Gut microbiome remains stable following COVID-19 vaccination in healthy and immuno-compromised individuals

The bidirectional interaction between the immune system and the gut microbiota is a key contributor to various host physiological functions. Immune-associated diseases such as cancer and autoimmunity, as well as the efficacy of immunomodulatory therapies, have been linked to microbiome variation. Here, we investigate the temporal impact of COVID-19 vaccination on the gut microbiome in healthy and immuno-compromised individuals; the latter included patients with primary immunodeficiency and cancer patients on immunosuppressive therapy. We find that the gut microbiome, assessed using shotgun metagenomics, remained stable post-vaccination irrespective of diverse immune status, vaccine response, and microbial composition spanned by the cohort. The stability is evident at all tested levels including phylum, species, and functional capacity. Our results show the resilience of the gut microbiome to host immune changes triggered by COVID-19 vaccination and suggest minimal, if any, impact on microbiome-mediated processes.

[1]  Q-G Yang,et al.  A Review of Gut Microbiota‐Derived Metabolites in Tumor Progression and Cancer Therapy , 2023, Advanced science.

[2]  Xiaomin Su,et al.  Gut-Microbiota-Derived Metabolites Maintain Gut and Systemic Immune Homeostasis , 2023, Cells.

[3]  F. Hollfelder,et al.  Age-Associated B cells predict impaired humoral immunity after COVID-19 vaccination in patients receiving immune checkpoint blockade , 2022, medRxiv.

[4]  H. Kitano,et al.  Human immune and gut microbial parameters associated with inter-individual variations in COVID-19 mRNA vaccine-induced immunity , 2022, bioRxiv.

[5]  C. Chakraborty,et al.  Immune Response to SARS-CoV-2 Vaccines , 2022, Biomedicines.

[6]  C. Lemere,et al.  Microbiota in neuroinflammation and synaptic dysfunction: a focus on Alzheimer’s disease , 2022, Molecular neurodegeneration.

[7]  Mark M. Davis,et al.  Immune imprinting, breadth of variant recognition, and germinal center response in human SARS-CoV-2 infection and vaccination , 2022, Cell.

[8]  A. Kostic,et al.  Reproducible and opposing gut microbiome signatures distinguish autoimmune diseases and cancers: a systematic review and meta-analysis , 2021, Microbiome.

[9]  D. Lewis,et al.  Immunogenicity and tolerability of COVID-19 messenger RNA vaccines in primary immunodeficiency patients with functional B-cell defects , 2021, Journal of Allergy and Clinical Immunology.

[10]  Anna V. Protasio,et al.  A protease-activatable luminescent biosensor and reporter cell line for authentic SARS-CoV-2 infection , 2021, bioRxiv.

[11]  C. Cunningham-Rundles,et al.  Circulating bioactive bacterial DNA is associated with immune activation and complications in common variable immunodeficiency , 2021, JCI insight.

[12]  Frances E. Muldoon,et al.  Age-related immune response heterogeneity to SARS-CoV-2 vaccine BNT162b2 , 2021, Nature.

[13]  M. Spitzer,et al.  Systemic immunity in cancer , 2021, Nature Reviews Cancer.

[14]  V. Moreno,et al.  Tumor-Associated Microbiome: Where Do We Stand? , 2021, International journal of molecular sciences.

[15]  L. Notarangelo,et al.  Gut Microbiota–Host Interactions in Inborn Errors of Immunity , 2021, International journal of molecular sciences.

[16]  J. Trent,et al.  Stool Microbiome Profiling of Patients with Metastatic Renal Cell Carcinoma Receiving Anti-PD-1 Immune Checkpoint Inhibitors. , 2020, European urology.

[17]  A. Aoun,et al.  The Influence of the Gut Microbiome on Obesity in Adults and the Role of Probiotics, Prebiotics, and Synbiotics for Weight Loss , 2020, Preventive nutrition and food science.

[18]  John A.G. Briggs,et al.  A thermostable, closed SARS-CoV-2 spike protein trimer , 2020, Nature Structural & Molecular Biology.

[19]  Edoardo Pasolli,et al.  Gut Bacteria Composition Drives Primary Resistance to Cancer Immunotherapy in Renal Cell Carcinoma Patients. , 2020, European urology.

[20]  Vito Adrian Cantu,et al.  PRINSEQ++, a multi-threaded tool for fast and efficient quality control and preprocessing of sequencing datasets , 2019 .

[21]  Davide Heller,et al.  eggNOG 5.0: a hierarchical, functionally and phylogenetically annotated orthology resource based on 5090 organisms and 2502 viruses , 2018, Nucleic Acids Res..

[22]  Peer Bork,et al.  Extensive impact of non-antibiotic drugs on human gut bacteria , 2018, Nature.

[23]  Laurence Zitvogel,et al.  Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors , 2018, Science.

[24]  Riyue Bao,et al.  The commensal microbiome is associated with anti–PD-1 efficacy in metastatic melanoma patients , 2018, Science.

[25]  E. Frenkel,et al.  Metagenomic Shotgun Sequencing and Unbiased Metabolomic Profiling Identify Specific Human Gut Microbiota and Metabolites Associated with Immune Checkpoint Therapy Efficacy in Melanoma Patients1 , 2017, Neoplasia.

[26]  P. Pevzner,et al.  metaSPAdes: a new versatile metagenomic assembler. , 2017, Genome research.

[27]  F. Bonilla,et al.  Vaccination in Primary Immunodeficiency Disorders. , 2016, The journal of allergy and clinical immunology. In practice.

[28]  Chang H. Kim,et al.  Gut Microbial Metabolites Fuel Host Antibody Responses. , 2016, Cell host & microbe.

[29]  P. Brigidi,et al.  The effect of short-chain fatty acids on human monocyte-derived dendritic cells , 2015, Scientific Reports.

[30]  M. Holubar,et al.  Immunodeficiency-related vaccine-derived poliovirus (iVDPV) cases: a systematic review and implications for polio eradication. , 2015, Vaccine.

[31]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[32]  Susan Holmes,et al.  phyloseq: An R Package for Reproducible Interactive Analysis and Graphics of Microbiome Census Data , 2013, PloS one.

[33]  Jung Ok Shim,et al.  Gut Microbiota in Inflammatory Bowel Disease , 2013, Pediatric gastroenterology, hepatology & nutrition.

[34]  Steven L Salzberg,et al.  Fast gapped-read alignment with Bowtie 2 , 2012, Nature Methods.

[35]  R. Peek,et al.  Helicobacter pylori: gastric cancer and beyond , 2010, Nature Reviews Cancer.

[36]  J. Adams,et al.  1 alpha,25-dihydroxyvitamin D3 suppresses proliferation and immunoglobulin production by normal human peripheral blood mononuclear cells. , 1984, The Journal of clinical investigation.

[37]  Dr. La´szlo´ 1,25-Dihydroxyvitamin D 3 Is an Autonomous Regulator of the Transcriptional Changes Leading to a Tolerogenic Dendritic Cell Phenotype , 2023 .

[38]  John S M Leung Interaction between gut microbiota and COVID-19 and its vaccines , 2022, World journal of gastroenterology.