1
|
Feng Y, de Jong SE, Oliveira APBN, Samaha H, Yang F, Hu M, Wang Y, Beydoun N, Xie X, Zhang H, Kazmin D, Fang Z, Zou J, Gewirtz AT, Boyd SD, Hagan T, Rouphael N, Pulendran B. Antibiotic-induced gut microbiome perturbation alters the immune responses to the rabies vaccine. Cell Host Microbe 2025:S1931-3128(25)00126-X. [PMID: 40252648 DOI: 10.1016/j.chom.2025.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/12/2025] [Accepted: 03/27/2025] [Indexed: 04/21/2025]
Abstract
The gut microbiome plays a crucial role in modulating human immunity. Previously, we reported that antibiotic-induced microbiome perturbation affects influenza vaccine responses, depending on pre-existing immunity levels. Here, we employed a systems biology approach to analyze the impact of antibiotic administration on both primary and secondary immune responses to the rabies vaccine in humans. Antibiotic administration reduced the gut bacterial load, with a long-lasting reduction in commensal diversity. This alteration was associated with reduced rabies-specific humoral responses. Multi-omics profiling revealed that antibiotic administration induced (1) an enhanced pro-inflammatory signature early after vaccination, (2) a shift in the balance of vaccine-specific T-helper 1 (Th1) to T-follicular-helper response toward Th1 phenotype, and (3) profound alterations in metabolites, particularly in secondary bile acids in the blood. By integrating multi-omics datasets, we generated a multiscale, multi-response network that revealed key regulatory nodes, including the microbiota, secondary bile acids, and humoral immunity to vaccination.
Collapse
Affiliation(s)
- Yupeng Feng
- Institute of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou 510440, China; Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Sanne E de Jong
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Ana Paula B N Oliveira
- Department of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Hady Samaha
- Hope Clinic of the Emory Vaccine Center, Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Decatur, GA 30030, USA
| | - Fan Yang
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Mengyun Hu
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Yanli Wang
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Nour Beydoun
- Hope Clinic of the Emory Vaccine Center, Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Decatur, GA 30030, USA
| | - Xia Xie
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Haibo Zhang
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Dmitri Kazmin
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Zhuoqing Fang
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Jun Zou
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Andrew T Gewirtz
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Scott D Boyd
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA; Sean N. Parker Center for Allergy and Immunology Research, Stanford University, Stanford, CA 94305, USA; Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Thomas Hagan
- Department of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Nadine Rouphael
- Hope Clinic of the Emory Vaccine Center, Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Decatur, GA 30030, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
2
|
Ryan FJ, Clarke M, Lynn MA, Benson SC, McAlister S, Giles LC, Choo JM, Rossouw C, Ng YY, Semchenko EA, Richard A, Leong LEX, Taylor SL, Blake SJ, Mugabushaka JI, Walker M, Wesselingh SL, Licciardi PV, Seib KL, Tumes DJ, Richmond P, Rogers GB, Marshall HS, Lynn DJ. Bifidobacteria support optimal infant vaccine responses. Nature 2025:10.1038/s41586-025-08796-4. [PMID: 40175554 DOI: 10.1038/s41586-025-08796-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/17/2025] [Indexed: 04/04/2025]
Abstract
Accumulating evidence indicates that antibiotic exposure may lead to impaired vaccine responses1-4; however, the mechanisms underlying this association remain poorly understood. Here we prospectively followed 191 healthy, vaginally born, term infants from birth to 15 months, using a systems vaccinology approach to assess the effects of antibiotic exposure on immune responses to vaccination. Exposure to direct neonatal but not intrapartum antibiotics was associated with significantly lower antibody titres against various polysaccharides in the 13-valent pneumococcal conjugate vaccine and the Haemophilus influenzae type b polyribosylribitol phosphate and diphtheria toxoid antigens in the combined 6-in-1 Infanrix Hexa vaccine at 7 months of age. Blood from infants exposed to neonatal antibiotics had an inflammatory transcriptional profile before vaccination; in addition, faecal metagenomics showed reduced abundance of Bifidobacterium species in these infants at the time of vaccination, which was correlated with reduced vaccine antibody titres 6 months later. In preclinical models, responses to the 13-valent pneumococcal conjugate vaccine were strongly dependent on an intact microbiota but could be restored in germ-free mice by administering a consortium of Bifidobacterium species or a probiotic already widely used in neonatal units. Our data suggest that microbiota-targeted interventions could mitigate the detrimental effects of early-life antibiotics on vaccine immunogenicity.
Collapse
Affiliation(s)
- Feargal J Ryan
- Precision Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Michelle Clarke
- Women's and Children's Health Network, North Adelaide, South Australia, Australia
- Adelaide Medical School and The Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
- Vaccinology and Immunology Research Trials Unit, Women's and Children's Health Network, Adelaide, South Australia, Australia
| | - Miriam A Lynn
- Precision Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Saoirse C Benson
- Precision Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Sonia McAlister
- Wesfarmers Centre for Vaccines and Infectious Diseases, The Kids Institute, Perth, Western Australia, Australia
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - Lynne C Giles
- School of Public Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Jocelyn M Choo
- Precision Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Charné Rossouw
- Precision Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Yan Yung Ng
- Vaccine Immunology, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Evgeny A Semchenko
- Institute for Biomedicine and Glycomics, Griffith University, Southport, Queensland, Australia
| | - Alyson Richard
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Lex E X Leong
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Steven L Taylor
- Precision Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Stephen J Blake
- Precision Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Joyce I Mugabushaka
- Precision Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Mary Walker
- Women's and Children's Health Network, North Adelaide, South Australia, Australia
| | - Steve L Wesselingh
- Precision Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Paul V Licciardi
- Vaccine Immunology, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Kate L Seib
- Institute for Biomedicine and Glycomics, Griffith University, Southport, Queensland, Australia
| | - Damon J Tumes
- Precision Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia
| | - Peter Richmond
- Wesfarmers Centre for Vaccines and Infectious Diseases, The Kids Institute, Perth, Western Australia, Australia
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia
- Department of Immunology and General Paediatrics, Perth Children's Hospital, Nedlands, Western Australia, Australia
| | - Geraint B Rogers
- Precision Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Helen S Marshall
- Women's and Children's Health Network, North Adelaide, South Australia, Australia
- Adelaide Medical School and The Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
- Vaccinology and Immunology Research Trials Unit, Women's and Children's Health Network, Adelaide, South Australia, Australia
| | - David J Lynn
- Precision Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia.
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia.
| |
Collapse
|
3
|
Fusco EM, Bower L, Polidoro R, Minns AM, Lindner SE, Schmidt NW. Microbiome-mediated modulation of immune memory to P. yoelii affects the resistance to secondary cerebral malaria challenge. Immunohorizons 2025; 9:vlaf009. [PMID: 40193560 DOI: 10.1093/immhor/vlaf009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/29/2025] [Indexed: 04/09/2025] Open
Abstract
Malaria is caused by protozoan parasites in the genus Plasmodium. Over time individuals slowly develop clinical immunity to malaria, but this process occurs at variable rates, and the mechanism of protection is not fully understood. We have recently demonstrated that in genetically identical C57BL/6N mice, gut microbiota composition dramatically impacts the quality of the humoral immune response to Plasmodium yoelii and subsequent protection against a lethal secondary challenge with Plasmodium berghei ANKA in C57BL/6N mice. Here, we utilize this genetically identical, gut microbiome-dependent model to investigate how the gut microbiota modulate immunological memory, hypothesizing that the gut microbiome impacts the formation and functionality of immune memory. In support of this hypothesis, P. yoelii hyperparasitemia-resistant C57BL/6N mice exhibit increased protection against P. berghei ANKA-induced experimental cerebral malaria (ECM) compared to P. yoelii hyperparasitemia-susceptible C57BL/6N mice. Despite differences in protection against ECM, P. yoelii-resistant and -susceptible mice accumulate similar numbers of memory B cells (MBCs) and memory T cells. Following challenge with P. berghei ANKA, P. yoelii-resistant mice generated more rapid germinal center reactions; however, P. yoelii-resistant and -susceptible mice had similar titers of P. yoelii- and P. berghei-specific antibodies. In contrast, P. yoelii-resistant mice had an increased number of regulatory T cells in response to secondary challenge with P. berghei ANKA, which may dampen the immune-mediated breakdown of the blood-brain barrier and susceptibility to P. berghei-induced ECM. These findings demonstrate the ability of the gut microbiome to shape immune memory and the potential to enhance resistance to severe malaria outcomes.
Collapse
Affiliation(s)
- Elizabeth M Fusco
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Layne Bower
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Rafael Polidoro
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Allen M Minns
- The Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA, United States
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, United States
- The Huck Center for Malaria Research, University Park, PA, United States
| | - Scott E Lindner
- The Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA, United States
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, United States
- The Huck Center for Malaria Research, University Park, PA, United States
| | - Nathan W Schmidt
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
4
|
Rossouw C, Ryan FJ, Lynn DJ. The role of the gut microbiota in regulating responses to vaccination: current knowledge and future directions. FEBS J 2025; 292:1480-1499. [PMID: 39102299 PMCID: PMC11927049 DOI: 10.1111/febs.17241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/13/2024] [Accepted: 07/24/2024] [Indexed: 08/07/2024]
Abstract
Antigen-specific B and T cell responses play a critical role in vaccine-mediated protection against infectious diseases, but these responses are highly variable between individuals and vaccine immunogenicity is frequently sub-optimal in infants, the elderly and in people living in low- and middle-income countries. Although many factors such as nutrition, age, sex, genetics, environmental exposures, and infections may all contribute to variable vaccine immunogenicity, mounting evidence indicates that the gut microbiota is an important and targetable factor shaping optimal immune responses to vaccination. In this review, we discuss evidence from human, preclinical and experimental studies supporting a role for a healthy gut microbiota in mediating optimal vaccine immunogenicity, including the immunogenicity of COVID-19 vaccines. Furthermore, we provide an overview of the potential mechanisms through which this could occur and discuss strategies that could be used to target the microbiota to boost vaccine immunogenicity where it is currently sub-optimal.
Collapse
Affiliation(s)
- Charné Rossouw
- Precision MedicineSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
- Flinders Health and Medical Research InstituteFlinders UniversityBedford ParkAustralia
| | - Feargal J. Ryan
- Precision MedicineSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
- Flinders Health and Medical Research InstituteFlinders UniversityBedford ParkAustralia
| | - David J. Lynn
- Precision MedicineSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
- Flinders Health and Medical Research InstituteFlinders UniversityBedford ParkAustralia
| |
Collapse
|
5
|
Ng HY, Liao Y, Cheung CL, Zhang R, Chan KH, Seto WK, Leung WK, Hung IFN, Lam TTY, Cheung KS. Gut microbiota is associated with persistence of longer-term BNT162b2 vaccine immunogenicity. Front Immunol 2025; 16:1534787. [PMID: 40083550 PMCID: PMC11903479 DOI: 10.3389/fimmu.2025.1534787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/07/2025] [Indexed: 03/16/2025] Open
Abstract
Introduction BNT162b2 immunogenicity wanes with time and we investigated association between gut microbiota and longer-term immunogenicity. Methods This cohort study prospectively recruited adult BNT162b2 two-dose recipients from three vaccination centers in Hong Kong. Blood samples were collected at baseline and day 180 after first dose, and tested for neutralizing antibodies (NAb) against receptor-binding domain (RBD) of wild type SARS-CoV-2 virus using chemiluminescence immunoassay. Shotgun DNA metagenomic sequencing was performed to characterize baseline stool microbiome. Baseline metabolites were measured by gas and liquid chromatography-tandem mass spectrometry (GC-MS/MS and LC-MS/MS). Primary outcome was persistent high NAb response (defined as top 25% of NAb level) at day 180. Putative bacterial species and metabolic pathways were identified using linear discriminant analysis [LDA] effect size analysis. Multivariable logistic regression adjusting for clinical factors was used to derive adjusted odds ratio (aOR) of outcome with bacterial species and metabolites. Results Of 242 subjects (median age: 50.2 years [IQR:42.5-55.6]; male:85 [35.1%]), 61 (25.2%) were high-responders while 33 (13.6%) were extreme-high responders (defined as NAb≥200AU/mL). None had COVID-19 at end of study. Ruminococcus bicirculans (log10LDA score=3.65), Parasutterella excrementihominis (score=2.82) and Streptococcus salivarius (score=2.31) were enriched in high-responders, while Bacteroides thetaiotaomicron was enriched in low-responders (score=-3.70). On multivariable analysis, bacterial species (R. bicirculans-aOR: 1.87, 95% CI: 1.02-3.51; P. excrementihominis-aOR: 2.2, 95% CI: 1.18-4.18; S. salivarius-aOR: 2.09, 95% CI: 1.13-3.94) but not clinical factors associated with high response. R. bicirculans positively correlated with most metabolic pathways enriched in high-responders, including superpathway of L-cysteine biosynthesis (score=2.25) and L-isoleucine biosynthesis I pathway (score=2.16) known to benefit immune system. Baseline serum butyrate (aOR:10.00, 95% CI:1.81-107.2) and isoleucine (aOR:1.17, 95% CI:1.04-1.35) significantly associated with extreme-high vaccine response. Conclusion Certain gut bacterial species, metabolic pathways and metabolites associate with longer-term COVID-19 vaccine immunogenicity.
Collapse
Affiliation(s)
- Ho Yu Ng
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yunshi Liao
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ching Lung Cheung
- Department of Pharmacology and Pharmacy, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
- Laboratory of Data Discovery for Health Limited, 19W Hong Kong Science & Technology Parks, Hong Kong, Hong Kong SAR, China
| | - Ruiqi Zhang
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
| | - Kwok Hung Chan
- Department of Microbiology, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
| | - Wai-Kay Seto
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
| | - Wai K. Leung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
| | - Ivan F. N. Hung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
| | - Tommy T. Y. Lam
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Laboratory of Data Discovery for Health Limited, 19W Hong Kong Science & Technology Parks, Hong Kong, Hong Kong SAR, China
- Centre for Immunology & Infection Limited, 17W Hong Kong Science & Technology Parks, Hong Kong, Hong Kong SAR, China
| | - Ka Shing Cheung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
6
|
Gutzeit C, Grasset EK, Matthews DB, Maglione PJ, Britton GJ, Miller H, Magri G, Tomalin L, Stapylton M, Canales-Herrerias P, Sominskaia M, Guzman M, Pybus M, Tejedor Vaquero S, Radigan L, Tachó-Piñot R, Martín Nalda A, García Prat M, Martinez Gallo M, Dieli-Crimi R, Clemente JC, Mehandru S, Suarez-Farinas M, Faith JJ, Cunningham-Rundles C, Cerutti A. Gut IgA functionally interacts with systemic IgG to enhance antipneumococcal vaccine responses. SCIENCE ADVANCES 2025; 11:eado9455. [PMID: 39937896 PMCID: PMC11817949 DOI: 10.1126/sciadv.ado9455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 01/13/2025] [Indexed: 02/14/2025]
Abstract
The gut microbiota enhances systemic immunoglobulin G (IgG) responses to vaccines, but it is unknown whether this effect involves IgA, which coats intestinal microbes. That IgA may amplify postimmune IgG production is suggested by the impaired IgG response to pneumococcal vaccines in some IgA-deficient patients. Here, we found that antipneumococcal but not total IgG production was impaired in mice with IgA deficiency. The positive effect of gut IgA on antipneumococcal IgG responses started very early in life and could implicate gut bacteria, as these responses were attenuated in germ-free mice recolonized with gut microbes from IgA-deficient donors. IgA could exert this effect by constraining the systemic translocation of gut antigens, which was associated with chronic immune activation, including T cell overexpression of programmed cell death protein 1 (PD-1). This inhibitory receptor may attenuate antipneumococcal IgG production by causing B cell hyporesponsiveness, which improved upon anti-PD-1 treatment. Thus, gut IgA functionally interacts with systemic IgG to enhance antipneumococcal vaccine responses.
Collapse
Affiliation(s)
- Cindy Gutzeit
- Department of Medicine, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Emilie K. Grasset
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10021, USA
- Gale and Ira Drukier Institute for Children’s Health, Weill Cornell Medicine, New York, NY 10021, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Dean B. Matthews
- Immunology Program of the Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | - Paul J. Maglione
- Pulmonary Center and Department of Medicine, Boston University, Boston, MA 02118, USA
| | - Graham J. Britton
- Precision Immunology Institute, Icahn Institute for Data Science and Genome Technology, School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Haley Miller
- Department of Medicine, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Giuliana Magri
- Program for Inflammatory and Cardiovascular Disorders, Institute Hospital del Mar for Medical Investigations (IMIM), 08003 Barcelona, Spain
| | - Lewis Tomalin
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Matthew Stapylton
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Pablo Canales-Herrerias
- Department of Medicine, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Musia Sominskaia
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10021, USA
| | - Mauricio Guzman
- Program for Inflammatory and Cardiovascular Disorders, Institute Hospital del Mar for Medical Investigations (IMIM), 08003 Barcelona, Spain
| | - Marc Pybus
- Molecular Biology Laboratory, Fundació Puigvert, Instituto de Investigaciones Biomédicas Sant Pau (IIB-Sant Pau), 02041 Barcelona, Spain
| | - Sonia Tejedor Vaquero
- Program for Inflammatory and Cardiovascular Disorders, Institute Hospital del Mar for Medical Investigations (IMIM), 08003 Barcelona, Spain
| | - Lin Radigan
- Departments of Medicine and Pediatrics, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Roser Tachó-Piñot
- Program for Inflammatory and Cardiovascular Disorders, Institute Hospital del Mar for Medical Investigations (IMIM), 08003 Barcelona, Spain
| | - Andrea Martín Nalda
- Infection in Immunocompromised Pediatric Patients Research Group, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron University Hospital (HUVH), Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Vall d’Hebron University Hospital (HUVH), Barcelona Autònoma University (UAB), 48201 Barcelona, Spain
- Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, 08035 Barcelona, Spain
| | - Marina García Prat
- Infection in Immunocompromised Pediatric Patients Research Group, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron University Hospital (HUVH), Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Vall d’Hebron University Hospital (HUVH), Barcelona Autònoma University (UAB), 48201 Barcelona, Spain
- Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, 08035 Barcelona, Spain
| | - Monica Martinez Gallo
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Vall d’Hebron University Hospital (HUVH), Barcelona Autònoma University (UAB), 48201 Barcelona, Spain
- Division of Immunology, Vall d’Hebron University Hospital (HUVH), Barcelona Autònoma University (UAB), 48201 Barcelona, Spain
| | - Romina Dieli-Crimi
- Division of Immunology, Vall d’Hebron University Hospital (HUVH), Barcelona Autònoma University (UAB), 48201 Barcelona, Spain
| | - José C. Clemente
- Department of Medicine, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Saurabh Mehandru
- Department of Medicine, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Division of Gastroenterology, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mayte Suarez-Farinas
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jeremiah J. Faith
- Department of Medicine, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Charlotte Cunningham-Rundles
- Departments of Medicine and Pediatrics, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Andrea Cerutti
- Department of Medicine, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Program for Inflammatory and Cardiovascular Disorders, Institute Hospital del Mar for Medical Investigations (IMIM), 08003 Barcelona, Spain
- Catalan Institute for Research and Advanced Studies (ICREA), 08003 Barcelona, Spain
| |
Collapse
|
7
|
Brown JA, Bashir H, Zeng MY. Lifelong partners: Gut microbiota-immune cell interactions from infancy to old age. Mucosal Immunol 2025:S1933-0219(25)00006-6. [PMID: 39862964 DOI: 10.1016/j.mucimm.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
Our immune system and gut microbiota are intricately coupled from birth, both going through maturation during early life and senescence during aging almost in a synchronized fashion. The symbiotic relationship between the human host and microbiota is critically dependent on a healthy immune system to keep our microbiota in check, while the microbiota provides essential functions to promote the development and fitness of our immune system. The partnership between our immune system and microbiota is particularly important during early life, when microbial ligands and metabolites shape the development of the immune cells and immune tolerance; during aging, having sufficient beneficial gut bacteria is critical for the maintenance of intact mucosal barriers, immune metabolic fitness, and strong immunity against pathogens. The immune system during childhood is programmed, with the support of the microbiota, to develop robust immune tolerance, and limit autoimmunity and metabolic dysregulation, which are prevalent during aging. This review comprehensively explores the mechanistic underpinnings of gut microbiota-immune cell interactions during infancy and old age, with the goal to gain a better understanding of potential strategies to leverage the gut microbiota to combat age-related immune decline.
Collapse
Affiliation(s)
- Julia A Brown
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States
| | - Hilal Bashir
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States
| | - Melody Y Zeng
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School, New York, NY 10065, United States.
| |
Collapse
|
8
|
Taitz JJ, Tan J, Ni D, Potier-Villette C, Grau G, Nanan R, Macia L. Antibiotic-mediated dysbiosis leads to activation of inflammatory pathways. Front Immunol 2025; 15:1493991. [PMID: 39850904 PMCID: PMC11754057 DOI: 10.3389/fimmu.2024.1493991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/19/2024] [Indexed: 01/25/2025] Open
Abstract
Introduction The gut microbiota plays a pivotal role in influencing host health, through the production of metabolites and other key signalling molecules. While the impact of specific metabolites or taxa on host cells is well-documented, the broader impact of a disrupted microbiota on immune homeostasis is less understood, which is particularly important in the context of the increasing overuse of antibiotics. Methods Female C57BL/6 mice were gavaged twice daily for four weeks with Vancomycin, Polymyxin B, or PBS (control). Caecal microbiota composition was assessed via 16S rRNA sequencing and caecal metabolites were quantified with NMR spectroscopy. Immune profiles of spleen and mesenteric lymph nodes (MLNs) were assessed by flow cytometry, and splenocytes assessed for ex vivo cytokine production. A generalised additive model approach was used to examine the relationship between global antibiotic consumption and IBD incidence. Results Antibiotics significantly altered gut microbiota composition, reducing alpha-diversity. Acetate and butyrate were significantly reduced in antibiotic groups, while propionate and succinate increased in Vancomycin and PmB-treated mice, respectively. The MLNs and spleen showed changes only to DC numbers. Splenocytes from antibiotic-treated mice stimulated ex vivo exhibited increased production of TNF. Epidemiological analysis revealed a positive correlation between global antibiotic consumption and IBD incidence. Discussion Our findings demonstrate that antibiotic-mediated dysbiosis results in significantly altered short-chain fatty acid levels but immune homeostasis in spleen and MLNs at steady state is mostly preserved. Non-specific activation of splenocytes ex vivo, however, revealed mice with perturbed microbiota had significantly elevated production of TNF. Thus, this highlights antibiotic-mediated disruption of the gut microbiota may program the host towards dysregulated immune responses, predisposing to the development of TNF-associated autoimmune or chronic inflammatory disease.
Collapse
Affiliation(s)
- Jemma J. Taitz
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Jian Tan
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Duan Ni
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Camille Potier-Villette
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Georges Grau
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Vascular Immunology Unit, Discipline of Pathology, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
| | - Ralph Nanan
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Sydney Medical School Nepean, The University of Sydney, Sydney, NSW, Australia
| | - Laurence Macia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Sydney Medical School Nepean, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
9
|
Eftekhari Z, Doroud D, Tajabadi-Ebrahimi M, Kazemi-Lomedasht F. Improving Vaccine Response through Probiotics and Micronutrient Supplementation: Evaluating the Role of TLR5 in Adult Female BALB/c Mice. Curr Pharm Des 2025; 31:233-242. [PMID: 39257145 DOI: 10.2174/0113816128310203240823053538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/29/2024] [Indexed: 09/12/2024]
Abstract
BACKGROUND The role of probiotics and micronutrients in improving immune system function and response to vaccination has been proven. Hence, this study aimed to investigate the effects of probiotics enriched with micronutrients on the immunogenicity of PastoCovac® vaccine. METHODS The probiotic supplement BioBoost® and PastoCovac® vaccine, which contain six expressed Receptor- binding Domains (RBD) and conjugated with tetanus toxin, were administered concurrently. The safety and efficacy were assessed by determining Immunoglobulin G (IgG) antibody titers to RBD and cytokines, mRNA expression of Toll-like Receptors (TLRs) 5, and clinical symptoms. RESULTS Results revealed that the administration of the probiotics enriched with micronutrients and vitamins for 14 days before the first vaccine dose, followed by continued supplementation for 14 days after the first dose, and in conjunction with the second vaccine dose, yielded the most significant elevation in Interleukin 4 (IL-4), Tumor Necrosis Factor-alpha (TNF alpha), Interferon-gamma (IFN-gamma), and anti-SARS-CoV-2 RBD IgG levels within the supernatant samples collected from spleen cultures with the highest expression of TLR5 genes in intestinal samples, compared to the control group. CONCLUSION Our results indicated that the inclusion of probiotics enriched with micronutrients and vitamins significantly enhanced the immunogenicity of the PastoCovac® vaccine. Based on the recommendation to administer third and fourth vaccine doses, particularly for vulnerable and elderly individuals, the utilization of supplements containing probiotics is expected to favorably influence immune responses.
Collapse
Affiliation(s)
- Zohre Eftekhari
- Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Delaram Doroud
- Research and Production Complex, Pasteur Institute of Iran, Tehran, Iran
| | | | - Fatemeh Kazemi-Lomedasht
- Venom and Bio-therapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
10
|
Cortese M, Hagan T, Rouphael N, Wu SY, Xie X, Kazmin D, Wimmers F, Gupta S, van der Most R, Coccia M, Aranuchalam PS, Nakaya HI, Wang Y, Coyle E, Horiuchi S, Wu H, Bower M, Mehta A, Gunthel C, Bosinger SE, Kotliarov Y, Cheung F, Schwartzberg PL, Germain RN, Tsang J, Li S, Albrecht R, Ueno H, Subramaniam S, Mulligan MJ, Khurana S, Golding H, Pulendran B. System vaccinology analysis of predictors and mechanisms of antibody response durability to multiple vaccines in humans. Nat Immunol 2025; 26:116-130. [PMID: 39747435 DOI: 10.1038/s41590-024-02036-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/11/2024] [Indexed: 01/04/2025]
Abstract
We performed a systems vaccinology analysis to investigate immune responses in humans to an H5N1 influenza vaccine, with and without the AS03 adjuvant, to identify factors influencing antibody response magnitude and durability. Our findings revealed a platelet and adhesion-related blood transcriptional signature on day 7 that predicted the longevity of the antibody response, suggesting a potential role for platelets in modulating antibody response durability. As platelets originate from megakaryocytes, we explored the effect of thrombopoietin (TPO)-mediated megakaryocyte activation on antibody response longevity. We found that TPO administration enhanced the durability of vaccine-induced antibody responses. TPO-activated megakaryocytes also promoted survival of human bone-marrow plasma cells through integrin β1/β2-mediated cell-cell interactions, along with survival factors APRIL and the MIF-CD74 axis. Using machine learning, we developed a classifier based on this platelet-associated signature, which predicted antibody response longevity across six vaccines from seven independent trials, highlighting a conserved mechanism for vaccine durability.
Collapse
Affiliation(s)
- Mario Cortese
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Thomas Hagan
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | - Sheng-Yang Wu
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Xia Xie
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Dmitri Kazmin
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Florian Wimmers
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | - Shakti Gupta
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | | | | | - Prabhu S Aranuchalam
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA
| | | | - Yating Wang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Elizabeth Coyle
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Shu Horiuchi
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hanchih Wu
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mary Bower
- Hope Clinic of the Emory Vaccine Center, Decatur, GA, USA
| | - Aneesh Mehta
- Hope Clinic of the Emory Vaccine Center, Decatur, GA, USA
| | | | - Steve E Bosinger
- Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA, USA
- Yerkes Genomics Core Laboratory, Yerkes National Primate Research Center, Atlanta, GA, USA
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Yuri Kotliarov
- NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD, USA
| | - Foo Cheung
- NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD, USA
| | - Pamela L Schwartzberg
- NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD, USA
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA
| | - Ronald N Germain
- NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD, USA
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA
| | - John Tsang
- NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD, USA
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA
| | - Shuzhao Li
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Randy Albrecht
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hideki Ueno
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology, Kyoto University, Kyoto, Japan
| | - Shankar Subramaniam
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Mark J Mulligan
- Division of Infectious Diseases and Immunology, Department of Medicine and NYU Langone Vaccine Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Hana Golding
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
11
|
Pan J, Zhang X, Shi D, Tian X, Xu L, Lu X, Dong M, Yao P, Pan Z, Ling Z, Wu N, Yao H. Short-chain fatty acids play a key role in antibody response to SARS-CoV-2 infection in people living with HIV. Sci Rep 2024; 14:31211. [PMID: 39732792 DOI: 10.1038/s41598-024-82596-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 12/06/2024] [Indexed: 12/30/2024] Open
Abstract
High SARS-CoV-2-specific antibody levels can protect against SARS-CoV-2 reinfection. The gut microbiome can affect a host's immune response. However, its role in the antibody response to SARS-CoV-2 in people living with HIV (PLWH) remains poorly understood. Here, we categorised PLWH and healthy individuals into high- and low-antibody-response groups. Shotgun metagenomic sequencing and targeted metabolomic assays were used to investigate the differences in the gut microbiome and metabolic functions between the high- and low-antibody-response groups. PLWH demonstrated a higher abundance of short-chain fatty acid (SCFA)-producing species, accompanied by high serum levels of several SCFAs, in the high-antibody-response group than in the low-antibody-response group. In contrast, healthy individuals demonstrated higher enrichment of pilus-bearing bacterial species, with flagella-expressing genes, in the high-antibody-response group than in the low-antibody-response group. Therefore, gut-microbiota-derived SCFAs play a key role in antibody responses in PLWH but not in healthy individuals. Our results afford a novel understanding of how the gut microbiome and its metabolites are associated with host immunity. Moreover, they may facilitate the exploration of modalities to prevent SARS-CoV-2 reinfection through various gut-microbiota-targeted interventions tailored to different populations.
Collapse
Affiliation(s)
- Jingying Pan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250000, China
| | - Xiaodi Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Danrong Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Xuebin Tian
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Lijun Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Xiangyun Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Mingqing Dong
- Department of Infectious Disease, Zhejiang Qingchun Hospital, Zhejiang University, Hangzhou, 310000, China
| | - Peng Yao
- Department of Infectious Disease, Zhejiang Qingchun Hospital, Zhejiang University, Hangzhou, 310000, China
| | - Zhaoyi Pan
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250000, China
| | - Zongxin Ling
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Nanping Wu
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250000, China
| | - Hangping Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250000, China.
| |
Collapse
|
12
|
Ardura-Garcia C, Curtis N, Zimmermann P. Systematic review of the impact of intestinal microbiota on vaccine responses. NPJ Vaccines 2024; 9:254. [PMID: 39706841 DOI: 10.1038/s41541-024-01000-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 10/17/2024] [Indexed: 12/23/2024] Open
Abstract
The intestinal microbiota plays a critical role in host immunity and might contribute to the significant variation between individuals' vaccine responses. A systematic search was done using MEDLINE and Embase to identify original human studies investigating the association between intestinal microbiota composition and humoral and cellular vaccine responses. In total, 30 publications (26 studies, 14 in infants, 12 in adults), were included. Of these, 26 publications found an association between intestinal microbiota composition and vaccine responses. A beneficial effect of Actynomycetota (particularly Bifidobacterium) and a detrimental effect of Pseudomonadota (particularly Gammaproteobacteria) were observed across studies. Study designs were highly heterogenous, with variation in vaccine type, outcome measure, timing of stool analysis and analysis methods. Overall, studies support the concept that the composition of the intestinal microbiota influences vaccine responses. Further adequately powered studies are needed to confirm this association and inform potential microbiota-targeted interventions to optimise vaccine responses.
Collapse
Affiliation(s)
- Cristina Ardura-Garcia
- Department of Paediatrics, Fribourg Hospital, Fribourg, Switzerland
- Cambodia Oxford Medical Research Unit, Angkor Hospital for Children, Siem Reap, Cambodia
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
- Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, VIC, Australia
- Infectious Diseases Unit, The Royal Children's Hospital Melbourne, Parkville, VIC, Australia
| | - Petra Zimmermann
- Department of Paediatrics, Fribourg Hospital, Fribourg, Switzerland.
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.
- Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, VIC, Australia.
- Department for Community Health, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
13
|
Kroemer G, Montégut L, Kepp O, Zitvogel L. The danger theory of immunity revisited. Nat Rev Immunol 2024; 24:912-928. [PMID: 39511426 DOI: 10.1038/s41577-024-01102-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2024] [Indexed: 11/15/2024]
Abstract
The danger theory of immunity, introduced by Polly Matzinger in 1994, posits that tissue stress, damage or infection has a decisive role in determining immune responses. Since then, a growing body of evidence has supported the idea that the capacity to elicit cognate immune responses (immunogenicity) relies on the combination of antigenicity (the ability to be recognized by T cell receptors or antibodies) and adjuvanticity (additional signals arising owing to tissue damage). Here, we discuss the molecular foundations of the danger theory while focusing on immunologically relevant damage-associated molecular patterns, microorganism-associated molecular patterns, and neuroendocrine stress-associated immunomodulatory molecules, as well as on their receptors. We critically evaluate patient-relevant evidence, examining how cancer cells and pathogenic viruses suppress damage-associated molecular patterns to evade immune recognition, how intestinal dysbiosis can reduce immunostimulatory microorganism-associated molecular patterns and compromise immune responses, and which hereditary immune defects support the validity of the danger theory. Furthermore, we incorporate the danger hypothesis into a close-to-fail-safe hierarchy of immunological tolerance mechanisms that also involve the clonal deletion and inactivation of immune cells.
Collapse
Affiliation(s)
- Guido Kroemer
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - Léa Montégut
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Oliver Kepp
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France.
- INSERM UMR 1015, ClinicObiome, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France.
- Université Paris-Saclay, Ile-de-France, Paris, France.
- Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS), Villejuif, France.
| |
Collapse
|
14
|
An S, Oh J, Shon HJ, Song J, Choi YS, Kim D. Co-adjuvanting Nod2-stimulating bacteria with a TLR7 agonist elicits potent protective immunity against respiratory virus infection. Int J Antimicrob Agents 2024; 64:107369. [PMID: 39477030 DOI: 10.1016/j.ijantimicag.2024.107369] [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: 05/10/2024] [Revised: 09/17/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024]
Abstract
OBJECTIVES This study investigates the synergistic effect of combining the TLR7 agonist Imiquimod with either the Nod2 agonist (muramyl dipeptide; MDP) or commensal bacteria as nasal vaccine adjuvants to enhance immunity against respiratory viruses. METHODS Mice assessed immune responses, including antibody and cytokine profiles, after intranasal immunization with antigen and adjuvant combinations. BMDCs were cultured with these components to measure cytokine production. Germinal center formation and hapten-specific antibodies were evaluated using OT-II T-cell transfer and hapten-ovalbumin. Commensal bacteria from healthy nasal cavities were screened for Nod2-stimulating activity using a reporter assay. Protective efficacy against viral pathogens was evaluated using an influenza A infection model and a pseudovirus system for SARS-CoV-2 neutralizing antibodies. RESULTS Screening identified Imiquimod as a potent enhancer of adaptive immune responses during nasal immunization, showing synergy with MDP. This combination elevated IL-12p40 and IL-6 levels, enhanced antibody production, and promoted T follicular helper cell differentiation. The Imiquimod-MDP combination provided robust protection against influenza and SARS-CoV-2. Screening of commensal bacteria revealed differential Nod2-stimulating capacities, with Staphylococcus aureus exhibiting superior synergy with Imiquimod compared to Staphylococcus epidermidis. Notably, this synergism was abolished in Nod2-deficient mice, and pretreatment with S. aureus significantly enhanced the protective efficacy of Imiquimod against influenza compared to S. epidermidis. CONCLUSIONS Combining Imiquimod with MDP or high Nod2-stimulating bacteria offers a promising strategy for nasal vaccine adjuvants. These combinations effectively boost humoral and cellular immune responses, providing strong protection against respiratory viruses.
Collapse
Affiliation(s)
- Sujin An
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jeein Oh
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hoh-Jeong Shon
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jaehwan Song
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Youn Soo Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Donghyun Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea; Institute of Endemic Diseases, Seoul National University Medical Research Center, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Dutta R, Stothers L, Ackerman AL. Manipulating the Gut Microbiome in Urinary Tract Infection-Prone Patients. Urol Clin North Am 2024; 51:525-536. [PMID: 39349020 DOI: 10.1016/j.ucl.2024.07.016] [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: 10/02/2024]
Abstract
Although antibiotics remain the mainstay of urinary tract infection treatment, many affected women can be caught in a vicious cycle in which antibiotics given to eradicate one infection predispose them to develop another. This effect is primarily mediated by disturbances in the gut microbiome that both directly enrich for uropathogenic overgrowth and induce systemic alterations in inflammation, tissue permeability, and metabolism that also decrease host resistance to infection recurrences. Here, we discuss nonantibiotic approaches to manipulating the gut microbiome to reverse the systemic consequences of antibiotics, including cranberry supplementation and other dietary approaches, probiotic administration, and fecal microbiota transplantation.
Collapse
Affiliation(s)
- Rahul Dutta
- Division of Urogynecology and Reconstructive Pelvic Surgery, David Geffen School of Medicine at UCLA, Box 951738, Los Angeles, CA 90095-1738, USA
| | - Lynn Stothers
- Division of Urogynecology and Reconstructive Pelvic Surgery, David Geffen School of Medicine at UCLA, Box 951738, Los Angeles, CA 90095-1738, USA
| | - A Lenore Ackerman
- Division of Urogynecology and Reconstructive Pelvic Surgery, David Geffen School of Medicine at UCLA, Box 951738, Los Angeles, CA 90095-1738, USA.
| |
Collapse
|
16
|
Li Y, Molleston JM, Lovato C, Wright J, Erickson I, Bui D, Kim AH, Ingle H, Aggarwal S, Nolan LS, Hassan AO, Foster L, Diamond MS, Baldridge MT. Sequential early-life viral infections modulate the microbiota and adaptive immune responses to systemic and mucosal vaccination. PLoS Pathog 2024; 20:e1012557. [PMID: 39356719 PMCID: PMC11472911 DOI: 10.1371/journal.ppat.1012557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/14/2024] [Accepted: 09/01/2024] [Indexed: 10/04/2024] Open
Abstract
Increasing evidence points to the microbial exposome as a critical factor in maturing and shaping the host immune system, thereby influencing responses to immune challenges such as infections or vaccines. To investigate the effect of early-life viral exposures on immune development and vaccine responses, we inoculated mice with six distinct viral pathogens in sequence beginning in the neonatal period, and then evaluated their immune signatures before and after intramuscular or intranasal vaccination against SARS-CoV-2. Sequential viral infection drove profound changes in all aspects of the immune system, including increasing circulating leukocytes, altering innate and adaptive immune cell lineages in tissues, and markedly influencing serum cytokine and total antibody levels. Beyond changes in the immune responses, these exposures also modulated the composition of the endogenous intestinal microbiota. Although sequentially-infected mice exhibited increased systemic immune activation and T cell responses after intramuscular and intranasal SARS-CoV-2 immunization, we observed decreased vaccine-induced antibody responses in these animals. These results suggest that early-life viral exposures are sufficient to diminish antibody responses to vaccination in mice, and highlight the potential importance of considering prior microbial exposures when investigating vaccine responses.
Collapse
Affiliation(s)
- Yuhao Li
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Jerome M. Molleston
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Crystal Lovato
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Jasmine Wright
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Isabel Erickson
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Duyen Bui
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Andrew H. Kim
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Harshad Ingle
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Somya Aggarwal
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Lila S. Nolan
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Ahmed O. Hassan
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Lynne Foster
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Michael S. Diamond
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Megan T. Baldridge
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
17
|
Luo C, Yang Y, Jiang C, Lv A, Zuo W, Ye Y, Ke J. Influenza and the gut microbiota: A hidden therapeutic link. Heliyon 2024; 10:e37661. [PMID: 39315196 PMCID: PMC11417228 DOI: 10.1016/j.heliyon.2024.e37661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/31/2024] [Accepted: 09/07/2024] [Indexed: 09/25/2024] Open
Abstract
Background The extensive community of gut microbiota significantly influences various biological functions throughout the body, making its characterization a focal point in biomedicine research. Over the past few decades, studies have revealed a potential link between specific gut bacteria, their associated metabolic pathways, and influenza. Bacterial metabolites can communicate directly or indirectly with organs beyond the gut via the intestinal barrier, thereby impacting the physiological functions of the host. As the microbiota increasingly emerges as a 'gut signature' in influenza, gaining a deeper understanding of its role may offer new insights into its pathophysiological relevance and open avenues for novel therapeutic targets. In this Review, we explore the differences in gut microbiota between healthy individuals and those with influenza, the relationship between gut microbiota metabolites and influenza, and potential strategies for preventing and treating influenza through the regulation of gut microbiota and its metabolites, including fecal microbiota transplantation and microecological preparations. Methods We utilized PubMed and Web of Science as our search databases, employing keywords such as "influenza," "gut microbiota," "traditional Chinese medicine," "metabolites," "prebiotics," "probiotics," and "machine learning" to retrieve studies examining the potential therapeutic connections between the modulation of gut microbiota and its metabolites in the treatment of influenza. The search encompassed literature from the inception of the databases up to December 2023. Results Fecal microbiota transplantation (FMT), microbial preparations (probiotics and prebiotics), and traditional Chinese medicine have unique advantages in regulating intestinal microbiota and its metabolites to improve influenza outcomes. The primary mechanism involves increasing beneficial intestinal bacteria such as Bacteroidetes and Bifidobacterium while reducing harmful bacteria such as Proteobacteria. These interventions act directly or indirectly on metabolites such as short-chain fatty acids (SCFAs), amino acids (AAs), bile acids, and monoamines to alleviate lung inflammation, reduce viral load, and exert anti-influenza virus effects. Conclusion The gut microbiota and its metabolites have direct or indirect therapeutic effects on influenza, presenting broad research potential for providing new directions in influenza research and offering references for clinical prevention and treatment. Future research should focus on identifying key strains, specific metabolites, and immune regulation mechanisms within the gut microbiota to accurately target microbiota interventions and prevent respiratory viral infections such as influenza.
Collapse
Affiliation(s)
- Cheng Luo
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610032, China
| | - Yi Yang
- Hubei Provincial Hospital of Traditional Chinese Medicine, Hubei Academy of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Wuhan, 430074, China
| | - Cheng Jiang
- Hubei Provincial Hospital of Traditional Chinese Medicine, Hubei Academy of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Wuhan, 430074, China
| | - Anqi Lv
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, 430061, China
| | - Wanzhao Zuo
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, 430061, China
| | - Yuanhang Ye
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, 430061, China
| | - Jia Ke
- Hubei Provincial Hospital of Traditional Chinese Medicine, Hubei Academy of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Wuhan, 430074, China
| |
Collapse
|
18
|
Rappuoli R, Alter G, Pulendran B. Transforming vaccinology. Cell 2024; 187:5171-5194. [PMID: 39303685 PMCID: PMC11736809 DOI: 10.1016/j.cell.2024.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 06/24/2024] [Accepted: 07/12/2024] [Indexed: 09/22/2024]
Abstract
The COVID-19 pandemic placed the field of vaccinology squarely at the center of global consciousness, emphasizing the vital role of vaccines as transformative public health tools. The impact of vaccines was recently acknowledged by the award of the 2023 Nobel Prize in Physiology or Medicine to Katalin Kariko and Drew Weissman for their seminal contributions to the development of mRNA vaccines. Here, we provide a historic perspective on the key innovations that led to the development of some 27 licensed vaccines over the past two centuries and recent advances that promise to transform vaccines in the future. Technological revolutions such as reverse vaccinology, synthetic biology, and structure-based design transformed decades of vaccine failures into successful vaccines against meningococcus B and respiratory syncytial virus (RSV). Likewise, the speed and flexibility of mRNA vaccines profoundly altered vaccine development, and the advancement of novel adjuvants promises to revolutionize our ability to tune immunity. Here, we highlight exciting new advances in the field of systems immunology that are transforming our mechanistic understanding of the human immune response to vaccines and how to predict and manipulate them. Additionally, we discuss major immunological challenges such as learning how to stimulate durable protective immune response in humans.
Collapse
Affiliation(s)
| | - Galit Alter
- Moderna Therapeutics, Cambridge, MA 02139, USA.
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA; Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
19
|
Anderson CJ, Boeckaerts L, Chin P, Cardas JB, Xie W, Gonçalves A, Blancke G, Benson S, Rogatti S, Simpson MS, Davey A, Choi SM, Desmet S, Bushman SD, Goeminne G, Vandenabeele P, Desai MS, Vereecke L, Ravichandran KS. Metabolite-based inter-kingdom communication controls intestinal tissue recovery following chemotherapeutic injury. Cell Host Microbe 2024; 32:1469-1487.e9. [PMID: 39197455 DOI: 10.1016/j.chom.2024.07.026] [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: 01/04/2024] [Revised: 06/12/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024]
Abstract
Cytotoxic chemotherapies have devastating side effects, particularly within the gastrointestinal tract. Gastrointestinal toxicity includes the death and damage of the epithelium and an imbalance in the intestinal microbiota, otherwise known as dysbiosis. Whether dysbiosis is a direct contributor to tissue toxicity is a key area of focus. Here, from both mammalian and bacterial perspectives, we uncover an intestinal epithelial cell death-Enterobacteriaceae signaling axis that fuels dysbiosis. Specifically, our data demonstrate that chemotherapy-induced epithelial cell apoptosis and the purine-containing metabolites released from dying cells drive the inter-kingdom transcriptional re-wiring of the Enterobacteriaceae, including fundamental shifts in bacterial respiration and promotion of purine utilization-dependent expansion, which in turn delays the recovery of the intestinal tract. Inhibition of epithelial cell death or restriction of the Enterobacteriaceae to homeostatic levels reverses dysbiosis and improves intestinal recovery. These findings suggest that supportive therapies that maintain homeostatic levels of Enterobacteriaceae may be useful in resolving intestinal disease.
Collapse
Affiliation(s)
- Christopher J Anderson
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
| | - Laura Boeckaerts
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Priscilla Chin
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Javier Burgoa Cardas
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Wei Xie
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Amanda Gonçalves
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; VIB BioImaging Core, Ghent, Belgium
| | - Gillian Blancke
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Sam Benson
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Sebastian Rogatti
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Mariska S Simpson
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Anna Davey
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Sze Men Choi
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | | | - Summer D Bushman
- Department of Infection and Immunity, Luxembourg Institute of Health, L-4354 Esch-sur-Alzette, Luxembourg
| | | | - Peter Vandenabeele
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Mahesh S Desai
- Department of Infection and Immunity, Luxembourg Institute of Health, L-4354 Esch-sur-Alzette, Luxembourg
| | - Lars Vereecke
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Kodi S Ravichandran
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
20
|
Seong H, Yoon JG, Nham E, Choi YJ, Noh JY, Cheong HJ, Kim WJ, Kim EH, Kim C, Han YH, Lim S, Song JY. The gut microbiota modifies antibody durability and booster responses after SARS-CoV-2 vaccination. J Transl Med 2024; 22:827. [PMID: 39242525 PMCID: PMC11380214 DOI: 10.1186/s12967-024-05637-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 08/26/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines are pivotal in combating coronavirus disease 2019 (COVID-19); however, the declining antibody titers postvaccination pose challenges for sustained protection and herd immunity. Although gut microbiome is reported to affect the early antibody response after vaccination, its impact on the longevity of vaccine-induced antibodies remains unexplored. METHODS A prospective cohort study was conducted involving 44 healthy adults who received two doses of either the BNT162b2 or ChAdOx1 vaccine, followed by a BNT162b2 booster at six months. The gut microbiome was serially analyzed using 16S rRNA and shotgun sequencing, while humoral immune response was assessed using a SARS-CoV-2 spike protein immunoassay. RESULTS Faecalibacterium prausnitzii was associated with robust and persistent antibody responses post-BNT162b2 vaccination. In comparison, Escherichia coli was associated with a slower antibody decay following ChAdOx1 vaccination. The booster immune response was correlated with metabolic pathways involving cellular functions and aromatic amino acid synthesis. CONCLUSIONS The findings of this study underscored the potential interaction between the gut microbiome and the longevity/boosting effect of antibodies following vaccination against SARS-CoV-2. The identification of specific microbial associations suggests the prospect of microbiome-based strategies for enhancing vaccine efficacy.
Collapse
Affiliation(s)
- Hye Seong
- Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Asia Pacific Influenza Institute, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Vaccine Innovation Center-KU Medicine, Seoul, Republic of Korea
| | - Jin Gu Yoon
- Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Vaccine Innovation Center-KU Medicine, Seoul, Republic of Korea
| | - Eliel Nham
- Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Vaccine Innovation Center-KU Medicine, Seoul, Republic of Korea
| | - Yu Jung Choi
- Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Vaccine Innovation Center-KU Medicine, Seoul, Republic of Korea
| | - Ji Yun Noh
- Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Asia Pacific Influenza Institute, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Vaccine Innovation Center-KU Medicine, Seoul, Republic of Korea
| | - Hee Jin Cheong
- Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Asia Pacific Influenza Institute, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Vaccine Innovation Center-KU Medicine, Seoul, Republic of Korea
| | - Woo Joo Kim
- Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Asia Pacific Influenza Institute, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Vaccine Innovation Center-KU Medicine, Seoul, Republic of Korea
| | - Eui Ho Kim
- Viral Immunology Laboratory, Institut Pasteur Korea, Seongnam, Republic of Korea
| | - Chulwoo Kim
- Vaccine Innovation Center-KU Medicine, Seoul, Republic of Korea
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Young-Hee Han
- Department of Food and Nutrition, Chungbuk National University, Cheongju, Republic of Korea
| | - Sooyeon Lim
- Asia Pacific Influenza Institute, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea.
- Vaccine Innovation Center-KU Medicine, Seoul, Republic of Korea.
| | - Joon Young Song
- Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea.
- Asia Pacific Influenza Institute, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea.
- Vaccine Innovation Center-KU Medicine, Seoul, Republic of Korea.
| |
Collapse
|
21
|
Cheung KS, Yan VKC, Ye X, Hung IFN, Chan EW, Leung WK. Proton pump inhibitors associated with severe COVID-19 among two-dose but not three-dose vaccine recipients. J Gastroenterol Hepatol 2024; 39:1837-1846. [PMID: 38705849 DOI: 10.1111/jgh.16601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/16/2024] [Accepted: 04/21/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND AND AIM Proton pump inhibitors (PPIs) may increase the risk of COVID-19 among non-vaccinated subjects via various mechanisms, including gut dysbiosis. We aimed to investigate whether PPIs also affect the clinical outcomes of COVID-19 among vaccine recipients. METHODS This was a territory-wide cohort study of 3 272 286 vaccine recipients (aged ≥ 18 years) of ≥ 2 doses of either BNT162b2 or CoronaVac. Exclusion criteria included prior gastrointestinal surgery, immunocompromised status, and prior COVID-19. The primary outcome was COVID-19, and secondary outcomes included COVID-19-related hospitalization and severe infection (composite of intensive care unit admission, ventilatory support, and/or death). Covariates include age, sex, the Charlson Comorbidity Index, comorbidities, and concomitant medication use. Subjects were followed from index date (first dose of vaccination) until outcome occurrence, death, additional dose of vaccination, or March 31, 2022. Exposure was pre-vaccination PPI use (any prescription within 90 days before the index date). Propensity score (PS) matching and a Poisson regression model were used to estimate the adjusted incidence rate ratio (aIRR) of outcomes with PPI use. RESULTS Among 439 154 PS-matched two-dose vaccine recipients (mean age: 65.3 years; male: 45.7%) with a median follow-up of 6.8 months (interquartile range: 2.6-7.9), PPI exposure was associated with a higher risk of COVID-19 (aIRR: 1.08; 95% confidence interval [95% CI]: 1.05-1.10), hospitalization (aIRR: 1.20; 95% CI: 1.08-1.33), and severe infection (aIRR: 1.57; 95% CI: 1.24-1.98). Among 188 360 PS-matched three-dose vaccine recipients (mean age: 62.5 years; male: 49.0%; median follow-up: 9.1 months [interquartile range: 8.0-10.9]), PPIs were associated with higher infection risk (aIRR: 1.11; 95% CI: 1.08-1.15) but not other outcomes. CONCLUSIONS Although PPI use was associated with a higher COVID-19 risk, severe infection was limited to two-dose but not three-dose vaccine recipients.
Collapse
Affiliation(s)
- Ka Shing Cheung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Vincent K C Yan
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Xuxiao Ye
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Ivan F N Hung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Esther W Chan
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong
- Department of Pharmacy, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- The University of Hong Kong Shenzhen Institute of Research and Innovation, Shenzhen, China
| | - Wai K Leung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| |
Collapse
|
22
|
Sugrue JA, Duffy D. Systems vaccinology studies - achievements and future potential. Microbes Infect 2024; 26:105318. [PMID: 38460935 DOI: 10.1016/j.micinf.2024.105318] [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: 07/02/2023] [Revised: 02/22/2024] [Accepted: 03/01/2024] [Indexed: 03/11/2024]
Abstract
Human immune responses to vaccination are variable both within and between populations. Systems vaccinology, which is the application of multi-omics technologies to vaccine studies, seeks to understand such variation and predict responses to optimise vaccine strategies. Here, we outline new approaches to systems vaccinology, focusing on the incorporation of additional cohorts, endpoints and technologies.
Collapse
Affiliation(s)
- Jamie A Sugrue
- Translational Immunology Unit, Institut Pasteur, Université de Paris Cité, F75015, Paris, France
| | - Darragh Duffy
- Translational Immunology Unit, Institut Pasteur, Université de Paris Cité, F75015, Paris, France.
| |
Collapse
|
23
|
Azarmi M, Seyed Toutounchi N, Hogenkamp A, Thijssen S, Overbeek SA, Garssen J, Folkerts G, Van't Land B, Braber S. Human Milk Oligosaccharides in Combination with Galacto- and Long-Chain Fructo-Oligosaccharides Enhance Vaccination Efficacy in a Murine Influenza Vaccination Model. Nutrients 2024; 16:2858. [PMID: 39275175 PMCID: PMC11397401 DOI: 10.3390/nu16172858] [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: 08/02/2024] [Revised: 08/23/2024] [Accepted: 08/24/2024] [Indexed: 09/16/2024] Open
Abstract
Early-life nutrition significantly impacts vaccination efficacy in infants, whose immune response to vaccines is weaker compared to adults. This study investigated vaccination efficacy in female C57Bl/6JOlaHsd mice (6 weeks old) fed diets with 0.7% galacto-oligosaccharides (GOS)/long-chain fructo-oligosaccharides (lcFOS) (9:1), 0.3% human milk oligosaccharides (HMOS), or a combination (GFH) for 14 days prior to and during vaccination. Delayed-type hypersensitivity (DTH) was measured by assessing ear swelling following an intradermal challenge. Influvac-specific IgG1 and IgG2a levels were assessed using ELISAs, while splenic T and B lymphocytes were analyzed for frequency and activation via flow cytometry. Additionally, cytokine production was evaluated using murine splenocytes co-cultured with influenza-loaded dendritic cells. Mice on the GFH diet showed a significantly enhanced DTH response (p < 0.05), increased serological IgG1 levels, and a significant rise in memory B lymphocytes (CD27+ B220+ CD19+). GFH-fed mice also exhibited more activated splenic Th1 cells (CD69+ CXCR3+ CD4+) and higher IFN-γ production after ex vivo restimulation (p < 0.05). These findings suggest that GOS/lcFOS and HMOS, particularly in combination, enhance vaccine responses by improving memory B cells, IgG production, and Th1 cell activation, supporting the potential use of these prebiotics in infant formula for better early-life immune development.
Collapse
Affiliation(s)
- Mehrdad Azarmi
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Science (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Negisa Seyed Toutounchi
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Science (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Astrid Hogenkamp
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Science (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Suzan Thijssen
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Science (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Saskia A Overbeek
- Danone Global Research and Innovation Center B.V., 3584 CT Utrecht, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Science (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands
- Danone Global Research and Innovation Center B.V., 3584 CT Utrecht, The Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Science (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Belinda Van't Land
- Danone Global Research and Innovation Center B.V., 3584 CT Utrecht, The Netherlands
- Department of Pediatric Immunology, Wilhelmina Children Hospital, University Medical Center Utrecht, 3584 EA Utrecht, The Netherlands
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Science (UIPS), Utrecht University, 3584 CG Utrecht, The Netherlands
- Danone Global Research and Innovation Center B.V., 3584 CT Utrecht, The Netherlands
| |
Collapse
|
24
|
Zhang LN, Tan JT, Ng HY, Liao YS, Zhang RQ, Chan KH, Hung IFN, Lam TTY, Cheung KS. Baseline Gut Microbiota Was Associated with Long-Term Immune Response at One Year Following Three Doses of BNT162b2. Vaccines (Basel) 2024; 12:916. [PMID: 39204040 PMCID: PMC11359560 DOI: 10.3390/vaccines12080916] [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: 06/25/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
BACKGROUND This study explored neutralizing IgG antibody levels against COVID-19 decline over time post-vaccination. We conducted this prospective cohort study to investigate the function of gut microbiota in the host immune response following three doses of BNT162b2. METHODS Subjects who received three doses of BNT162b2 were recruited from three centers in Hong Kong. Blood samples were obtained before the first dose and at the one-year timepoint for IgG ELISA to determine the level of neutralizing antibody (NAb). The primary outcome was a high immune response (NAb > 600 AU/mL). We performed shotgun DNA metagenomic sequencing on baseline fecal samples to identify bacterial species and metabolic pathways associated with high immune response using linear discriminant analysis effect size analysis. RESULTS A total of 125 subjects were recruited (median age: 52 years [IQR: 46.2-59.0]; male: 43 [34.4%]), and 20 were regarded as low responders at the one-year timepoint. Streptococcus parasanguinis (log10LDA score = 2.38, p = 0.003; relative abundance of 2.97 × 10-5 vs. 0.03%, p = 0.001), Bacteroides stercoris (log10LDA score = 4.29, p = 0.024; relative abundance of 0.14% vs. 2.40%, p = 0.014) and Haemophilus parainfluenzae (log10LDA score = 2.15, p = 0.022; relative abundance of 0.01% vs. 0, p = 0.010) were enriched in low responders. Bifidobacterium pseudocatenulatum (log10LDA score = 2.99, p = 0.048; relative abundance of 0.09% vs. 0.36%, p = 0.049) and Clostridium leptum (log10LDA score = 2.38, p = 0.014; relative abundance of 1.2 × 10-5% vs. 0, p = 0.044) were enriched in high responders. S. parasanguinis was negatively correlated with the superpathway of pyrimidine ribonucleotides de novo biosynthesis (log10LDA score = 2.63), which contributes to inflammation and antibody production. H. parainfluenzae was positively correlated with pathways related to anti-inflammatory processes, including the superpathway of histidine, purine, and pyrimidine biosynthesis (log10LDA score = 2.14). CONCLUSION Among three-dose BNT162b2 recipients, S. parasanguinis, B. stercoris and H. parainfluenzae were associated with poorer immunogenicity at one year, while B. pseudocatenulatum and C. leptum was associated with a better response.
Collapse
Affiliation(s)
- Li-Na Zhang
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China (J.-T.T.); (R.-Q.Z.); (I.F.-N.H.)
| | - Jing-Tong Tan
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China (J.-T.T.); (R.-Q.Z.); (I.F.-N.H.)
| | - Ho-Yu Ng
- School of Clinical Medicine, The University of Hong Kong, Hong Kong, China;
| | - Yun-Shi Liao
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, The University of Hong Kong, Hong Kong, China; (Y.-S.L.); (T.T.-Y.L.)
- Centre for Immunology & Infection Limited, 17W Hong Kong Science & Technology Parks, Hong Kong, China
| | - Rui-Qi Zhang
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China (J.-T.T.); (R.-Q.Z.); (I.F.-N.H.)
| | - Kwok-Hung Chan
- Department of Microbiology, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China;
| | - Ivan Fan-Ngai Hung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China (J.-T.T.); (R.-Q.Z.); (I.F.-N.H.)
| | - Tommy Tsan-Yuk Lam
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, The University of Hong Kong, Hong Kong, China; (Y.-S.L.); (T.T.-Y.L.)
| | - Ka-Shing Cheung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China (J.-T.T.); (R.-Q.Z.); (I.F.-N.H.)
- Department of Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518000, China
| |
Collapse
|
25
|
Liu Y, Zhou J, Yang Y, Chen X, Chen L, Wu Y. Intestinal Microbiota and Its Effect on Vaccine-Induced Immune Amplification and Tolerance. Vaccines (Basel) 2024; 12:868. [PMID: 39203994 PMCID: PMC11359036 DOI: 10.3390/vaccines12080868] [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: 06/27/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 09/03/2024] Open
Abstract
This review provides the potential of intestinal microbiota in vaccine design and application, exploring the current insights into the interplay between the intestinal microbiota and the immune system, with a focus on its intermediary function in vaccine efficacy. It summarizes families and genera of bacteria that are part of the intestinal microbiota that may enhance or diminish vaccine efficacy and discusses the foundational principles of vaccine sequence design and the application of gut microbial characteristics in vaccine development. Future research should further investigate the use of multi-omics technologies to elucidate the interactive mechanisms between intestinal microbiota and vaccine-induced immune responses, aiming to optimize and improve vaccine design.
Collapse
Affiliation(s)
- Yixin Liu
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China;
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; (J.Z.); (L.C.)
| | - Jianfeng Zhou
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; (J.Z.); (L.C.)
| | - Yushang Yang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; (J.Z.); (L.C.)
| | - Xiangzheng Chen
- Department of Liver Surgery & Liver Transplantation, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Longqi Chen
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; (J.Z.); (L.C.)
| | - Yangping Wu
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China;
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Chengdu 610041, China
| |
Collapse
|
26
|
Caetano‐Silva ME, Shrestha A, Duff AF, Kontic D, Brewster PC, Kasperek MC, Lin C, Wainwright DA, Hernandez‐Saavedra D, Woods JA, Bailey MT, Buford TW, Allen JM. Aging amplifies a gut microbiota immunogenic signature linked to heightened inflammation. Aging Cell 2024; 23:e14190. [PMID: 38725282 PMCID: PMC11320341 DOI: 10.1111/acel.14190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 08/15/2024] Open
Abstract
Aging is associated with low-grade inflammation that increases the risk of infection and disease, yet the underlying mechanisms remain unclear. Gut microbiota composition shifts with age, harboring microbes with varied immunogenic capacities. We hypothesized the gut microbiota acts as an active driver of low-grade inflammation during aging. Microbiome patterns in aged mice strongly associated with signs of bacterial-induced barrier disruption and immune infiltration, including marked increased levels of circulating lipopolysaccharide (LPS)-binding protein (LBP) and colonic calprotectin. Ex vivo immunogenicity assays revealed that both colonic contents and mucosa of aged mice harbored increased capacity to activate toll-like receptor 4 (TLR4) whereas TLR5 signaling was unchanged. We found patterns of elevated innate inflammatory signaling (colonic Il6, Tnf, and Tlr4) and endotoxemia (circulating LBP) in young germ-free mice after 4 weeks of colonization with intestinal contents from aged mice compared with young counterparts, thus providing a direct link between aging-induced shifts in microbiota immunogenicity and host inflammation. Additionally, we discovered that the gut microbiota of aged mice exhibited unique responses to a broad-spectrum antibiotic challenge (Abx), with sustained elevation in Escherichia (Proteobacteria) and altered TLR5 immunogenicity 7 days post-Abx cessation. Together, these data indicate that old age results in a gut microbiota that differentially acts on TLR signaling pathways of the innate immune system. We found that these age-associated microbiota immunogenic signatures are less resilient to challenge and strongly linked to host inflammatory status. Gut microbiota immunogenic signatures should be thus considered as critical factors in mediating chronic inflammatory diseases disproportionally impacting older populations.
Collapse
Affiliation(s)
- Maria Elisa Caetano‐Silva
- Department of Health and KinesiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Division of Nutritional SciencesUniversity of Illinois at Urbana ChampaignUrbanaIllinoisUSA
| | - Akriti Shrestha
- Division of Nutritional SciencesUniversity of Illinois at Urbana ChampaignUrbanaIllinoisUSA
| | - Audrey F. Duff
- Center for Microbial PathogenesisNationwide Children's HospitalColumbusOhioUSA
| | - Danica Kontic
- Center for Microbial PathogenesisNationwide Children's HospitalColumbusOhioUSA
| | - Patricia C. Brewster
- Department of Health and KinesiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Mikaela C. Kasperek
- Division of Nutritional SciencesUniversity of Illinois at Urbana ChampaignUrbanaIllinoisUSA
| | - Chia‐Hao Lin
- Department of Health and KinesiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Derek A. Wainwright
- Departments of Cancer Biology and Neurological SurgeryLoyola University Chicago, Stritch School of MedicineMaywoodIllinoisUSA
| | - Diego Hernandez‐Saavedra
- Department of Health and KinesiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Division of Nutritional SciencesUniversity of Illinois at Urbana ChampaignUrbanaIllinoisUSA
| | - Jeffrey A. Woods
- Department of Health and KinesiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Division of Nutritional SciencesUniversity of Illinois at Urbana ChampaignUrbanaIllinoisUSA
| | - Michael T. Bailey
- Center for Microbial PathogenesisNationwide Children's HospitalColumbusOhioUSA
| | - Thomas W. Buford
- Division of Gerontology, Geriatrics and Palliative Care, Department of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
- Birmingham/Atlanta VA GRECCBirmingham VA Medical CenterBirminghamAlabamaUSA
| | - Jacob M. Allen
- Department of Health and KinesiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Division of Nutritional SciencesUniversity of Illinois at Urbana ChampaignUrbanaIllinoisUSA
| |
Collapse
|
27
|
Wolska M, Wypych TP, Rodríguez-Viso P. The Influence of Premature Birth on the Development of Pulmonary Diseases: Focus on the Microbiome. Metabolites 2024; 14:382. [PMID: 39057705 PMCID: PMC11279213 DOI: 10.3390/metabo14070382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Globally, around 11% of neonates are born prematurely, comprising a highly vulnerable population with a myriad of health problems. Premature births are often accompanied by an underdeveloped immune system biased towards a Th2 phenotype and microbiota dysbiosis. Typically, a healthy gut microbiota interacts with the host, driving the proper maturation of the host immunity. However, factors like cesarean section, formula milk feeding, hospitalization in neonatal intensive care units (NICU), and routine antibiotic treatments compromise microbial colonization and increase the risk of developing related diseases. This, along with alterations in the innate immune system, could predispose the neonates to the development of respiratory diseases later in life. Currently, therapeutic strategies are mainly focused on restoring gut microbiota composition using probiotics and prebiotics. Understanding the interactions between the gut microbiota and the immature immune system in premature neonates could help to develop novel therapeutic strategies for treating or preventing gut-lung axis disorders.
Collapse
Affiliation(s)
| | - Tomasz Piotr Wypych
- Laboratory of Host-Microbiota Interactions, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Ludwika Pasteura 3, 02-093 Warsaw, Poland; (M.W.); (P.R.-V.)
| | | |
Collapse
|
28
|
Tian K, Jing D, Lan J, Lv M, Wang T. Commensal microbiome and gastrointestinal mucosal immunity: Harmony and conflict with our closest neighbor. Immun Inflamm Dis 2024; 12:e1316. [PMID: 39023417 PMCID: PMC11256888 DOI: 10.1002/iid3.1316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/06/2024] [Accepted: 06/03/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND The gastrointestinal tract contains a wide range of microorganisms that have evolved alongside the immune system of the host. The intestinal mucosa maintains balance within the intestines by utilizing the mucosal immune system, which is controlled by the complex gut mucosal immune network. OBJECTIVE This review aims to comprehensively introduce current knowledge of the gut mucosal immune system, focusing on its interaction with commensal bacteria. RESULTS The gut mucosal immune network includes gut-associated lymphoid tissue, mucosal immune cells, cytokines, and chemokines. The connection between microbiota and the immune system occurs through the engagement of bacterial components with pattern recognition receptors found in the intestinal epithelium and antigen-presenting cells. This interaction leads to the activation of both innate and adaptive immune responses. The interaction between the microbial community and the host is vital for maintaining the balance and health of the host's mucosal system. CONCLUSION The gut mucosal immune network maintains a delicate equilibrium between active immunity, which defends against infections and damaging non-self antigens, and immunological tolerance, which allows for the presence of commensal microbiota and dietary antigens. This balance is crucial for the maintenance of intestinal health and homeostasis. Disturbance of gut homeostasis leads to enduring or severe gastrointestinal ailments, such as colorectal cancer and inflammatory bowel disease. Utilizing these factors can aid in the development of cutting-edge mucosal vaccines that have the ability to elicit strong protective immune responses at the primary sites of pathogen invasion.
Collapse
Affiliation(s)
- Kexin Tian
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
| | - Dehong Jing
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
| | - Junzhe Lan
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
| | - Mingming Lv
- Department of BreastWomen's Hospital of Nanjing Medical University, Nanjing Maternity, and Child Health Care HospitalNanjingChina
| | - Tingting Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
| |
Collapse
|
29
|
Labeur-Iurman L, Harker JA. Mechanisms of antibody mediated immunity - Distinct in early life. Int J Biochem Cell Biol 2024; 172:106588. [PMID: 38768890 DOI: 10.1016/j.biocel.2024.106588] [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: 11/17/2023] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/22/2024]
Abstract
Immune responses in early life are characterized by a failure to robustly generate long-lasting protective responses against many common pathogens or upon vaccination. This is associated with a reduced ability to generate T-cell dependent high affinity antibodies. This review highlights the differences in T-cell dependent antibody responses observed between infants and adults, in particular focussing on the alterations in immune cell function that lead to reduced T follicular helper cell-B cell crosstalk within germinal centres in early life. Understanding the distinct functional characteristics of early life humoral immunity, and how these are regulated, will be critical in guiding age-appropriate immunological interventions in the very young.
Collapse
Affiliation(s)
- Lucia Labeur-Iurman
- National Heart & Lung Institute, Imperial College London, London, United Kingdom.
| | - James A Harker
- National Heart & Lung Institute, Imperial College London, London, United Kingdom; Centre for Paediatrics and Child Health, Imperial College London, London, United Kingdom.
| |
Collapse
|
30
|
Norton T, Lynn MA, Rossouw C, Abayasingam A, Perkins G, Hissaria P, Bull RA, Lynn DJ. B and T cell responses to the BNT162b2 COVID-19 mRNA vaccine are not impaired in germ-free or antibiotic-treated mice. Gut 2024; 73:1222-1224. [PMID: 37500502 PMCID: PMC11185899 DOI: 10.1136/gutjnl-2023-329810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 07/15/2023] [Indexed: 07/29/2023]
Affiliation(s)
- Todd Norton
- Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Miriam Anne Lynn
- Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Infection and Immunity, Flinders Health and Medical Research Institute, Bedford Park, South Australia, Australia
| | - Charné Rossouw
- Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Infection and Immunity, Flinders Health and Medical Research Institute, Bedford Park, South Australia, Australia
| | - Arunasingam Abayasingam
- The Viral Immunology Systems Program, The Kirby Institute, Sydney, New South Wales, Australia
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Griffith Perkins
- Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, South Australia, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- Immunology Directorate, SA Pathology, Adelaide, South Australia, Australia
| | - Pravin Hissaria
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- Immunology Directorate, SA Pathology, Adelaide, South Australia, Australia
- Immunology Department, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Rowena Anne Bull
- The Viral Immunology Systems Program, The Kirby Institute, Sydney, New South Wales, Australia
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - David John Lynn
- Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Infection and Immunity, Flinders Health and Medical Research Institute, Bedford Park, South Australia, Australia
| |
Collapse
|
31
|
Boncheva I, Poudrier J, Falcone EL. Role of the intestinal microbiota in host defense against respiratory viral infections. Curr Opin Virol 2024; 66:101410. [PMID: 38718575 DOI: 10.1016/j.coviro.2024.101410] [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: 01/17/2024] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 06/07/2024]
Abstract
Viral infections, including those affecting the respiratory tract, can alter the composition of the intestinal microbiota, which, in turn, can significantly influence both innate and adaptive immune responses, resulting in either enhanced pathogen clearance or exacerbation of the infection, possibly leading to inflammatory complications. A deeper understanding of the interplay between the intestinal microbiota and host immune responses in the context of respiratory viral infections (i.e. the gut-lung axis) is necessary to develop new treatments. This review highlights key mechanisms by which the intestinal microbiota, including its metabolites, can act locally or at distant organs to combat respiratory viruses. Therapeutics aimed at harnessing the microbiota to prevent and/or help treat respiratory viral infections represent a promising avenue for future investigation.
Collapse
Affiliation(s)
- Idia Boncheva
- Center for Immunity, Inflammation and Infectious Diseases, Montreal Clinical Research Institute/Institut de recherches cliniques de Montréal (IRCM), Montreal, QC, Canada
| | - Johanne Poudrier
- Center for Immunity, Inflammation and Infectious Diseases, Montreal Clinical Research Institute/Institut de recherches cliniques de Montréal (IRCM), Montreal, QC, Canada; Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada
| | - Emilia L Falcone
- Center for Immunity, Inflammation and Infectious Diseases, Montreal Clinical Research Institute/Institut de recherches cliniques de Montréal (IRCM), Montreal, QC, Canada; Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada; Department of Medicine, Université de Montréal, Montreal, QC, Canada; Department of Microbiology and Infectious Diseases, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada.
| |
Collapse
|
32
|
Kelly AM, McCarthy KN, Claxton TJ, Carlile SR, O'Brien EC, Vozza EG, Mills KH, McLoughlin RM. IL-10 inhibition during immunization improves vaccine-induced protection against Staphylococcus aureus infection. JCI Insight 2024; 9:e178216. [PMID: 38973612 PMCID: PMC11383370 DOI: 10.1172/jci.insight.178216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/22/2024] [Indexed: 07/09/2024] Open
Abstract
Staphylococcus aureus is a major human pathogen. An effective anti-S. aureus vaccine remains elusive as the correlates of protection are ill-defined. Targeting specific T cell populations is an important strategy for improving anti-S. aureus vaccine efficacy. Potential bottlenecks that remain are S. aureus-induced immunosuppression and the impact this might have on vaccine-induced immunity. S. aureus induces IL-10, which impedes effector T cell responses, facilitating persistence during both colonization and infection. Thus, it was hypothesized that transient targeting of IL-10 might represent an innovative way to improve vaccine efficacy. In this study, IL-10 expression was elevated in the nares of persistent carriers of S. aureus, and this was associated with reduced systemic S. aureus-specific Th1 responses. This suggests that systemic responses are remodeled because of commensal exposure to S. aureus, which negatively implicates vaccine function. To provide proof of concept that targeting immunosuppressive responses during immunization may be a useful approach to improve vaccine efficacy, we immunized mice with T cell-activating vaccines in combination with IL-10-neutralizing antibodies. Blocking IL-10 during vaccination enhanced effector T cell responses and improved bacterial clearance during subsequent systemic and subcutaneous infection. Taken together, these results reveal a potentially novel strategy for improving anti-S. aureus vaccine efficacy.
Collapse
Affiliation(s)
| | - Karen N McCarthy
- Host-Pathogen Interactions Group and
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | | | | | - Kingston Hg Mills
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | |
Collapse
|
33
|
Shannon CP, Lee AH, Tebbutt SJ, Singh A. A Commentary on Multi-omics Data Integration in Systems Vaccinology. J Mol Biol 2024; 436:168522. [PMID: 38458605 DOI: 10.1016/j.jmb.2024.168522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 03/10/2024]
Affiliation(s)
| | - Amy Hy Lee
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Scott J Tebbutt
- PROOF Centre of Excellence, Vancouver, Canada; Department of Medicine, The University of British Columbia, Vancouver, Canada; Centre for Heart Lung Innovation, Vancouver, Canada
| | - Amrit Singh
- Centre for Heart Lung Innovation, Vancouver, Canada; Department of Anesthesiology, Pharmacology and Therapeutics, The University of British Columbia, Vancouver, Canada.
| |
Collapse
|
34
|
Zhang LN, Tan JT, Ng HY, Liao YS, Zhang RQ, Chan KH, Hung IFN, Lam TTY, Cheung KS. Association between Gut Microbiota Composition and Long-Term Vaccine Immunogenicity following Three Doses of CoronaVac. Vaccines (Basel) 2024; 12:365. [PMID: 38675747 PMCID: PMC11055114 DOI: 10.3390/vaccines12040365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/15/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Neutralizing antibody level wanes with time after COVID-19 vaccination. We aimed to study the relationship between baseline gut microbiota and immunogenicity after three doses of CoronaVac. METHODS This was a prospective cohort study recruiting three-dose CoronaVac recipients from two centers in Hong Kong. Blood samples were collected at baseline and one year post-first dose for virus microneutralization (vMN) assays to determine neutralization titers. The primary outcome was high immune response (defined as with vMN titer ≥ 40). Shotgun DNA metagenomic sequencing of baseline fecal samples identified potential bacterial species and metabolic pathways using Linear Discriminant Analysis Effect Size (LEfSe) analysis. Univariate and multivariable logistic regression models were used to identify high response predictors. RESULTS In total, 36 subjects were recruited (median age: 52.7 years [IQR: 47.9-56.4]; male: 14 [38.9%]), and 18 had low immune response at one year post-first dose vaccination. Eubacterium rectale (log10LDA score = 4.15, p = 0.001; relative abundance of 1.4% vs. 0, p = 0.002), Collinsella aerofaciens (log10LDA score = 3.31, p = 0.037; 0.39% vs. 0.18%, p = 0.038), and Streptococcus salivarius (log10LDA score = 2.79, p = 0.021; 0.05% vs. 0.02%, p = 0.022) were enriched in low responders. The aOR of high immune response with E. rectale, C. aerofaciens, and S. salivarius was 0.03 (95% CI: 9.56 × 10-4-0.32), 0.03 (95% CI: 4.47 × 10-4-0.59), and 10.19 (95% CI: 0.81-323.88), respectively. S. salivarius had a positive correlation with pathways enriched in high responders like incomplete reductive TCA cycle (log10LDA score = 2.23). C. aerofaciens similarly correlated with amino acid biosynthesis-related pathways. These pathways all showed anti-inflammation functions. CONCLUSION E. rectale,C. aerofaciens, and S. salivarius correlated with poorer long-term immunogenicity following three doses of CoronaVac.
Collapse
Affiliation(s)
- Li-Na Zhang
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Jing-Tong Tan
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Ho-Yu Ng
- School of Clinical Medicine, The University of Hong Kong, Hong Kong
| | - Yun-Shi Liao
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, The University of Hong Kong, Hong Kong
- Centre for Immunology & Infection Limited, 17W Hong Kong Science & Technology Parks, Hong Kong
| | - Rui-Qi Zhang
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Kwok-Hung Chan
- Centre for Immunology & Infection Limited, 17W Hong Kong Science & Technology Parks, Hong Kong
| | - Ivan Fan-Ngai Hung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Tommy Tsan-Yuk Lam
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, The University of Hong Kong, Hong Kong
| | - Ka-Shing Cheung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| |
Collapse
|
35
|
Rio P, Caldarelli M, Chiantore M, Ocarino F, Candelli M, Gasbarrini A, Gambassi G, Cianci R. Immune Cells, Gut Microbiota, and Vaccines: A Gender Perspective. Cells 2024; 13:526. [PMID: 38534370 PMCID: PMC10969451 DOI: 10.3390/cells13060526] [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: 02/17/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 03/28/2024] Open
Abstract
The development of preventive and therapeutic vaccines has played a crucial role in preventing infections and treating chronic and non-communicable diseases, respectively. For a long time, the influence of sex differences on modifying health and disease has not been addressed in clinical and preclinical studies. The interaction of genetic, epigenetic, and hormonal factors plays a role in the sex-related differences in the epidemiology of diseases, clinical manifestations, and the response to treatment. Moreover, sex is one of the leading factors influencing the gut microbiota composition, which could further explain the different predisposition to diseases in men and women. In the same way, differences between sexes occur also in the immune response to vaccines. This narrative review aims to highlight these differences, focusing on the immune response to vaccines. Comparative data about immune responses, vaccine effectiveness, and side effects are reviewed. Hence, the intricate interplay between sex, immunity, and the gut microbiota will be discussed for its potential role in the response to vaccination. Embracing a sex-oriented perspective in research may improve the efficacy of the immune response and allow the design of tailored vaccine schedules.
Collapse
Affiliation(s)
- Pierluigi Rio
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (P.R.); (M.C.); (M.C.); (F.O.); (A.G.); (G.G.)
| | - Mario Caldarelli
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (P.R.); (M.C.); (M.C.); (F.O.); (A.G.); (G.G.)
| | - Monica Chiantore
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (P.R.); (M.C.); (M.C.); (F.O.); (A.G.); (G.G.)
| | - Francesca Ocarino
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (P.R.); (M.C.); (M.C.); (F.O.); (A.G.); (G.G.)
| | - Marcello Candelli
- Department of Emergency, Anesthesiological and Reanimation Sciences, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy;
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (P.R.); (M.C.); (M.C.); (F.O.); (A.G.); (G.G.)
| | - Giovanni Gambassi
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (P.R.); (M.C.); (M.C.); (F.O.); (A.G.); (G.G.)
| | - Rossella Cianci
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (P.R.); (M.C.); (M.C.); (F.O.); (A.G.); (G.G.)
| |
Collapse
|
36
|
Pearson JA, Hu Y, Peng J, Wong FS, Wen L. TLR5-deficiency controls dendritic cell subset development in an autoimmune diabetes-susceptible model. Front Immunol 2024; 15:1333967. [PMID: 38482010 PMCID: PMC10935730 DOI: 10.3389/fimmu.2024.1333967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/06/2024] [Indexed: 03/17/2024] Open
Abstract
Introduction The incidence of the autoimmune disease, type 1 diabetes (T1D), has been increasing worldwide and recent studies have shown that the gut microbiota are associated with modulating susceptibility to T1D. Toll-like receptor 5 (TLR5) recognizes bacterial flagellin and is widely expressed on many cells, including dendritic cells (DCs), which are potent antigen-presenting cells (APCs). TLR5 modulates susceptibility to obesity and alters metabolism through gut microbiota; however, little is known about the role TLR5 plays in autoimmunity, especially in T1D. Methods To fill this knowledge gap, we generated a TLR5-deficient non-obese diabetic (NOD) mouse, an animal model of human T1D, for study. Results We found that TLR5-deficiency led to a reduction in CD11c+ DC development in utero, prior to microbial colonization, which was maintained into adulthood. This was associated with a bias in the DC populations expressing CD103, with or without CD8α co-expression, and hyper-secretion of different cytokines, both in vitro (after stimulation) and directly ex vivo. We also found that TLR5-deficient DCs were able to promote polyclonal and islet antigen-specific CD4+ T cell proliferation and proinflammatory cytokine secretion. Interestingly, only older TLR5-deficient NOD mice had a greater risk of developing spontaneous T1D compared to wild-type mice. Discussion In summary, our data show that TLR5 modulates DC development and enhances cytokine secretion and diabetogenic CD4+ T cell responses. Further investigation into the role of TLR5 in DC development and autoimmune diabetes may give additional insights into the pathogenesis of Type 1 diabetes.
Collapse
Affiliation(s)
- James Alexander Pearson
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, United States
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Youjia Hu
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, United States
| | - Jian Peng
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, United States
| | - F. Susan Wong
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Li Wen
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
37
|
Ng HY, Liao Y, Zhang R, Chan KH, To WP, Hui CH, Seto WK, Leung WK, Hung IFN, Lam TTY, Cheung KS. The Predictive Value of Gut Microbiota Composition for Sustained Immunogenicity following Two Doses of CoronaVac. Int J Mol Sci 2024; 25:2583. [PMID: 38473829 DOI: 10.3390/ijms25052583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/14/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
CoronaVac immunogenicity decreases with time, and we aimed to investigate whether gut microbiota associate with longer-term immunogenicity of CoronaVac. This was a prospective cohort study recruiting two-dose CoronaVac recipients from three centres in Hong Kong. We collected blood samples at baseline and day 180 after the first dose and used chemiluminescence immunoassay to test for neutralizing antibodies (NAbs) against the receptor-binding domain (RBD) of wild-type SARS-CoV-2 virus. We performed shotgun metagenomic sequencing performed on baseline stool samples. The primary outcome was the NAb seroconversion rate (seropositivity defined as NAb ≥ 15AU/mL) at day 180. Linear discriminant analysis [LDA] effect size analysis was used to identify putative bacterial species and metabolic pathways. A univariate logistic regression model was used to derive the odds ratio (OR) of seropositivity with bacterial species. Of 119 CoronaVac recipients (median age: 53.4 years [IQR: 47.8-61.3]; male: 39 [32.8%]), only 8 (6.7%) remained seropositive at 6 months after vaccination. Bacteroides uniformis (log10LDA score = 4.39) and Bacteroides eggerthii (log10LDA score = 3.89) were significantly enriched in seropositive than seronegative participants. Seropositivity was associated with B. eggerthii (OR: 5.73; 95% CI: 1.32-29.55; p = 0.022) and B. uniformis with borderline significance (OR: 3.27; 95% CI: 0.73-14.72; p = 0.110). Additionally, B. uniformis was positively correlated with most enriched metabolic pathways in seropositive vaccinees, including the superpathway of adenosine nucleotide de novo biosynthesis I (log10LDA score = 2.88) and II (log10LDA score = 2.91), as well as pathways related to vitamin B biosynthesis, all of which are known to promote immune functions. In conclusion, certain gut bacterial species (B. eggerthii and B. uniformis) and metabolic pathways were associated with longer-term CoronaVac immunogenicity.
Collapse
Affiliation(s)
- Ho-Yu Ng
- School of Clinical Medicine, The University of Hong Kong, Hong Kong
| | - Yunshi Liao
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Ruiqi Zhang
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Kwok-Hung Chan
- Department of Microbiology, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Wai-Pan To
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Chun-Him Hui
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Wai-Kay Seto
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Wai K Leung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Ivan F N Hung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Tommy T Y Lam
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, The University of Hong Kong, Hong Kong
- Centre for Immunology & Infection Ltd., 17W Hong Kong Science & Technology Parks, Hong Kong
- Laboratory of Data Discovery for Health Ltd., 19W Hong Kong Science & Technology Parks, Hong Kong
- School of Public Health, The University of Hong Kong, Hong Kong
| | - Ka-Shing Cheung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| |
Collapse
|
38
|
Jesus GFA, Galvani NC, Abel JDS, Scussel R, Fagundes MĹ, Córneo EDS, Rossetto M, Sargiani D, de Ávila RAM, Michels M. Nuxcell Neo ® improves vaccine efficacy in antibody response. Front Vet Sci 2024; 11:1248811. [PMID: 38414656 PMCID: PMC10898353 DOI: 10.3389/fvets.2024.1248811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 01/22/2024] [Indexed: 02/29/2024] Open
Abstract
Current vaccination protocols raise concerns about the efficacy of immunization. There is evidence that changes in the gut microbiota can impact immune response. The formation of the gut microbiota in newborns plays a crucial role in immunity. Probiotic bacteria and prebiotics present important health-promoting and immunomodulatory properties. Thus, we hypothesize that pro and prebiotic supplementation can improve the efficacy of vaccination in newborns. In this protocol, newborn mice were used and treated with a single-dose rabies vaccine combined with Nuxcell Neo® (2 g/animal/week) for 3 weeks. Samples were collected on days 7, 14, and 21 after vaccination for analysis of cytokines and concentration of circulating antibodies. Our results show an increased concentration of antibodies in animals vaccinated against rabies and simultaneously treated with Nuxcell Neo® on days 14 and 21 when compared to the group receiving only the vaccine. In the cytokine levels analysis, it was possible to observe that there weren't relevant and significant changes between the groups, which demonstrates that the health of the animal remains stable. The results of our study confirm the promising impact of the use of Nuxcell Neo® on the immune response after vaccination.
Collapse
Affiliation(s)
| | - Nathalia Coral Galvani
- Laboratory of Experimental Pathophysiology, UNESC—University of Southern Santa Catarina, Criciúma, Brazil
| | - Jéssica da Silva Abel
- Laboratory of Experimental Pathophysiology, UNESC—University of Southern Santa Catarina, Criciúma, Brazil
| | - Rahisa Scussel
- Laboratory of Experimental Pathophysiology, UNESC—University of Southern Santa Catarina, Criciúma, Brazil
| | - Mírian ĺvens Fagundes
- Laboratory of Experimental Pathophysiology, UNESC—University of Southern Santa Catarina, Criciúma, Brazil
| | - Emily da Silva Córneo
- Laboratory of Experimental Pathophysiology, UNESC—University of Southern Santa Catarina, Criciúma, Brazil
| | | | | | | | - Monique Michels
- Biohall Consulting, Research and Innovation, Itajaí, Santa Catarina, Brazil
| |
Collapse
|
39
|
Aghighi F, Salami M. What we need to know about the germ-free animal models. AIMS Microbiol 2024; 10:107-147. [PMID: 38525038 PMCID: PMC10955174 DOI: 10.3934/microbiol.2024007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 03/26/2024] Open
Abstract
The gut microbiota (GM), as a forgotten organ, refers to the microbial community that resides in the gastrointestinal tract and plays a critical role in a variety of physiological activities in different body organs. The GM affects its targets through neurological, metabolic, immune, and endocrine pathways. The GM is a dynamic system for which exogenous and endogenous factors have negative or positive effects on its density and composition. Since the mid-twentieth century, laboratory animals are known as the major tools for preclinical research; however, each model has its own limitations. So far, two main models have been used to explore the effects of the GM under normal and abnormal conditions: the isolated germ-free and antibiotic-treated models. Both methods have strengths and weaknesses. In many fields of host-microbe interactions, research on these animal models are known as appropriate experimental subjects that enable investigators to directly assess the role of the microbiota on all features of physiology. These animal models present biological model systems to either study outcomes of the absence of microbes, or to verify the effects of colonization with specific and known microbial species. This paper reviews these current approaches and gives advantages and disadvantages of both models.
Collapse
Affiliation(s)
| | - Mahmoud Salami
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I. R. Iran
| |
Collapse
|
40
|
Zhao L, Wang X, Li Z. A novel chimeric recombinant FliC-Pgp3 vaccine promotes immunoprotection against Chlamydia muridarum infection in mice. Int J Biol Macromol 2024; 258:128723. [PMID: 38101679 DOI: 10.1016/j.ijbiomac.2023.128723] [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: 11/03/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
The Pgp3 subunit vaccine elicits immune protection against Chlamydia trachomatis infection, but additional adjuvants are still required to enhance its immunoprotective efficacy. Flagellin can selectively stimulate immunity and act as an adjuvant. In this research, the FliC-Pgp3 recombinant was successfully expressed and purified. Tri-immunization with the FliC-Pgp3 vaccine in Balb/C mice induced rapid and persistent germinal center B-cell response and Tfh differentiation, promoting a significantly higher IgG antibody titer compared to the Pgp3 group. FliC-Pgp3 immunization primarily induced Th1-type cellular immunity, leading to higher levels of IFN-γ, TNF-α, and IL-2 secreted by CD4+ T cells than in Pgp3-vaccinated mice. Chlamydia muridarum challenge results showed that FliC-Pgp3-vaccinated mice exhibited more rapid clearance of Chlamydia muridarum colonization in the lower genital tract, ensuring a lower hydrosalpinx rate and cumulative score. Histological analysis showed reduced dilation and inflammatory infiltration in the oviduct and uterine horn of FliC-Pgp3-vaccinated mice compared to the PBS and Pgp3 control. Importantly, tri-immunization with FliC-Pgp3 effectively activated CD4+ T cells and dendritic cells, as confirmed by the adoptive transfer, resulting in better immune protection in recipient mice. In summary, the novel FliC-Pgp3 chimeric is hoped to be a novel vaccine with improved immunoprotection against Chlamydia muridarum.
Collapse
Affiliation(s)
- Lanhua Zhao
- Institute of Pathogenic Biology, School of Nursing, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province, University of South China, Hengyang 421001, Hunan, People's Republic of China
| | - Xinglv Wang
- Institute of Pathogenic Biology, School of Nursing, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province, University of South China, Hengyang 421001, Hunan, People's Republic of China
| | - Zhongyu Li
- Institute of Pathogenic Biology, School of Nursing, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province, University of South China, Hengyang 421001, Hunan, People's Republic of China.
| |
Collapse
|
41
|
Arifuzzaman M, Collins N, Guo CJ, Artis D. Nutritional regulation of microbiota-derived metabolites: Implications for immunity and inflammation. Immunity 2024; 57:14-27. [PMID: 38198849 PMCID: PMC10795735 DOI: 10.1016/j.immuni.2023.12.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024]
Abstract
Nutrition profoundly shapes immunity and inflammation across the lifespan of mammals, from pre- and post-natal periods to later life. Emerging insights into diet-microbiota interactions indicate that nutrition has a dominant influence on the composition-and metabolic output-of the intestinal microbiota, which in turn has major consequences for host immunity and inflammation. Here, we discuss recent findings that support the concept that dietary effects on microbiota-derived metabolites potently alter immune responses in health and disease. We discuss how specific dietary components and metabolites can be either pro-inflammatory or anti-inflammatory in a context- and tissue-dependent manner during infection, chronic inflammation, and cancer. Together, these studies emphasize the influence of diet-microbiota crosstalk on immune regulation that will have a significant impact on precision nutrition approaches and therapeutic interventions for managing inflammation, infection, and cancer immunotherapy.
Collapse
Affiliation(s)
- Mohammad Arifuzzaman
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA.
| | - Nicholas Collins
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Chun-Jun Guo
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; Allen Discovery Center for Neuroimmune Interactions, New York, NY 10021, USA.
| |
Collapse
|
42
|
Ray S, Narayanan A, Vesterbacka J, Blennow O, Chen P, Gao Y, Gabarrini G, Ljunggren HG, Buggert M, Manoharan L, Chen MS, Aleman S, Sönnerborg A, Nowak P. Impact of the gut microbiome on immunological responses to COVID-19 vaccination in healthy controls and people living with HIV. NPJ Biofilms Microbiomes 2023; 9:104. [PMID: 38123600 PMCID: PMC10733305 DOI: 10.1038/s41522-023-00461-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023] Open
Abstract
Although mRNA SARS-CoV-2 vaccines are generally safe and effective, in certain immunocompromised individuals they can elicit poor immunogenic responses. Among these individuals, people living with HIV (PLWH) have poor immunogenicity to several oral and parenteral vaccines. As the gut microbiome is known to affect vaccine immunogenicity, we investigated whether baseline gut microbiota predicts immune responses to the BNT162b2 mRNA SARS-CoV-2 vaccine in healthy controls and PLWH after two doses of BNT162b2. Individuals with high spike IgG titers and high spike-specific CD4+ T-cell responses against SARS-CoV-2 showed low α-diversity in the gut. Here, we investigated and presented initial evidence that the gut microbial composition influences the response to BNT162b2 in PLWH. From our predictive models, Bifidobacterium and Faecalibacterium appeared to be microbial markers of individuals with higher spike IgG titers, while Cloacibacillus was associated with low spike IgG titers. We therefore propose that microbiome modulation could optimize immunogenicity of SARS-CoV-2 mRNA vaccines.
Collapse
Affiliation(s)
- Shilpa Ray
- Department of Medicine Huddinge, Division of Infectious Diseases, Karolinska Institutet, Stockholm, Sweden.
| | - Aswathy Narayanan
- Department of Medicine Huddinge, Division of Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - Jan Vesterbacka
- Department of Medicine Huddinge, Division of Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Ola Blennow
- Department of Medicine Huddinge, Division of Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Puran Chen
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Yu Gao
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Giorgio Gabarrini
- Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Hans-Gustaf Ljunggren
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marcus Buggert
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lokeshwaran Manoharan
- National Bioinformatics Infrastructure Sweden (NBIS), SciLifeLab, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | | | - Soo Aleman
- Department of Medicine Huddinge, Division of Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Anders Sönnerborg
- Department of Medicine Huddinge, Division of Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Laboratory Medicine, Division of Clinical Microbiology, ANA Futura, Karolinska Institutet, Stockholm, 141 52, Sweden
| | - Piotr Nowak
- Department of Medicine Huddinge, Division of Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
43
|
Jiang D, Goswami R, Dennis M, Heimsath H, Kozlowski PA, Ardeshir A, Van Rompay KKA, De Paris K, Permar SR, Surana NK. Sutterella and its metabolic pathways positively correlate with vaccine-elicited antibody responses in infant rhesus macaques. Front Immunol 2023; 14:1283343. [PMID: 38124733 PMCID: PMC10731017 DOI: 10.3389/fimmu.2023.1283343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
Introduction It is becoming clearer that the microbiota helps drive responses to vaccines; however, little is known about the underlying mechanism. In this study, we aimed to identify microbial features that are associated with vaccine immunogenicity in infant rhesus macaques. Methods We analyzed 16S rRNA gene sequencing data of 215 fecal samples collected at multiple timepoints from 64 nursery-reared infant macaques that received various HIV vaccine regimens. PERMANOVA tests were performed to determine factors affecting composition of the gut microbiota throughout the first eight months of life in these monkeys. We used DESeq2 to identify differentially abundant bacterial taxa, PICRUSt2 to impute metagenomic information, and mass spectrophotometry to determine levels of fecal short-chain fatty acids and bile acids. Results Composition of the early-life gut microbial communities in nursery-reared rhesus macaques from the same animal care facility was driven by age, birth year, and vaccination status. We identified a Sutterella and a Rodentibacter species that positively correlated with vaccine-elicited antibody responses, with the Sutterella species exhibiting more robust findings. Analysis of Sutterella-related metagenomic data revealed five metabolic pathways that significantly correlated with improved antibody responses following HIV vaccination. Given these pathways have been associated with short-chain fatty acids and bile acids, we quantified the fecal concentration of these metabolites and found several that correlated with higher levels of HIV immunogen-elicited plasma IgG. Discussion Our findings highlight an intricate bidirectional relationship between the microbiota and vaccines, where multiple aspects of the vaccination regimen modulate the microbiota and specific microbial features facilitate vaccine responses. An improved understanding of this microbiota-vaccine interplay will help develop more effective vaccines, particularly those that are tailored for early life.
Collapse
Affiliation(s)
- Danting Jiang
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
- Program in Computational Biology and Bioinformatics, Duke University School of Medicine, Durham, NC, United States
| | - Ria Goswami
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Maria Dennis
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Holly Heimsath
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Pamela A. Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Amir Ardeshir
- California National Primate Research Center, University of California, Davis, Davis, CA, United States
| | - Koen K. A. Van Rompay
- California National Primate Research Center, University of California, Davis, Davis, CA, United States
| | - Kristina De Paris
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Neeraj K. Surana
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, United States
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
44
|
Wang X, Sun L, Li P, Zhang S. Changes in the gut microbiome can predict and decrease Epstein-Barr virus infection risk in children after liver transplantation. Transpl Infect Dis 2023; 25:e14114. [PMID: 37639316 DOI: 10.1111/tid.14114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/02/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2023]
Abstract
OBJECTIVE Primary Epstein-Barr virus (EBV) infection is observed in 60% of children during the first year after liver transplantation as usage of imm-unosuppressant. Finding predictive indicators of EBV infection is important to reduce the morbidity and mortality of EBV infection-related diseases by suggesting a dose reduction of immunosuppressant. METHODS We compared and analysed the gut microbiome of EBV-infected children with an asymptomatic virus-carrying status and EBV-uninfected children after liver transplantation using high-throughput sequencing. RESULTS Significant differences in gut microbiome composition in two groups were detected. In detail, Firmicutes and Lactobacillus were increased in EBV-infected group, while Clostridium was increased in EBV-uninfected group. Furthermore, CD4 percentage in T cells of blood showed a significant positive correlation with the content of Clostridium sp. CAG: 127 in EBV-uninfected group. CONCLUSION Changes in the gut microbiome could predict and decrease the EBV infection risk of children after liver transplantation.
Collapse
Affiliation(s)
- Xu Wang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Liying Sun
- Department of Liver Transplantation, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Peng Li
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Shutian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| |
Collapse
|
45
|
Johnson AMF, Hager K, Alameh MG, Van P, Potchen N, Mayer-Blackwell K, Fiore-Gartland A, Minot S, Lin PJC, Tam YK, Weissman D, Kublin JG. The Regulation of Nucleic Acid Vaccine Responses by the Microbiome. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1680-1692. [PMID: 37850965 PMCID: PMC10656434 DOI: 10.4049/jimmunol.2300196] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 09/19/2023] [Indexed: 10/19/2023]
Abstract
Nucleic acid vaccines, including both RNA and DNA platforms, are key technologies that have considerable promise in combating both infectious disease and cancer. However, little is known about the extrinsic factors that regulate nucleic acid vaccine responses and which may determine their effectiveness. The microbiome is recognized as a significant regulator of immune development and response, whose role in regulating some traditional vaccine platforms has recently been discovered. Using germ-free and specific pathogen-free mouse models in combination with different protein, DNA, and mRNA vaccine regimens, we demonstrate that the microbiome is a significant regulator of nucleic acid vaccine immunogenicity. Although the presence of the microbiome enhances CD8+ T cell responses to mRNA lipid nanoparticle immunization, the microbiome suppresses Ig and CD4+ T cell responses to DNA-prime, DNA-protein-boost immunization, indicating contrasting roles for the microbiome in the regulation of these different nucleic acid vaccine platforms. In the case of mRNA lipid nanoparticle vaccination, germ-free mice display reduced dendritic cell/macrophage activation that may underlie the deficient vaccine response. Our study identifies the microbiome as a relevant determinant of nucleic acid vaccine response with implications for continued therapeutic development and deployment of these vaccines.
Collapse
Affiliation(s)
- Andrew M. F. Johnson
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Kevin Hager
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | | | - Phuong Van
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Nicole Potchen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | | | | | - Samuel Minot
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | | | | | - Drew Weissman
- Penn Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA
| | - James G. Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| |
Collapse
|
46
|
Nehar-Belaid D, Sokolowski M, Ravichandran S, Banchereau J, Chaussabel D, Ucar D. Baseline immune states (BIS) associated with vaccine responsiveness and factors that shape the BIS. Semin Immunol 2023; 70:101842. [PMID: 37717525 DOI: 10.1016/j.smim.2023.101842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/11/2023] [Indexed: 09/19/2023]
Abstract
Vaccines are among the greatest inventions in medicine, leading to the elimination or control of numerous diseases, including smallpox, polio, measles, rubella, and, most recently, COVID-19. Yet, the effectiveness of vaccines varies among individuals. In fact, while some recipients mount a robust response to vaccination that protects them from the disease, others fail to respond. Multiple clinical and epidemiological factors contribute to this heterogeneity in responsiveness. Systems immunology studies fueled by advances in single-cell biology have been instrumental in uncovering pre-vaccination immune cell types and genomic features (i.e., the baseline immune state, BIS) that have been associated with vaccine responsiveness. Here, we review clinical factors that shape the BIS, and the characteristics of the BIS associated with responsiveness to frequently studied vaccines (i.e., influenza, COVID-19, bacterial pneumonia, malaria). Finally, we discuss potential strategies to enhance vaccine responsiveness in high-risk groups, focusing specifically on older adults.
Collapse
Affiliation(s)
| | - Mark Sokolowski
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | | | | | - Damien Chaussabel
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA; Institute for Systems Genomics, University of Connecticut Health Center, Farmington, CT, USA.
| |
Collapse
|
47
|
Zhang T, Lv Y, Zhao Y, Yang J, Qian B, Zhu Y, Zhao W, Zhu M. Changes in intestinal flora of mice induced by rEg.P29 epitope peptide vaccines. Immun Inflamm Dis 2023; 11:e1082. [PMID: 38018604 PMCID: PMC10664826 DOI: 10.1002/iid3.1082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/20/2023] [Accepted: 10/26/2023] [Indexed: 11/30/2023] Open
Abstract
OBJECTIVE Cystic echinococcosis (CE), a zoonotic parasitic disease caused by Echinococcus granulosus, remains a public health and socioeconomic issue worldwide, making its prevention and treatment of vital importance. The aim of this study was to investigate changes in the intestinal microbiota of mice immunized with three peptide vaccines based on the recombinant antigen of E. granulosus, P29 (rEg.P29), with the hope of providing more valuable information for the development of vaccines against CE. METHODS Three peptide vaccines, rEg.P29T , rEg.P29B , and rEg.P29T + B , were prepared based on rEg.P29, and a subcutaneous immunization model was established. The intestinal floras of mice in the different immunization groups were analyzed by 16 S rRNA gene sequencing. RESULTS The intestinal microbiota analysis at both immunization time points revealed that Firmicutes, Bacteroidota, and Verrucomicrobiota were the predominant flora at the phylum level, while at the genus level, Akkermansia, unclassified_Muribaculaceae, Lachnospiraceae_NK4A136_group, and uncultured_rumen bacterium were the dominant genera. Some probiotics in the intestines of mice were significantly increased after immunization with the peptide vaccines, such as Lactobacillus_taiwanensis, Lactobacillus_reuteri, Lachnospiraceae_NK4A136_group, Bacteroides_acidifaciens, and so forth. Meanwhile, some harmful or conditionally pathogenic bacteria were decreased, such as Turicibacter sanguinis, Desulfovibrio_fairfieldensis, Clostridium_sp, and so forth, most of which are associated with inflammatory or infectious diseases. Kyoto Encyclopaedia of Genes and Genomes enrichment analysis revealed that the differential flora were enriched in multiple metabolic pathways, primarily biological systems, human diseases, metabolism, cellular processes, and environmental information processing. CONCLUSION In this study, we comprehensively analyzed and compared changes in the intestinal microbiota of mice immunized with three peptide vaccines as well as their related metabolic pathways, providing a theoretical background for the development of novel vaccines against E. granulosus.
Collapse
Affiliation(s)
- Tingting Zhang
- School of Clinical MedicineNingxia Medical UniversityYinchuanChina
- Key Laboratory of Common Infectious Disease Prevention and Control in NingxiaYinchuanChina
| | - Yongxue Lv
- Key Laboratory of Common Infectious Disease Prevention and Control in NingxiaYinchuanChina
- School of Basic Medical SciencesNingxia Medical UniversityYinchuanChina
| | - Yinqi Zhao
- Key Laboratory of Common Infectious Disease Prevention and Control in NingxiaYinchuanChina
- Science and Technology Center of Ningxia Medical UniversityYinchuanChina
| | - Jihui Yang
- Key Laboratory of Common Infectious Disease Prevention and Control in NingxiaYinchuanChina
- Science and Technology Center of Ningxia Medical UniversityYinchuanChina
| | - Bingshuo Qian
- General Hospital of Ningxia Medical UniversityYinchuanChina
| | - Yazhou Zhu
- Key Laboratory of Common Infectious Disease Prevention and Control in NingxiaYinchuanChina
| | - Wei Zhao
- Key Laboratory of Common Infectious Disease Prevention and Control in NingxiaYinchuanChina
- School of Basic Medical SciencesNingxia Medical UniversityYinchuanChina
| | - Mingxing Zhu
- Key Laboratory of Common Infectious Disease Prevention and Control in NingxiaYinchuanChina
- Science and Technology Center of Ningxia Medical UniversityYinchuanChina
| |
Collapse
|
48
|
Pichichero ME. Variability of vaccine responsiveness in early life. Cell Immunol 2023; 393-394:104777. [PMID: 37866234 DOI: 10.1016/j.cellimm.2023.104777] [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: 06/15/2023] [Revised: 09/18/2023] [Accepted: 10/14/2023] [Indexed: 10/24/2023]
Abstract
Vaccinations in early life elicit variable antibody and cellular immune responses, sometimes leaving fully vaccinated children unprotected against life-threatening infectious diseases. Specific immune cell populations and immune networks may have a critical period of development and calibration in a window of opportunity occurring during the first 100 days of early life. Among the early life determinants of vaccine responses, this review will focus on modifiable factors involving development of the infant microbiota and metabolome: antibiotic exposure, breast versus formula feeding, and Caesarian section versus vaginal delivery of newborns. How microbiota may serve as natural adjuvants for vaccine responses and how microbiota-derived metabolites influence vaccine responses are also reviewed. Early life poor vaccine responsiveness can be linked to increased infection susceptibility because both phenotypes share similar immunity dysregulation profiles. An early life pre-vaccination endotype, when interventions have the highest potential for success, should be sought that predicts vaccine response trajectories.
Collapse
Affiliation(s)
- Michael E Pichichero
- Center for Infectious Diseases and Immunology, Research Institute, Rochester General Hospital, 1425 Portland Ave, Rochester, NY 14621, USA.
| |
Collapse
|
49
|
Elizalde-Torrent A, Borgognone A, Casadellà M, Romero-Martin L, Escribà T, Parera M, Rosales-Salgado Y, Díaz-Pedroza J, Català-Moll F, Noguera-Julian M, Brander C, Paredes R, Olvera A. Vaccination with an HIV T-Cell Immunogen (HTI) Using DNA Primes Followed by a ChAdOx1-MVA Boost Is Immunogenic in Gut Microbiota-Depleted Mice despite Low IL-22 Serum Levels. Vaccines (Basel) 2023; 11:1663. [PMID: 38005995 PMCID: PMC10675013 DOI: 10.3390/vaccines11111663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
Despite the important role of gut microbiota in the maturation of the immune system, little is known about its impact on the development of T-cell responses to vaccination. Here, we immunized C57BL/6 mice with a prime-boost regimen using DNA plasmid, the Chimpanzee Adenovirus, and the modified Vaccinia Ankara virus expressing a candidate HIV T-cell immunogen and compared the T-cell responses between individuals with an intact or antibiotic-depleted microbiota. Overall, the depletion of the gut microbiota did not result in significant differences in the magnitude or breadth of the immunogen-specific IFNγ T-cell response after vaccination. However, we observed marked changes in the serum levels of four cytokines after vaccinating microbiota-depleted animals, particularly a significant reduction in IL-22 levels. Interestingly, the level of IL-22 in serum correlated with the abundance of Roseburia in the large intestine of mice in the mock and vaccinated groups with intact microbiota. This short-chain fatty acid (SCFA)-producing bacterium was significantly reduced in the vaccinated, microbiota-depleted group. Therefore, our results indicate that, although microbiota depletion reduces serum levels of IL-22, the powerful vaccine regime used could have overcome the impact of microbiota depletion on IFNγ-producing T-cell responses.
Collapse
Affiliation(s)
- Aleix Elizalde-Torrent
- Irsicaixa—AIDS Research Institute, 08916 Barcelona, Spain; (A.E.-T.); (A.B.); (M.C.); (L.R.-M.); (T.E.); (M.P.); (F.C.-M.); (M.N.-J.); (C.B.); (R.P.)
| | - Alessandra Borgognone
- Irsicaixa—AIDS Research Institute, 08916 Barcelona, Spain; (A.E.-T.); (A.B.); (M.C.); (L.R.-M.); (T.E.); (M.P.); (F.C.-M.); (M.N.-J.); (C.B.); (R.P.)
| | - Maria Casadellà
- Irsicaixa—AIDS Research Institute, 08916 Barcelona, Spain; (A.E.-T.); (A.B.); (M.C.); (L.R.-M.); (T.E.); (M.P.); (F.C.-M.); (M.N.-J.); (C.B.); (R.P.)
| | - Luis Romero-Martin
- Irsicaixa—AIDS Research Institute, 08916 Barcelona, Spain; (A.E.-T.); (A.B.); (M.C.); (L.R.-M.); (T.E.); (M.P.); (F.C.-M.); (M.N.-J.); (C.B.); (R.P.)
- Departament de Biologia Cellular, Fisiologia i Immunologia, Universitat Autonoma de Barcelona (UAB), 08193 Cerdanyola del Valles, Spain
| | - Tuixent Escribà
- Irsicaixa—AIDS Research Institute, 08916 Barcelona, Spain; (A.E.-T.); (A.B.); (M.C.); (L.R.-M.); (T.E.); (M.P.); (F.C.-M.); (M.N.-J.); (C.B.); (R.P.)
| | - Mariona Parera
- Irsicaixa—AIDS Research Institute, 08916 Barcelona, Spain; (A.E.-T.); (A.B.); (M.C.); (L.R.-M.); (T.E.); (M.P.); (F.C.-M.); (M.N.-J.); (C.B.); (R.P.)
| | - Yaiza Rosales-Salgado
- Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), 08916 Badalona, Spain; (Y.R.-S.); (J.D.-P.)
| | - Jorge Díaz-Pedroza
- Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), 08916 Badalona, Spain; (Y.R.-S.); (J.D.-P.)
| | - Francesc Català-Moll
- Irsicaixa—AIDS Research Institute, 08916 Barcelona, Spain; (A.E.-T.); (A.B.); (M.C.); (L.R.-M.); (T.E.); (M.P.); (F.C.-M.); (M.N.-J.); (C.B.); (R.P.)
| | - Marc Noguera-Julian
- Irsicaixa—AIDS Research Institute, 08916 Barcelona, Spain; (A.E.-T.); (A.B.); (M.C.); (L.R.-M.); (T.E.); (M.P.); (F.C.-M.); (M.N.-J.); (C.B.); (R.P.)
- Facultat de Medicina, Universitat de Vic—Universitat Central de Catalunya (UVic-UCC), 08500 Vic, Spain
- CIBERINFEC—ISCIII, 28029 Madrid, Spain
| | - Christian Brander
- Irsicaixa—AIDS Research Institute, 08916 Barcelona, Spain; (A.E.-T.); (A.B.); (M.C.); (L.R.-M.); (T.E.); (M.P.); (F.C.-M.); (M.N.-J.); (C.B.); (R.P.)
- Facultat de Medicina, Universitat de Vic—Universitat Central de Catalunya (UVic-UCC), 08500 Vic, Spain
- CIBERINFEC—ISCIII, 28029 Madrid, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
- Aelix Therapeutics, 08028 Barcelona, Spain
| | - Roger Paredes
- Irsicaixa—AIDS Research Institute, 08916 Barcelona, Spain; (A.E.-T.); (A.B.); (M.C.); (L.R.-M.); (T.E.); (M.P.); (F.C.-M.); (M.N.-J.); (C.B.); (R.P.)
- Facultat de Medicina, Universitat de Vic—Universitat Central de Catalunya (UVic-UCC), 08500 Vic, Spain
- CIBERINFEC—ISCIII, 28029 Madrid, Spain
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
- Fight AIDS Foundation, Infectious Diseases Department, Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
- Department of Infectious Diseases Service, Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
| | - Alex Olvera
- Irsicaixa—AIDS Research Institute, 08916 Barcelona, Spain; (A.E.-T.); (A.B.); (M.C.); (L.R.-M.); (T.E.); (M.P.); (F.C.-M.); (M.N.-J.); (C.B.); (R.P.)
- CIBERINFEC—ISCIII, 28029 Madrid, Spain
- Facultat de Ciències, Tecnologia i Enginyeries, Universitat de Vic—Universitat Central de Catalunya (UVic-UCC), 08500 Vic, Spain
| |
Collapse
|
50
|
Qu S, Gao Y, Ma J, Yan Q. Microbiota-derived short-chain fatty acids functions in the biology of B lymphocytes: From differentiation to antibody formation. Biomed Pharmacother 2023; 168:115773. [PMID: 39491858 DOI: 10.1016/j.biopha.2023.115773] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/15/2023] [Accepted: 10/20/2023] [Indexed: 11/05/2024] Open
Abstract
Gut bacteria produce various metabolites from dietary fiber, the most abundant of which are short-chain fatty acids (SCFAs) such as acetate, propionate, and butyrate. Many biological functions, such as host metabolism and the immune system, are regulated by SCFAs because they act on a wide variety of cell types. A growing body of documents has shown that microbiota SCFAs directly regulate B-cell growth, proliferation, and immunoglobulin (Ig) production. As histone deacetylase (HDAC) inhibitors, SCFAs alter gene expression to enhance the expression of critical regulators of B cell growth. In particular, microbiota SCFAs increase the production of acetyl coenzyme A (acetyl-CoA), adenosine triphosphate (ATP), and fatty acids in B cells, which provide the energy and building blocks needed for the growth of plasma B cells. SCFAs play a significant role in promoting the involvement of B cells in host immunity during both homeostatic conditions and disease states. In this context, SCFAs stimulate B-cell activation and promote the differentiation of plasma B cells in response to B cell receptor (BCR)-activating antigens or co-stimulatory receptor ligands. The result may be increased production of IgA. Microbiota SCFAs were found to lower both overall and antigen-specific IgE levels, indicating their potential to mitigate IgE-related allergic reactions, much like their effect on class-switch recombination (CSR) towards IgG and IgA. Therefore, in the future, the therapeutic advantage should be to use specific and diffusible chemicals, such as SCFAs, which show a strong immunoregulatory function of B cells. This review focuses on the role of microbiota-produced SCFAs in regulating B cell development and antibody production, both in health and diseases.
Collapse
Affiliation(s)
- Shengming Qu
- Department of Dermatology, the Second Hospital of Jilin University, Changchun 130000, China
| | - Yihang Gao
- Department of Clinical Laboratory, the Second Hospital of Jilin University, Changchun 130000, China.
| | - Jingru Ma
- Department of Clinical Laboratory, the Second Hospital of Jilin University, Changchun 130000, China
| | - Qingzhu Yan
- Department of Ultrasound Medicine, the Second Hospital of Jilin University, Changchun 130000, China
| |
Collapse
|