Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Resource
  • Published:

Mouse transcriptome reveals potential signatures of protection and pathogenesis in human tuberculosis

Abstract

Although mouse infection models have been extensively used to study the host response to Mycobacterium tuberculosis, their validity in revealing determinants of human tuberculosis (TB) resistance and disease progression has been heavily debated. Here, we show that the modular transcriptional signature in the blood of susceptible mice infected with a clinical isolate of M. tuberculosis resembles that of active human TB disease, with dominance of a type I interferon response and neutrophil activation and recruitment, together with a loss in B lymphocyte, natural killer and T cell effector responses. In addition, resistant but not susceptible strains of mice show increased lung B cell, natural killer and T cell effector responses in the lung upon infection. Notably, the blood signature of active disease shared by mice and humans is also evident in latent TB progressors before diagnosis, suggesting that these responses both predict and contribute to the pathogenesis of progressive M. tuberculosis infection.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Human TB blood transcriptional signature is preserved in blood of TB-susceptible mice.
Fig. 2: Mouse lung disease modules tested in lungs from diverse mouse TB models.
Fig. 3: Histological analysis of mouse lungs from M. tuberculosis-infected mice.
Fig. 4: Gene networks of specific TB modules in human blood from patients with TB and blood and lung from M. tuberculosis-infected mice.
Fig. 5: Histological analysis of mouse lungs from M. tuberculosis-infected mice for neutrophils, T cells and B cells.
Fig. 6: Quantitation of specific blood modular signatures against extent of lung pathology in mouse models and human TB.
Fig. 7: Quantitation of specific blood modular signatures in blood of healthy controls, LTBI, LTBI-progressors and patients with active TB.
Fig. 8: Quantitation of IFN-, neutrophil- and lymphocyte-specific gene expression in blood of healthy controls, LTBI, LTBI-progressors and patients with active TB.

Similar content being viewed by others

Data availability

The materials, data and any associated protocols that support the findings of this study are available from the corresponding author upon request. The RNA-seq datasets have been deposited in the NCBI Gene Expression Omnibus database with the primary accession number GSE140945 (TB mouse blood and lung). Publicly available datasets used in this study include GSE107995 (human TB datasets from Singhania et al.16) and GSE79362 (human TB dataset from Zak et al.38).

References

  1. WHO. Global Tuberculosis Report 2017 (World Health Organization, 2017).

  2. Dowdy, D. W., Basu, S. & Andrews, J. R. Is passive diagnosis enough? The impact of subclinical disease on diagnostic strategies for tuberculosis. Am. J. Resp. Crit. Care Med. 187, 543–551 (2013).

    PubMed  PubMed Central  Google Scholar 

  3. Casanova, J. L. & Abel, L. Genetic dissection of immunity to mycobacteria: the human model. Annu. Rev. Immunol. 20, 581–620 (2002).

    CAS  PubMed  Google Scholar 

  4. Cooper, A. M., Mayer-Barber, K. D. & Sher, A. Role of innate cytokines in mycobacterial infection. Mucosal Immunol. 4, 252–260 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Flynn, J. L. & Chan, J. Immunology of tuberculosis. Annu. Rev. Immunol. 19, 93–129 (2001).

    CAS  PubMed  Google Scholar 

  6. O’Garra, A. et al. The immune response in tuberculosis. Annu. Rev. Immunol. 31, 475–527 (2013).

    PubMed  Google Scholar 

  7. Mayer-Barber, K. D. et al. Host-directed therapy of tuberculosis based on interleukin-1 and type I interferon crosstalk. Nature 511, 99–103 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Berry, M. P. et al. An interferon-inducible neutrophil-driven blood transcriptional signature in human tuberculosis. Nature 466, 973–977 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Blankley, S. et al. A 380-gene meta-signature of active tuberculosis compared with healthy controls. Eur. Resp. J. 47, 1873–1876 (2016).

    Google Scholar 

  10. Blankley, S. et al. The transcriptional signature of active tuberculosis reflects symptom status in extra-pulmonary and pulmonary tuberculosis. PloS One 11, e0162220 (2016).

