermore, they establish a generalizable framework to define the contextual cues contributing to the "incomplete penetrance" of microbial impacts on human disease.
Collapse
Affiliation(s)
- Tyler A. Rice
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Agata A. Bielecka
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA,Present address: Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Mytien T. Nguyen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Connor E. Rosen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA,Present address: Arcus Biosciences, Hayward, CA 94545, USA
| | - Deguang Song
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Nicole D. Sonnert
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Yi Yang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Yiyun Cao
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Varnica Khetrapal
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jason R. Catanzaro
- Section of Pulmonology, Allergy, Immunology, and Sleep Medicine, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Anjelica L. Martin
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Saleh A. Rashed
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA,Present address: Center for Precision Cancer Medicine, Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA
| | - Shana R. Leopold
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Liming Hao
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Xuezhu Yu
- Department of Computer Science, Yale University, New Haven, CT 06520, USA
| | - David van Dijk
- Department of Computer Science, Yale University, New Haven, CT 06520, USA
| | - Aaron M. Ring
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Richard A. Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Marcel R. de Zoete
- Department of Medical Microbiology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Noah W. Palm
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA,Lead Contact: Noah W. Palm:
| |
Collapse
|
26
|
Michaud E, Waeckel L, Gayet R, Goguyer-Deschaumes R, Chanut B, Jospin F, Bathany K, Monnoye M, Genet C, Prier A, Tokarski C, Gérard P, Roblin X, Rochereau N, Paul S. Alteration of microbiota antibody-mediated immune selection contributes to dysbiosis in inflammatory bowel diseases. EMBO Mol Med 2022; 14:e15386. [PMID: 35785473 PMCID: PMC9358401 DOI: 10.15252/emmm.202115386] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 06/15/2022] [Accepted: 06/15/2022] [Indexed: 11/09/2022] Open
Abstract
Human secretory immunoglobulins (SIg) A1 and SIgA2 guide mucosal responses toward tolerance or inflammation, notably through reverse-transcytosis, the apical-to-basal transport of IgA2 immune complexes via M cells of gut Peyer's patches. As such, the maintenance of a diverse gut microbiota requires broad affinity IgA and glycan-glycan interaction. Here, we asked whether IgA1 and IgA2-microbiota interactions might be involved in dysbiosis induction during inflammatory bowel diseases. Using stool HPLC-purified IgA, we show that reverse-transcytosis is abrogated in ulcerative colitis (UC) while it is extended to IgA1 in Crohn's disease (CD). 16S RNA sequencing of IgA-bound microbiota in CD and UC showed distinct IgA1- and IgA2-associated microbiota; the IgA1+ fraction of CD microbiota was notably enriched in beneficial commensals. These features were associated with increased IgA anti-glycan reactivity in CD and an opposite loss of reactivity in UC. Our results highlight previously unknown pathogenic properties of IgA in IBD that could support dysbiosis.
Collapse
Affiliation(s)
- Eva Michaud
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, Saint-Etienne, France
| | - Louis Waeckel
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, Saint-Etienne, France
| | - Rémi Gayet
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, Saint-Etienne, France
| | - Roman Goguyer-Deschaumes
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, Saint-Etienne, France
| | - Blandine Chanut
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, Saint-Etienne, France
| | - Fabienne Jospin
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, Saint-Etienne, France
| | - Katell Bathany
- Chimie et Biologie des Membranes et des Nano-objets (UMR 5248), Université de Bordeaux, CNRS, Bordeaux INP, Pessac, France
| | - Magali Monnoye
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Coraline Genet
- Inserm UMR 1098 Right, Université Bourgogne Franche-Comté, Besançon, France
| | - Amelie Prier
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, Saint-Etienne, France
| | - Caroline Tokarski
- Chimie et Biologie des Membranes et des Nano-objets (UMR 5248), Université de Bordeaux, CNRS, Bordeaux INP, Pessac, France
| | - Philippe Gérard
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Xavier Roblin
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, Saint-Etienne, France
| | - Nicolas Rochereau
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, Saint-Etienne, France
| | - Stéphane Paul
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, Saint-Etienne, France
| |
Collapse
|
27
|
Rodrigues MO, Isoppo VG, Moro AV, Rodembusch FS. Photoactive organic-inorganic hybrid materials: From silylated compounds to optical applications. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY C: PHOTOCHEMISTRY REVIEWS 2022. [DOI: 10.1016/j.jphotochemrev.2021.100474] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
28
|
Bourgonje AR, Vogl T, Segal E, Weersma RK. Antibody signatures in inflammatory bowel disease: current developments and future applications. Trends Mol Med 2022; 28:693-705. [DOI: 10.1016/j.molmed.2022.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/01/2022] [Accepted: 05/03/2022] [Indexed: 11/25/2022]
|
29
|
Goncalves P, Doisne JM, Eri T, Charbit B, Bondet V, Posseme C, Llibre A, Casrouge A, Lenoir C, Neven B, Duffy D, Fischer A, Di Santo JP, The Milieu Intérieur Consortium. Defects in mucosal immunity and nasopharyngeal dysbiosis in HSC-transplanted SCID patients with IL2RG/JAK3 deficiency. Blood 2022; 139:2585-2600. [PMID: 35157765 PMCID: PMC11022929 DOI: 10.1182/blood.2021014654] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/31/2022] [Indexed: 11/20/2022] Open
Abstract
Both innate and adaptive lymphocytes have critical roles in mucosal defense that contain commensal microbial communities and protect against pathogen invasion. Here we characterize mucosal immunity in patients with severe combined immunodeficiency (SCID) receiving hematopoietic stem cell transplantation (HSCT) with or without myeloablation. We confirmed that pretransplant conditioning had an impact on innate (natural killer and innate lymphoid cells) and adaptive (B and T cells) lymphocyte reconstitution in these patients with SCID and now show that this further extends to generation of T helper 2 and type 2 cytotoxic T cells. Using an integrated approach to assess nasopharyngeal immunity, we identified a local mucosal defect in type 2 cytokines, mucus production, and a selective local immunoglobulin A (IgA) deficiency in HSCT-treated SCID patients with genetic defects in IL2RG/GC or JAK3. These patients have a reduction in IgA-coated nasopharyngeal bacteria and exhibit microbial dysbiosis with increased pathobiont carriage. Interestingly, intravenous immunoglobulin replacement therapy can partially normalize nasopharyngeal immunoglobulin profiles and restore microbial communities in GC/JAK3 patients. Together, our results suggest a potential nonredundant role for type 2 immunity and/or of local IgA antibody production in the maintenance of nasopharyngeal microbial homeostasis and mucosal barrier function.
Collapse
Affiliation(s)
- Pedro Goncalves
- Institut Pasteur, Université de Paris Cité, Inserm U1223, Innate Immunity Unit, Paris, France
| | - Jean-Marc Doisne
- Institut Pasteur, Université de Paris Cité, Inserm U1223, Innate Immunity Unit, Paris, France
| | - Toshiki Eri
- Institut Pasteur, Université de Paris Cité, Inserm U1223, Innate Immunity Unit, Paris, France
| | - Bruno Charbit
- Institut Pasteur, Université de Paris Cité, Center for Translational Science, Paris, France
| | - Vincent Bondet
- Institut Pasteur, Université de Paris Cité, Translational Immunology Unit, Paris, France
| | - Celine Posseme
- Institut Pasteur, Université de Paris Cité, Translational Immunology Unit, Paris, France
| | - Alba Llibre
- Institut Pasteur, Université de Paris Cité, Translational Immunology Unit, Paris, France
| | - Armanda Casrouge
- Institut Pasteur, Université de Paris Cité, Inserm U1223, Innate Immunity Unit, Paris, France
| | - Christelle Lenoir
- Inserm Unité Mixte de Recherche 1163, Paris, France
- Imagine Institut, Université de Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Bénédicte Neven
- Inserm Unité Mixte de Recherche 1163, Paris, France
- Department of Pediatric Immunology, Hematology and Rheumatology, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Darragh Duffy
- Institut Pasteur, Université de Paris Cité, Translational Immunology Unit, Paris, France
| | - Alain Fischer
- Inserm Unité Mixte de Recherche 1163, Paris, France
- Collège de France, Paris, France
| | - James P. Di Santo
- Institut Pasteur, Université de Paris Cité, Inserm U1223, Innate Immunity Unit, Paris, France
| | - The Milieu Intérieur Consortium
- Institut Pasteur, Université de Paris Cité, Inserm U1223, Innate Immunity Unit, Paris, France
- Institut Pasteur, Université de Paris Cité, Center for Translational Science, Paris, France
- Institut Pasteur, Université de Paris Cité, Translational Immunology Unit, Paris, France
- Inserm Unité Mixte de Recherche 1163, Paris, France
- Imagine Institut, Université de Paris Descartes Sorbonne Paris Cité, Paris, France
- Department of Pediatric Immunology, Hematology and Rheumatology, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
- Collège de France, Paris, France
| |
Collapse
|
30
|
Application of antibiotic-derived fluorescent probes to bacterial studies. Methods Enzymol 2022; 665:1-28. [DOI: 10.1016/bs.mie.2021.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
31
|
Silverman MA, Green JL. Insight Into Host-Microbe Interactions Using Microbial Flow Cytometry Coupled to Next-Generation Sequencing. J Pediatric Infect Dis Soc 2021; 10:S106-S111. [PMID: 34951471 PMCID: PMC8703255 DOI: 10.1093/jpids/piab092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Antibody-based assays have been a cornerstone of infectious disease diagnostics for over 100 years [1]. These assays rely on the exquisite sensitivity and specificity of humoral response to almost all infections. While next-generation sequencing (NGS) has tremendous potential to improve diagnostics and uncover host-microbial relationships by directly identifying nucleic acids from infectious microbes, challenges and opportunities for new approaches remain. Here, we review a group of cutting-edge techniques that couple antibody responses with flow cytometry of antibody tagged microbes and NGS. These studies are bringing into focus the dynamic relationship between our immune systems and endogenous microbial communities, which are an important source of pathogens. For simplicity, we use the umbrella term mFLOW-Seq (microbial flow cytometry coupled to NGS) to describe these approaches.
