Gastrointestinal microbiota composition predicts peripheral inflammatory state during treatment of human tuberculosis

Matthew F Wipperman, Shakti K Bhattarai, Charles Kyriakos Vorkas, Venkata Suhas Maringati, Ying Taur, Laurent Mathurin, Katherine McAulay, Stalz Charles Vilbrun, Daphie Francois, James Bean, Kathleen F Walsh, Carl Nathan, Daniel W Fitzgerald, Michael S Glickman, Vanni Bucci, Matthew F Wipperman, Shakti K Bhattarai, Charles Kyriakos Vorkas, Venkata Suhas Maringati, Ying Taur, Laurent Mathurin, Katherine McAulay, Stalz Charles Vilbrun, Daphie Francois, James Bean, Kathleen F Walsh, Carl Nathan, Daniel W Fitzgerald, Michael S Glickman, Vanni Bucci

Abstract

The composition of the gastrointestinal microbiota influences systemic immune responses, but how this affects infectious disease pathogenesis and antibiotic therapy outcome is poorly understood. This question is rarely examined in humans due to the difficulty in dissociating the immunologic effects of antibiotic-induced pathogen clearance and microbiome alteration. Here, we analyze data from two longitudinal studies of tuberculosis (TB) therapy (35 and 20 individuals) and a cross sectional study from 55 healthy controls, in which we collected fecal samples (for microbiome analysis), sputum (for determination of Mycobacterium tuberculosis (Mtb) bacterial load), and peripheral blood (for transcriptomic analysis). We decouple microbiome effects from pathogen sterilization by comparing standard TB therapy with an experimental TB treatment that did not reduce Mtb bacterial load. Random forest regression to the microbiome-transcriptome-sputum data from the two longitudinal datasets reveals that renormalization of the TB inflammatory state is associated with Mtb pathogen clearance, increased abundance of Clusters IV and XIVa Clostridia, and decreased abundance of Bacilli and Proteobacteria. We find similar associations when applying machine learning to peripheral gene expression and microbiota profiling in the independent cohort of healthy individuals. Our findings indicate that antibiotic-induced reduction in pathogen burden and changes in the microbiome are independently associated with treatment-induced changes of the inflammatory response of active TB, and the response to antibiotic therapy may be a combined effect of pathogen killing and microbiome driven immunomodulation.

Trial registration: ClinicalTrials.gov NCT02684240.

Conflict of interest statement

M.F.W. is currently an employee and shareholder of Regeneron Pharmaceuticals, Inc. M.S.G. reports consulting fees and equity in Vedanta Biosciences, Inc., consulting fees from Takeda, and is on the SAB of PRL-NYC. V.B. is supported by a Sponsored Research Agreement from Vedanta Biosciences, Inc. The remaining authors declare no competing interests.

Figures

Fig. 1. Overview of cohorts, subjects, timepoints,…
Fig. 1. Overview of cohorts, subjects, timepoints, samples, and hypotheses in this study.
A This study investigates microbiome-transcriptome relationships in three separate cohorts of individuals in Haiti. Cohort 1 (2-week longitudinal and interventional clinical trial) consists of secondary analysis of a randomized clinical trial of study volunteers, where we collected disease severity measurements (Mtb bacterial load, TTP), microbiome profiling, and peripheral transcriptomics in active TB patients at baseline, before randomization to either HRZE (arm 1) (standard of care TB treatment), or Nitazoxanide (NTZ) (arm 2). Cohort 2 (6 month longitudinal and observational study) consists of study volunteers who were followed throughout the course of 6 months of TB treatment, where we collected TTP, microbiome, and transcriptomics data. Finally, Cohort 3 (cross sectional and observational) consists of healthy volunteers. These healthy volunteers were enrolled separately. Around half are healthy and TB-negative household or family contacts (FC) of active TB-patients, and the other half are community controls (CC), with no know TB exposure. We performed microbiome profiling and peripheral transcriptomics on these individuals as well. B Numbers of individuals in this study. C Diagram showing the major questions investigated in this study. Supplementary Data 1 provides a table with dates of first and last enrollment for every Cohort.
Fig. 2. Both HRZE and NTZ perturb…
Fig. 2. Both HRZE and NTZ perturb the gut microbiota after two weeks of therapy, but only HRZE reduces M. tuberculosis bacterial load.
A Schematic of the clinical trial comparing bactericidal effect of HRZE and NTZ. B Paired M. tuberculosis sputum time to positivity (TTP) at day 0 and day 14 for the NTZ treatment arm and HRZE treatment arm. Data are displayed as range (minimum and maximum) of two-three technical replicates; n = 16 biologically independent individuals for the HRZE arm and n = 19 biologically independent individuals for the NTZ arm. Linear mixed effect modeling was used to determine significance of difference in post/pre treatment in each arm as TTP~1+Sex+Age+Treatment+Time+Treatment:Time+1∣ID, where Treatment indicates the arm (NTZ or HRZE), Time indicates Pre or Post antibiotic administration, and : indicates the interaction term. For each individual we use as TTP measurement the average of the relative technical measurements. NTZ treatment is associated with no difference in TTP between day 0 and day 14 (p > 0.05 for the coefficient of variable Time, see Supplementary Data 2), whereas HRZE significantly reduces bacterial load (p < 0.05 for the coefficient of variable Treatment : Time see Supplementary Data 2). Data for TTP were obtained from Walsh et al.. C Principal Coordinate analysis (PCoA) with Bray–Curtis distance showing differences in microbiome community structure between individuals before and after 14 days of either HRZE or NTZ treatment. The gray line connects baseline and day 14 treatment paired samples. PCoA1 clearly discriminates samples post NTZ treatment (pink triangles) from those at baseline or after HRZE treatment. PERMANOVA analysis was used to reject (p = 0.001, see Supplementary Data 4) the null hypothesis that the centroids and dispersion of the groups (pretreatment, after NTZ and after HRZE) are equivalent for all groups (see Supplementary Data 4). D Microbiota alpha diversity plotted using the Inverse Simpson Diversity index; n = 16 biologically independent individuals for the HRZE arm and n = 19 biologically independent individuals for the NTZ arm. Linear mixed effect modeling was used to determine the significance of difference of treatment on diversity. We fitted the model Diversity~1+Sex+Age+Treatment:Time+1∣ID. The symbol: indicates the interaction term. HRZE was used as the reference level. No significant difference between the two treatment at baseline was observed. Both groups (p < 0.05 for the coefficient of variable Time corresponding to HRZE treatment and p < 0.05 for the coefficient of variable Treatment:Time corresponding to NTZ treatment in this model) display significantly reduced Inverse Simpson diversity after 14 days of treatment (see Supplementary Data 3). Source data are provided as a Source Data file.
Fig. 3. Overlapping and distinct microbiome perturbation…
Fig. 3. Overlapping and distinct microbiome perturbation induced by NTZ and HRZE.
A Volcano plots indicating the post (day 14) vs pretreatment (baseline) differences at the ASV level for HRZE and NTZ. The color of each ASV is according to the phylogenetic Order. A single linear mixed effect model for each ASV of the form ASVicounts~Sex+Batch+Group+1∣ID was fitted to determine differences due to treatment while accounting for sequencing batch and sex. ASVs significantly affected by the treatment were those determined to have a Benjamini-Hochberg false discovery rate (FDR) adjusted p-value less than 0.05 for the variable Group in the limma/voom model (see “Methods” section). The horizontal dotted lines indicate FDR < 0.05 and vertical dotted lines indicate |log2FC| > 1.5. B Within-arm unsupervised hierarchical clustering of the abundances of 404 ASVs found to be significantly affected by HRZE or NTZ treatment (FDR < 0.05, see Supplementary 5). The heatmap columns are split by arm membership (including baseline randomization group), and the heatmap rows are split by ASV phylogenetic Phylum, and within the Phylum, the Order is colored as in A. The right annotations (HRZE and NTZ) indicate whether each ASV was significantly perturbed by treatment. P value in y axis is adjusted according to Benjamini–Hochberg (FDR). Source data are provided as a Source Data file.
Fig. 4. Hallmark pathway gene set enrichment…
Fig. 4. Hallmark pathway gene set enrichment analysis and gene expression comparison in HRZE and NTZ treated arms.
A Hallmark gene pathway changes associated with 2 weeks of HRZE (A) or NTZ (B). Positive are pathways overrepresented at 2 weeks of therapy (up), and negative are pathways underrepresented at 2 weeks (down), both compared to baseline. All pathways are significant (FDR < 0.05, see Supplementary Data 6) with the size of the arrow indicating level of significance. Only pathways from MiSigDB Hallmark pathway set found to be significantly altered in this analysis are shown. Here NES stands for Normalized Enrichment Score. B, C TB-associated peripheral blood transcripts from ref. , highlighting post treatment vs baseline changes in gene expression for HRZE (B) or NTZ (C). Notably, HRZE renormalizes (i.e., towards a healthy control state) the expression of 144 validated TB inflammatory transcripts and exacerbates only 13 (see Supplementary Data 7). NTZ is found only to exacerbate four (see Supplementary Data 8). TB-associated peripheral blood transcripts significantly affected by each treatment were those determined to have a Benjamini–Hochberg false discovery rate (FDR) adjusted p-value less than 0.05 for the variable Group in the limma/voom model (see “Methods” section). D, E Effect of HRZE and NTZ on blood gene expression for a set of IBD-associated genes from Palmer, et al.. Both HRZE (D) and NTZ (E) cause different genes to either renormalize (HRZE 66, NTZ 34) (see Supplementary Data 9) or exacerbate (HRZE 55, NTZ 21) (see Supplementary Data 10). IBD-associated peripheral blood transcripts significantly affected by each treatment were those determined to have a Benjamini–Hochberg false discovery rate (FDR) adjusted p-value less than 0.05 for the variable Group in the limma/voom model (see “Methods” section) P value in y axis is adjusted according to Benjamini–Hochberg (FDR). Source data are provided as a Source Data file.
Fig. 5. Longitudinal profiling of HRZE treatment…
Fig. 5. Longitudinal profiling of HRZE treatment induced changes of microbiome composition and peripheral gene inflammatory expression.
A. Schematic diagram of Cohort. B. Time to positivity was measured at baseline (n = 19 biologically independent samples), day 14 (n = 14 biologically independent samples), one month (n = 5 biologically independent samples), and two months (n = 9 biologically independent samples). To determine statistical significance of differences in TTP at different time points we fit the linear mixed-effect model TTP~Sex+Age+Time+1∣ID. We inspected the p-value associated by running contrasts for the variable Time to determine significant (p-value for the contrast <0.05) differences in TTP. C Microbiome diversity was computed for each study volunteer at baseline (n = 20 biologically independent samples), day 7 (n = 7 biologically independent samples), day 14 (n = 19 biologically independent samples), one month (n = 13 biologically independent samples), two months (n = 13 biologically independent samples), and 6 months (n = 13 biologically independent samples). Microbiome α diversity was measured using the Inverse Simpson index. To determine statistical significance of differences in α diversity at different time points we fit the linear mixed-effect model Diversity~Sex+Age+Time+1∣ID. We inspected the p-value associated by running contrasts for the variable Time to determine significant (p-value for the contrast <0.05) differences in microbiota diversity D Volcano plots showing significance of differences in microbiome composition vs. fold change from baseline at Day 7, Day 14, Day 30, Day 56, and Day 180. E Normalized enrichment scores calculated for the Hallmark Pathway list for Day 14 vs. Baseline, Day 56 vs Day 14, and Day 56 vs. Baseline. FH Volcano plot showing TB transcripts from ref. at Day 14 vs. Baseline at Day 56 vs. Day 14, and at Day 14 vs. Baseline. IK Volcano plot showing transcripts from Palmer et al. of IBD cases vs. controls detected in this study for Day 14 vs. Baseline, Day 56 vs. Day 14, and Day 56 vs. Baseline. Source data are provided as a Source Data file.
Fig. 6. Use of Random Forest Regression…
Fig. 6. Use of Random Forest Regression Modeling to search for associations between immune-related peripheral blood gene signatures and changes in gastrointestinal microbiota and TTP.
The heatmap displays the sign of the derivative of the ALE curve (See Text). Blue/orange entries indicate features found to significantly associate with changes in a specific inflammatory pathway. Blue indicates a negative relationship, while orange a positive. For each immune pathway, a pathway-ASV association was determined significant if the Benjamini–Hochberg false discovery rate (FDR) adjusted p-value from the permutated importance analysis was found to be less than 0.05. Black symbols are used to identify the modeling-identified top important predictor (i.e., the predictor that if missing would lead to highest increase in mean squared error between model predictions and observations) for each specific host pathway. This analysis shows that reduction in TB burden and increased abundance of health-associated Cluster IV and XIVa Clostridia predicts inflammatory dampening. In contrast, increased abundance of oxygen-tolerant pathobionts including Enterococcus, Streptococcus, and E. coli predicts inflammatory exacerbation. A table reporting variable importance values, slope and intercepts from the ALE plot calculation and the related p values is provided in Supplementary Data 18. Source data are provided as a Source Data file.
Fig. 7. Analysis of microbiome and blood…
Fig. 7. Analysis of microbiome and blood peripheral gene expression in an independent healthy control human cohort validates association between specific microbiome members and host peripheral gene expression.
A NES scores of 50 Hallmark pathways from the MiSigDB on a per-sample basis for all cohorts in this study. NES score was calculated using the variance stabilized transformed counts from DSEeq, calculated with the GSVA package in R, and plotted after scaling (Z score) across all samples. Columns are split based on arm or group membership and rows are split based on Hallmark pathway categorization. B Random forest regression results associating specific microbial taxa with Hallmark pathways. Only pathways identified in the RFR model are shown. The ‘Relation’, calculated by taking the first derivative of the ALE plot for each relationship, is positive if the pathway positively associates with a particular ASV, or negative if the pathway negatively associates with a particular ASV. Source data are provided as a Source Data file.

References

    1. Martin CR, Osadchiy V, Kalani A, Mayer EA. The brain-gut-microbiome axis. Cell Mol. Gastroenterol. Hepatol. 2018;6:133–148. doi: 10.1016/j.jcmgh.2018.04.003.
    1. Atarashi K, et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature. 2013;500:232–236. doi: 10.1038/nature12331.
    1. Atarashi K, et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science. 2011;331:337–341.
    1. Tanoue T, et al. A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature. 2019;565:600–605. doi: 10.1038/s41586-019-0878-z.
    1. Vorkas CK, et al. Mucosal-associated invariant and gammadelta T cell subsets respond to initial Mycobacterium tuberculosis infection. JCI Insight. 2018;3:e121899. doi: 10.1172/jci.insight.121899.
    1. Campbell C, et al. Extrathymically generated regulatory t cells establish a niche for intestinal border-dwelling bacteria and affect physiologic metabolite balance. Immunity. 2018;48:1245–1257 e1249. doi: 10.1016/j.immuni.2018.04.013.
    1. Levan, S. R. et al. Elevated faecal 12,13-diHOME concentration in neonates at high risk for asthma is produced by gut bacteria and impedes immune tolerance. Nat. Microbiol.4, 1851–1861 (2019).
    1. Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell. 2014;157:121–141. doi: 10.1016/j.cell.2014.03.011.
    1. Ryan FJ, et al. Changes in the composition of the gut microbiota and the blood transcriptome in preterm infants at less than 29 weeks gestation diagnosed with bronchopulmonary dysplasia. mSystems. 2019;4:e00484–00419. doi: 10.1128/mSystems.00484-19.
    1. Grigg JB, Sonnenberg GF. Host-microbiota interactions shape local and systemic inflammatory diseases. J. Immunol. 2017;198:564–571. doi: 10.4049/jimmunol.1601621.
    1. Wang Z, Arat S, Magid-Slav M, Brown JR. Meta-analysis of human gene expression in response to Mycobacterium tuberculosis infection reveals potential therapeutic targets. BMC Syst. Biol. 2018;12:3. doi: 10.1186/s12918-017-0524-z.
    1. Kaforou M, et al. Detection of tuberculosis in HIV-infected and -uninfected African adults using whole blood RNA expression signatures: a case-control study. PLoS Med. 2013;10:e1001538. doi: 10.1371/journal.pmed.1001538.
    1. Zak DE, et al. A blood RNA signature for tuberculosis disease risk: a prospective cohort study. Lancet. 2016;387:2312–2322. doi: 10.1016/S0140-6736(15)01316-1.
    1. Berry MP, et al. An interferon-inducible neutrophil-driven blood transcriptional signature in human tuberculosis. Nature. 2010;466:973–977. doi: 10.1038/nature09247.
    1. Lesho E, et al. Transcriptional responses of host peripheral blood cells to tuberculosis infection. Tuberculosis. 2011;91:390–399. doi: 10.1016/j.tube.2011.07.002.
    1. Ottenhoff TH, et al. Genome-wide expression profiling identifies type 1 interferon response pathways in active tuberculosis. PLoS ONE. 2012;7:e45839. doi: 10.1371/journal.pone.0045839.
    1. Bloom CI, et al. Transcriptional blood signatures distinguish pulmonary tuberculosis, pulmonary sarcoidosis, pneumonias and lung cancers. PLoS ONE. 2013;8:e70630. doi: 10.1371/journal.pone.0070630.
    1. Cliff JM, et al. Distinct phases of blood gene expression pattern through tuberculosis treatment reflect modulation of the humoral immune response. J. Infect. Dis. 2013;207:18–29. doi: 10.1093/infdis/jis499.
    1. Singhania A, et al. A modular transcriptional signature identifies phenotypic heterogeneity of human tuberculosis infection. Nat. Commun. 2018;9:2308. doi: 10.1038/s41467-018-04579-w.
    1. Aljayyoussi G, et al. Pharmacokinetic-Pharmacodynamic modelling of intracellular Mycobacterium tuberculosis growth and kill rates is predictive of clinical treatment duration. Sci. Rep. 2017;7:502–502. doi: 10.1038/s41598-017-00529-6.
    1. Namasivayam S, et al. Longitudinal profiling reveals a persistent intestinal dysbiosis triggered by conventional anti-tuberculosis therapy. Microbiome. 2017;5:71. doi: 10.1186/s40168-017-0286-2.
    1. Wipperman MF, et al. Antibiotic treatment for Tuberculosis induces a profound dysbiosis of the microbiome that persists long after therapy is completed. Sci. Rep. 2017;7:10767. doi: 10.1038/s41598-017-10346-6.
    1. Faith JJ, Ahern PP, Ridaura VK, Cheng J, Gordon JI. Identifying gut microbe-host phenotype relationships using combinatorial communities in gnotobiotic mice. Sci. Transl. Med. 2014;6:220ra211. doi: 10.1126/scitranslmed.3008051.
    1. Geva-Zatorsky N, et al. Mining the human gut microbiota for immunomodulatory organisms. Cell. 2017;168:928–943 e911. doi: 10.1016/j.cell.2017.01.022.
    1. Shigyo K, et al. Efficacy of nitazoxanide against clinical isolates of Mycobacterium tuberculosis. Antimicrob. Agents Chemother. 2013;57:2834–2837. doi: 10.1128/AAC.02542-12.
    1. Walsh KF, et al. Early bactericidal activity trial of nitazoxanide for pulmonary tuberculosis. Antimicrob. Agents Chemother. 2020;64:e01956-19. doi: 10.1128/AAC.01956-19.
    1. Harausz EP, et al. Activity of nitazoxanide and tizoxanide against Mycobacterium tuberculosis in vitro and in whole blood culture. Tuberculosis. 2016;98:92–96. doi: 10.1016/j.tube.2016.03.002.
    1. Wagner, B. D. et al. On the use of diversity measures in longitudinal sequencing studies of microbial communities. Front. Microbiol.9, 1037 (2018).
    1. Ritchie ME, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47–e47. doi: 10.1093/nar/gkv007.
    1. Parada Venegas, D. et al. Short chain fatty acids (SCFAs)-mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front. Immunol.10, 277 (2019).
    1. Heinken A, et al. Systematic assessment of secondary bile acid metabolism in gut microbes reveals distinct metabolic capabilities in inflammatory bowel disease. Microbiome. 2019;7:75. doi: 10.1186/s40168-019-0689-3.
    1. Kim S, Covington A, Pamer EG. The intestinal microbiota: antibiotics, colonization resistance, and enteric pathogens. Immunol. Rev. 2017;279:90–105. doi: 10.1111/imr.12563.
    1. Rivera-Chávez F, et al. Depletion of butyrate-producing clostridia from the gut microbiota drives an aerobic luminal expansion of Salmonella. Cell Host Microbe. 2016;19:443–454. doi: 10.1016/j.chom.2016.03.004.
    1. Kelly CJ, et al. Crosstalk between microbiota-derived short-chain fatty acids and intestinal epithelial hif augments tissue barrier function. Cell Host Microbe. 2015;17:662–671. doi: 10.1016/j.chom.2015.03.005.
    1. Reese AT, et al. Antibiotic-induced changes in the microbiota disrupt redox dynamics in the gut. Elife. 2018;7:e35987. doi: 10.7554/eLife.35987.
    1. Subramanian A, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA. 2005;102:15545–15550. doi: 10.1073/pnas.0506580102.
    1. Korotkevich, G., Sukhov, V. & Sergushichev, A. Fast gene set enrichment analysis. Preprint at bioRxiv 060012 (2019).
    1. Liberzon A, et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 2015;1:417–425.
    1. Glickman MS, Cox JS, Jacobs WR., Jr. A novel mycolic acid cyclopropane synthetase is required for cording, persistence, and virulence of Mycobacterium tuberculosis. Mol. Cell. 2000;5:717–727. doi: 10.1016/S1097-2765(00)80250-6.
    1. Krzywinska, E. & Stockmann, C. Hypoxia, metabolism and immune cell function. Biomedicines6, 56 (2018).
    1. Palmer NP, et al. Concordance between gene expression in peripheral whole blood and colonic tissue in children with inflammatory bowel disease. PLoS ONE. 2019;14:e0222952–e0222952. doi: 10.1371/journal.pone.0222952.
    1. Knox NC, Forbes JD, Van Domselaar G, Bernstein CN. The gut microbiome as a target for IBD treatment: are we there yet? Curr. Treat. Options Gastroenterol. 2019;17:115–126. doi: 10.1007/s11938-019-00221-w.
    1. Magurran, A. E. Measuring Biological Diversity. (John Wiley & Sons, 2013).
    1. Haran JP, et al. Alzheimer’s disease microbiome is associated with dysregulation of the anti-inflammatory P-glycoprotein pathway. MBio. 2019;10:e00632-19. doi: 10.1128/mBio.00632-19.
    1. Johnstone IM, Titterington DM. Statistical challenges of high-dimensional data. Philos. Trans. A Math. Phys. Eng. Sci. 2009;367:4237–4253.
    1. Altmann A, Tolosi L, Sander O, Lengauer T. Permutation importance: a corrected feature importance measure. Bioinformatics. 2010;26:1340–1347. doi: 10.1093/bioinformatics/btq134.
    1. Apley, D. W. & Zhu, J. Visualizing the effects of predictor variables in black box supervised learning models. J. R. Statis. Soc.82, 1059–1086 (2016).
    1. Collins MD, et al. The phylogeny of the genus Clostridium: proposal of five new genera and eleven new species combinations. Int J. Syst. Bacteriol. 1994;44:812–826. doi: 10.1099/00207713-44-4-812.
    1. Lopetuso LR, Scaldaferri F, Petito V, Gasbarrini A. Commensal Clostridia: leading players in the maintenance of gut homeostasis. Gut Pathog. 2013;5:23–23. doi: 10.1186/1757-4749-5-23.
    1. Garrett WS, et al. Enterobacteriaceae act in concert with the gut microbiota to induce spontaneous and maternally transmitted colitis. Cell Host Microbe. 2010;8:292–300. doi: 10.1016/j.chom.2010.08.004.
    1. Seishima J, et al. Gut-derived Enterococcus faecium from ulcerative colitis patients promotes colitis in a genetically susceptible mouse host. Genome Biol. 2019;20:252. doi: 10.1186/s13059-019-1879-9.
    1. Peled JU, et al. Microbiota as predictor of mortality in allogeneic hematopoietic-cell transplantation. N. Engl. J. Med. 2020;382:822–834. doi: 10.1056/NEJMoa1900623.
    1. Freedberg DE, et al. Pathogen colonization of the gastrointestinal microbiome at intensive care unit admission and risk for subsequent death or infection. Intensive Care Med. 2018;44:1203–1211. doi: 10.1007/s00134-018-5268-8.
    1. Zeng MY, Inohara N, Nuñez G. Mechanisms of inflammation-driven bacterial dysbiosis in the gut. Mucosal Immunol. 2017;10:18–26. doi: 10.1038/mi.2016.75.
    1. Hanzelmann S, Castelo R, Guinney J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinforma. 2013;14:7. doi: 10.1186/1471-2105-14-7.
    1. Dethlefsen L, Relman DA. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc. Natl Acad. Sci. USA. 2011;108(Suppl 1):4554–4561. doi: 10.1073/pnas.1000087107.
    1. Lavelle A, et al. Baseline microbiota composition modulates antibiotic-mediated effects on the gut microbiota and host. Microbiome. 2019;7:111. doi: 10.1186/s40168-019-0725-3.
    1. Morgun A, et al. Uncovering effects of antibiotics on the host and microbiota using transkingdom gene networks. Gut. 2015;64:1732–1743. doi: 10.1136/gutjnl-2014-308820.
    1. Mayer-Barber KD, et al. Host-directed therapy of tuberculosis based on interleukin-1 and type I interferon crosstalk. Nature. 2014;511:99–103. doi: 10.1038/nature13489.
    1. Pirofski, L.-A. & Casadevall, A. Antimicrobial therapy in the context of the Damage-response framework: the prospect of optimizing therapy by reducing host damage. Antimicrob. Agents Chemother.64, 1800-01819 (2019).
    1. Namasivayam S, Sher A, Glickman MS, Wipperman MF. The microbiome and tuberculosis: early evidence for cross talk. MBio. 2018;9:e01420-18. doi: 10.1128/mBio.01420-18.
    1. Arpaia N, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504:451–455. doi: 10.1038/nature12726.
    1. Gold MC, et al. Human mucosal associated invariant T cells detect bacterially infected cells. PLoS Biol. 2010;8:e1000407. doi: 10.1371/journal.pbio.1000407.
    1. Skelly AN, Sato Y, Kearney S, Honda K. Mining the microbiota for microbial and metabolite-based immunotherapies. Nat. Rev. Immunol. 2019;19:305–323. doi: 10.1038/s41577-019-0144-5.
    1. Schirmer M, Garner A, Vlamakis H, Xavier RJ. Microbial genes and pathways in inflammatory bowel disease. Nat. Rev. Microbiol. 2019;17:497–511. doi: 10.1038/s41579-019-0213-6.
    1. Rutjes AW, Reitsma JB, Vandenbroucke JP, Glas AS, Bossuyt PM. Case–control and two-gate designs in diagnostic accuracy studies. Clin. Chem. 2005;51:1335–1341. doi: 10.1373/clinchem.2005.048595.
    1. Diacon AH, et al. Time to positivity in liquid culture predicts colony forming unit counts of Mycobacterium tuberculosis in sputum specimens. Tuberculosis. 2014;94:148–151. doi: 10.1016/j.tube.2013.12.002.
    1. Harris PA, et al. Research electronic data capture (REDCap)–a metadata-driven methodology and workflow process for providing translational research informatics support. J. Biomed. Inf. 2009;42:377–381. doi: 10.1016/j.jbi.2008.08.010.
    1. Callahan BJ, et al. DADA2: High-resolution sample inference from Illumina amplicon data. Nat. Methods. 2016;13:581–583. doi: 10.1038/nmeth.3869.
    1. Dupnik KM, et al. Blood transcriptomic markers of Mycobacterium tuberculosis load in sputum. Int. J. Tuberc. Lung Dis. 2018;22:950–958. doi: 10.5588/ijtld.17.0855.
    1. Dobin A, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2012;29:15–21. doi: 10.1093/bioinformatics/bts635.
    1. Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30:923–930.
    1. Hartley SW, Mullikin JC. QoRTs: a comprehensive toolset for quality control and data processing of RNA-Seq experiments. BMC Bioinforma. 2015;16:224–224. doi: 10.1186/s12859-015-0670-5.
    1. Barbie DA, et al. Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1. Nature. 2009;462:108–112. doi: 10.1038/nature08460.

Source: PubMed

3
Tilaa