    PubMed  PubMed Central  Google Scholar 

  11. Bloom, C. I. et al. Transcriptional blood signatures distinguish pulmonary tuberculosis, pulmonary sarcoidosis, pneumonias and lung cancers. PloS One 8, e70630 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Joosten, S. A., Fletcher, H. A. & Ottenhoff, T. H. A helicopter perspective on TB biomarkers: pathway and process based analysis of gene expression data provides new insight into TB pathogenesis. PloS One 8, e73230 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. 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. 10, e1001538 (2013).

    PubMed  PubMed Central  Google Scholar 

  14. Maertzdorf, J. et al. Human gene expression profiles of susceptibility and resistance in tuberculosis. Genes Immun. 12, 15–22 (2011).

    CAS  PubMed  Google Scholar 

  15. Scriba, T. J. et al. Sequential inflammatory processes define human progression from M. tuberculosis infection to tuberculosis disease. PLoS Pathog. 13, e1006687 (2017).

    PubMed  PubMed Central  Google Scholar 

  16. Singhania, A. et al. A modular transcriptional signature identifies phenotypic heterogeneity of human tuberculosis infection. Nat. Commun. 9, 2308 (2018).

    PubMed  PubMed Central  Google Scholar 

  17. Behar, S. M., Divangahi, M. & Remold, H. G. Evasion of innate immunity by Mycobacterium tuberculosis: is death an exit strategy? Nat. Rev. Microbiol. 8, 668–674 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Moreira-Teixeira, L., Mayer-Barber, K., Sher, A. & O’Garra, A. Type I interferons in tuberculosis: foe and occasionally friend. J. Exp. Med. 215, 1273–1285 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Carmona, J. et al. Mycobacterium tuberculosis strains are differentially recognized by TLRs with an impact on the immune response. PloS One 8, e67277 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Dorhoi, A. et al. Type I IFN signaling triggers immunopathology in tuberculosis-susceptible mice by modulating lung phagocyte dynamics. Eur. J. Immunol. 44, 2380–2393 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Manca, C. et al. Virulence of a Mycobacterium tuberculosis clinical isolate in mice is determined by failure to induce Th1 type immunity and is associated with induction of IFN-α/β. Proc. Natl Acad. Sci. USA 98, 5752–5757 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Manca, C. et al. Hypervirulent M. tuberculosis W/Beijing strains upregulate type I IFNs and increase expression of negative regulators of the Jak–Stat pathway. J. Interferon Cytokine Res. 25, 694–701 (2005).

    CAS  PubMed  Google Scholar 

  23. Ordway, D. et al. The hypervirulent Mycobacterium tuberculosis strain HN878 induces a potent TH1 response followed by rapid down-regulation. J. Immunol. 179, 522–531 (2007).

    CAS  PubMed  Google Scholar 

  24. McNab, F. W. et al. TPL-2-ERK1/2 signaling promotes host resistance against intracellular bacterial infection by negative regulation of type I IFN production. J. Immunol. 191, 1732–1743 (2013).

    CAS  PubMed  Google Scholar 

  25. Bogunovic, D. et al. Mycobacterial disease and impaired IFN-γ immunity in humans with inherited ISG15 deficiency. Science 337, 1684–1688 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Antonelli, L. R. et al. Intranasal poly-IC treatment exacerbates tuberculosis in mice through the pulmonary recruitment of a pathogen-permissive monocyte/macrophage population. J. Clin. Invest. 120, 1674–1682 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Redford, P. S. et al. Influenza A virus impairs control of Mycobacterium tuberculosis coinfection through a type I interferon receptor-dependent pathway. J. Infect. Dis. 209, 270–274 (2014).

    CAS  PubMed  Google Scholar 

  28. Barry, C. E. III et al. The spectrum of latent tuberculosis: rethinking the biology and intervention strategies. Nat. Rev. Microbiol. 7, 845–855 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Kramnik, I. & Beamer, G. Mouse models of human TB pathology: roles in the analysis of necrosis and the development of host-directed therapies. Semin. Immunopathol. 38, 221–237 (2016).

    PubMed  Google Scholar 

  30. Domaszewska, T. et al. Concordant and discordant gene expression patterns in mouse strains identify best-fit animal model for human tuberculosis. Sci. Rep. 7, 12094 (2017).

    PubMed  PubMed Central  Google Scholar 

  31. Singhania, A. et al. Transcriptional profiling unveils type I and II interferon networks in blood and tissues across diseases. Nat. Commun. 10, 2887 (2019).

    PubMed  PubMed Central  Google Scholar 

  32. Ley, K. et al. Neutrophils: new insights and open questions. Sci. Immunol. 3, eaat4579 (2018).

    PubMed  Google Scholar 

  33. Dorhoi, A. et al. The adaptor molecule CARD9 is essential for tuberculosis control. J. Exp. Med. 207, 777–792 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Eruslanov, E. B. et al. Neutrophil responses to Mycobacterium tuberculosis infection in genetically susceptible and resistant mice. Infect. Immun. 73, 1744–1753 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Nandi, B. & Behar, S. M. Regulation of neutrophils by interferon-γ limits lung inflammation during tuberculosis infection. J. Exp. Med. 208, 2251–2262 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Achkar, J. M., Chan, J. & Casadevall, A. B cells and antibodies in the defense against Mycobacterium tuberculosis infection. Immunol. Rev. 264, 167–181 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Lu, L. L. et al. A functional role for antibodies in tuberculosis. Cell 167, 433–443 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Zak, D. E. et al. A blood RNA signature for tuberculosis disease risk: a prospective cohort study. Lancet 387, 2312–2322 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Feng, C. G. et al. NK cell-derived IFN-γ differentially regulates innate resistance and neutrophil response in T cell-deficient hosts infected with Mycobacterium tuberculosis. J. Immunol. 177, 7086–7093 (2006).

    CAS  PubMed  Google Scholar 

  40. Chowdhury, R. R. et al. A multi-cohort study of the immune factors associated with M. tuberculosis infection outcomes. Nature 560, 644–648 (2018).

    PubMed Central  Google Scholar 

  41. Aly, S. et al. Oxygen status of lung granulomas in Mycobacterium tuberculosis-infected mice. J. Pathol. 210, 298–305 (2006).

    CAS  PubMed  Google Scholar 

  42. Gopal, R. et al. S100A8/A9 proteins mediate neutrophilic inflammation and lung pathology during tuberculosis. Am. J. Resp. Crit. Care Med. 188, 1137–1146 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Eum, S.-Y. et al. Neutrophils are the predominant infected phagocytic cells in the airways of patients with active pulmonary TB. Chest 137, 122–128 (2010).

    PubMed  Google Scholar 

  44. Verma, R. et al. A novel high sensitivity bacteriophage-based assay identifies low level M. tuberculosis bacteraemia in immunocompetent patients with active and incipient TB. Clin. Infect. Dis. 70, 933–936 (2019).

    Google Scholar 

  45. Cooper, A. M., Pearl, J. E., Brooks, J. V., Ehlers, S. & Orme, I. M. Expression of the nitric oxide synthase 2 gene is not essential for early control of Mycobacterium tuberculosis in the murine lung. Infect. Immun. 68, 6879–6882 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Moreira-Teixeira, L. et al. T Cell-derived IL-10 impairs host resistance to Mycobacterium tuberculosis infection. J. Immunol. 199, 613–623 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Pichugin, A. V., Yan, B. S., Sloutsky, A., Kobzik, L. & Kramnik, I. Dominant role of the sst1 locus in pathogenesis of necrotizing lung granulomas during chronic tuberculosis infection and reactivation in genetically resistant hosts. Am. J. Pathol. 174, 2190–2201 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Ji, D. X. et al. Type I interferon-driven susceptibility to Mycobacterium tuberculosis is mediated by IL-1Ra. Nat. Microbiol. 4, 2128–2135 (2019).

    PubMed  PubMed Central  Google Scholar 

  49. Esmail, H. et al. Complement pathway gene activation and rising circulating immune complexes characterize early disease in HIV-associated tuberculosis. Proc. Natl Acad. Sci. USA 115, E964–E973 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Irwin, S. M. et al. Presence of multiple lesion types with vastly different microenvironments in C3HeB/FeJ mice following aerosol infection with Mycobacterium tuberculosis. Dis. Models Mech. 8, 591–602 (2015).

    Google Scholar 

  51. Ewels, P., Magnusson, M., Lundin, S. & Kaller, M. MultiQC: summarize analysis results for multiple tools and samples in a single report. Bioinformatics 32, 3047–3048 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Anders, S., Pyl, P. T. & Huber, W. HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    CAS  PubMed  Google Scholar 

  55. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    PubMed  PubMed Central  Google Scholar 

  56. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. B Stat. Methodol. 57, 289–300 (1995).

    Google Scholar 

  57. Hadley, W. Ggplot2: Elegant Graphics for Data Analysis 2nd edn (Springer International Publishing, 2016).

  58. Newman, A. M. et al. Robust enumeration of cell subsets from tissue expression profiles. Nat. Methods 12, 453–457 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Chen, Z. et al. Inference of immune cell composition on the expression profiles of mouse tissue. Sci. Rep. 7, 40508 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Heng, T. S. & Painter, M. W. The Immunological Genome Project: networks of gene expression in immune cells. Nat. Immunol. 9, 1091–1094 (2008).

    CAS  PubMed  Google Scholar 

  61. Yoshida, H. et al. The cis-regulatory atlas of the mouse immune system. Cell 176, 897–912 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Langfelder, P. & Horvath, S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 9, 559 (2008).

    PubMed  PubMed Central  Google Scholar 

  63. Yip, A. M. & Horvath, S. Gene network interconnectedness and the generalized topological overlap measure. BMC Bioinformatics 8, 22 (2007).

    PubMed  PubMed Central  Google Scholar 

  64. Langfelder, P., Zhang, B. & Horvath, S. Defining clusters from a hierarchical cluster tree: the Dynamic Tree Cut package for R. Bioinformatics 24, 719–720 (2008).

    CAS  PubMed  Google Scholar 

  65. Yaari, G., Bolen, C. R., Thakar, J. & Kleinstein, S. H. Quantitative set analysis for gene expression: a method to quantify gene set differential expression including gene–gene correlations. Nucleic Acids Res. 41, e170 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank S. Hadebe (Francis Crick Institute UK, now UCT, South Africa) and J. Pitt (Medical Research Council (MRC), National Institute for Medical Research (NIMR), UK) for some of the early mouse M. tuberculosis infections and samples and sample processing leading up to the current study and K. Potempa (MRC, NIMR, UK) for early analysis of some of the microarray data leading up to the current study. The authors also thank L. Gabryšová (Francis Crick Institute UK, now Novartis, Basel, Switzerland) for her intellectual contribution to discussion of the project. The authors thank X. Wu for her help in organization of mice for TB experiments. The authors thank the Francis Crick Institute Science Technology Platforms: Biological Services for breeding and maintenance of the mice used for the early mouse M. tuberculosis infections and samples leading up to the current study; Advanced Sequencing Facility, Bioinformatics and Biostatistics Science Technology Platforms for their contribution to our sequencing processing and R. Goldstone for excellent project management of sequencing and D. Jackson for support of sequencing; and Experimental Histopathology for their excellent work in preparing lung sections for histological analyses. This study was funded by the Francis Crick Institute, which receives its core funding from Cancer Research UK (FC001126), the UK MRC (FC001126) and the Wellcome Trust (FC001126); before that by the UK MRC (U117565642); and by the European Research Council (294682-TB-PATH). A.O’G., L.M-T., O.T., C.M.G., A. Singhania and E.S. were supported by the Francis Crick Institute, which receives its core funding as described above; L.M.-T., and A. Singhania, were additionally funded by the European Research Council (294682-TB-PATH). S.L.P., A.S.-B. and E.H. were funded by the Royal Veterinary College and the Francis Crick Institute. K.D.M.-B. and A. Sher were funded by the Intramural Research Program of the National Institutes of Allergy and Infectious Disease. M.S. was funded by FEDER-Fundo Europeu de Desenvolvimento Regional funds through the COMPETE 2020 – Operational Programme for Competitiveness and Internationalization (POCI), Portugal 2020 and by Portuguese funds through Fundação para a Ciência e a Tecnologia (FCT) in the framework of the project ‘Institute for Research and Innovation in Health Sciences’ (POCI-01-0145-FEDER-007274) and by FCT through Estimulo Individual ao Emprego Científico. K.L.F. and B.C. are funded by FCT PhD scholarships SFRH/BD/114405/2016 and SFRH/BD/114403/2016, respectively. P.H. and R.V. were supported by NIHR Leicester Biomedical Research Centre and the University of Leicester.

Author information

Authors and Affiliations

Authors

Contributions

A.O’G. conceived and designed the study with input from L.M.-T., M.S., C.M.G. and P.S.R. and led and coordinated the study with help from L.M.-T. A.O’G. cowrote the manuscript together with L.M.-T. O.T. performed the major part of the bioinformatics analyses with considerable input from A. Singhania, supervised by A.O’G. P.C. provided technical bioinformatics support. L.M.-T. coordinated the logistics of the study. C.M.G. isolated RNA and prepared sequencing libraries from the TB mouse models. E.S. assisted in TB mouse model experiments executed by K.L.F., J.S., B.C. and P.S.R. and designed by A.O’G., L.M.-T. and M.S. E.S. contributed to all early mouse model TB experiments executed with P.S.R. and L.M.-T., supervised by A.O’G., L.M.-T. and M.S. S.L.P., A.S.-B. and E.H. performed histopathological analysis and interpretation. K.D.M.-B. and A.Sh. provided TB samples from early mouse TB experiments leading up to the current study. M.S., K.D.M.-B. and A. Sher also provided important discussion for the project throughout and critical feedback on the manuscript. R.V. and P.H. provided clinical analysis of patients with TB and also provided critical feedback on the manuscript and important discussion for the project. All coauthors read, reviewed and approved the manuscript.

Corresponding author

Correspondence to Anne O’Garra.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Zoltan Fehervari was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Information

Supplementary Figs. 1–9.

Reporting Summary

Supplementary Table 1

Human blood TB modules annotation. Annotation and number of genes of each human blood TB module (HB1–HB23) from Singhania et al.16. Module name indicates biological processes associated with the genes included in the module. Gene of interest lists a subset of genes of interest included in each module, Canonical Pathway (IPA) shows the top five canonical pathways found with IPA and Pathways (Acumenta) shows the top five pathways found with Acumenta (Singhania et al.16).

Supplementary Table 2

Human blood data in human blood TB modules, from Singhania et al.16, separated in 12 tabs depicting: London individuals raw, the raw read counts for all genes in all individual samples from the London dataset; London groups raw, the mean raw counts per group from the London dataset for all genes; London individuals norm, the normalized read counts of filtered genes in all individual samples from the London dataset; London groups norm, the mean normalized read counts per group from the London dataset for filtered genes; SA individuals raw, the raw read counts of all genes in all individual samples from the South Africa (SA) dataset; SA groups raw, the mean raw counts per group from the SA dataset for all genes; SA individuals norm, the normalized read counts of filtered genes in all individual samples of SA dataset; SA groups norm, the mean normalized read counts per group from the SA dataset for filtered genes; Leicester individuals raw, the raw read counts of all genes in all individual samples from the Leicester dataset; Leicester groups raw, the mean raw counts per group from the Leicester dataset for all genes; Leicester individuals norm, the normalized read counts of filtered genes in all individual samples from the Leicester dataset; Leicester groups norm, the mean normalized read counts per group from the Leicester dataset for filtered genes. For each gene, the corresponding human blood TB module is detailed if applicable. Normalized read counts have been processed using the variance stabilizing transformation from DESeq2 R library, and genes with low read counts have been filtered out, as described in Singhania et al.16.

Supplementary Table 3

Mouse blood data in human blood TB modules, from Singhania et al.16, separated in six tabs depicting: mouse blood individuals raw, the raw read counts of all genes in all individual samples of mouse blood RNA-seq dataset; mouse blood groups raw, the mean raw counts per group in the mouse blood RNA-seq dataset for all genes; mouse blood individuals norm, the normalized read counts of all genes in all individual samples of mouse blood RNA-seq dataset; mouse blood groups norm, the mean normalized read counts per group in the mouse blood RNA-seq dataset for all genes; Supplementary Fig. 2a mouse blood microarray, the mouse blood microarray data corresponding to Supplementary Fig. 2a; Supplementary Fig. 3a mouse blood RNA-seq, the mouse blood normalized RNA-seq data corresponding to Supplementary Fig. 3a. For each gene, the corresponding human blood TB module is detailed if applicable. Normalized read counts from RNA-seq data represent read counts processed using the variance stabilizing transformation from DESeq2 R library.

Supplementary Table 4

Mouse lung data in mouse lung disease modules, from Singhania et al.31, separated in six tabs depicting: mouse lung individuals raw, the raw read counts of all genes in all individual samples of mouse lung RNA-seq dataset; mouse lung groups raw, the mean raw counts per group in the mouse lung RNA-seq dataset for all genes; mouse lung individuals norm, the normalized read counts of all genes in all individual samples of lung blood RNA-seq dataset; mouse lung groups norm, the mean normalized read counts per group in the mouse lung RNA-seq dataset for all genes; Supplementary Fig. 2b mouse lung microarray, the mouse lung microarray data corresponding to Supplementary Fig. 2b; Supplementary Fig. 3b mouse lung RNA-seq, the mouse lung normalized RNA-seq data corresponding to Supplementary Fig. 3b. For each gene, the corresponding disease lung module is detailed if applicable. Normalized read counts from RNA-seq data represent read counts processed using the variance stabilizing transformation from DESeq2 R library.

Supplementary Table 5

Mouse lung TB modules annotation. Annotation and the number of genes for each mouse lung TB module (ML1–ML27). Module name indicates biological processes associated with the genes within the module. Gene of interest lists a subset of genes of interest included in each module and all the other columns represent an output from either IPA or Metacore.

Supplementary Table 6

Mouse lung data in mouse lung TB modules, separated in four tabs depicting: mouse lung individuals raw, the raw read counts of all genes in all individual samples of mouse lung RNA-seq dataset; Mouse lung groups raw, the mean raw counts per group in the mouse lung RNA-seq dataset for all genes; Mouse lung individuals norm, the normalized read counts of all genes in all samples of mouse lung RNA-seq dataset; Mouse lung groups norm, the mean normalized read counts per group in the mouse lung RNA-seq dataset for all genes. For each gene, the corresponding TB lung module is detailed if applicable. Normalized read counts from RNA-seq data represent read counts processed using the variance stabilizing transformation from DESeq2 R library.

Supplementary Table 7

X-ray scores for human TB cohort from Leicester. X-ray classification (CXR classification) for each patient evaluated from the Leicester cohort and their associated disease group (TB subgroup).

Supplementary Table 8

Mouse to human gene mapping. The table lists the correspondence between mouse Ensembl gene ID and human Ensembl gene ID (ortholog genes), found using biomaRt package in R.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Moreira-Teixeira, L., Tabone, O., Graham, C.M. et al. Mouse transcriptome reveals potential signatures of protection and pathogenesis in human tuberculosis. Nat Immunol 21, 464–476 (2020). https://doi.org/10.1038/s41590-020-0610-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-020-0610-z

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing