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:

Spatial transcriptome uncovers rich coordination of metabolism in E. coli K12 biofilm

Abstract

Microbial communities often display region-specific properties, which give rise to complex interactions and emergent behaviors that are critical to the homeostasis and stress response of the communities. However, systems-level understanding of these properties still remains elusive. In this study, we established RAINBOW-seq and profiled the transcriptome of Escherichia coli biofilm communities with high spatial resolution and high gene coverage. We uncovered three modes of community-level coordination, including cross-regional resource allocation, local cycling and feedback signaling, which were mediated by strengthened transmembrane transport and spatially specific activation of metabolism. As a consequence of such coordination, the nutrient-limited region of the community maintained an unexpectedly high level of metabolism, enabling it to express many signaling genes and functionally unknown genes with potential sociality functions. Our work provides an extended understanding of the metabolic interplay in biofilms and presents a new approach of investigating complex interactions in bacterial communities on the systems level.

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: Development and validation of RAINBOW-seq.
Fig. 2: Spatial transcriptome of E. coli biofilm.
Fig. 3: Strengthened transmembrane transport process and its landscape within bacterial community.
Fig. 4: Cross-regional allocation of arginine from biofilm periphery to interior.
Fig. 5: Polyamine-mediated feedback signaling in the biofilm.
Fig. 6: Local cycling of nucleotide and metabolic division of labor at biofilm periphery.

Similar content being viewed by others

Data availability

The raw data were deposited to the Gene Expression Omnibus (GSE197541 for RAINBOW-seq data and GSE213531 for RNA-seq ΔsapD biofilm periphery and pyrimidine biosynthesis heterogeneity). Other materials that support the findings of this study are available upon reasonable request. Source data are provided with this paper.

Code availability

MATLAB codes for RAINBOW-seq (image and FACS data processing) have been deposited to GitHub (https://github.com/Shenpinggg/RainbowSeq.git). The core scripts to process raw NGS data in this work have also been deposited to GitHub (https://github.com/wtmbiohacker/miniBac-seq.git).

References

  1. Flemming, H.-C. & Wuertz, S. Bacteria and archaea on Earth and their abundance in biofilms. Nat. Rev. Microbiol. 17, 247–260 (2019).

    CAS  PubMed  Google Scholar 

  2. Arnaouteli, S., Bamford, N. C., Stanley-Wall, N. R. & Kovács, Á. T. Bacillus subtilis biofilm formation and social interactions. Nat. Rev. Microbiol. 19, 600–614 (2021).

    CAS  PubMed  Google Scholar 

  3. Nadell, C. D., Drescher, K. & Foster, K. R. Spatial structure, cooperation and competition in biofilms. Nat. Rev. Microbiol. 14, 589–600 (2016).

    CAS  PubMed  Google Scholar 

  4. Liu, W., Cremer, J., Li, D., Hwa, T. & Liu, C. An evolutionarily stable strategy to colonize spatially extended habitats. Nature 575, 664–668 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Yan, J., Sharo, A. G., Stone, H. A., Wingreen, N. S. & Bassler, B. L. Vibrio cholerae biofilm growth program and architecture revealed by single-cell live imaging. Proc. Natl Acad. Sci. USA 113, E5337–E5343 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Stewart, P. S. & Franklin, M. J. Physiological heterogeneity in biofilms. Nat. Rev. Microbiol. 6, 199–210 (2008).

    CAS  PubMed  Google Scholar 

  7. Jo, J., Price-Whelan, A. & Dietrich, L. E. P. Gradients and consequences of heterogeneity in biofilms. Nat. Rev. Microbiol. 20, 593–607 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Wessel, A. K. et al. Oxygen limitation within a bacterial aggregate. MBio 5, e00992 (2014).

    PubMed  PubMed Central  Google Scholar 

  9. Blair, K. M., Turner, L., Winkelman, J. T., Berg, H. C. & Kearns, D. B. A molecular clutch disables flagella in the Bacillus subtilis biofilm. Science 320, 1636–1638 (2008).

    CAS  PubMed  Google Scholar 

  10. Schreiber, F. et al. Phenotypic heterogeneity driven by nutrient limitation promotes growth in fluctuating environments. Nat. Microbiol. 1, 16055 (2016).

    CAS  PubMed  Google Scholar 

  11. Liu, J. et al. Metabolic co-dependence gives rise to collective oscillations within biofilms. Nature 523, 550–554 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Prindle, A. et al. Ion channels enable electrical communication in bacterial communities. Nature 527, 59–63 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Yan, J. & Bassler, B. L. Surviving as a community: antibiotic tolerance and persistence in bacterial biofilms. Cell Host Microbe 26, 15–21 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Schiessl, K. T. et al. Phenazine production promotes antibiotic tolerance and metabolic heterogeneity in Pseudomonas aeruginosa biofilms. Nat. Commun. 10, 762 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Chiang, W. C. et al. Extracellular DNA shields against aminoglycosides in Pseudomonas aeruginosa biofilms. Antimicrob. Agents Chemother. 57, 2352–2361 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Tseng, B. S. et al. The extracellular matrix protects Pseudomonas aeruginosa biofilms by limiting the penetration of tobramycin. Environ. Microbiol. 15, 2865–2878 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Costerton, J. W., Stewart, P. S. & Greenberg, E. P. Bacterial biofilms: a common cause of persistent infections. Science 284, 1318–1322 (1999).

    CAS  PubMed  Google Scholar 

  18. Azimi, S., Lewin, G. R. & Whiteley, M. The biogeography of infection revisited. Nat. Rev. Microbiol. 20, 579–592 (2022).

    CAS  PubMed  Google Scholar 

  19. Whiteley, M. et al. Gene expression in Pseudomonas aeruginosa biofilms. Nature 413, 860–864 (2001).

    CAS  PubMed  Google Scholar 

  20. Yang, L., Nilsson, M., Gjermansen, M., Givskov, M. & Tolker-Nielsen, T. Pyoverdine and PQS mediated subpopulation interactions involved in Pseudomonas aeruginosa biofilm formation. Mol. Microbiol. 74, 1380–1392 (2009).

    CAS  PubMed  Google Scholar 

  21. Co, A. D., Van Vliet, S. & Ackermann, M. Emergent microscale gradients give rise to metabolic cross-feeding and antibiotic tolerance in clonal bacterial populations. Philos. Trans. R. Soc. B Biol. Sci. 374, 20190080 (2019).

    Google Scholar 

  22. Passos da Silva, D. et al. The Pseudomonas aeruginosa lectin LecB binds to the exopolysaccharide Psl and stabilizes the biofilm matrix. Nat. Commun. 10, 2183 (2019).

    PubMed  PubMed Central  Google Scholar 

  23. Srinivasan, S. et al. Matrix production and sporulation in Bacillus subtilis biofilms localize to propagating wave fronts. Biophys. J. 114, 1490–1498 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Nadezhdin, E., Murphy, N., Dalchau, N., Phillips, A. & Locke, J. C. W. Stochastic pulsing of gene expression enables the generation of spatial patterns in Bacillus subtilis biofilms. Nat. Commun. 11, 950 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Dar, D., Dar, N., Cai, L. & Newman, D. K. Spatial transcriptomics of planktonic and sessile bacterial populations at single-cell resolution. Science 373, eabi4882 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Heacock-Kang, Y. et al. Spatial transcriptomes within the Pseudomonas aeruginosa biofilm architecture. Mol. Microbiol. 106, 976–985 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Díaz-Pascual, F. et al. Spatial alanine metabolism determines local growth dynamics of Escherichia coli colonies. eLife 10, e70794 (2021).

    PubMed  PubMed Central  Google Scholar 

  28. Kuru, E. et al. In situ probing of newly synthesized peptidoglycan in live bacteria with fluorescent d-amino acids. Angew. Chem. Int. Ed. 51, 12519–12523 (2012).

    CAS  Google Scholar 

  29. Zhang, Y. et al. A microfluidic approach for quantitative study of spatial heterogeneity in bacterial biofilms. Small Sci. 2, 2200047 (2022).

    CAS  Google Scholar 

  30. Wang, T., Shen, P., Chai, R., He, Y. & Liu, J. Profiling of bacterial transcriptome from ultra-low input with MiniBac-seq. Environ. Microbiol. 24, 5774–5787 (2022).

