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.

  • Protocol
  • Published:

INVADEseq to identify cell-adherent or invasive bacteria and the associated host transcriptome at single-cell-level resolution

Abstract

Single-cell RNA sequencing (scRNAseq) technologies have been beneficial in revealing and describing cellular heterogeneity within mammalian tissues, including solid tumors. However, many of these techniques apply poly(A) selection of RNA, and thus have primarily focused on determining the gene signatures of eukaryotic cellular components of the tumor microenvironment. Microbiome analysis has revealed the presence of microbial ecosystems, including bacteria and fungi, within human tumor tissues from major cancer types. Imaging data have revealed that intratumoral bacteria may be located within epithelial and immune cell types. However, as bacterial RNA typically lacks a poly(A) tail, standard scRNAseq approaches have limited ability to capture this microbial component of the tumor microenvironment. To overcome this, we describe the invasion–adhesion-directed expression sequencing (INVADEseq) approach, whereby we adapt 10x Genomics 5′ scRNAseq protocol by introducing a primer that targets a conserved region of the bacterial 16S ribosomal RNA gene in addition to the standard primer for eukaryotic poly(A) RNA selection. This ‘add-on’ approach enables the generation of eukaryotic and bacterial DNA libraries at eukaryotic single-cell level resolution, utilizing the 10x barcode to identify single cells with intracellular bacteria. The INVADEseq method takes 30 h to complete, including tissue processing, sequencing and computational analysis. As an output, INVADEseq has shown to be a reliable tool in human cancer cell lines and patient tumor specimens by detecting the proportion of human cells that harbor bacteria and the identities of human cells and intracellular bacteria, along with identifying host transcriptional programs that are modulated on the basis of associated bacteria.

Key points

  • Invasion–adhesion-directed expression sequencing uses a modified 10x Genomics 5′ single-cell RNA sequencing protocol, introducing a primer targeting the bacterial 16S ribosomal RNA gene as well as the standard primer for eukaryotic poly(A) RNA selection to identify cell-associated bacteria and the host transcriptome.

  • Invasion–adhesion-directed expression thus overcomes the limited ability of standard single-cell RNA sequencing approaches to capture the microbial component of the tumor microenvironment, facilitating analysis of host–bacterial interactions at the single-cell 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: Tumor processing for single-cell RNAseq acquisition and computational pipeline for host and bacteria cell annotations, host-associated transcriptome and GSEA pathway enrichment analysis.
Fig. 2: INVADEseq specificity and bacterial 16S rRNA gene enrichment libraries following cDNA amplification using a nested bacterial 16S primer.
Fig. 3: Single-cell RNAseq analysis of a OSCC tumor sample using the INVADEseq method.
Fig. 4: Single-cell RNAseq analysis in specific cell clusters from OSCC tumor samples containing a relatively high load of cell-associated bacteria (High_Bac cohort) following data integration.
Fig. 5: Single-cell RNAseq analysis in specific cell clusters from two OSCC tumor samples containing low bacteria load (Low_Bac) following data integration.

Similar content being viewed by others

Data availability

Raw sequences data from INVADEseq bacterial 16S rRNA and human (GEX) gene libraries are available in the NCBI Sequence Read Archive repository under the Bioproject accession number PRJNA811533.

Code availability

Code for data processing and analysis of single-cell RNAseq data is available at https://github.com/FredHutch/Galeano-Nino-Bullman-Intratumoral-Microbiota_2022.

References

  1. Anderson, N. M. & Simon, M. C. The tumor microenvironment. Curr. Biol. 30, R921–R925 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Kostic, A. D. et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe 14, 207–215 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Nejman, D. et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science 368, 973–980 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Narunsky-Haziza, L. et al. Pan-cancer analyses reveal cancer-type-specific fungal ecologies and bacteriome interactions. Cell 185, 3789–3806 e3717 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Dohlman, A. B. et al. A pan-cancer mycobiome analysis reveals fungal involvement in gastrointestinal and lung tumors. Cell 185, 3807–3822 e3812 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Bullman, S. et al. Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science 358, 1443–1448 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Kalaora, S. et al. Identification of bacteria-derived HLA-bound peptides in melanoma. Nature 592, 138–143 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Galeano Nino, J. L. et al. Effect of the intratumoral microbiota on spatial and cellular heterogeneity in cancer. Nature 611, 810–817 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Chen, Z. et al. Single-cell RNA sequencing highlights the role of inflammatory cancer-associated fibroblasts in bladder urothelial carcinoma. Nat. Commun. 11, 5077 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Yeo, A. T. et al. Single-cell RNA sequencing reveals evolution of immune landscape during glioblastoma progression. Nat. Immunol. 23, 971–984 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Joanito, I. et al. Single-cell and bulk transcriptome sequencing identifies two epithelial tumor cell states and refines the consensus molecular classification of colorectal cancer. Nat. Genet. 54, 963–975 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Jovic, D. et al. Single-cell RNA sequencing technologies and applications: a brief overview. Clin. Transl. Med. 12, e694 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Hajnsdorf, E. & Kaberdin, V. R. RNA polyadenylation and its consequences in prokaryotes. Philos. Trans. R Soc. Lond. B https://doi.org/10.1098/rstb.2018.0166 (2018).

  14. Bullman, S. INVADEseq to study the intratumoural microbiota at host single-cell resolution. Nat. Rev. Cancer 23, 189 (2023).

    Article  CAS  PubMed  Google Scholar 

  15. Poore, G. D. et al. Microbiome analyses of blood and tissues suggest cancer diagnostic approach. Nature 579, 567–574 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. James, K. R., Elmentaite, R., Teichmann, S. A. & Hold, G. L. Correction to: redefining intestinal immunity with single-cell transcriptomics. Mucosal. Immunol. 15, 526 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Jaeger, N. et al. Single-cell analyses of Crohn’s disease tissues reveal intestinal intraepithelial T cells heterogeneity and altered subset distributions. Nat. Commun. 12, 1921 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Blaalid, R. et al. ITS1 versus ITS2 as DNA metabarcodes for fungi. Mol. Ecol. Resour. 13, 218–224 (2013).

    Article  CAS  PubMed  Google Scholar 

  19. Ihrmark, K. et al. New primers to amplify the fungal ITS2 region—evaluation by 454-sequencing of artificial and natural communities. FEMS Microbiol. Ecol. 82, 666–677 (2012).

    Article  CAS  PubMed  Google Scholar 

  20. Wang, X., He, Y., Zhang, Q., Ren, X. & Zhang, Z. Direct comparative analyses of 10X Genomics Chromium and Smart-seq2. Genomics Proteom. Bioinforma. 19, 253–266 (2021).

    Article  CAS  Google Scholar 

  21. Dreyfus, M. & Regnier, P. The poly(A) tail of mRNAs: bodyguard in eukaryotes, scavenger in bacteria. Cell 111, 611–613 (2002).

    Article  CAS  PubMed  Google Scholar 

  22. Ghaddar, B. et al. Tumor microbiome links cellular programs and immunity in pancreatic cancer. Cancer Cell 40, 1240–1253 e1245 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Jin, C. et al. Commensal microbiota promote lung cancer development via gammadelta T cells. Cell 176, 998–1013 e1016 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Imdahl, F., Vafadarnejad, E., Homberger, C., Saliba, A. E. & Vogel, J. Single-cell RNA-sequencing reports growth-condition-specific global transcriptomes of individual bacteria. Nat. Microbiol. 5, 1202–1206 (2020).

    Article  CAS  PubMed  Google Scholar 

  25. Johnston, C. D. & Bullman, S. The tumour-associated microbiome. Nat. Rev. Gastroenterol. Hepatol. 19, 347–348 (2022).

    Article  PubMed  Google Scholar 

  26. Haas, B. J., Chin, M., Nusbaum, C., Birren, B. W. & Livny, J. How deep is deep enough for RNA-seq profiling of bacterial transcriptomes? BMC Genomics 13, 734 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Salter, S. J. et al. Reagent and laboratory contamination can critically impact sequence-based microbiome analyses. BMC Biol. 12, 87 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Biesbroek, G. et al. Deep sequencing analyses of low density microbial communities: working at the boundary of accurate microbiota detection. PLoS ONE 7, e32942 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Davis, N. M., Proctor, D. M., Holmes, S. P., Relman, D. A. & Callahan, B. J. Simple statistical identification and removal of contaminant sequences in marker-gene and metagenomics data. Microbiome 6, 226 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Jervis-Bardy, J. et al. Deriving accurate microbiota profiles from human samples with low bacterial content through post-sequencing processing of Illumina MiSeq data. Microbiome 3, 19 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Aran, D. et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat. Immunol. 20, 163–172 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Wu, Y. E., Pan, L., Zuo, Y., Li, X. & Hong, W. Detecting activated cell populations using single-cell RNA-seq. Neuron 96, 313–329 e316 (2017).

    Article  CAS  PubMed  Google Scholar 

  34. Kennedy, K. M. et al. Questioning the fetal microbiome illustrates pitfalls of low-biomass microbial studies. Nature 613, 639–649 (2023).

    Article  CAS  PubMed  Google Scholar 

  35. LaCourse, K. D. et al. The cancer chemotherapeutic 5-fluorouracil is a potent Fusobacterium nucleatum inhibitor and its activity is modified by intratumoral microbiota. Cell Rep. 41, 111625 (2022).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This research was supported by the Genomics and Bioinformatics Shared Resource of the Fred Hutch/University of Washington Cancer Consortium (P30 CA015704) and the Scientific Computing Infrastructure at Fred Hutch funded by ORIP grant S10OD028685. Research reported in this publication was supported by the National Institute of Dental and Craniofacial Research of the National Institutes of Health under award number R01 DE027850 (to C.D.J.), the National Cancer Institute under award number R00 CA229984-03 (to S.B.), the Interdisciplinary Training Grant in Cancer T32 CA080416 (to J.L.G.N.) and the Cancer Research Institute Irvington Postdoctoral Fellowship (CRI Award CRI4208 to J.L.G.N.). Special thanks to H. Johnston for guidance and helpful discussions. We thank A. Baryiames for help with bacterial co-culture experiments. The use of patient specimens for this work was approved by the Fred Hutchinson Cancer Center Institutional Review Board under the following protocol numbers: RG 1006552 and 1006974.

Author information

Authors and Affiliations

Authors

Contributions

J.L.G.N., C.D.J. and S.B. designed the study and wrote the paper. H.W. performed the computational analysis of INVADEseq data, S.S.M., M.F. and H.S. provided computational guidance. K.D.L. performed INVADEseq experiments and C.S. performed library preparation and sequencing. K.D.L. performed bacteria and cell line co-cultures. J.L.G.N. and H.W. performed gene expression pathway analysis of INVADEseq data. J.L.G.N. and H.W. performed statical analysis. C.S., M.F., H.S., S.S.M., K.D.L. and H.W. provided edits to the paper. All authors contributed to the final version of the paper.

Corresponding authors

Correspondence to Christopher D. Johnston or Susan Bullman.

Ethics declarations

Competing interests

S.B. has consulted for GlaxoSmithKline and BiomX. C.D.J. has consulted for Series Therapeutics and Azitra. S.B. is an inventor on US Patent Application no: PCT/US2018/042966, submitted by the Broad Institute and Dana-Farber Cancer Institute, that covers targeting of Fusobacterium for treatment of CRC cancer.

Peer review

Peer review information

Nature Protocols thanks Wen Lu and Antoine-Emmanuel Saliba 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.

Related links

Key references using this protocol:

Galeano Niño, J. L. et al. Nature 611, 810–817 (2022): https://doi.org/10.1038/s41586-022-05435-0

Supplementary information

Supplementary Information

Supplementary Fig. 1.

Reporting Summary

Supplementary Table

Supplementary Table 1: scRNAseq: cell-associated microbiome composition in an individual OSCC patient. The table indicates the microbiome composition in a OSCC sample, including the number of UMI transcripts for each bacterium per cell. The relative abundance (%) for each bacterium is plotted in Fig. 3b. The data used to generate this table has been published previously8. Supplementary Table 2: scRNAseq: gene expression profile for an individual OSCC patient. All cluster analysis. Differential expression analysis between cellular groups as shown in Fig. 3c. The analysis was performed using the FindMarker function from the Seurat package based on the Wilcoxon rank sum test. The log2 fold change is calculated based on the difference in the gene expression average between two groups. Adjusted P values were calculated using the Bonferroni correction test for each gene in the dataset. P values <0.05 indicate significant differential gene regulation between experimental conditions. The data used to generate this table have been published previously8. Supplementary Table 3: scRNAseq: GSEA for an individual OSCC patient. All cluster analysis. GSEA between cellular groups as shown in Fig. 3d. The analysis was performed using the FindMarker function from the Seurat package based on the Wilcoxon rank sum test. The log2 fold change is calculated based on the difference in the gene expression average between two groups. Adjusted P values were calculated using the Bonferroni correction test for each gene in the dataset. P values <0.05 indicate significant differential gene regulation between experimental conditions. The data used to generate this table have been published previously8. Supplementary Table 4: scRNAseq. cell-associated microbiome composition from the high bacteria load OSCC cohort (High_Bac). The table indicates the microbiome composition in samples from the High_Bac cohort including the number of UMI transcripts for each bacterium. Relative abundance for each bacterium was plotted in Fig. 4b. The data used to generate this table have been published previously8. Supplementary Table 5: scRNAseq: gene expression profile in specific cell clusters from the high bacteria load OSCC cohort (High_Bac). Integrated data. Differential expression analysis between cellular groups as shown in Fig. 4c. The analysis was performed using the FindMarker function from the Seurat package based on the Wilcoxon rank sum test. The log2 fold change is calculated based on the difference in the gene expression average between two groups. Adjusted P values were calculated using the Bonferroni correction test for each gene in the dataset. P values <0.05 indicate significant differential gene regulation between experimental conditions. The data used to generate this table have been published previously8. Supplementary Table 6: scRNAseq: GSEA for specific cell clusters from the high bacteria load OSCC cohort (High_Bac). Integrated data. GSEA between cellular groups as shown in Fig. 4d. The analysis was performed using the FindMarker function from the Seurat package based on the Wilcoxon Rank Sum test. The log2 fold change is calculated based on the difference in the gene expression average between two groups. Adjusted P values were calculated using the Bonferroni correction test for each gene in the dataset. P values <0.05 indicate significant differential gene regulation between experimental conditions. The data used to generate this table have been published previously8. Supplementary Table 7: scRNAseq: cell-associated microbiome composition from the low bacterial load OSCC cohort (Low_Bac). The table indicates the microbiome composition in samples from the Low_Bac cohort including the number of UMI transcripts for each bacterium. The relative abundance (%) for each bacterium was plotted in Fig. 5b. The data used to generate this table have been published previously8. Supplementary Table 8: scRNAseq: gene expression profile in specific cell clusters from the low bacteria load OSCC cohort (Low_Bac). Integrated data. Differential expression analysis between cellular groups as shown in Fig. 5c. The analysis was performed using the FindMarker function from the Seurat package based on the Wilcoxon rank sum test. The log2 fold change is calculated based on the difference in the gene expression average between two groups. Adjusted P values were calculated using the Bonferroni correction test for each gene in the dataset. P values <0.05 indicate significant differential gene regulation between experimental conditions. The data used to generate this table have been published previously8.

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

Galeano Niño, J.L., Wu, H., LaCourse, K.D. et al. INVADEseq to identify cell-adherent or invasive bacteria and the associated host transcriptome at single-cell-level resolution. Nat Protoc 18, 3355–3389 (2023). https://doi.org/10.1038/s41596-023-00888-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-023-00888-7

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Cancer

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

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer