Content area
The expression of co-inhibitory receptors, such as CTLA-4 and PD-1, on effector T cells is a key mechanism for ensuring immune homeostasis. Dysregulated expression of co-inhibitory receptors on CD4<sup>+</sup> T cells promotes autoimmunity, whereas sustained overexpression on CD8<sup>+</sup> T cells promotes T cell dysfunction or exhaustion, leading to impaired ability to clear chronic viral infections and diseases such as cancer1,2. Here, using RNA and protein expression profiling at single-cell resolution in mouse cells, we identify a module of co-inhibitory receptors that includes not only several known co-inhibitory receptors (PD-1, TIM-3, LAG-3 and TIGIT) but also many new surface receptors. We functionally validated two new co-inhibitory receptors, activated protein C receptor (PROCR) and podoplanin (PDPN). The module of coinhibitory receptors is co-expressed in both CD4<sup>+</sup> and CD8<sup>+</sup> T cells and is part of a larger co-inhibitory gene program that is shared by non-responsive T cells in several physiological contexts and is driven by the immunoregulatory cytokine IL-27. Computational analysis identified the transcription factors PRDM1 and c-MAF as cooperative regulators of the co-inhibitory module, and this was validated experimentally. This molecular circuit underlies the co-expression of co-inhibitory receptors in T cells and identifies regulators of T cell function with the potential to control autoimmunity and tumour immunity.
The expression of co-inhibitory receptors, such as CTLA-4 and PD-1, on effector T cells is a key mechanism for ensuring immune homeostasis. Dysregulated expression of co-inhibitory receptors on CD4<sup>+</sup> T cells promotes autoimmunity, whereas sustained overexpression on CD8<sup>+</sup> T cells promotes T cell dysfunction or exhaustion, leading to impaired ability to clear chronic viral infections and diseases such as cancer1,2. Here, using RNA and protein expression profiling at single-cell resolution in mouse cells, we identify a module of co-inhibitory receptors that includes not only several known co-inhibitory receptors (PD-1, TIM-3, LAG-3 and TIGIT) but also many new surface receptors. We functionally validated two new co-inhibitory receptors, activated protein C receptor (PROCR) and podoplanin (PDPN). The module of coinhibitory receptors is co-expressed in both CD4<sup>+</sup> and CD8<sup>+</sup> T cells and is part of a larger co-inhibitory gene program that is shared by non-responsive T cells in several physiological contexts and is driven by the immunoregulatory cytokine IL-27. Computational analysis identified the transcription factors PRDM1 and c-MAF as cooperative regulators of the co-inhibitory module, and this was validated experimentally. This molecular circuit underlies the co-expression of co-inhibitory receptors in T cells and identifies regulators of T cell function with the potential to control autoimmunity and tumour immunity.
We used single-cell RNA sequencing (scRNA-seq) to analyse coinhibitory and co-stimulatory receptor expression in 588 CD8+ and 316 CD4+ tumour-infiltrating lymphocytes (TILs) from B16F10 mouse melanoma3. We found that the expression of Pdcd1 (also known as PD-1), Tim3 (Havcr2), Lag3, Ctla4, 4-1BB (Tnfrsf9) and Tigit strongly co-vary in CD8+ TILs. CD4+ TILs showed a similar pattern with the additional co-expression of Icos, Gitr (also known as Tnfrsf18) and Ox40 (Tnfrsf4) (Fig. 1a, top). Single-cell mass cytometry (cytometry by time of flight, CyTOF) confirmed the surface co-expression of these receptors (Fig. 1a, bottom, Supplementary Information 1). The expression of PD-1, LAG-3, TIM-3 and TIGIT was tightly correlated on both CD8+ and CD4+ TILs (Fig. 1a, bottom). Clustering analysis (t-stochastic neighbourhood embedding (t-SNE)4, Methods) showed two groups of CD8+ TILs (clusters 1 and 2) (Fig. 1b, Extended Data Fig. 1a, c), with PD-1, LAG-3, TIM-3 and TIGIT mainly expressed in cluster 1 cells (Fig. 1b, Extended Data Fig. 1c), in addition to LILRB4 (Extended Data Fig. 1a) and co-stimulatory receptors of the TNF receptor family, 4-1BB, OX40 and GITR. By contrast, ICOS and CD226 were less restricted to cluster 1 (Extended Data Fig. 1a). We further observed two discrete clusters of CD4+ TILs (clusters 3 and 4), with co-expression of PD-1, TIM-3, LAG-3 and TIGIT restricted to cluster 3 (Fig. 1b, Extended Data Fig. 1c).
The co-expression of co-inhibitory receptors on CD8+ and CD4+ T cells suggests a common trigger. One candidate is IL-27, a heterodimeric member of the IL-12 cytokine family that suppresses autoimmunity5, induces IL-10-secreting type 1 regulatory T (Treg) cells6,7 and induces expression of TIM-3 and PD-L1 on CD4+ and CD8+ T cells8,9. Activation of CD4+ and CD8+ T cells in the presence of IL-27 induced the expression of TIM-3, LAG-3 and TIGIT at both the mRNA (Fig. 1c) and protein levels (Extended Data Fig. 2a). mRNA expression of Tim3 (Havcr2), Lag3 and Tigit was reduced in IL-27RAdeficient T cells, whereas Pdcd1 expression was unaffected by IL-27 in vitro (Fig. 1c, Extended Data Fig. 2a).
CyTOF analysis showed that the loss of IL-27RA resulted in the loss of cells in cluster 1 of CD8+ TILs and cluster 3 of CD4+ TILs (Fig. 1d, P = 5 × 10-23 and 6.8 × 10-7 for CD8+ and CD4+, respectively, hypergeometric test; Extended Data Fig. 1b-d), indicating a key role for IL-27 in driving co-inhibitory receptor co-expression in both CD4+ and CD8+ T cells in vivo. Although PD-1 expression was not dependent on IL-27 in vitro, it was dependent on IL-27RA signalling in vivo. Consistent with the induction of IL-10 by IL-275-7, we observed reduced IL-10 in IL-27RA-knockout CD8+ TILs (Extended Data Fig. 2b).
scRNA-seq of CD8+ and CD4+ TILs from wild-type and IL-27RAknockout mice (Fig. 1e, Extended Data Fig. 3a, b, Methods) revealed distinct clusters of CD8+ (cluster 5) and CD4+ (cluster 4) TILs that highly expressed the co-inhibitory receptors Pdcd1, Tim3, Lag3 and Tigit. The expression of these genes was decreased in CD8+ TILs from IL-27RA-knockout mice, whereas the expression of only Tim3 and Lag3 was decreased in CD4+ TILs from IL-27RA-knockout mice (Fig. 1e). Thus, IL-27 drives a module of co-inhibitory receptors that are strongly co-expressed in vivo together with IL-10.
The co-inhibitory receptor module could be part of a larger IL-27- driven inhibitory gene program. We analysed the mRNA profiles of CD4+ and CD8+ T cells stimulated in the presence or absence of IL-27. IL-27 induced similar expression programs in CD4+ and CD8+ T cells (Extended Data Fig. 4a, b). We identified 1,201 genes with IL-27- dependent expression (Methods). We compared the IL-27-driven gene program to the gene signatures for four different states of T cell nonresponsiveness: CD8+ T cell exhaustion in both cancer3 and chronic viral infection10, and antigen-specific11 and non-specific (anti-CD3 antibody12) CD4+ T cell tolerance. We found a significant overlap with all of these signatures (Methods, Extended Data Fig. 4c-f).
Projection of the IL-27 and CD8+ cancer T cell exhaustion overlap signature onto the single-cell profiles of CD8+ TILs marked a distinct subset of cells (Fig. 2a, panel I). This subset scored highly for the overlap signatures between the IL-27-driven gene program and each of the other three states of T cell non-responsiveness (Fig. 2a, panels II-IV). The transcriptional program induced in IL-27RA-knockout TILs was active in a complimentary subset of TILs (Fig. 2a, panel V, Methods). The control signature from cells stimulated with IL-27 in vitro showed bimodal distribution and by itself did not detect the same population of cells (Fig. 2a, panel VI). From these analyses, we identified a co-inhibitory gene module (272 genes) that is shared across several states of T cell non-responsiveness (Supplementary Table 2). Within this module, we identified a set of 57 genes that encode cell-surface receptors and cytokines, including TIM-3, LAG-3, TIGIT and IL-10 (Fig. 2b), which we further stratified by their expression in cancer and chronic viral infections (Fig. 2c). Two surface molecules, PROCR and PDPN, were highly expressed in the setting of cancer (Fig. 2c). Activation of naive CD4+ and CD8+ T cells in vitro in the presence of IL-27 induced the expression of PROCR and PDPN (Extended Data Fig. 5a). In vivo, PROCR and PDPN exhibited IL-27-dependent coexpression with PD-1 and TIM-3 on CD8+ TILs (Extended Data Fig. 5b).
