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.

  • Article
  • Published:

A molecular network regulating the proinflammatory phenotype of human memory T lymphocytes

Abstract

Understanding the mechanisms that modulate helper T lymphocyte functions is crucial to decipher normal and pathogenic immune responses in humans. To identify molecular determinants influencing the pathogenicity of T cells, we separated ex vivo-isolated primary human memory T lymphocytes on the basis of their ability to produce high levels of inflammatory cytokines. We found that the inflammatory, cytokine-producing phenotype of memory T lymphocytes was defined by a specific core gene signature and was mechanistically regulated by the constitutive activation of the NF-κB pathway and by the expression of the transcriptional repressor BHLHE40. BHLHE40 attenuated the expression of anti-inflammatory factors, including miR-146a, a negative regulator of NF-κB activation and ZC3H12D, an RNase of the Regnase-1 family able to degrade inflammatory transcripts. Our data reveal a molecular network regulating the proinflammatory phenotype of human memory T lymphocytes, with the potential to contribute to disease.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Transcriptomic analysis of GM-CSF+ and GM-CSF cells.
Fig. 2: A general gene signature linked to a high cytokine-producing phenotype.
Fig. 3: Identification of TFs regulating the inflammatory cytokine-producing phenotype.
Fig. 4: BHLHE40 affects inflammatory cytokine production in human memory T cells.
Fig. 5: Phosphorylation of NF-κB p65 in GM-CSF+ and GM-CSF T cells.
Fig. 6: miR-146a expression is regulated by BHLHE40 and inhibits NF-κB p65 phosphorylation.
Fig. 7: ChIP-seq of BHLHE40 identifies specific binding loci in the human genome.
Fig. 8: Direct regulation of ZC3H12D by BHLHE40 and effect on inflammatory cytokine expression in human T lymphocytes.

Similar content being viewed by others

Data availability

All Nanostring, RNA-seq, ChIP-seq and ATAC-seq datasets are available in GEO (accession number GSE122946). Source data are provided for Figs. 4e and 5b. Other supporting raw data are available from the corresponding author upon request.

References

  1. Newell, E. W., Sigal, N., Bendall, S. C., Nolan, G. P. & Davis, M. M. Cytometry by time-of-flight shows combinatorial cytokine expression and virus-specific cell niches within a continuum of CD8+ T cell phenotypes. Immunity 36, 142–152 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Helmstetter, C. et al. Individual T helper cells have a quantitative cytokine memory. Immunity 42, 108–122 (2015).

    CAS  PubMed  Google Scholar 

  3. Cao, Y. et al. Functional inflammatory profiles distinguish myelin-reactive T cells from patients with multiple sclerosis. Sci. Transl. Med. 7, 287ra274 (2015).

    Google Scholar 

  4. Galli, E. et al. GM-CSF and CXCR4 define a T helper cell signature in multiple sclerosis. Nat. Med. 25, 1290–1300 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Hartmann, F. J. et al. Multiple sclerosis-associated IL2RA polymorphism controls GM-CSF production in human TH cells. Nat. Commun. 5, 5056 (2014).

    CAS  PubMed  Google Scholar 

  6. Hirota, K. et al. Autoimmune Th17 cells induced synovial stromal and innate lymphoid cell secretion of the cytokine GM-CSF to initiate and augment autoimmune arthritis. Immunity 48, 1220–1232 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Behrens, F. et al. MOR103, a human monoclonal antibody to granulocyte–macrophage colony-stimulating factor, in the treatment of patients with moderate rheumatoid arthritis: results of a phase Ib/IIa randomised, double-blind, placebo-controlled, dose-escalation trial. Ann. Rheum. Dis. 74, 1058–1064 (2015).

    CAS  PubMed  Google Scholar 

  8. Burmester, G. R. et al. Efficacy and safety of mavrilimumab in subjects with rheumatoid arthritis. Ann. Rheum. Dis. 72, 1445–1452 (2013).

    CAS  PubMed  Google Scholar 

  9. de Vries, E. G. et al. Flare-up of rheumatoid arthritis during GM-CSF treatment after chemotherapy. Lancet 338, 517–518 (1991).

    PubMed  Google Scholar 

  10. Codarri, L. et al. RORγT drives production of the cytokine GM-CSF in helper T cells, which is essential for the effector phase of autoimmune neuroinflammation. Nat. Immunol. 12, 560–567 (2011).

    CAS  PubMed  Google Scholar 

  11. Sonderegger, I. et al. GM-CSF mediates autoimmunity by enhancing IL-6-dependent Th17 cell development and survival. J. Exp. Med. 205, 2281–2294 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. McQualter, J. L. et al. Granulocyte–macrophage colony-stimulating factor: a new putative therapeutic target in multiple sclerosis. J. Exp. Med. 194, 873–882 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Campbell, I. K. et al. Protection from collagen-induced arthritis in granulocyte–macrophage colony-stimulating factor-deficient mice. J. Immunol. 161, 3639–3644 (1998).

    CAS  PubMed  Google Scholar 

  14. Wawro, M., Kochan, J., Krzanik, S., Jura, J. & Kasza, A. Intact NYN/PIN-like domain is crucial for the degradation of inflammation-related transcripts by ZC3H12D. J. Cell Biochem. 118, 487–498 (2017).

    CAS  PubMed  Google Scholar 

  15. Taganov, K. D., Boldin, M. P., Chang, K. J. & Baltimore, D. NF-κB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc. Natl Acad. Sci. USA 103, 12481–12486 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. McGeachy, M. J. et al. The interleukin 23 receptor is essential for the terminal differentiation of interleukin 17-producing effector T helper cells in vivo. Nat. Immunol. 10, 314–324 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Sakaguchi, S., Miyara, M., Costantino, C. M. & Hafler, D. A. FOXP3+ regulatory T cells in the human immune system. Nat. Rev. Immunol. 10, 490–500 (2010).

    CAS  PubMed  Google Scholar 

  18. Ascherio, A., Munger, K. L. & Simon, K. C. Vitamin D and multiple sclerosis. Lancet Neurol. 9, 599–612 (2010).

    PubMed  Google Scholar 

  19. Kongsbak, M., Levring, T. B., Geisler, C. & von Essen, M. R. The vitamin D receptor and T cell function. Front. Immunol. 4, 148 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Annunziato, F. et al. Phenotypic and functional features of human Th17 cells. J. Exp. Med. 204, 1849–1861 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Lin, C. C. et al. IL-1-induced Bhlhe40 identifies pathogenic T helper cells in a model of autoimmune neuroinflammation. J. Exp. Med. 213, 251–271 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Sun, H., Lu, B., Li, R. Q., Flavell, R. A. & Taneja, R. Defective T cell activation and autoimmune disorder in Stra13-deficient mice. Nat. Immunol. 2, 1040–1047 (2001).

    CAS  PubMed  Google Scholar 

  23. Martinez-Llordella, M. et al. CD28-inducible transcription factor DEC1 is required for efficient autoreactive CD4+ T cell response. J. Exp. Med. 210, 1603–1619 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Lin, C. C. et al. Bhlhe40 controls cytokine production by T cells and is essential for pathogenicity in autoimmune neuroinflammation. Nat. Commun. 5, 3551 (2014).

    PubMed  Google Scholar 

  25. Huynh, J. P. et al. Bhlhe40 is an essential repressor of IL-10 during Mycobacterium tuberculosis infection. J. Exp. Med. 215, 1823–1838 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Yu, F. et al. The transcription factor Bhlhe40 is a switch of inflammatory versus anti-inflammatory Th1 cell fate determination. J. Exp. Med. 215, 1813–1821 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Miyazaki, K. et al. The role of the basic helix-loop-helix transcription factor Dec1 in the regulatory T cells. J. Immunol. 185, 7330–7339 (2010).

    CAS  PubMed  Google Scholar 

  28. Ran, F. A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Geiger, R., Duhen, T., Lanzavecchia, A. & Sallusto, F. Human naive and memory CD4+ T cell repertoires specific for naturally processed antigens analyzed using libraries of amplified T cells. J. Exp. Med. 206, 1525–1534 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Mele, F. et al. ERK phosphorylation and miR-181a expression modulate activation of human memory TH17 cells. Nat. Commun. 6, 6431 (2015).

    CAS  PubMed  Google Scholar 

  31. St-Pierre, B., Flock, G., Zacksenhaus, E. & Egan, S. E. Stra13 homodimers repress transcription through class B E-box elements. J. Biol. Chem. 277, 46544–46551 (2002).

    CAS  PubMed  Google Scholar 

  32. Sun, H. & Taneja, R. Stra13 expression is associated with growth arrest and represses transcription through histone deacetylase (HDAC)-dependent and HDAC-independent mechanisms. Proc. Natl Acad. Sci. USA 97, 4058–4063 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Ivanova, A. V. et al. STRA13 interacts with STAT3 and modulates transcription of STAT3-dependent targets. J. Mol. Biol. 340, 641–653 (2004).

    CAS  PubMed  Google Scholar 

  34. Li, Y. et al. DEC1 negatively regulates the expression of DEC2 through binding to the E-box in the proximal promoter. J. Biol. Chem. 278, 16899–16907 (2003).

    CAS  PubMed  Google Scholar 

  35. Housley, W. J. et al. Genetic variants associated with autoimmunity drive NF-κB signaling and responses to inflammatory stimuli. Sci. Transl. Med. 7, 291ra293 (2015).

    Google Scholar 

  36. Monticelli, S., Aijo, T. & Trifari, S. Approaches to detect microRNA expression in T cell subsets and T cell differentiation. Methods Mol. Biol. 1514, 153–172 (2017).

    CAS  PubMed  Google Scholar 

  37. Matsushita, K. et al. Zc3h12a is an RNase essential for controlling immune responses by regulating mRNA decay. Nature 458, 1185–1190 (2009).

    CAS  PubMed  Google Scholar 

  38. Liang, J. et al. MCP-induced protein 1 deubiquitinates TRAF proteins and negatively regulates JNK and NF-κB signaling. J. Exp. Med. 207, 2959–2973 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Minagawa, K. et al. Post-transcriptional modulation of cytokine production in T cells for the regulation of excessive inflammation by TFL. J. Immunol. 192, 1512–1524 (2014).

    CAS  PubMed  Google Scholar 

  40. Ferber, I. A. et al. Mice with a disrupted IFN-γ gene are susceptible to the induction of experimental autoimmune encephalomyelitis (EAE). J. Immunol. 156, 5–7 (1996).

    CAS  PubMed  Google Scholar 

  41. Bettelli, E. et al. Loss of T-bet, but not STAT1, prevents the development of experimental autoimmune encephalomyelitis. J. Exp. Med. 200, 79–87 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Oestreich, K. J. & Weinmann, A. S. Master regulators or lineage-specifying? Changing views on CD4+ T cell transcription factors. Nat. Rev. Immunol. 12, 799–804 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Kanda, M. et al. Transcriptional regulator Bhlhe40 works as a cofactor of T-bet in the regulation of IFN-γ production in iNKT cells. Proc. Natl Acad. Sci. USA 113, e3394–e3402 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Zhang, L. et al. Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature 564, 268–272 (2018).

    CAS  PubMed  Google Scholar 

  45. Hosokawa, H. et al. Transcription factor PU.1 represses and activates gene expression in early T cells by redirecting partner transcription factor binding. Immunity 48, 1119–1134 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Oestreich, K. J. & Weinmann, A. S. T-bet employs diverse regulatory mechanisms to repress transcription. Trends Immunol. 33, 78–83 (2012).

    CAS  PubMed  Google Scholar 

  47. Curina, A. et al. High constitutive activity of a broad panel of housekeeping and tissue-specific cis-regulatory elements depends on a subset of ETS proteins. Genes Dev. 31, 399–412 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Balestrieri, C. et al. Co-optation of tandem DNA repeats for the maintenance of mesenchymal identity. Cell 173, 1150–1164 (2018).

    CAS  PubMed  Google Scholar 

  49. 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 

  50. Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y. & Greenleaf, W. J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

    PubMed  PubMed Central  Google Scholar 

  52. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Ross-Innes, C. S. et al. Differential oestrogen receptor binding is associated with clinical outcome in breast cancer. Nature 481, 389–393 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Diaferia, G. R. et al. Dissection of transcriptional and cis-regulatory control of differentiation in human pancreatic cancer. EMBO J. 35, 595–617 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Zambelli, F., Pesole, G. & Pavesi, G. Pscan: finding over-represented transcription factor binding site motifs in sequences from co-regulated or co-expressed genes. Nucleic Acids Res. 37, W247–W252 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Zambelli, F., Pesole, G. & Pavesi, G. PscanChIP: finding over-represented transcription factor-binding site motifs and their correlations in sequences from ChIP-Seq experiments. Nucleic Acids Res. 41, W535–W543 (2013).

    PubMed  PubMed Central  Google Scholar 

  59. Mahony, S. & Benos, P. V. STAMP: a web tool for exploring DNA-binding motif similarities. Nucleic Acids Res. 35, W253–W258 (2007).

    PubMed  PubMed Central  Google Scholar 

  60. Letunic, I. & Bork, P. Interactive tree of life (iTOL) v3: an online tool for the display and annotation of phylogenetic and other trees. Nucleic Acids Res. 44, W242–W245 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    PubMed  PubMed Central  Google Scholar 

  62. Delaleu, N., Nguyen, C. Q., Tekle, K. M., Jonsson, R. & Peck, A. B. Transcriptional landscapes of emerging autoimmunity: transient aberrations in the targeted tissue’s extracellular milieu precede immune responses in Sjogren’s syndrome. Arthritis Res. Ther. 15, R174 (2013).

    PubMed  PubMed Central  Google Scholar 

  63. Subramanian, A. et al. Gene-set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Su, G., Morris, J. H., Demchak, B. & Bader, G. D. Biological network exploration with Cytoscape 3. Curr. Protoc. Bioinformatics 47, 11–24 (2014).

    Google Scholar 

  65. Merico, D., Isserlin, R., Stueker, O., Emili, A. & Bader, G. D. Enrichment map: a network-based method for gene-set enrichment visualization and interpretation. PloS One 5, e13984 (2010).

    PubMed  PubMed Central  Google Scholar 

  66. Lindner, S. E. et al. The miR-15 family reinforces the transition from proliferation to differentiation in pre-B cells. EMBO Rep. 18, 1604–1617 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank D. Jarossay, S. Notarbartolo and F. Mele for invaluable technical support and input; M. Perez for help with Fig. 1; L. Vincenzetti for pilot CRISPR-Cas9 experiments and E. Džafo for help with experiments involving Regnase-4. This work was supported by the Swiss National Science Foundation (grant number 156875 and 175569), the NCCR ‘RNA & Disease’, the Swiss MS Society, the Ceresio Foundation, the Vontobel Stiftung and the Kurt und Senta Herrmann Stiftung (all to S. Monticelli). S. Montagner was supported by a Marie Heim-Vögtlin postdoctoral fellowship (number 164489). This work was also partially supported by the Italian Ministry of Health with Ricerca Corrente and 5×1000 funds (to G.N.).

Author information

Authors and Affiliations

Authors

Contributions

S.E., N.B., C.L., S. Montagner and M.C. designed and performed experiments and analyzed data; S.P., C.B. and G.N. performed and analyzed all the sequencing experiments; N.D. analyzed data; S. Monticelli oversaw the project and analyzed data. S. Monticelli and G.N. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Silvia Monticelli.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Extended data

Extended Data Fig. 1 Sorting scheme and principal component analysis.

a, Sorting scheme for GM-CSF secretion assay. TEM cells were separated by sorting and further divided by secretion assay and sorting. b, Principal component analysis of RNA-seq data. Cells from nine individual donors were separated by GM-CSF secretion assay and pooled in three pools of three donors each prior to RNA-seq. This analysis showed robust results, with differences due to sample-to-sample variability accounting for only 15.45% (PC#2) of the variance, while the majority of the observed differences were due to the phenotype of the cells (65.03%, GM-CSFpos vs. neg, PC#1).

Extended Data Fig. 2 Expression of specific markers in T cell subsets.

a, Cytokine expression in different T cell subsets. Each indicated cytokine (IL-22, IL-17A and IFN-γ) is expressed with or without concomitant co-expression of GM-CSF. Also, the IL-22+ cells are not simply a subset of the larger GM-CSF+ pool of cells. Mean ± SD. Each dot represents one donor (n = 5). b, Expression of IL23R. Expression of the IL23R gene in GM-CSF+ and GM-CSF cells, as determined by RNA-seq (n = 9). FDR = False discovery rate calculated for RNA-seq samples. c, FOXP3 expression. Comparison of FOXP3 expression in Treg cells (CD4+CD25highCD127low), GM-CSF+ and GM-CSF cells, as assessed by intracellular staining. Representative of n = 2 experiments.

Extended Data Fig. 3 Differences in gene expression patterns between GM-CSF+ and GM-CSF TEM cells.

Systematic delineation of coordinated changes in gene expression within specific gene sets (that is, pathways) was achieved by applying the data analysis and data visualization pipeline described in the Methods. 119 gene sets yielded significant enrichment in GM-CSF+ vs. GM-CSF cells (false-discovery-rate (FDR)-q < 0.01, nom. p-value of < 0.005 and TAGS > 50) whereas none were found depleted. Defining the network’s organization is the degree of overlap in leading edge-genes (genes contributing to each gene set’s significant enrichment) two gene sets (nodes) linked by a connection (edge) share (threshold connectivity 0.05). Node-size corresponds to FDR-q value threshold passed by the gene set (small < 0.01, medium < 0.001, large < 0.0001). Node color denotes MCL-cluster membership that subsequently would be summarized within 5 biological themes that is color-coded network areas. Their FDR-q value distribution is displayed in Fig. 1f.

Extended Data Fig. 4 RNA-seq, Nanostring, ATAC-seq and ChIP-seq analyses.

a, Comparison of RNA-seq and Nanostring data. Concordant results of gene expression profiling obtained by RNA-seq of TEM cells and Nanostring profiling of TCM cells. b, Expression of BHLHE40 in different T cell subsets. Expression of BHLHE40 was determined by qRT-PCR in different T cell subsets freshly sorted from peripheral blood. Each dot represents one donor (n = 6). Mean ± SD; paired t-test, two-tailed. c, ATAC-seq and ChIP-seq analyses. Representative snapshots of ChIP-seq and ATAC-seq tracks for selected genomic loci.

Extended Data Fig. 5 Optimization of CRISP-Cas9-mediated deletion and screening.

a, As a proof-of-principle and test of efficiency, Jurkat T cells were transfected using the Neon transfection system with ribonucleoparticles of Cas9 and one gRNA against the TCRα chain. After three days the cells were stained for surface TCR expression, showing a loss of expression in 62% of the cells (green). Grey: mock transfected control cells. Representative of at least n = 3 independent experiments. b, Schematic representation of the BHLHE40 locus with indicated the location of gRNAs and primers used for screening of the clones. c, Optimization of screening procedure by mismatch cleavage assay. After transfection with two gRNAs against BHLHE40, Jurkat cells were single cell-cloned by seeding in a 384-well plate in 20% FBS. Genomic DNA was extracted from 10 clones and from the entire population (no cloning) and negative controls. The first ‘long’ PCR showed already the presence of indels in most clones (left panel) and even at the level of whole population. This was further confirmed by denaturing, reannealing and T7 endonuclease digestion (middle panel). The presence of a homozygous deletion was further confirmed using a ‘short’ PCR that cannot provide an amplification product if the region between the two gRNAs is deleted. The highlighted Clone 7 (black box) is one of several clones in which the deletion was most likely identical on both alleles, leading to a shorter PCR product that cannot be digested by the T7 endonuclease because of the absence of mismatches. Representative of at least n = 2 independent transfections and cloning procedures. d, The genomic DNA from Clone 7 was sequenced in the BHLHE40 locus region, revealing a deletion of 196 nucleotides involving part of exon 1 and exon 2, as well as the intron. e, Schematic representation of the ZC3H12D locus with indicated the location of gRNAs and primers used for screening of the clones in Fig. 8f.

Extended Data Fig. 6 Analysis of primary human T cell clones transfected with Cas9 ribonucleoparticles directed against the BHLHE40 gene.

Primary human memory T lymphocytes were transfected with ribonucleoparticles of Cas9 and gRNAs against exon 1/2 and exon 5 of BHLHE40. After single cell cloning and expansion, individual clones were tested for the presence of the deletion by PCR. Median; Mann-Whitney test. Each dot represents one clone (n = 77 for KO and n = 90 for control clones).

Extended Data Fig. 7 MiRNA analysis.

a, Transduction with a miR-146a sponge of primary human T lymphocytes leads to increased NF-kB p65 phosphorylation. Primary memory T lymphocytes were transduced with a lentivirus expressing a miR-146a sponge and GFP as an independent reporter of the efficiency of transduction (schematic representation on top); a few days after transduction cells were stimulated with PMA and ionomycin for 5 min and levels of p65 phosphorylation were determined by intracellular staining. Cells were gated as GFPneg (non-expressing the sponge) and GFPpos (expressing the sponge) and compared also to untransduced cells. Representative of n = 2 experiments. b, Same as (c) except that the results (expressed as mean fluorescence intensity, MFI) of two independent experiments in which cells were stimulated for the indicated times are shown. c, Effect of BHLHE40 expression on miR-181a in Jurkat T cells. Jurkat T cells were transduced with a BHLHE40-expressing lentivirus. After selection and expansion of the transduced cells, expression of miR-181a was measured by qRT-PCR. Each dot represents one independent experiment (n = 4). Mean ± SD; paired t-test, two-tailed. d, Phylogenetic analysis of the top 50 bHLH matrices recovered from the BHLHE40 ChIP-seq data.

Extended Data Fig. 8 Schematic diagram describing the regulatory module identified in this study.

Using GM-CSF secretion as a proxy for an inflammatory phenotype of primary human memory TH lymphocytes, we identified a regulatory module that influences the activity of these pro-inflammatory and potentially pathogenic cells. In GM-CSF cells, mechanisms are in place to actively repress the expression of inflammatory cytokines, including high levels of miR-146a expression (a negative regulator of NF-κB activation), and relatively higher expression of ZC3H12D, an RNase enzyme involved in the negative regulation of cytokine expression. Conversely, in GM-CSF+ cells, the expression of the transcriptional repressor BHLHE40 leads to the direct downregulation of ZC3H12D expression, and indirectly affects also miR-146a. This in turn allows full-blown NF-κB activation and expression of inflammatory cytokines.

Supplementary information

Source data

Source Data Fig. 4

Uncropped images of western blots in Fig. 4. After transfer, the membrane was cut using the marker as a reference and the two parts of the same membrane were blotted with the indicated antibody. The boxes represent the parts of the gel shown in the figure.

Source Data Fig. 5

Uncropped images of western blots in Fig. 4. After transfer, the membrane was cut using the marker as a reference and the two parts of the same membrane were blotted with the indicated antibody. For each experiment, the same lysate was loaded twice on the same gel, the membrane was cut and part of the membrane was blotted with an anti-phosphorylated-p65 antibody, while the other part was blotted with a total anti-p65 antibody. The boxes represent the parts of the gels shown in the figure.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Emming, S., Bianchi, N., Polletti, S. et al. A molecular network regulating the proinflammatory phenotype of human memory T lymphocytes. Nat Immunol 21, 388–399 (2020). https://doi.org/10.1038/s41590-020-0622-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-020-0622-8

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