    CAS  PubMed  Google Scholar 

  31. Beebout, C. J. et al. Respiratory heterogeneity shapes biofilm formation and host colonization in uropathogenic Escherichia coli. MBio 10, e02400–e02418 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Borriello, G. et al. Oxygen limitation contributes to antibiotic tolerance of Pseudomonas aeruginosa in biofilms. Antimicrob. Agents Chemother. 48, 2659–2664 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Flemming, H.-C. et al. Biofilms: an emergent form of bacterial life. Nat. Rev. Microbiol. 14, 563–575 (2016).

    CAS  PubMed  Google Scholar 

  34. Li, J. et al. Quorum sensing in Escherichia coli is signaled by AI-2/LsrR: effects on small RNA and biofilm architecture. J. Bacteriol. 189, 6011–6020 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Hengge, R. Principles of c-di-GMP signalling in bacteria. Nat. Rev. Microbiol. 7, 263–273 (2009).

    CAS  PubMed  Google Scholar 

  36. Ghatak, S., King, Z. A., Sastry, A. & Palsson, B. O. The y-ome defines the 35% of Escherichia coli genes that lack experimental evidence of function. Nucleic Acids Res. 47, 2446–2454 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Burrell, M., Hanfrey, C. C., Murray, E. J., Stanley-Wall, N. R. & Michael, A. J. Evolution and multiplicity of arginine decarboxylases in polyamine biosynthesis and essential role in Bacillus subtilis biofilm formation. J. Biol. Chem. 285, 39224–39238 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Thongbhubate, K., Nakafuji, Y., Matsuoka, R., Kakegawa, S. & Suzuki, H. Effect of spermidine on biofilm formation in Escherichia coli K-12. J. Bacteriol. 203, e00652–20 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Berg, N. I. et al. Ecological modelling approaches for predicting emergent properties in microbial communities. Nat. Ecol. Evol. 6, 855–865 (2022).

    PubMed  PubMed Central  Google Scholar 

  40. Artzi, L. et al. Dormant spores sense amino acids through the B subunits of their germination receptors. Nat. Commun. 12, 6842 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Robinson, C. D. et al. Host-emitted amino acid cues regulate bacterial chemokinesis to enhance colonization. Cell Host Microbe 29, 1221–1234 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Mills, E., Petersen, E., Kulasekara, B. R. & Miller, S. I. A direct screen for c-di-GMP modulators reveals a Salmonella Typhimurium periplasmic l-arginine-sensing pathway. Sci. Signal. 8, ra57 (2015).

    PubMed  Google Scholar 

  43. Bridges, A. A. & Bassler, B. L. Inverse regulation of Vibrio cholerae biofilm dispersal by polyamine signals. eLife 10, e65487 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Lee, H. H., Molla, M. N., Cantor, C. R. & Collins, J. J. Bacterial charity work leads to population-wide resistance. Nature 467, 82–85 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Wang, W. et al. Assessing the viability of transplanted gut microbiota by sequential tagging with d-amino acid-based metabolic probes. Nat. Commun. 10, 1317 (2019).

    PubMed  PubMed Central  Google Scholar 

  46. Datsenko, K. A. & Wanner, B. L. One-step inactivation of chromosomal genes in Escherichia coli K-12 using PCR products. Proc. Natl Acad. Sci. USA 97, 6640–6645 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Wang, T. et al. Dynamics of transcription–translation coordination tune bacterial indole signaling. Nat. Chem. Biol. 16, 440–449 (2020).

    CAS  PubMed  Google Scholar 

  48. Bittihn, P., Didovyk, A., Tsimring, L. S. & Hasty, J. Genetically engineered control of phenotypic structure in microbial colonies. Nat. Microbiol. 5, 697–705 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Kuru, E., Tekkam, S., Hall, E., Brun, Y. V. & Van Nieuwenhze, M. S. Synthesis of fluorescent d-amino acids and their use for probing peptidoglycan synthesis and bacterial growth in situ. Nat. Protoc. 10, 33–52 (2015).

    CAS  PubMed  Google Scholar 

  50. Hsu, Y.-P. et al. Full color palette of fluorescent d-amino acids for in situ labeling of bacterial cell walls. Chem. Sci. 8, 6313–6321 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Xu, M. et al. Structural insights into the regulatory mechanism of the Pseudomonas aeruginosa YfiBNR system. Protein Cell 7, 403–416 (2016).

    PubMed  PubMed Central  Google Scholar 

  52. Malone, J. G. et al. YfiBNR mediates cyclic di-GMP dependent small colony variant formation and persistence in Pseudomonas aeruginosa. PLoS Pathog. 6, e1000804 (2010).

    PubMed  PubMed Central  Google Scholar 

  53. Serra, D. O. & Hengge, R. A c-di-GMP-based switch controls local heterogeneity of extracellular matrix synthesis which is crucial for integrity and morphogenesis of Escherichia coli macrocolony biofilms. J. Mol. Biol. 431, 4775–4793 (2019).

    CAS  PubMed  Google Scholar 

  54. Hengge, R. High-specificity local and global c-di-GMP signaling. Trends Microbiol. 29, 993–1003 (2021).

    CAS  PubMed  Google Scholar 

  55. Schmidt, A. et al. The quantitative and condition-dependent Escherichia coli proteome. Nat. Biotechnol. 34, 104–110 (2015).

    PubMed  PubMed Central  Google Scholar 

  56. Zhang, Z. et al. From coarse to fine: the absolute Escherichia coli proteome under diverse growth conditions. Mol. Syst. Biol. 17, e9536 (2021).

    PubMed  PubMed Central  Google Scholar 

  57. Serra, D. O., Richter, A. M. & Hengge, R. Cellulose as an architectural element in spatially structured Escherichia coli biofilms. J. Bacteriol. 195, 5540–5554 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Gualdi, L. et al. Cellulose modulates biofilm formation by counteracting curli-mediated colonization of solid surfaces in Escherichia coli. Microbiology 154, 2017–2024 (2008).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank M. Asally, Y. Wang, G. Liang, J. Liu and X. Tan for helpful discussions; R. Chai for help with RNA extraction and library preparation; and B. Yu for assistance with cell sorting. J.L. was supported by the National Natural Science Foundation of China (32170099), the Tsinghua-Peking Center for Life Sciences and the Tsinghua University Independent Research Program (20197030008). T.W. was supported by the National Natural Science Foundation of China (32100071), the Chinese Postdoctoral Science Foundation (BX20190176 and 2020M670288) and the Tsinghua-Peking Center for Life Sciences. Y.Z. was supported by the National Natural Science Foundation of China (21908129), the Chinese Postdoctoral Science Foundation (2018M631481) and the Tsinghua-Peking Center for Life Sciences.

Author information

Authors and Affiliations

Authors

Contributions

T.W., P.S. and J.L. designed the research. T.W. and P.S. performed all experiments. Y.H. participated in validation experiments. Y.Z. contributed to the microfluidic aspect of the experiments. T.W., P.S., Y.H. and J.L. analyzed the data. T.W., P.S. and J.L. wrote the manuscript. All authors discussed the manuscript.

Corresponding author

Correspondence to Jintao Liu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks Maria Hadjifrangiskou and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 Molecular structures and fluorescent spectrums of FDAAs used in spatially resolved labeling of biofilm.

Molecular structures and fluorescent spectrums of FDAAs (Fluorescent D-alanine) used in spatially resolved labeling of biofilm. Shown in (a), (b) and (c) are molecular structures of HADA, NADA and TADA, respectively. The fluorescent spectrums of these three FDAAs are shown in (d); here dotted lines represent excitation spectrums and solid lines represent emission spectrums; the spectrum was measured by a microplate reader system (Tecan, Spark); FDAAs were at working concentration in M63B1 medium. (e) Labeling of FDAAs onto actively growing and dormant E. coli cells; here the seed culture was transferred to M63B1 medium with and without glucose to modulate the growth of the bacteria; and the FDAAs were added to stain the bacteria for three hours; shown is the merged image of phase contrast, CFP, GFP and mCherry channels under a 40× objective len; scale bar, 10 μm. Two biologically independent experiments were repeated with similar results.

Source data

Extended Data Fig. 2 Perturbation of FDAAs and cell sorting on bacterial transcriptome.

Perturbation of FDAAs and cell sorting on bacterial transcriptome. (a) Workflow to test the impact of FDAAs on E. coli transcriptome. (b) Comparison of transcriptomes when treated with control (DMSO) or FDAAs at working concentrations; shown here is the consistency of transcriptomes in terms of expression (log2(TPM + 1)) correlation (n = 4,508; Pearson’s r = 0.97 for 10 µM HADA, Pearson’s r = 0.95 for 10 µM NADA, Pearson’s r = 0.96 for 25 µM TADA, Pearson’s r = 0.97 for 25 µM CADA). (c) Workflow to test whether cell sorting perturbs E. coli transcriptome. (d) Agreement between control (non-sorting) and sorting group, shown is the consistency of transcriptomes in terms of expression (log2(TPM + 1)) correlation (n = 4,508; Pearson’s r = 0.98).

Source data

Extended Data Fig. 3 Sequential staining of the biofilm and the subsequent spatial segmentation according to the final fluorescent image.

Sequential staining of the biofilm and the subsequent spatial segmentation according to the final fluorescent image. (a) Fluorescence images of a biofilm after labeling at different stages. Shown are composite images of the biofilm (phase contrast; mCherry, red; GFP, green; CFP, blue) right after the sequential labeling by TADA, NADA and HADA; Scale bar is 200 µm. Note that TADA molecules were absorbed by the microfluidic chamber (PDMS) during labeling, giving rise to the background signal; Further cultivation using medium without TADA washed out these absorbed dyes and reduced the background signal to the normal level. Actually, that is the main reason we used TADA as the first dye in the sequential labeling. (b) Extracting the fluorescent signals for each pixel of the biofilm in microscopic image; the pixel-fluorescence matrix is presented in the three-dimensional fluorescent space. Next, k-means clustering is executed to assign each pixel to one particular group according to its fluorescent pattern; shown are clustering of all pixels into (c) 4, (d) 8, (e) 12, (f) 20 and (g) 30 groups. According to the clustering result for all pixels, we segmented the biofilm into (h) 4, (i) 8, (j) 12, (k) 20 and (l) 30 groups (scale bar: 500 μm). Note that the number of groups to be clustered (N) is a manual parameter in k-means clustering; when it is set below a threshold (N ≤ 20), the biofilm can be perfectly segmented into multiple concentric rings from the periphery to interior of biofilm—the major direction of spatial heterogeneity (as shown in (h), (i), (j) and (k). However, when N is set too big, the rainbow structure in biofilm segmentation will be disrupted, as shown in (l) N = 30. This gives rise to an upper limit (N = 20) of computational segment of stained biofilm, which further limits the spatial resolution of current version of RAINBOW-seq.

Source data

Extended Data Fig. 4 Overall workflow to sort bacterial cells.

Overall workflow to sort bacterial cells. (a) The framework of the experiment (orange step) and computation (green step) during cell sorting stage in RAINBOW-seq; here, the schematics of two core algorithms in automatic gating calculation is presented in (b) the optimal transport and (c) CART tree algorithm to partition the 3D fluorescent space. (b) In optimal transport algorithm, a distance matrix is firstly computed based on the distance between each cell and each pixel in high-dimensional fluorescent space; another assignment matrix is subsequently calculated to assign one cell to one and only one pixel in biofilm image, via minimizing the overall transport distance based on the abovementioned distance matrix. (c) Shown here is a simplified 2D space partition via CART tree algorithm; in RAINBOW-seq experiments reported in this work, 3D space is partitioned via CART tree algorithm based on the group label of each cell mapped from the previous optimal transport step. Shown in (d), (e), (f) and (g) are four real examples of final gates, each of them represents a gate for cells from a particular region in biofilm. In each example, two rectangle gates are combined to generate a final gate. The cells to be sorted in each example are highlighted by corresponding colors of the gate, while all other cells are labeled with grey. Counter-clockwise rotation of the gates and cells are presented for each example.

Extended Data Fig. 5 Overview of spatially resolved gene expressions participating in energy metabolism, biosynthesis, ribosome, pili and curli biogenesis.

Overview of spatially resolved gene expressions participating in (a) energy metabolism, (b) biosynthesis, (c) ribosome, (d) pili and (e) curli biogenesis. Shown is log2(fold change) relative to average of each profile; abbreviations: TCA, tricarboxylic acid cycle; glycerol-3P, glycerol-3-phosphate; PPP, pentose phosphate pathway (provides NADPH for biosynthesis).

Source data

Extended Data Fig. 6 Overview of spatially resolved gene expressions participating in amino acid catabolism, fatty acid catabolism, stress response and two-component signaling systems.

Overview of spatially resolved gene expressions participating in (a) amino acid catabolism, (b) fatty acid catabolism, (c) stress response and (d) two-component signaling systems. Shown is log2(fold change) relative to average of each profile.

Source data

Extended Data Fig. 7 Spatial profiles of AI-2-QS-related genes in biofilm.

Spatial profiles of AI-2-QS-related genes in biofilm. (a) AI-2-mediated QS in E. coli. (b) The log2(fold change) between biofilm periphery (n = 5) and exponential phase of planktonic culture (n = 2) are presented as heatmaps for AI-2 related genes. The spatial profile (n = 5; solid line and shaded area indicate mean ± std) of AI-2 (c) biosynthesis, (d) import and (e) processing in biofilm. (f) Utilization of secondary metabolites at intermediate region within biofilm. Shown is log2(fold change) relative to average of each profile; abbreviations: GABA, gamma-aminobutyric acid. See ‘Signaling mediated by AI-2 QS and c-di-GMP in biofilms uncovered by RAINBOW-seq’ subsection in Methods for more discussions about AI-2 quorum sensing in biofilm learned from RAINBOW-seq data.

Source data

Extended Data Fig. 8 Signaling mediated by c-di-GMP in E. coli biofilm.

Signaling mediated by c-di-GMP in E. coli biofilm. (a) Differential expression of genes encoding c-di-GMP processing enzymes in biofilm relative to planktonic bacteria; Shown is log2(fold change) between the maximum in biofilm (n = 24) and the maximum in planktonic culture (n = 14). (b) Bulk profiling cannot identify the local activation of c-di-GMP processing genes in biofilm. Shown is the log2(fold change) of genes encoding c-di-GMP processing enzymes in biofilm relative to planktonic lifestyle; a computationally synthetic biofilm transcriptome by averaging all 24 spatially resolved samples was generated to mimic a bulk biofilm sample, which was compared with the maximum from planktonic culture (n = 14). (c) Regulatory logic of PdeR/DgcM cascade via local c-di-GMP signaling to modulate MlrA activity, which drives the transcription of csgD; note that PdeR inhibits MlrA, either directly or indirectly via DgcM; and DgcM activates MlrA, either directly or indirectly via PdeR. (d) Gene expression levels of csgD at the interior of biofilm (n = 5) and the stationary phase of planktonic culture (n = 2). Data are presented as mean ± std (P = 0.0009, two tailed Mann-Whitley U test). (e) Different responses of DGCs and PDEs between biofilm and planktonic culture when stepping into famine from feast; x axis presents the log2(fold change) between famine (interior) and feast (periphery) in biofilm; y axis presents the log2(fold change) between famine (stationary stage) and feast (exponential stage) in planktonic culture; blue dots are genes encoding DGCs and red for PDEs; diagonal line highlights the equivalent feast-to-famine expression change between biofilm and planktonic culture; the region between y = x + 1 and y = x − 1 is shaded to highlight those outlier genes; these genes exhibit different expression change from feast to famine between biofilm and planktonic culture. (f) Above are the spatial expression profiles (log2(TPM + 1)) of dgcM and pdeR in biofilm; below are the temporal profiles of these two genes in planktonic culture. See ‘Signaling mediated by AI-2 QS and c-di-GMP in biofilms uncovered by RAINBOW-seq’ subsection in Methods for more discussions about c-di-GMP signaling in biofilm learned from RAINBOW-seq data.

Source data

Extended Data Fig. 9 Activation of y-genes in biofilm.

Activation of y-genes in biofilm. (a) Ratio of y-genes in four spatial patterns as shown in Fig. 2j. Y-genes are labeled as dark grey while others kept light grey; pattern 1: 162 y-genes out of 962 genes; pattern 2: 586 y-genes out of 1,702 genes; pattern 3: 61 y-genes out of 184 genes; pattern 4: 304 y-genes out of 823 genes. (b) The spatial expression profiles of 10 representative y-genes with ascending spatial patterns (n = 5; red line and shaded area indicate mean ± std); blue dot is the expression level at the stationary phase of planktonic culture (n = 2, error bar: mean ± std). (c) Activation of sociality protein domains carried by y-genes in biofilm; the fold of activation of the associated y-genes in biofilm relative to planktonic culture are presented as circles; The colors encode cellular locations; Genes previously shown to be related to biofilm formation are highlighted as squares. (d) Occurrence of the activated sociality Pfam-A domains of (c) in prokaryotic genomes. Occurrence are counted based on the distribution of these Pfam-A domains in relevant phyla of bacteria and archaea. Red color indicates that a given domain is present in at least one genome of the relevant class. (e) and (f) Genomic neighborhoods between representative y-genes activated in biofilm and genes of known sociality functions, aligned with the phylogenetic tree of relevant bacteria. For (e), 1: Betaproteobacteria, 2: Alphaproteobacteria, 3: Gammaproteobacteria, 3.1: Aeromonadales, 3.2: Pseudomonadales, 3.3: Enterobacterales, 3.3.1: Enterobacteriaceae; For (f), 1: Firmicutes, 1.1: Clostridia, 1.2: Bacilli, 1.3: Lactobacillales, 2: Proteobacteria, 2.1: Alphaproteobacteria, 2.2: Gammaproteobacteria. Genomic neighborhoods are identified from STRING database according to gene interaction scores. The homologues of y-genes are shown in red and neighborhood genes with known sociality functions are shown in blue; The color bars indicate the degree of identity between homologues in relevant bacteria and in E. coli K-12 MG1655, as calculated by BLASTP. Note that the dgcN homologue in S. flexneri is a pseudogene due to a premature stop codon; and yfcC’ in (f) indicates a paralog of yfcC in (e) encoded by E. coli K12 BW25113.

Source data

Extended Data Fig. 10 Polyamine metabolism and the spatial profiles of the relevant genes in biofilm.

Polyamine metabolism and the spatial profiles of the relevant genes in biofilm. (a) Schematics of intracellular metabolism and transmembrane transport for polyamines (putrescine and spermidine) in E. coli. Spatial profiles for (b) polyamine biosynthesis, (c) putrescine degradation and (d) spermidine export. Solid line and shaded area indicate mean ± std (n = 5).

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–13.

Reporting Summary

Supplementary Data

Supplementary Table 1: Biofilm information. Supplementary Table 2: y-genes list. Supplementary Table 3: Genes involved in amino acid utilization and synthesis. Supplementary Table 4: Oligos for rRNA depletion. Supplementary Table 5: Fitted spatial expression profiles of biofilm (log2(TPM + 1)). Supplementary Table 6: Transcriptome of biofilm and planktonic bacteria (TPM). Supplementary Table 7: Pattern of spatial expression profile. Supplementary Table 8: Transporter-coding genes list. Supplementary Table 9: Pfam-A domains of protein-coding y-genes. Supplementary Table 10: Sociality Pfam-A domain in biofilm. Supplementary Table 11: y-genes upregulated in biofilm. Supplementary Table 12: Genome and protein homology information used in phylogenetic analysis. Supplementary Table 13: Pattern-enriched Pfam-A domain of y-genes.

Source data

Source Data Fig. 1

Statistical Source Data

Source Data Fig. 1

Unprocessed image data

Source Data Fig. 2

Statistical Source Data

Source Data Fig. 3

Statistical Source Data

Source Data Fig. 4

Statistical Source Data

Source Data Fig. 4

Unprocessed image data

Source Data Fig. 5

Statistical Source Data

Source Data Fig. 5

Unprocessed image data

Source Data Fig. 6

Statistical Source Data

Source Data Fig. 6

Unprocessed image data

Source Data Extended Data Fig. 1

Statistical Source Data

Source Data Extended Data Fig. 2

Statistical Source Data

Source Data Extended Data Fig. 3

Unprocessed image Data

Source Data Extended Data Fig. 5

Statistical Source Data

Source Data Extended Data Fig. 6

Statistical Source Data

Source Data Extended Data Fig. 7

Statistical Source Data

Source Data Extended Data Fig. 8

Statistical Source Data

Source Data Extended Data Fig. 9

Statistical Source Data

Source Data Extended Data Fig. 10

Statistical Source Data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, T., Shen, P., He, Y. et al. Spatial transcriptome uncovers rich coordination of metabolism in E. coli K12 biofilm. Nat Chem Biol 19, 940–950 (2023). https://doi.org/10.1038/s41589-023-01282-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-023-01282-w

This article is cited by

Search

Quick links

Nature Briefing Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

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