PROCR+CD8+ TILs exhibited an exhausted phenotype, producing less TNF and IL-2 and more IL-10 than PROCR-CD8+ TILs (Extended Data Fig. 5c). The growth of B16F10 melanoma was inhibited in PROCR hypomorph (Procrdelta/delta, hereafter Procrd/d)13 mice (Fig. 2d), and Procrd/d CD8+ TILs mice exhibited enhanced production of TNF, but no difference in the production of IL-2, IFN-γ or IL-10 (Fig. 2e). Procrd/d TILs exhibited a decreased frequency of TIM-3high and PD-1high CD8+ T cells, suggesting that PROCR signalling promotes a severely exhausted phenotype in CD8+ T cells14 (Fig. 2f). Adoptive transfer of CD8+ T cells that lack PROCR revealed a T cell-specific role for PROCR in constraining tumour growth (Extended Data Fig. 5d).
Although PDPN can limit CD4+ T cell survival in inflamed tissues15, its role in T cell exhaustion is unknown. We observed a significant delay in B16F10 tumour growth in mice with PDPN deficiency in T cells (PDPN conditional knockout (cKO)) (Fig. 2g). PDPN-deficient CD8+ TILs exhibited enhanced TNF production but no significant difference in IL-2, IFN-γ or IL-10 (Fig. 2h). The frequency of TIM-3high and PD-1high CD8+ TILs was decreased, indicating a reduced accumulation of T cells with a severely exhausted phenotype in PDPN cKO mice14 (Fig. 2i). Consistent with previous data15, PDPN-deficient PD-1+TIM-3+ CD8+ TILs had higher expression of IL-7RA, indicating that PDPN may limit the survival of CD8+ TILs in the tumour microenvironment (Extended Data Fig. 5e, f).
We identified the transcription factor PRDM1 as a candidate regulator of the co-inhibitory module. PRDM1 is induced in vitro by IL-27 in CD4+ and CD8+ T cells (Extended Data Fig. 6a), is enriched in TILs with high expression of the IL-27 co-inhibitory module (Extended Data Figs. 3c-f, 6b, c, Methods), and is overexpressed in exhausted CD8+ TILs (P = 0.0004, t-test, Extended Data Fig. 6d). Network analysis based on profiling of naive CD8+ T cells from mice with a T cell-specific deletion of PRDM1 (PRDM1 cKO) stimulated with IL-27, showed that PRDM1 regulates several genes in the IL-27 co-inhibitory module (Extended Data Fig. 6e, P = 2.32 × 10-12; hypergeometric test; Methods). This was further supported by PRDM1 chromatin immunoprecipitation followed by sequencing (ChIP-seq) data16 (P = 2.9 × 10-8, hypergeometric test; Extended Data Fig. 6e, Methods).
CD8+ TILs from B16F10 tumour-bearing PRDM1 cKO mice expressed lower levels of TIM-3, PD-1 and PROCR (Fig. 3a); however, there was no difference in tumour growth compared to wildtype controls (Fig. 3b), indicating that the reduction of co-inhibitory receptor expression in PRDM1 cKO mice was insufficient to promote effective anti-tumour immunity. We therefore examined whether other transcription factors may regulate the co-inhibitory module and compensate for the absence of PRDM1. We analysed CD8+ TILs from PRDM1 cKO mice for the expression of genes from the IL-27-driven gene signature and the signature for exhausted CD8+ TILs (Methods, Supplementary Table 3). We found that only a few genes were upregulated in PRDM1 cKO CD8+ T cells, including one transcription factor, c-MAF (P < 0.05; Fig. 3c). Indeed, c-MAF is induced by IL-27, is coexpressed with PRDM1 in T cells after IL-27 stimulation (Extended Data Fig. 6a), and can regulate IL-10 expression17 and T cell exhaustion18. In addition, many genes (226 genes, P = 5.34 × 10-5, hypergeometric test) in the co-inhibitory gene module have a binding motif and a reported binding event for c-MAF within their promoter regions19.
CD8+ TILs from c-MAF cKO mice exhibited decreased expression of several co-inhibitory receptors (Fig. 3d). PRDM1 and c-MAF each affected co-inhibitory receptor expression only partially (Fig. 3e). As in the PRDM1 cKO mice, c-MAF cKO mice did not show any differences in tumour growth relative to controls (Fig. 3f). Notably, PRDM1 expression in c-MAF cKO TILs was similar to that in wild-type TILs, indicating that PRDM1 might drive the expression of the co-inhibitory gene module in the absence of c-MAF.
We addressed whether PRDM1 and c-MAF could act cooperatively to regulate co-inhibitory receptor expression. We found no evidence for a physical interaction between PRDM1 and c-MAF (data not shown); we therefore examined whether they shared targets. We combined the network analysis for PRDM1 (Extended Data Fig. 6e) with c-MAF ChIP-seq data19 and c-MAF targets (Methods). We observed 121 genes in the co-inhibitory module that are affected (RNA-seq) or have a direct binding event (ChIP-seq) for both PRDM1 and c-MAF (Fig. 4a), but that are not affected in either individual knockout. This is consistent, among other possibilities, with compensatory (for example, 'OR' gate logic) regulation20. Examination of ATAC-seq (assay for transposase-accessible chromatin using sequencing)21,22 and ChIP- seq data for PD-1, TIM-3, LAG-3 and TIGIT shows that PRDM1 and c-MAF can bind both overlapping and non-overlapping sites in the loci of these receptors and can synergistically trans-activate TIM-3 expression (Extended Data Fig. 7).
Mice with a T cell-specific deletion in both PRDM1 and c-MAF (PRDM1/c-MAF conditional double-knockout (cDKO)) showed normal development of CD4+ and CD8+ T cells in terms of frequency and expression of memory or activation markers, although the frequency of FOXP3+ Treg cells was increased (Extended Data Fig. 8a). CD4+ and CD8+ TILs from cDKO mice bearing B16F10 melanomas exhibited a near absence of PD-1, TIM-3, LAG-3, TIGIT, PDPN and PROCR expression (Fig. 4b, Extended Data Fig. 8b). Moreover, cDKO CD8+ TILs exhibited enhanced IL-2 and TNF production (Extended Data Fig. 8c). In contrast to singly deficient mice, cDKO mice showed significant control of B16F10 tumour growth despite the increased frequency of Treg cells (Fig. 4c). We addressed whether PRDM1 and c-MAF have a cell-intrinsic role in CD8+ and CD4+ T cells in controlling tumour growth by using an adoptive transfer model. Although CD8+ T cells from cDKO were able to inhibit tumour growth with decreased expression of co-inhibitory molecules, these effects were stronger when PRDM1 and c-MAF were lacking in both CD4+ and CD8+ T cells (Fig. 4d, Extended Data Fig. 8d). We examined the roles of PRDM-1 and c-MAF in tumour antigen-specific T cell responses using the MC38-OVA tumour model. We observed a significant reduction in tumour growth in mice receiving cDKO T cells as compared to mice receiving wild-type T cells (Extended Data Fig. 8e). We also observed an increase in ovalbumin (OVA)-specific T cells in the tumour draining lymph nodes and in OVA-specific IFN-γ- and TNF-producing CD8+ T cells in both the tumour infiltrate and the periphery in mice receiving double-knockout T cells (Fig. 4e, f, Extended Data Fig. 8f). Lastly, we observed an increase in CD8+Ki67+ T cells in the periphery of mice receiving double-knockout T cells (Fig. 4f).
We tested for non-additive effects between PRDM1 and c-MAF by using a binomial generalized linear model to compare the effect of single knockouts to the cDKO, and found that 149 out of 940 differentially expressed genes (adjusted P < 0.05, likelihood ratio test and false discovery rate (FDR) correction) between wild-type and cDKO CD8+ TILs have non-additive (that is, synergistic) effects (Extended Data Fig. 9, Methods).
Examination of the transcriptional signatures of cDKO CD8+ TILs showed significant overlap with those of CD8+TIM-3-PD-1- TILs (Fig. 4g, P = 2.8 × 10-7, one-sample Kolmogorov-Smirnov test; Extended Data Fig. 10a-c), suggesting that the loss of both c-MAF and PRDM1 increases the proportion of non-exhausted CD8+ effectors that exist normally in tumours. We scored the individual scRNA-seq profiles of CD8+ TILs for the cDKO 940 gene signature and found that the expression of the cDKO gene signature and the co-inhibitory gene module signature mark mutually exclusive populations of TILs (Extended Data Fig. 10e). The cDKO signature showed significant overlap with PD-1+CXCR5+CD8+ T cells, which may represent precursors for functional effectors in chronic lymphocytic choriomeningitis virus (LCMV) infection23 (Extended Data Fig. 10d, e, P = 1 × 10-13, one-sample Kolmogorov-Smirnov test). Furthermore, the IL-27RA-knockout TIL signature also showed significant overlap with this PD-1+CXCR5+CD8+ T cell signature (P < 2.2 × 10-16, one-sample Kolmogorov-Smirnov test; Fig. 2a, Extended Data Fig. 10e). Collectively, our data indicate that the loss of c-MAF and PRDM1 preferentially results in loss of the co-inhibitory gene module expression and acquisition of a more responsive effector T cell state.
In conclusion, we identified a co-inhibitory gene module, which is expressed in several settings of both CD4+ and CD8+ T cell nonresponsiveness, along with its transcriptional regulators. The discovery of this module provides a basis for the identification of novel co-inhibitory and co-stimulatory receptors that may have an important role in T cell regulation.
Online content
Any Methods, including any statements of data availability and Nature Research reporting summaries, along with any additional references and Source Data files, are available in the online version of the paper at https://doi.org/10.1038/s41586- 018-0206-z.
Received: 16 August 2016; Accepted: 27 April 2018;
Published online 13 June 2018.
1. Wherry, E. J. & Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 15, 486-499 (2015).
2. Anderson, A. C., Joller, N. & Kuchroo, V. K. Lag-3, Tim-3, and TIGIT: co-inhibitory receptors with specialized functions in immune regulation. Immunity 44, 989-1004 (2016).
3. Singer, M. et al. A distinct gene module for dysfunction uncoupled from activation in tumor-infiltrating T cells. Cell 166, 1500-1511 (2016).
4. Maaten, L. H. G. Visualizing data using t-SNE. J. Mach. Learn. Res. 9, 2579-2605 (2008).
5. Fitzgerald, D. C. et al. Suppression of autoimmune inflammation of the central nervous system by interleukin 10 secreted by interleukin 27-stimulated T cells. Nat. Immunol. 8, 1372-1379 (2007).
6. Awasthi, A. et al. A dominant function for interleukin 27 in generating interleukin 10-producing anti-inflammatory T cells. Nat. Immunol. 8, 1380-1389 (2007).
7. Stumhofer, J. S. et al. Interleukins 27 and 6 induce STAT3-mediated T cell production of interleukin 10. Nat. Immunol. 8, 1363-1371 (2007).
8. Zhu, C. et al. An IL-27/NFIL3 signalling axis drives Tim-3 and IL-10 expression and T-cell dysfunction. Nat. Commun. 6, 6072 (2015).
9. Hirahara, K. et al. Interleukin-27 priming of T cells controls IL-17 production in trans via induction of the ligand PD-L1. Immunity 36, 1017-1030 (2012).
10. Doering, T. A. et al. Network analysis reveals centrally connected genes and pathways involved in CD8+ T cell exhaustion versus memory. Immunity 37, 1130-1144 (2012).
11. Burton, B. R. et al. Sequential transcriptional changes dictate safe and effective antigen-specific immunotherapy. Nat. Commun. 5, 4741 (2014).