Collapse
Affiliation(s)
- Michael A Silverman
- Department of Pediatrics, Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA,Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA,Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA,Corresponding Author: Michael A. Silverman, MD, PhD, Department of Pediatrics, The Children’s Hospital of Philadelphia, 3615 Civic Center Blvd. Abramson Research Center, 12th floor, Room 1202, Philadelphia, PA 19104, USA. E-mail:
| | - Jamal L Green
- Department of Pediatrics, Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA,Biomedical Graduate Studies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
32
|
Trindade BC, Ceglia S, Berthelette A, Raso F, Howley K, Muppidi JR, Reboldi A. The cholesterol metabolite 25-hydroxycholesterol restrains the transcriptional regulator SREBP2 and limits intestinal IgA plasma cell differentiation. Immunity 2021; 54:2273-2287.e6. [PMID: 34644558 DOI: 10.1016/j.immuni.2021.09.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/10/2021] [Accepted: 09/08/2021] [Indexed: 12/24/2022]
Abstract
Diets high in cholesterol alter intestinal immunity. Here, we examined how the cholesterol metabolite 25-hydroxycholesterol (25-HC) impacts the intestinal B cell response. Mice lacking cholesterol 25-hydroxylase (CH25H), the enzyme generating 25-HC, had higher frequencies of immunoglobulin A (IgA)-secreting antigen-specific B cells upon immunization or infection. 25-HC did not affect class-switch recombination but rather restrained plasma cell (PC) differentiation. 25-HC was produced by follicular dendritic cells and increased in response to dietary cholesterol. Mechanistically, 25-HC restricted activation of the sterol-sensing transcription factor SREBP2, thereby regulating B cell cholesterol biosynthesis. Ectopic expression of SREBP2 in germinal center B cells induced rapid PC differentiation, whereas SREBP2 deficiency reduced PC output in vitro and in vivo. High-cholesterol diet impaired, whereas Ch25h deficiency enhanced, the IgA response against Salmonella and the resulting protection from systemic bacterial dissemination. Thus, a 25-HC-SREBP2 axis shapes the humoral response at the intestinal barrier, providing insight into the effect of high dietary cholesterol in intestinal immunity.
Collapse
Affiliation(s)
- Bruno C Trindade
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Simona Ceglia
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Alyssa Berthelette
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Fiona Raso
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Kelsey Howley
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jagan R Muppidi
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andrea Reboldi
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
33
|
Population-wide diversity and stability of serum antibody epitope repertoires against human microbiota. Nat Med 2021; 27:1442-1450. [PMID: 34282338 DOI: 10.1038/s41591-021-01409-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022]
Abstract
Serum antibodies can recognize both pathogens and commensal gut microbiota. However, our current understanding of antibody repertoires is largely based on DNA sequencing of the corresponding B-cell receptor genes, and actual bacterial antigen targets remain incompletely characterized. Here we have profiled the serum antibody responses of 997 healthy individuals against 244,000 rationally selected peptide antigens derived from gut microbiota and pathogenic and probiotic bacteria. Leveraging phage immunoprecipitation sequencing (PhIP-Seq) based on phage-displayed synthetic oligo libraries, we detect a wide breadth of individual-specific as well as shared antibody responses against microbiota that associate with age and gender. We also demonstrate that these antibody epitope repertoires are more longitudinally stable than gut microbiome species abundances. Serum samples of more than 200 individuals collected five years apart could be accurately matched and could serve as an immunologic fingerprint. Overall, our results suggest that systemic antibody responses provide a non-redundant layer of information about microbiota beyond gut microbial species composition.
Collapse
|
34
|
Diard M, Bakkeren E, Lentsch V, Rocker A, Bekele NA, Hoces D, Aslani S, Arnoldini M, Böhi F, Schumann-Moor K, Adamcik J, Piccoli L, Lanzavecchia A, Stadtmueller BM, Donohue N, van der Woude MW, Hockenberry A, Viollier PH, Falquet L, Wüthrich D, Bonfiglio F, Loverdo C, Egli A, Zandomeneghi G, Mezzenga R, Holst O, Meier BH, Hardt WD, Slack E. A rationally designed oral vaccine induces immunoglobulin A in the murine gut that directs the evolution of attenuated Salmonella variants. Nat Microbiol 2021; 6:830-841. [PMID: 34045711 PMCID: PMC7611113 DOI: 10.1038/s41564-021-00911-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 04/14/2021] [Indexed: 12/12/2022]
Abstract
The ability of gut bacterial pathogens to escape immunity by antigenic variation-particularly via changes to surface-exposed antigens-is a major barrier to immune clearance1. However, not all variants are equally fit in all environments2,3. It should therefore be possible to exploit such immune escape mechanisms to direct an evolutionary trade-off. Here, we demonstrate this phenomenon using Salmonella enterica subspecies enterica serovar Typhimurium (S.Tm). A dominant surface antigen of S.Tm is its O-antigen: a long, repetitive glycan that can be rapidly varied by mutations in biosynthetic pathways or by phase variation4,5. We quantified the selective advantage of O-antigen variants in the presence and absence of O-antigen-specific immunoglobulin A and identified a set of evolutionary trajectories allowing immune escape without an associated fitness cost in naive mice. Through the use of rationally designed oral vaccines, we induced immunoglobulin A responses blocking all of these trajectories. This selected for Salmonella mutants carrying deletions of the O-antigen polymerase gene wzyB. Due to their short O-antigen, these evolved mutants were more susceptible to environmental stressors (detergents or complement) and predation (bacteriophages) and were impaired in gut colonization and virulence in mice. Therefore, a rationally induced cocktail of intestinal antibodies can direct an evolutionary trade-off in S.Tm. This lays the foundations for the exploration of mucosal vaccines capable of setting evolutionary traps as a prophylactic strategy.
Collapse
Affiliation(s)
- Médéric Diard
- Biozentrum, University of Basel, Basel, Switzerland.
| | - Erik Bakkeren
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.,Department of Zoology, University of Oxford, Oxford, UK
| | - Verena Lentsch
- Institute of Food, Nutrition and Health, D-HEST, ETH Zürich, Zürich, Switzerland
| | | | | | - Daniel Hoces
- Institute of Food, Nutrition and Health, D-HEST, ETH Zürich, Zürich, Switzerland
| | - Selma Aslani
- Institute of Food, Nutrition and Health, D-HEST, ETH Zürich, Zürich, Switzerland
| | - Markus Arnoldini
- Institute of Food, Nutrition and Health, D-HEST, ETH Zürich, Zürich, Switzerland
| | - Flurina Böhi
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.,Department of Molecular Mechanisms of Disease, University of Zürich, Zürich, Switzerland
| | - Kathrin Schumann-Moor
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.,Division of Surgical Research, University Hospital of Zürich, Zürich, Switzerland
| | - Jozef Adamcik
- Institute of Food, Nutrition and Health, D-HEST, ETH Zürich, Zürich, Switzerland
| | - Luca Piccoli
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Antonio Lanzavecchia
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Beth M Stadtmueller
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Nicholas Donohue
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK.,Department of Orthopedics and Trauma, Medical University of Graz, Graz, Austria
| | - Marjan W van der Woude
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK
| | - Alyson Hockenberry
- Department of Environmental Microbiology, Eawag, Dubendorf, Switzerland.,Department of Environmental Sciences, ETH Zürich, Zürich, Switzerland
| | - Patrick H Viollier
- Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Laurent Falquet
- Department of Biology, University of Fribourg, Fribourg, Switzerland.,Swiss Institute of Bioinformatics, Fribourg, Switzerland
| | - Daniel Wüthrich
- Infection Biology, University Hospital of Basel, Basel, Switzerland
| | | | - Claude Loverdo
- Department of Materials, ETH Zürich, Zürich, Switzerland
| | - Adrian Egli
- Infection Biology, University Hospital of Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Raffaele Mezzenga
- Institute of Food, Nutrition and Health, D-HEST, ETH Zürich, Zürich, Switzerland.,Department of Materials, ETH Zürich, Zürich, Switzerland
| | - Otto Holst
- Forschungszentrum Borstel, Borstel, Germany
| | - Beat H Meier
- Institute for Physical Chemistry, ETH Zürich, Zürich, Switzerland
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.
| | - Emma Slack
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland. .,Institute of Food, Nutrition and Health, D-HEST, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
35
|
Richards A, Baranova DE, Pizzuto MS, Jaconi S, Willsey GG, Torres-Velez FJ, Doering JE, Benigni F, Corti D, Mantis NJ. Recombinant Human Secretory IgA Induces Salmonella Typhimurium Agglutination and Limits Bacterial Invasion into Gut-Associated Lymphoid Tissues. ACS Infect Dis 2021; 7:1221-1235. [PMID: 33728898 PMCID: PMC8154420 DOI: 10.1021/acsinfecdis.0c00842] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Indexed: 12/11/2022]
Abstract
As the predominant antibody type in mucosal secretions, human colostrum, and breast milk, secretory IgA (SIgA) plays a central role in safeguarding the intestinal epithelium of newborns from invasive enteric pathogens like the Gram-negative bacterium Salmonella enterica serovar Typhimurium (STm). SIgA is a complex molecule, consisting of an assemblage of two or more IgA monomers, joining (J)-chain, and secretory component (SC), whose exact functions in neutralizing pathogens are only beginning to be elucidated. In this study, we produced and characterized a recombinant human SIgA variant of Sal4, a well-characterized monoclonal antibody (mAb) specific for the O5-antigen of STm lipopolysaccharide (LPS). We demonstrate by flow cytometry, light microscopy, and fluorescence microscopy that Sal4 SIgA promotes the formation of large, densely packed bacterial aggregates in vitro. In a mouse model, passive oral administration of Sal4 SIgA was sufficient to entrap STm within the intestinal lumen and reduce bacterial invasion into gut-associated lymphoid tissues by several orders of magnitude. Bacterial aggregates induced by Sal4 SIgA treatment in the intestinal lumen were recalcitrant to immunohistochemical staining, suggesting the bacteria were encased in a protective capsule. Indeed, a crystal violet staining assay demonstrated that STm secretes an extracellular matrix enriched in cellulose following even short exposures to Sal4 SIgA. Collectively, these results demonstrate that recombinant human SIgA recapitulates key biological activities associated with mucosal immunity and raises the prospect of oral passive immunization to combat enteric diseases.
Collapse
Affiliation(s)
- Angelene
F. Richards
- Department
of Biomedical Sciences, University at Albany
School of Public Health, Albany, New York 12208, United States
- Division
of Infectious Diseases, Wadsworth Center,
New York State Department of Health, Albany, New York 12208, United States
| | - Danielle E. Baranova
- Division
of Infectious Diseases, Wadsworth Center,
New York State Department of Health, Albany, New York 12208, United States
| | - Matteo S. Pizzuto
- Humabs
BioMed SA a Subsidiary of Vir Biotechnology Inc., 6500 Bellinzona, Switzerland
| | - Stefano Jaconi
- Humabs
BioMed SA a Subsidiary of Vir Biotechnology Inc., 6500 Bellinzona, Switzerland
| | - Graham G. Willsey
- Division
of Infectious Diseases, Wadsworth Center,
New York State Department of Health, Albany, New York 12208, United States
| | - Fernando J. Torres-Velez
- Division
of Infectious Diseases, Wadsworth Center,
New York State Department of Health, Albany, New York 12208, United States
| | - Jennifer E. Doering
- Division
of Infectious Diseases, Wadsworth Center,
New York State Department of Health, Albany, New York 12208, United States
| | - Fabio Benigni
- Humabs
BioMed SA a Subsidiary of Vir Biotechnology Inc., 6500 Bellinzona, Switzerland
| | - Davide Corti
- Humabs
BioMed SA a Subsidiary of Vir Biotechnology Inc., 6500 Bellinzona, Switzerland
| | - Nicholas J. Mantis
- Department
of Biomedical Sciences, University at Albany
School of Public Health, Albany, New York 12208, United States
- Division
of Infectious Diseases, Wadsworth Center,
New York State Department of Health, Albany, New York 12208, United States
| |
Collapse
|
36
|
Sterlin D, Larsen M, Fadlallah J, Parizot C, Vignes M, Autaa G, Dorgham K, Juste C, Lepage P, Aboab J, Vicart S, Maillart E, Gout O, Lubetzki C, Deschamps R, Papeix C, Gorochov G. Perturbed Microbiota/Immune Homeostasis in Multiple Sclerosis. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/4/e997. [PMID: 33975914 PMCID: PMC8114833 DOI: 10.1212/nxi.0000000000000997] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 03/09/2021] [Indexed: 12/21/2022]
Abstract
Objective Based on animal models and human studies, there is now strong suspicion that host/microbiota mutualism in the context of gut microbial dysbiosis could influence immunity and multiple sclerosis (MS) evolution. Our goal was to seek evidence of deregulated microbiota-induced systemic immune responses in patients with MS. Methods We investigated gut and systemic commensal-specific antibody responses in healthy controls (n = 32), patients with relapsing-remitting MS (n = 30), and individuals with clinically isolated syndromes (CISs) (n = 15). Gut microbiota composition and diversity were compared between controls and patients by analysis of 16S ribosomal ribonucleic acid (rRNA) sequencing. Autologous microbiota and cultivable bacterial strains were used in bacterial flow cytometry assays to quantify autologous serum IgG and secretory IgA responses to microbiota. IgG-bound bacteria were sorted by flow cytometry and identified using 16S rRNA sequencing. Results We show that commensal-specific gut IgA responses are drastically reduced in patients with severe MS, disease severity being correlated with the IgA-coated fecal microbiota fraction (r = −0.647, p < 0.0001). At the same time, IgA-unbound bacteria elicit qualitatively broad and quantitatively increased serum IgG responses in patients with MS and CIS compared with controls (4.1% and 2.5% vs 1.9%, respectively, p < 0.001). Conclusions Gut and systemic microbiota/immune homeostasis are perturbed in MS. Our results argue that defective IgA responses in MS are linked to a breakdown of systemic tolerance to gut microbiota leading to an enhanced triggering of systemic IgG immunity against gut commensals occurring early in MS.
Collapse
Affiliation(s)
- Delphine Sterlin
- From the Sorbonne Université (D.S., M.L., J.F., C.P., M.V., G.A., K.D., G.G.), Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), AP-HP Hôpital Pitié-Salpêtrière, France; Université Paris-Saclay (C.J., P.L.), INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France; Hôpital Ophtalmologique Adolphe de Rothschild (J.A., O.G., R.D.), Département de Neurologie, Paris, France; and Sorbonne Université (S.V., E.M., C.L., C.P.), Département de Neurologie, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| | - Martin Larsen
- From the Sorbonne Université (D.S., M.L., J.F., C.P., M.V., G.A., K.D., G.G.), Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), AP-HP Hôpital Pitié-Salpêtrière, France; Université Paris-Saclay (C.J., P.L.), INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France; Hôpital Ophtalmologique Adolphe de Rothschild (J.A., O.G., R.D.), Département de Neurologie, Paris, France; and Sorbonne Université (S.V., E.M., C.L., C.P.), Département de Neurologie, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| | - Jehane Fadlallah
- From the Sorbonne Université (D.S., M.L., J.F., C.P., M.V., G.A., K.D., G.G.), Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), AP-HP Hôpital Pitié-Salpêtrière, France; Université Paris-Saclay (C.J., P.L.), INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France; Hôpital Ophtalmologique Adolphe de Rothschild (J.A., O.G., R.D.), Département de Neurologie, Paris, France; and Sorbonne Université (S.V., E.M., C.L., C.P.), Département de Neurologie, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| | - Christophe Parizot
- From the Sorbonne Université (D.S., M.L., J.F., C.P., M.V., G.A., K.D., G.G.), Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), AP-HP Hôpital Pitié-Salpêtrière, France; Université Paris-Saclay (C.J., P.L.), INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France; Hôpital Ophtalmologique Adolphe de Rothschild (J.A., O.G., R.D.), Département de Neurologie, Paris, France; and Sorbonne Université (S.V., E.M., C.L., C.P.), Département de Neurologie, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| | - Marina Vignes
- From the Sorbonne Université (D.S., M.L., J.F., C.P., M.V., G.A., K.D., G.G.), Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), AP-HP Hôpital Pitié-Salpêtrière, France; Université Paris-Saclay (C.J., P.L.), INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France; Hôpital Ophtalmologique Adolphe de Rothschild (J.A., O.G., R.D.), Département de Neurologie, Paris, France; and Sorbonne Université (S.V., E.M., C.L., C.P.), Département de Neurologie, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| | - Gaëlle Autaa
- From the Sorbonne Université (D.S., M.L., J.F., C.P., M.V., G.A., K.D., G.G.), Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), AP-HP Hôpital Pitié-Salpêtrière, France; Université Paris-Saclay (C.J., P.L.), INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France; Hôpital Ophtalmologique Adolphe de Rothschild (J.A., O.G., R.D.), Département de Neurologie, Paris, France; and Sorbonne Université (S.V., E.M., C.L., C.P.), Département de Neurologie, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| | - Karim Dorgham
- From the Sorbonne Université (D.S., M.L., J.F., C.P., M.V., G.A., K.D., G.G.), Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), AP-HP Hôpital Pitié-Salpêtrière, France; Université Paris-Saclay (C.J., P.L.), INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France; Hôpital Ophtalmologique Adolphe de Rothschild (J.A., O.G., R.D.), Département de Neurologie, Paris, France; and Sorbonne Université (S.V., E.M., C.L., C.P.), Département de Neurologie, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| | - Catherine Juste
- From the Sorbonne Université (D.S., M.L., J.F., C.P., M.V., G.A., K.D., G.G.), Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), AP-HP Hôpital Pitié-Salpêtrière, France; Université Paris-Saclay (C.J., P.L.), INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France; Hôpital Ophtalmologique Adolphe de Rothschild (J.A., O.G., R.D.), Département de Neurologie, Paris, France; and Sorbonne Université (S.V., E.M., C.L., C.P.), Département de Neurologie, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| | - Patricia Lepage
- From the Sorbonne Université (D.S., M.L., J.F., C.P., M.V., G.A., K.D., G.G.), Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), AP-HP Hôpital Pitié-Salpêtrière, France; Université Paris-Saclay (C.J., P.L.), INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France; Hôpital Ophtalmologique Adolphe de Rothschild (J.A., O.G., R.D.), Département de Neurologie, Paris, France; and Sorbonne Université (S.V., E.M., C.L., C.P.), Département de Neurologie, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| | - Jennifer Aboab
- From the Sorbonne Université (D.S., M.L., J.F., C.P., M.V., G.A., K.D., G.G.), Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), AP-HP Hôpital Pitié-Salpêtrière, France; Université Paris-Saclay (C.J., P.L.), INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France; Hôpital Ophtalmologique Adolphe de Rothschild (J.A., O.G., R.D.), Département de Neurologie, Paris, France; and Sorbonne Université (S.V., E.M., C.L., C.P.), Département de Neurologie, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| | - Savine Vicart
- From the Sorbonne Université (D.S., M.L., J.F., C.P., M.V., G.A., K.D., G.G.), Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), AP-HP Hôpital Pitié-Salpêtrière, France; Université Paris-Saclay (C.J., P.L.), INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France; Hôpital Ophtalmologique Adolphe de Rothschild (J.A., O.G., R.D.), Département de Neurologie, Paris, France; and Sorbonne Université (S.V., E.M., C.L., C.P.), Département de Neurologie, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| | - Elisabeth Maillart
- From the Sorbonne Université (D.S., M.L., J.F., C.P., M.V., G.A., K.D., G.G.), Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), AP-HP Hôpital Pitié-Salpêtrière, France; Université Paris-Saclay (C.J., P.L.), INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France; Hôpital Ophtalmologique Adolphe de Rothschild (J.A., O.G., R.D.), Département de Neurologie, Paris, France; and Sorbonne Université (S.V., E.M., C.L., C.P.), Département de Neurologie, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| | - Olivier Gout
- From the Sorbonne Université (D.S., M.L., J.F., C.P., M.V., G.A., K.D., G.G.), Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), AP-HP Hôpital Pitié-Salpêtrière, France; Université Paris-Saclay (C.J., P.L.), INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France; Hôpital Ophtalmologique Adolphe de Rothschild (J.A., O.G., R.D.), Département de Neurologie, Paris, France; and Sorbonne Université (S.V., E.M., C.L., C.P.), Département de Neurologie, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| | - Catherine Lubetzki
- From the Sorbonne Université (D.S., M.L., J.F., C.P., M.V., G.A., K.D., G.G.), Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), AP-HP Hôpital Pitié-Salpêtrière, France; Université Paris-Saclay (C.J., P.L.), INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France; Hôpital Ophtalmologique Adolphe de Rothschild (J.A., O.G., R.D.), Département de Neurologie, Paris, France; and Sorbonne Université (S.V., E.M., C.L., C.P.), Département de Neurologie, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| | - Romain Deschamps
- From the Sorbonne Université (D.S., M.L., J.F., C.P., M.V., G.A., K.D., G.G.), Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), AP-HP Hôpital Pitié-Salpêtrière, France; Université Paris-Saclay (C.J., P.L.), INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France; Hôpital Ophtalmologique Adolphe de Rothschild (J.A., O.G., R.D.), Département de Neurologie, Paris, France; and Sorbonne Université (S.V., E.M., C.L., C.P.), Département de Neurologie, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| | - Caroline Papeix
- From the Sorbonne Université (D.S., M.L., J.F., C.P., M.V., G.A., K.D., G.G.), Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), AP-HP Hôpital Pitié-Salpêtrière, France; Université Paris-Saclay (C.J., P.L.), INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France; Hôpital Ophtalmologique Adolphe de Rothschild (J.A., O.G., R.D.), Département de Neurologie, Paris, France; and Sorbonne Université (S.V., E.M., C.L., C.P.), Département de Neurologie, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| | - Guy Gorochov
- From the Sorbonne Université (D.S., M.L., J.F., C.P., M.V., G.A., K.D., G.G.), Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), AP-HP Hôpital Pitié-Salpêtrière, France; Université Paris-Saclay (C.J., P.L.), INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France; Hôpital Ophtalmologique Adolphe de Rothschild (J.A., O.G., R.D.), Département de Neurologie, Paris, France; and Sorbonne Université (S.V., E.M., C.L., C.P.), Département de Neurologie, AP-HP Hôpital Pitié-Salpêtrière, Paris, France.
| |
Collapse
|
37
|
Pugia M, Bose T, Tjioe M, Frabutt D, Baird Z, Cao Z, Vorsilak A, McLuckey I, Barron MR, Barron M, Denys G, Carpenter J, Das A, Kaur K, Roy S, Sen CK, Deiss F. Multiplexed Signal Ion Emission Reactive Release Amplification (SIERRA) Assay for the Culture-Free Detection of Gram-Negative and Gram-Positive Bacteria and Antimicrobial Resistance Genes. Anal Chem 2021; 93:6604-6612. [PMID: 33819029 PMCID: PMC9097648 DOI: 10.1021/acs.analchem.0c00453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The global prevalence of antibiotic-resistant bacteria has increased the risk of dangerous infections, requiring rapid diagnosis and treatment. The standard method for diagnosis of bacterial infections remains dependent on slow culture-based methods, carried out in central laboratories, not easily extensible to rapid identification of organisms, and thus not optimal for timely treatments at the point-of-care (POC). Here, we demonstrate rapid detection of bacteria by combining electrochemical immunoassays (EC-IA) for pathogen identification with confirmatory quantitative mass spectral immunoassays (MS-IA) based on signal ion emission reactive release amplification (SIERRA) nanoparticles with unique mass labels. This diagnostic method uses compatible reagents for all involved assays and standard fluidics for automatic sample preparation at POC. EC-IA, based on alkaline phosphatase-conjugated pathogen-specific antibodies, quantified down to 104 bacteria per sample when testing Staphylococcus aureus, Escherichia coli, and Pseudomonas aeruginosa lysates. EC-IA quantitation was also obtained for wound samples. The MS-IA using nanoparticles against S. aureus, E. coli, Klebsiella pneumoniae, and P. aeruginosa allowed selective quantitation of ∼105 bacteria per sample. This method preserves bacterial cells allowing extraction and amplification of 16S ribosomal RNA genes and antibiotic resistance genes, as was demonstrated through identification and quantitation of two strains of E. coli, resistant and nonresistant due to β-lactamase cefotaximase genes. Finally, the combined immunoassays were compared against culture using remnant deidentified patient urine samples. The sensitivities for these immunoassays were 83, 95, and 92% for the prediction of S. aureus, P. aeruginosa, and E. coli or K. pneumoniae positive culture, respectively, while specificities were 85, 92, and 97%. The diagnostic platform presented here with fluidics and combined immunoassays allows for pathogen isolation within 5 min and identification in as little as 15 min to 1 h, to help guide the decision for additional testing, optimally only on positive samples, such as multiplexed or resistance gene assays (6 h).
Collapse
Affiliation(s)
- Michael Pugia
- Bioanalytical Technologies, Indiana Biosciences Research Institute (IBRI), 1345 W. 16th Street, Suite #300, Indianapolis, Indiana 46202, United States
| | - Tiyash Bose
- Bioanalytical Technologies, Indiana Biosciences Research Institute (IBRI), 1345 W. 16th Street, Suite #300, Indianapolis, Indiana 46202, United States
| | - Marco Tjioe
- Bioanalytical Technologies, Indiana Biosciences Research Institute (IBRI), 1345 W. 16th Street, Suite #300, Indianapolis, Indiana 46202, United States
| | - Dylan Frabutt
- Bioanalytical Technologies, Indiana Biosciences Research Institute (IBRI), 1345 W. 16th Street, Suite #300, Indianapolis, Indiana 46202, United States
| | - Zane Baird
- Bioanalytical Technologies, Indiana Biosciences Research Institute (IBRI), 1345 W. 16th Street, Suite #300, Indianapolis, Indiana 46202, United States
| | - Zehui Cao
- Bioanalytical Technologies, Indiana Biosciences Research Institute (IBRI), 1345 W. 16th Street, Suite #300, Indianapolis, Indiana 46202, United States
| | - Anna Vorsilak
- Bioanalytical Technologies, Indiana Biosciences Research Institute (IBRI), 1345 W. 16th Street, Suite #300, Indianapolis, Indiana 46202, United States
| | - Ian McLuckey
- Bioanalytical Technologies, Indiana Biosciences Research Institute (IBRI), 1345 W. 16th Street, Suite #300, Indianapolis, Indiana 46202, United States
| | - M Regina Barron
- Bioanalytical Technologies, Indiana Biosciences Research Institute (IBRI), 1345 W. 16th Street, Suite #300, Indianapolis, Indiana 46202, United States
- Department of Chemistry & Chemical Biology, Indiana University-Purdue University Indianapolis (IUPUI), 402 N Blackford Street, LD326, Indianapolis, Indiana 46202, United States
| | - Monica Barron
- Bioanalytical Technologies, Indiana Biosciences Research Institute (IBRI), 1345 W. 16th Street, Suite #300, Indianapolis, Indiana 46202, United States
- Department of Chemistry & Chemical Biology, Indiana University-Purdue University Indianapolis (IUPUI), 402 N Blackford Street, LD326, Indianapolis, Indiana 46202, United States
| | - Gerald Denys
- Division of Clinical Microbiology, Department of Pathology and Laboratory Medicine, IU Health Pathology Laboratory, Indiana University School of Medicine, 350 W. 11th Street, Room 6027B, Indianapolis, Indiana 46202, United States
| | - Jessica Carpenter
- Division of Clinical Microbiology, Department of Pathology and Laboratory Medicine, IU Health Pathology Laboratory, Indiana University School of Medicine, 350 W. 11th Street, Room 6027B, Indianapolis, Indiana 46202, United States
| | - Amitava Das
- Indiana Center for Regenerative Medicine and Engineering (ICRME), IU Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine, 975 W. Walnut Street, Suite #444, Indianapolis, Indiana 46202,United States
| | - Karamjeet Kaur
- Indiana Center for Regenerative Medicine and Engineering (ICRME), IU Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine, 975 W. Walnut Street, Suite #444, Indianapolis, Indiana 46202,United States
| | - Sashwati Roy
- Indiana Center for Regenerative Medicine and Engineering (ICRME), IU Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine, 975 W. Walnut Street, Suite #444, Indianapolis, Indiana 46202,United States
| | - Chandan K Sen
- Indiana Center for Regenerative Medicine and Engineering (ICRME), IU Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine, 975 W. Walnut Street, Suite #444, Indianapolis, Indiana 46202,United States
| | - Frédérique Deiss
- Department of Chemistry & Chemical Biology, Indiana University-Purdue University Indianapolis (IUPUI), 402 N Blackford Street, LD326, Indianapolis, Indiana 46202, United States
| |
Collapse
|
38
|
Shielding Effect of Escherichia coli O-Antigen Polysaccharide on J5-Induced Cross-Reactive Antibodies. mSphere 2021; 6:6/1/e01227-20. [PMID: 33504665 PMCID: PMC7885324 DOI: 10.1128/msphere.01227-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Despite intensive research, mastitis remains an important disease in dairy cattle with a significant impact on animal welfare, use of antibiotics, and, in the end, the economy of dairy farms. Although vaccines available so far have shown limited efficacy against coliform mastitis, vaccination is considered one of the measures that could limit the consequences of mastitis. Escherichia coli is the leading cause of severe mastitis in dairy farms. As E. coli mastitis is refractory to the hygienic control measures adapted to contagious mastitis, efficient vaccines are in demand. Existing mastitis vaccines, based on the use of killed rough E. coli J5 as the antigen, aim at inducing phagocytosis by neutrophils. We assessed the binding of J5-induced antibodies to isogenic rough and smooth strains along with a panel of mastitis-associated E. coli. Analysis by enzyme-linked immunosorbent assay revealed that antibodies to OmpA or killed J5 bind readily to rough E. coli but poorly to smooth strains. Flow cytometry analysis indicated that immunization with J5 induced antibodies that cross-reacted with rough E. coli strains but with only a small subpopulation of smooth strains. We identified type 1 fimbriae as the target of most antibodies cross-reacting with the smooth strains. These results suggest that the O-polysaccharide of lipopolysaccharide shields the outer membrane antigens and that only fiber antigens protruding at the bacterial surface can elicit antibodies reacting with mastitis-associated E. coli. We evaluated J5-induced antibodies in an opsonophagocytic killing assay with bovine neutrophils. J5 immune serum was not more efficient than preimmune serum, showing that immunization did not improve on the already high efficiency of naturally acquired antibodies to E. coli. In conclusion, it is unlikely that the efficiency of J5 vaccines is related to the induction of opsonic antibodies. Consequently, other research directions, such as cell-mediated immunity, should be explored to improve E. coli mastitis vaccines. IMPORTANCE Despite intensive research, mastitis remains an important disease in dairy cattle with a significant impact on animal welfare, use of antibiotics, and, in the end, the economy of dairy farms. Although vaccines available so far have shown limited efficacy against coliform mastitis, vaccination is considered one of the measures that could limit the consequences of mastitis. One reason for the lack of efficiency of current vaccines likely stems from the current evaluation of vaccines that relies mostly on measuring antibody production against vaccine antigens. This report clearly shows that vaccine-induced antibodies fail to bind to most mastitis-associated E. coli strains because of the presence of an O-antigen and, thus, do not allow for improved phagocytosis of pathogens. As a consequence, this report calls for revised criteria for the evaluation of vaccines and suggests that cell-mediated immunity should be targeted by new vaccinal strategies. More generally, these results could be extended to other vaccine development strategies targeting coliform bacteria.
Collapse
|
39
|
Abstract
Antibodies against Streptococcus pneumoniae (pneumococcus) following vaccination are crucial for host protection against invasive pneumococcal infections. The antibodies induced by pneumococcal vaccines act as opsonins to mediate bacterial uptake and killing by host phagocytic cells, especially polymorphonuclear leukocytes (PMNs) also called neutrophils. Therefore, it is important to measure not only the levels of antibodies induced by a pneumococcal vaccine candidate but their actual functional capacity in mediating bacterial opsonization and killing by PMNs. Here, we describe a protocol to demonstrate effective deposition of vaccine-induced antibodies on the surface of S. pneumoniae by flow cytometry and subsequent opsonophagocytic killing (OPH) by murine bone-marrow derived PMNs.
Collapse
|
40
|
Ramakrishnan A, Joseph SS, Reynolds ND, Poncet D, Maciel M, Nunez G, Espinoza N, Nieto M, Castillo R, Royal JM, Poole S, McVeigh A, Rollenhagen JE, Heinrichs J, Prouty MG, Simons MP, Renauld-Mongénie G, Savarino SJ. Evaluation of the immunogenicity and protective efficacy of a recombinant CS6-based ETEC vaccine in an Aotus nancymaae CS6 + ETEC challenge model. Vaccine 2020; 39:487-494. [PMID: 33357957 DOI: 10.1016/j.vaccine.2020.12.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/30/2020] [Accepted: 12/09/2020] [Indexed: 10/22/2022]
Abstract
Colonization factors or Coli surface antigens (CFs or CS) are important virulence factors of Enterotoxigenic E. coli (ETEC) that mediate intestinal colonization and accordingly are targets of vaccine development efforts. CS6 is a highly prevalent CF associated with symptomatic ETEC infection both in endemic populations and amongst travelers. In this study, we used an Aotus nancymaae non-human primate ETEC challenge model with a CS6 + ETEC strain, B7A, to test the immunogenicity and protective efficacy (PE) of a recombinant CS6-based subunit vaccine. Specifically, we determined the ability of dscCssBA, the donor strand complemented recombinant stabilized fusion of the two subunits of the CS6 fimbriae, CssA and CssB, to elicit protection against CS6 + ETEC mediated diarrhea when given intradermally (ID) with the genetically attenuated double mutant heat-labile enterotoxin LT(R192G/L211A) (dmLT). ID vaccination with dscCssBA + dmLT induced strong serum antibody responses against CS6 and LT. Importantly, vaccination with dscCssBA + dmLT resulted in no observed diarrheal disease (PE = 100%, p = 0.03) following B7A challenge as compared to PBS immunized animals, with an attack rate of 62.5%. These data demonstrate the potential role that CS6 may play in ETEC infection and that recombinant dscCssBA antigen can provide protection against challenge with the homologous CS6 + ETEC strain, B7A, in the Aotus nancymaae diarrheal challenge model. Combined, these data indicate that CS6, and more specifically, a recombinant engineered derivative should be considered for further clinical development.
Collapse
Affiliation(s)
- A Ramakrishnan
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA
| | - S S Joseph
- Henry M Jackson Foundation for the Advancement of Military Medicine, 6720 Rockledge Drive, Bethesda, MD 20817, USA
| | - N D Reynolds
- Department of Bacteriology, Naval Medical Research Unit No. 6, Venezuela Ave. Block 36, Bellavista, Callao, Peru
| | - D Poncet
- Sanofi Pasteur, Research and External Innovation, 1541 Av. Marcel Mérieux, 69280 Marcy L'Etoile
| | - M Maciel
- Henry M Jackson Foundation for the Advancement of Military Medicine, 6720 Rockledge Drive, Bethesda, MD 20817, USA
| | - G Nunez
- Department of Bacteriology, Naval Medical Research Unit No. 6, Venezuela Ave. Block 36, Bellavista, Callao, Peru
| | - N Espinoza
- Department of Bacteriology, Naval Medical Research Unit No. 6, Venezuela Ave. Block 36, Bellavista, Callao, Peru
| | - M Nieto
- Department of Bacteriology, Naval Medical Research Unit No. 6, Venezuela Ave. Block 36, Bellavista, Callao, Peru
| | - R Castillo
- Department of Bacteriology, Naval Medical Research Unit No. 6, Venezuela Ave. Block 36, Bellavista, Callao, Peru
| | - J M Royal
- Department of Veterinary Services, Naval Medical Research Unit No. 6, Venezuela Ave. Block 36, Bellavista, Callao, Peru
| | - S Poole
- Henry M Jackson Foundation for the Advancement of Military Medicine, 6720 Rockledge Drive, Bethesda, MD 20817, USA
| | - A McVeigh
- Henry M Jackson Foundation for the Advancement of Military Medicine, 6720 Rockledge Drive, Bethesda, MD 20817, USA
| | - J E Rollenhagen
- Henry M Jackson Foundation for the Advancement of Military Medicine, 6720 Rockledge Drive, Bethesda, MD 20817, USA
| | | | - M G Prouty
- Enteric Diseases Department, Naval Medical Research Center, 503 Robert Grant Ave, Silver Spring, MD 20910, USA.
| | - M P Simons
- Department of Bacteriology, Naval Medical Research Unit No. 6, Venezuela Ave. Block 36, Bellavista, Callao, Peru
| | - G Renauld-Mongénie
- Sanofi Pasteur, Research and External Innovation, 1541 Av. Marcel Mérieux, 69280 Marcy L'Etoile
| | - S J Savarino
- Enteric Diseases Department, Naval Medical Research Center, 503 Robert Grant Ave, Silver Spring, MD 20910, USA
| |
Collapse
|
41
|
Pröbstel AK, Zhou X, Baumann R, Wischnewski S, Kutza M, Rojas OL, Sellrie K, Bischof A, Kim K, Ramesh A, Dandekar R, Greenfield AL, Schubert RD, Bisanz JE, Vistnes S, Khaleghi K, Landefeld J, Kirkish G, Liesche-Starnecker F, Ramaglia V, Singh S, Tran EB, Barba P, Zorn K, Oechtering J, Forsberg K, Shiow LR, Henry RG, Graves J, Cree BAC, Hauser SL, Kuhle J, Gelfand JM, Andersen PM, Schlegel J, Turnbaugh PJ, Seeberger PH, Gommerman JL, Wilson MR, Schirmer L, Baranzini SE. Gut microbiota-specific IgA + B cells traffic to the CNS in active multiple sclerosis. Sci Immunol 2020; 5:5/53/eabc7191. [PMID: 33219152 DOI: 10.1126/sciimmunol.abc7191] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 10/29/2020] [Indexed: 01/04/2023]
Abstract
Changes in gut microbiota composition and a diverse role of B cells have recently been implicated in multiple sclerosis (MS), a central nervous system (CNS) autoimmune disease. Immunoglobulin A (IgA) is a key regulator at the mucosal interface. However, whether gut microbiota shape IgA responses and what role IgA+ cells have in neuroinflammation are unknown. Here, we identify IgA-bound taxa in MS and show that IgA-producing cells specific for MS-associated taxa traffic to the inflamed CNS, resulting in a strong, compartmentalized IgA enrichment in active MS and other neuroinflammatory diseases. Unlike previously characterized polyreactive anti-commensal IgA responses, CNS IgA cross-reacts with surface structures on specific bacterial strains but not with brain tissue. These findings establish gut microbiota-specific IgA+ cells as a systemic mediator in MS and suggest a critical role of mucosal B cells during active neuroinflammation with broad implications for IgA as an informative biomarker and IgA-producing cells as an immune subset to harness for therapeutic interventions.
Collapse
Affiliation(s)
- Anne-Katrin Pröbstel
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA. .,Neurologic Clinic and Policlinic and Research Center for Clinical Neuroimmunology and Neuroscience Basel, Departments of Medicine, Biomedicine, and Clinical Research, University Hospital of Basel, University of Basel, 4031 Basel, Switzerland
| | - Xiaoyuan Zhou
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ryan Baumann
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sven Wischnewski
- Department of Neurology and Mannheim Center for Translational Neurosciences, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Michael Kutza
- Department of Neurology and Mannheim Center for Translational Neurosciences, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Olga L Rojas
- Department of Immunology, University of Toronto, Toronto, ON M5S 18A, Canada
| | - Katrin Sellrie
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14776 Potsdam, Germany
| | - Antje Bischof
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kicheol Kim
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Akshaya Ramesh
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ravi Dandekar
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ariele L Greenfield
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ryan D Schubert
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jordan E Bisanz
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.,Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Stephanie Vistnes
- Eli and Edythe Broad Center for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Khashayar Khaleghi
- Department of Immunology, University of Toronto, Toronto, ON M5S 18A, Canada
| | - James Landefeld
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Gina Kirkish
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Friederike Liesche-Starnecker
- Department of Neuropathology, School of Medicine, Institute of Pathology, Technical University Munich, 81675 Munich, Germany
| | - Valeria Ramaglia
- Department of Neurology and Mannheim Center for Translational Neurosciences, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Sneha Singh
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Edwina B Tran
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Patrick Barba
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kelsey Zorn
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Johanna Oechtering
- Neurologic Clinic and Policlinic and Research Center for Clinical Neuroimmunology and Neuroscience Basel, Departments of Medicine, Biomedicine, and Clinical Research, University Hospital of Basel, University of Basel, 4031 Basel, Switzerland
| | - Karin Forsberg
- Department of Clinical Science, Neurosciences, Umeå University, 90185 Umeå, Sweden
| | - Lawrence R Shiow
- Eli and Edythe Broad Center for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.,Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Roland G Henry
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jennifer Graves
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Bruce A C Cree
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Stephen L Hauser
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jens Kuhle
- Neurologic Clinic and Policlinic and Research Center for Clinical Neuroimmunology and Neuroscience Basel, Departments of Medicine, Biomedicine, and Clinical Research, University Hospital of Basel, University of Basel, 4031 Basel, Switzerland
| | - Jeffrey M Gelfand
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Peter M Andersen
- Department of Clinical Science, Neurosciences, Umeå University, 90185 Umeå, Sweden
| | - Jürgen Schlegel
- Department of Neuropathology, School of Medicine, Institute of Pathology, Technical University Munich, 81675 Munich, Germany
| | - Peter J Turnbaugh
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.,Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Peter H Seeberger
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14776 Potsdam, Germany
| | | | - Michael R Wilson
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lucas Schirmer
- Department of Neurology and Mannheim Center for Translational Neurosciences, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany.,Interdisciplinary Center for Neurosciences, University of Heidelberg, 69117 Heidelberg, Germany
| | - Sergio E Baranzini
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA. .,Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA.,Graduate Program in Bioinformatics, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
42
|
Moreno-Sabater A, Autaa G, Sterlin D, Jerbi A, Villette R, Holm JB, Parizot C, Selim S, Senghor Y, Ghillani-Dalbin P, Bachmeyer C, Hennequin C, Gorochov G, Larsen M. Systemic anti-commensal response to fungi analyzed by flow cytometry is related to gut mycobiome ecology. MICROBIOME 2020; 8:159. [PMID: 33190643 PMCID: PMC7667786 DOI: 10.1186/s40168-020-00924-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/15/2020] [Indexed: 05/08/2023]
Abstract
BACKGROUND Interest for the study of gut mycobiota in relation with human health and immune homeostasis has increased in the last years. From this perspective, new tools to study the immune/fungal interface are warranted. Systemic humoral immune responses could reflect the dynamic relationships between gut mycobiota and immunity. Using a novel flow cytometry technology (Fungi-Flow) to determine immunoglobulin (Ig) responses to fungi, we studied the relationships between gut mycobiota and systemic humoral anti-commensal immunity. RESULTS The Fungi-Flow method allows a sensitive and specific measurement of systemic IgG responses against 17 commensal and environmental fungi from the two main divisions; Ascomycota and Basidiomycota. IgG responses exhibited a high inter-individual variability. Anti-commensal IgG responses were contrasted with the relative abundance, alpha-diversity, and intra-genus richness of fungal species in gut mycobiota of twenty healthy donors. Categorization of gut mycobiota composition revealed two differentiated fungal ecosystems. Significant difference of anti-Saccharomyces systemic IgG responses were observed in healthy donors stratified according to the fungal ecosystem colonizing their gut. A positive and significant correlation was observed between the variety of IgG responses against fungal commensals and intestinal alpha-diversity. At the level of intra-genus species richness, intense IgG responses were associated with a low intra-genus richness for known pathobionts, but not commensals. CONCLUSIONS Fungi-Flow allows an easy and reliable measure of personalized humoral responses against commensal fungi. Combining sequencing technology with our novel Fungi-Flow immunological method, we propose that there are at least two defined ecosystems in the human gut mycobiome associated with systemic humoral responses. Fungi-Flow opens new opportunities to improve our knowledge about the impact of mycobiota in humoral anti-commensal immunity and homeostasis. Video Abstract.
Collapse
Affiliation(s)
- Alicia Moreno-Sabater
- Sorbonne Université, Inserm U1135, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013 Paris, France
- Service de Parasitologie-Mycologie AP-HP, Hôpital Saint-Antoine, 75012 Paris, France
| | - Gaelle Autaa
- Sorbonne Université, Inserm U1135, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013 Paris, France
| | - Delphine Sterlin
- Sorbonne Université, Inserm U1135, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013 Paris, France
- Service d’immunologie, AP-HP, Hôpital Pitié-Salpêtrière, 75013 Paris, France
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR1222 Inserm, 75015 Paris, France
| | - Amenie Jerbi
- Service d’immunologie, AP-HP, Hôpital Pitié-Salpêtrière, 75013 Paris, France
| | - Remy Villette
- Sorbonne Université, Inserm U1135, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013 Paris, France
| | - Johanna B. Holm
- Institute for Genome Sciences and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD USA
| | - Christophe Parizot
- Service d’immunologie, AP-HP, Hôpital Pitié-Salpêtrière, 75013 Paris, France
| | - Sameh Selim
- College of Agricultural Sciences AGHYLE Res, Unit. Institut Polytechnique UniLaSalle, 60026 Beauvais, France
| | - Yaye Senghor
- Service de Parasitologie-Mycologie AP-HP, Hôpital Saint-Antoine, 75012 Paris, France
| | | | | | - Christophe Hennequin
- Service de Parasitologie-Mycologie AP-HP, Hôpital Saint-Antoine, 75012 Paris, France
- Centre de Recherche Saint-Antoine, CRSA, AP-HP, Sorbonne Université, Inserm, 75012 Paris, France
| | - Guy Gorochov
- Sorbonne Université, Inserm U1135, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013 Paris, France
- Service d’immunologie, AP-HP, Hôpital Pitié-Salpêtrière, 75013 Paris, France
| | - Martin Larsen
- Sorbonne Université, Inserm U1135, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013 Paris, France
| |
Collapse
|
43
|
Sterlin D, Fadlallah J, Adams O, Fieschi C, Parizot C, Dorgham K, Rajkumar A, Autaa G, El-Kafsi H, Charuel JL, Juste C, Jönsson F, Candela T, Wardemann H, Aubry A, Capito C, Brisson H, Tresallet C, Cummings RD, Larsen M, Yssel H, von Gunten S, Gorochov G. Human IgA binds a diverse array of commensal bacteria. J Exp Med 2020; 217:133553. [PMID: 31891367 PMCID: PMC7062531 DOI: 10.1084/jem.20181635] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 05/10/2019] [Accepted: 11/22/2019] [Indexed: 11/23/2022] Open
Abstract
In humans, several grams of IgA are secreted every day in the intestinal lumen. While only one IgA isotype exists in mice, humans secrete IgA1 and IgA2, whose respective relations with the microbiota remain elusive. We compared the binding patterns of both polyclonal IgA subclasses to commensals and glycan arrays and determined the reactivity profile of native human monoclonal IgA antibodies. While most commensals are dually targeted by IgA1 and IgA2 in the small intestine, IgA1+IgA2+ and IgA1−IgA2+ bacteria coexist in the colon lumen, where Bacteroidetes is preferentially targeted by IgA2. We also observed that galactose-α terminated glycans are almost exclusively recognized by IgA2. Although bearing signs of affinity maturation, gut-derived IgA monoclonal antibodies are cross-reactive in the sense that they bind to multiple bacterial targets. Private anticarbohydrate-binding patterns, observed at clonal level as well, could explain these apparently opposing features of IgA, being at the same time cross-reactive and selective in its interactions with the microbiota.
Collapse
Affiliation(s)
- Delphine Sterlin
- Sorbonne Université, Institut national de la santé et de la recherche médicale, Centre d'Immunologie et des Maladies Infectieuses, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - Jehane Fadlallah
- Sorbonne Université, Institut national de la santé et de la recherche médicale, Centre d'Immunologie et des Maladies Infectieuses, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - Olivia Adams
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Claire Fieschi
- Université Paris Diderot Paris 7, Department of Clinical Immunology, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris, EA 3518, Paris, France
| | - Christophe Parizot
- Sorbonne Université, Institut national de la santé et de la recherche médicale, Centre d'Immunologie et des Maladies Infectieuses, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - Karim Dorgham
- Sorbonne Université, Institut national de la santé et de la recherche médicale, Centre d'Immunologie et des Maladies Infectieuses, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - Asok Rajkumar
- Sorbonne Université, Institut national de la santé et de la recherche médicale, Centre d'Immunologie et des Maladies Infectieuses, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - Gaëlle Autaa
- Sorbonne Université, Institut national de la santé et de la recherche médicale, Centre d'Immunologie et des Maladies Infectieuses, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - Hela El-Kafsi
- Sorbonne Université, Institut national de la santé et de la recherche médicale, Centre d'Immunologie et des Maladies Infectieuses, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - Jean-Luc Charuel
- Sorbonne Université, Institut national de la santé et de la recherche médicale, Centre d'Immunologie et des Maladies Infectieuses, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - Catherine Juste
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Friederike Jönsson
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR1222 Institut national de la santé et de la recherche médicale, Paris, France
| | - Thomas Candela
- EA 4043, Unité Bactéries Pathogènes et Santé, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Hedda Wardemann
- Division of B Cell Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Alexandra Aubry
- Sorbonne Université, Institut national de la santé et de la recherche médicale, Centre d'Immunologie et des Maladies Infectieuses, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - Carmen Capito
- EA 4043, Unité Bactéries Pathogènes et Santé, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Hélène Brisson
- Sorbonne Université, Institut national de la santé et de la recherche médicale, Centre d'Immunologie et des Maladies Infectieuses, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - Christophe Tresallet
- Sorbonne Université, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Martin Larsen
- Sorbonne Université, Institut national de la santé et de la recherche médicale, Centre d'Immunologie et des Maladies Infectieuses, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - Hans Yssel
- Sorbonne Université, Institut national de la santé et de la recherche médicale, Centre d'Immunologie et des Maladies Infectieuses, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | | | - Guy Gorochov
- Sorbonne Université, Institut national de la santé et de la recherche médicale, Centre d'Immunologie et des Maladies Infectieuses, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| |
Collapse
|
44
|
Heo M, Chenon G, Castrillon C, Bibette J, Bruhns P, Griffiths AD, Baudry J, Eyer K. Deep phenotypic characterization of immunization-induced antibacterial IgG repertoires in mice using a single-antibody bioassay. Commun Biol 2020; 3:614. [PMID: 33106526 PMCID: PMC7589517 DOI: 10.1038/s42003-020-01296-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Antibodies with antibacterial activity need to bind to the bacterial surface with affinity, specificity, and sufficient density to induce efficient elimination. To characterize the anti-bacterial antibody repertoire, we developed an in-droplet bioassay with single-antibody resolution. The assay not only allowed us to identify whether the secreted antibodies recognized a bacterial surface antigen, but also to estimate the apparent dissociation constant (KD app) of the interaction and the density of the recognized epitope on the bacteria. Herein, we found substantial differences within the KD app/epitope density profiles in mice immunized with various species of heat-killed bacteria. The experiments further revealed a high cross-reactivity of the secreted IgG repertoires, binding to even unrelated bacteria with high affinity. This application confirmed the ability to quantify the anti-bacterial antibody repertoire and the utility of the developed bioassay to study the interplay between bacteria and the humoral response.
Collapse
Affiliation(s)
- Millie Heo
- 'Laboratoire Colloïdes et Matériaux Divisés' (LCMD), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, F-75005, Paris, France
| | - Guilhem Chenon
- 'Laboratoire Colloïdes et Matériaux Divisés' (LCMD), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, F-75005, Paris, France
| | - Carlos Castrillon
- Unit of Antibodies in Therapy and Pathology, Institute Pasteur, UMR1222 INSERM, F-75015, Paris, France
- 'Laboratoire de Biochimie' (LBC), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, F-75005, Paris, France
| | - Jérôme Bibette
- 'Laboratoire Colloïdes et Matériaux Divisés' (LCMD), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, F-75005, Paris, France
| | - Pierre Bruhns
- Unit of Antibodies in Therapy and Pathology, Institute Pasteur, UMR1222 INSERM, F-75015, Paris, France
| | - Andrew D Griffiths
- 'Laboratoire de Biochimie' (LBC), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, F-75005, Paris, France
| | - Jean Baudry
- 'Laboratoire Colloïdes et Matériaux Divisés' (LCMD), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, F-75005, Paris, France
| | - Klaus Eyer
- 'Laboratoire Colloïdes et Matériaux Divisés' (LCMD), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, F-75005, Paris, France.
- Laboratory for Functional Immune Repertoire Analysis, Institute of Pharmaceutical Sciences, D-CHAB, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
45
|
Pfister SP, Schären OP, Beldi L, Printz A, Notter MD, Mukherjee M, Li H, Limenitakis JP, Werren JP, Tandon D, Cuenca M, Hagemann S, Uster SS, Terrazos MA, Gomez de Agüero M, Schürch CM, Coelho FM, Curtiss R, Slack E, Balmer ML, Hapfelmeier S. Uncoupling of invasive bacterial mucosal immunogenicity from pathogenicity. Nat Commun 2020; 11:1978. [PMID: 32332737 PMCID: PMC7181798 DOI: 10.1038/s41467-020-15891-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 03/30/2020] [Indexed: 11/17/2022] Open
Abstract
There is the notion that infection with a virulent intestinal pathogen induces generally stronger mucosal adaptive immunity than the exposure to an avirulent strain. Whether the associated mucosal inflammation is important or redundant for effective induction of immunity is, however, still unclear. Here we use a model of auxotrophic Salmonella infection in germ-free mice to show that live bacterial virulence factor-driven immunogenicity can be uncoupled from inflammatory pathogenicity. Although live auxotrophic Salmonella no longer causes inflammation, its mucosal virulence factors remain the main drivers of protective mucosal immunity; virulence factor-deficient, like killed, bacteria show reduced efficacy. Assessing the involvement of innate pathogen sensing mechanisms, we show MYD88/TRIF, Caspase-1/Caspase-11 inflammasome, and NOD1/NOD2 nodosome signaling to be individually redundant. In colonized animals we show that microbiota metabolite cross-feeding may recover intestinal luminal colonization but not pathogenicity. Consequent immunoglobulin A immunity and microbial niche competition synergistically protect against Salmonella wild-type infection.
Collapse
Affiliation(s)
- Simona P Pfister
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
- Graduate School GCB, University of Bern, Bern, Switzerland
| | - Olivier P Schären
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
- Graduate School GCB, University of Bern, Bern, Switzerland
| | - Luca Beldi
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Andrea Printz
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Matheus D Notter
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
- Graduate School GCB, University of Bern, Bern, Switzerland
| | - Mohana Mukherjee
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
- Graduate School GCB, University of Bern, Bern, Switzerland
| | - Hai Li
- Maurice Müller Laboratories (DBMR), Universitätsklinik für Viszerale Chirurgie und Medizin (UVCM) Inselspital, Bern, Switzerland
| | - Julien P Limenitakis
- Maurice Müller Laboratories (DBMR), Universitätsklinik für Viszerale Chirurgie und Medizin (UVCM) Inselspital, Bern, Switzerland
| | - Joel P Werren
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
- Graduate School GCB, University of Bern, Bern, Switzerland
| | - Disha Tandon
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
- Graduate School GCB, University of Bern, Bern, Switzerland
| | - Miguelangel Cuenca
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Stefanie Hagemann
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Stephanie S Uster
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Miguel A Terrazos
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Mercedes Gomez de Agüero
- Maurice Müller Laboratories (DBMR), Universitätsklinik für Viszerale Chirurgie und Medizin (UVCM) Inselspital, Bern, Switzerland
| | - Christian M Schürch
- Institute of Pathology, University of Bern, Bern, Switzerland
- Institute of Pathology and Neuropathology and Comprehensive Cancer Center, University Hospital Tübingen, Tübingen, Germany
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Fernanda M Coelho
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Roy Curtiss
- Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Emma Slack
- Institute for Food, Nutrition and Health, D-HEST, ETH Zürich, Switzerland
| | - Maria L Balmer
- Department of Biomedicine, Immunobiology, University of Basel, Basel, Switzerland
| | | |
Collapse
|
46
|
Paun A, Yau C, Meshkibaf S, Daigneault MC, Marandi L, Mortin-Toth S, Bar-Or A, Allen-Vercoe E, Poussier P, Danska JS. Association of HLA-dependent islet autoimmunity with systemic antibody responses to intestinal commensal bacteria in children. Sci Immunol 2020; 4:4/32/eaau8125. [PMID: 30709843 DOI: 10.1126/sciimmunol.aau8125] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 12/13/2018] [Indexed: 12/11/2022]
Abstract
Microbiome sequence analyses have suggested that changes in gut bacterial composition are associated with autoimmune disease in humans and animal models. However, little is known of the mechanisms through which the gut microbiota influences autoimmune responses to distant tissues. Here, we evaluated systemic antibody responses against cultured human gut bacterial strains to determine whether observed patterns of anticommensal antibody (ACAb) responses are associated with type 1 diabetes (T1D) in two cohorts of pediatric study participants. In the first cohort, ACAb responses in sera collected from participants within 6 months of T1D diagnosis were compared with age-matched healthy controls and also with patients with recent onset Crohn's disease. ACAb responses against multiple bacterial species discriminated among these three groups. In the second cohort, we asked whether ACAb responses present before diagnosis were associated with later T1D development and with HLA genotype in participants who were discordant for subsequent progression to diabetes. Serum IgG2 antibodies against Roseburia faecis and against a bacterial consortium were associated with future T1D diagnosis in an HLA DR3/DR4 haplotype-dependent manner. These analyses reveal associations between antibody responses to intestinal microbes and HLA-DR genotype and islet autoantibody specificity and with a future diagnosis of T1D. Further, we present a platform to investigate antibacterial antibodies in biological fluids that is applicable to studies of autoimmune diseases and responses to therapeutic interventions.
Collapse
Affiliation(s)
- Alexandra Paun
- Hospital for Sick Children, Toronto, ON, Canada. .,Department of Immunology, University of Toronto, Faculty of Medicine, Toronto, ON, Canada
| | - Christopher Yau
- Hospital for Sick Children, Toronto, ON, Canada. .,Department of Immunology, University of Toronto, Faculty of Medicine, Toronto, ON, Canada
| | | | - Michelle C Daigneault
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | | | | | - Amit Bar-Or
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada.,Department of Neurology, Perelman Center for Advanced Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, U.S.A
| | - Emma Allen-Vercoe
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Philippe Poussier
- Department of Immunology, University of Toronto, Faculty of Medicine, Toronto, ON, Canada.,Sunnybrook Research Institute, Toronto, ON Canada
| | - Jayne S Danska
- Hospital for Sick Children, Toronto, ON, Canada. .,Department of Immunology, University of Toronto, Faculty of Medicine, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Faculty of Medicine, Toronto, ON, Canada
| |
Collapse
|
47
|
Antibody-coated microbiota in nasopharynx of healthy individuals and IVIg-treated patients with hypogammaglobulinemia. J Allergy Clin Immunol 2020; 145:1686-1690.e4. [PMID: 31954779 DOI: 10.1016/j.jaci.2020.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/02/2020] [Accepted: 01/07/2020] [Indexed: 11/23/2022]
|
48
|
Cell wall hydrolase as a surface-associated protein target for the specific detection of Lactobacillus rhamnosus using flow cytometry. INNOV FOOD SCI EMERG 2020. [DOI: 10.1016/j.ifset.2019.102240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
49
|
FRET-based fluorescent nanoprobe platform for sorting of active microorganisms by functional properties. Biosens Bioelectron 2019; 148:111832. [PMID: 31706173 DOI: 10.1016/j.bios.2019.111832] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/24/2019] [Accepted: 10/29/2019] [Indexed: 12/19/2022]
Abstract
Fluorescence-activated cell sorting (FACS) has rarely been applied to screening of microorganisms because of poor detection resolution, which is compromised by poor stability, toxicity, or interference from background fluorescence of the fluorescence sensors used. Here, a fluorescence-based rapid high-throughput cell sorting method was first developed using a fluorescence resonance energy transfer (FRET) fluorescent nanoprobe NP-RA, which was constructed by coating a silica nanoparticle with Rhodamine B and methyl-red (an azo dye). Rhodamine B (inner layer) is the FRET donor and methyl-red (outer layer) is the acceptor. This ready-to-use NP-RA is non-fluorescent, but fluoresces once the outer layer is degraded by microorganisms. In our experiment, NP-RA was ultrasensitive to model strain Shewanella decolorationis S12, showing a broad detection range from 8.0 cfu/mL to 8.7 × 108 cfu/mL under confocal laser scanning microscopy, and from 1.1 × 107 to 9.36 × 108 cfu/mL under a fluorometer. In addition, NP-RA bioimaging can clearly identify other azo-respiring cells in the microbial community, including Bosea thiooxidans DSM 9653 and Lysinibacillus pakistanensis NCCP-54. Furthermore, the fluorescent probe NP-RA is compatible with downstream FACS so that azo-respiring cells can be rapidly sorted out directly from an artificial microbial community. To our knowledge, no fluorescent nanoprobe has yet been designed for tracking and sorting azo-respiration functional microorganisms.
Collapse
|
50
|
Fadlallah J, El Kafsi H, Sterlin D, Juste C, Parizot C, Dorgham K, Autaa G, Gouas D, Almeida M, Lepage P, Pons N, Le Chatelier E, Levenez F, Kennedy S, Galleron N, de Barros JPP, Malphettes M, Galicier L, Boutboul D, Mathian A, Miyara M, Oksenhendler E, Amoura Z, Doré J, Fieschi C, Ehrlich SD, Larsen M, Gorochov G. Microbial ecology perturbation in human IgA deficiency. Sci Transl Med 2019; 10:10/439/eaan1217. [PMID: 29720448 DOI: 10.1126/scitranslmed.aan1217] [Citation(s) in RCA: 204] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 12/07/2017] [Accepted: 03/12/2018] [Indexed: 12/14/2022]
Abstract
Paradoxically, loss of immunoglobulin A (IgA), one of the most abundant antibodies, does not irrevocably lead to severe infections in humans but rather is associated with relatively mild respiratory infections, atopy, and autoimmunity. IgA might therefore also play covert roles, not uniquely associated with control of pathogens. We show that human IgA deficiency is not associated with massive quantitative perturbations of gut microbial ecology. Metagenomic analysis highlights an expected pathobiont expansion but a less expected depletion in some typically beneficial symbionts. Gut colonization by species usually present in the oropharynx is also reminiscent of spatial microbiota disorganization. IgM only partially rescues IgA deficiency because not all typical IgA targets are efficiently bound by IgM in the intestinal lumen. Together, IgA appears to play a nonredundant role at the forefront of the immune/microbial interface, away from the intestinal barrier, ranging from pathobiont control and regulation of systemic inflammation to preservation of commensal diversity and community networks.
Collapse
Affiliation(s)
- Jehane Fadlallah
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses-Paris (CIMI-Paris), 75013 Paris, France
| | - Hela El Kafsi
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses-Paris (CIMI-Paris), 75013 Paris, France
| | - Delphine Sterlin
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses-Paris (CIMI-Paris), 75013 Paris, France.,Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, 75013 Paris, France
| | - Catherine Juste
- UMR1319 Micalis, Institut National de la Recherche Agronomique (INRA), Jouy-en-Josas, France
| | - Christophe Parizot
- Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, 75013 Paris, France
| | - Karim Dorgham
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses-Paris (CIMI-Paris), 75013 Paris, France
| | - Gaëlle Autaa
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses-Paris (CIMI-Paris), 75013 Paris, France
| | - Doriane Gouas
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses-Paris (CIMI-Paris), 75013 Paris, France
| | - Mathieu Almeida
- Center for Bioinformatics and Computational Biology, University of Maryland, Paint Branch Road, College Park, MD 20742, USA
| | - Patricia Lepage
- UMR1319 Micalis, Institut National de la Recherche Agronomique (INRA), Jouy-en-Josas, France
| | - Nicolas Pons
- INRA, US1367 MetaGenoPolis, 78350 Jouy en Josas, France
| | | | | | - Sean Kennedy
- INRA, US1367 MetaGenoPolis, 78350 Jouy en Josas, France
| | | | - Jean-Paul Pais de Barros
- INSERM, LNC UMR866, University Bourgogne Franche-Comté, F-21000 Dijon, France.,LIPoprotéines et Santé prévention & Traitement des maladies Inflammatoires et du Cancer (LipSTIC) LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, F-21000 Dijon, France
| | - Marion Malphettes
- Département d'Immunologie Clinique, Hôpital Saint-Louis, AP-HP, 75010 Paris, France
| | - Lionel Galicier
- Département d'Immunologie Clinique, Hôpital Saint-Louis, AP-HP, 75010 Paris, France
| | - David Boutboul
- Département d'Immunologie Clinique, Hôpital Saint-Louis, AP-HP, 75010 Paris, France.,INSERM U1126, Université Paris Diderot Paris 7, 75010 Paris, France
| | - Alexis Mathian
- Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Service de Médecine Interne 2, Institut E3M, 75013 Paris, France
| | - Makoto Miyara
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses-Paris (CIMI-Paris), 75013 Paris, France.,Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, 75013 Paris, France
| | - Eric Oksenhendler
- Département d'Immunologie Clinique, Hôpital Saint-Louis, AP-HP, 75010 Paris, France.,Université Paris Diderot Paris 7, EA3518, 75010 Paris, France
| | - Zahir Amoura
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses-Paris (CIMI-Paris), 75013 Paris, France.,Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Service de Médecine Interne 2, Institut E3M, 75013 Paris, France
| | - Joel Doré
- UMR1319 Micalis, Institut National de la Recherche Agronomique (INRA), Jouy-en-Josas, France.,INRA, US1367 MetaGenoPolis, 78350 Jouy en Josas, France
| | - Claire Fieschi
- Département d'Immunologie Clinique, Hôpital Saint-Louis, AP-HP, 75010 Paris, France.,INSERM U1126, Université Paris Diderot Paris 7, 75010 Paris, France
| | - S Dusko Ehrlich
- INRA, US1367 MetaGenoPolis, 78350 Jouy en Josas, France.,King's College London, Centre for Host-Microbiome Interactions, Dental Institute Central Office, Guy's Hospital, London, UK
| | - Martin Larsen
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses-Paris (CIMI-Paris), 75013 Paris, France. .,Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, 75013 Paris, France
| | - Guy Gorochov
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses-Paris (CIMI-Paris), 75013 Paris, France. .,Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, 75013 Paris, France
| |
Collapse
|