12. Mayo, L. et al. IL-10-dependent Tr1 cells attenuate astrocyte activation and ameliorate chronic central nervous system inflammation. Brain 139, 1939-1957 (2016).
13. Castellino, F. J. et al. Mice with a severe deficiency of the endothelial protein C receptor gene develop, survive, and reproduce normally, and do not present with enhanced arterial thrombosis after challenge. Thromb. Haemost. 88, 462-472 (2002).
14. Sakuishi, K. et al. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J. Exp. Med. 207, 2187-2194 (2010).
15. Peters, A. et al. Podoplanin negatively regulates CD4+ effector T cell responses. J. Clin. Invest. 125, 129-140 (2015).
16. Mackay, L. K. et al. Hobit and Blimp1 instruct a universal transcriptional program of tissue residency in lymphocytes. Science 352, 459-463 (2016).
17. Apetoh, L. et al. The aryl hydrocarbon receptor interacts with c-Maf to promote the differentiation of type 1 regulatory T cells induced by IL-27. Nat. Immunol. 11, 854-861 (2010).
18. Giordano, M. et al. Molecular profiling of CD8 T cells in autochthonous melanoma identifies Maf as driver of exhaustion. EMBO J. 34, 2042-2058 (2015).
19. Ciofani, M. et al. A validated regulatory network for Th17 cell specification. Cell 151, 289-303 (2012).
20. Capaldi, A. P. et al. Structure and function of a transcriptional network activated by the MAPK Hog1. Nat. Genet. 40, 1300-1306 (2008).
21. Karwacz, K. et al. Critical role of IRF1 and BATF in forming chromatin landscape during type 1 regulatory cell differentiation. Nat. Immunol. 18, 412-421 (2017).
22. Sen, D. R. et al. The epigenetic landscape of T cell exhaustion. Science 354, 1165-1169 (2016).
23. Im, S. J. et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417-421 (2016).
24. Chen, L. & Flies, D. B. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat. Rev. Immunol. 13, 227-242 (2013).
25. Wende, H. et al. The transcription factor c-Maf controls touch receptor development and function. Science 335, 1373-1376 (2012).
26. Smyth, G. K. Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat. Appl. Genet. Mol. Biol. 3, Article3 (2004).
27. Reich, M. et al. GenePattern 2.0. Nat. Genet. 38, 500-501 (2006).
28. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189-196 (2016).
29. Trapnell, C., Pachter, L. & Salzberg, S. L. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25, 1105-1111 (2009).
30. Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics 12, 323 (2011).
31. Anders, S. & Huber, W. Differential expression analysis for sequence count data. Genome Biol. 11, R106 (2010).
32. Shalek, A. K. et al. Single-cell transcriptomics reveals bimodality in expression and splicing in immune cells. Nature 498, 236-240 (2013).
33. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protocols 9, 171-181 (2014).
34. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memoryefficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).
35. Shekhar, K. et al. Comprehensive classification of retinal bipolar neurons by single-cell transcriptomics. Cell 166, 1308-1323 (2016).
36. Johnson, W. E., Li, C. & Rabinovic, A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics 8, 118-127 (2007).
37. Blondel, V. D., Guillaume, J. L., Lambiotte, R. & Lefebvre, E. Fast unfolding of communities in large networks. J. Stat. Mech. https://doi.org/10.1088/1742- 5468/2008/10/P10008 (2008).
38. Levine, J. H. et al. Data-driven phenotypic dissection of AML reveals progenitorlike cells that correlate with prognosis. Cell 162, 184-197 (2015).
39. Smyth, G. K. in Bioinformatics and Computational Biology Solutions using R and Bioconductor. Statistics for Biology and Health (eds. Gentleman, R. et al.) 397-420 (Springer, New York, 2005).
40. Davis, S. & Meltzer, P. S. GEOquery: a bridge between the Gene Expression Omnibus (GEO) and Bi°Conductor. Bioinformatics 23, 1846-1847 (2007).
41. Lopes, C. T. et al. Cytoscape Web: an interactive web-based network browser. Bioinformatics 26, 2347-2348 (2010).
Copyright Nature Publishing Group Jun 21, 2018