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:

Visualization of metabolites and microbes at high spatial resolution using MALDI mass spectrometry imaging and in situ fluorescence labeling

Abstract

Label-free molecular imaging techniques such as matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI-MSI) enable the direct and simultaneous mapping of hundreds of different metabolites in thin sections of biological tissues. However, in host–microbe interactions it remains challenging to localize microbes and to assign metabolites to the host versus members of the microbiome. We therefore developed a correlative imaging approach combining MALDI-MSI with fluorescence in situ hybridization (FISH) on the same section to identify and localize microbial cells. Here, we detail metaFISH as a robust and easy method for assigning the spatial distribution of metabolites to microbiome members based on imaging of nucleic acid probes, down to single-cell resolution. We describe the steps required for tissue preparation, on-tissue hybridization, fluorescence microscopy, data integration into a correlative image dataset, matrix application and MSI data acquisition. Using metaFISH, we map hundreds of metabolites and several microbial species to the micrometer scale on a single tissue section. For example, intra- and extracellular bacteria, host cells and their associated metabolites can be localized in animal tissues, revealing their complex metabolic interactions. We explain how we identify low-abundance bacterial infection sites as regions of interest for high-resolution MSI analysis, guiding the user to a trade-off between metabolite signal intensities and fluorescence signals. MetaFISH is suitable for a broad range of users from environmental microbiologists to clinical scientists. The protocol requires ~2 work days.

Key points

  • A procedure for spatial metabolomics of host–microbe interactions, including tissue preparation, matrix application, MSI data acquisition, on-tissue hybridization using nucleic acid probes, fluorescence microscopy and data integration into a correlative image dataset.

  • MALDI-MSI enables single-cell-level mapping of metabolites by revealing their spatial distribution. Alternatively, laser-capture microdissection can be combined with LC–MS, or metaFISH combines spatial metabolomics with FISH.

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: Workflow for visualization of metabolites and microbes at high spatial resolution using MALDI–MSI and in situ fluorescence labeling (metaFISH).
Fig. 2: Combined analysis of spatial metabolome and microscopy data to associate metabolites to host or microbes.
Fig. 3: Trimming of CMC block before cryosectioning.
Fig. 4: Tissue section and microscopy slide documentation to follow the protocol.
Fig. 5: Laser intensity and shot number influence MSI and FISH signal intensity.
Fig. 6: Identifying bacterial infection sites for high-resolution MALDI–MSI.
Fig. 7: Metabolites co-localize with a patch of bacterial cells in earthworm tissue.
Fig. 8: metaFISH reveals metabolite distributions correlated with the presence of symbionts and parasites at 10 µm and 3 µm resolution.

Similar content being viewed by others

Data availability

All data presented in this paper have been deposited in the METASPACE project protocol (https://metaspace2020.eu/project/metaFISH). Individual datasets are deposited as follows: Fig. 1, MPIMM_193_QE_P_BC_CF (https://metaspace2020.eu/dataset/2019-11-28_11h08m15s); Fig. 5, 20210518_b_child_nix_s1_dhap_maldi2_tof_5um_laser90%_50shots (https://metaspace2020.eu/dataset/2021-07-08_13h54m26s), 20210518_b_child_nix_s1_dhap_laser70%_shots50 (https://metaspace2020.eu/dataset/2021-05-30_18h51m15s), 20210518_b_child_nix_s1_dhap_5um_maldi2_tof_laser50%_100 (https://metaspace2020.eu/dataset/2021-05-30_18h08m20s), 20210518_b_child_nix_s1_dhap_5um_maldi2_tof_laser30%_150 (https://metaspace2020.eu/dataset/2021-05-30_18h08m14s), and MPIMM_299_TTF_M2_Grid (https://metaspace2020.eu/dataset/2023-01-26_10h37m44s); Fig. 6, 20210706_bchild_nix_n25_tm_sl108 (https://metaspace2020.eu/dataset/2021-07-08_15h03m30s); Fig. 7, MTBLS2639; and Fig. 8, MPIMM_054_QE_P_BP_CF (https://metaspace2020.eu/dataset/2017-03-28_16h40m57s) and MPIMM_193_QE_P_BC_CF (https://metaspace2020.eu/dataset/2019-11-28_11h08m15s).

Code availability

Open-source scripts for the implementation of the MSI and microscopy co-registration have been published previously6 and are available on GitHub (R scripts, https://github.com/esogin/miniature-octo-fiesta; MATLAB, https://github.com/BenediktSenorDingDong/MALDI-FISHregistration).

References

  1. Moses, L. & Pachter, L. Museum of spatial transcriptomics. Nat. Methods 19, 534–546 (2022).

    CAS  PubMed  Google Scholar 

  2. Mund, A., Brunner, A.-D. & Mann, M. Unbiased spatial proteomics with single-cell resolution in tissues. Mol. Cell 82, 2335–2349 (2022).

    CAS  PubMed  Google Scholar 

  3. Bauermeister, A., Mannochio-Russo, H., Costa-Lotufo, L. V., Jarmusch, A. K. & Dorrestein, P. C. Mass spectrometry-based metabolomics in microbiome investigations. Nat. Rev. Microbiol. 20, 143–160 (2022).

    CAS  PubMed  Google Scholar 

  4. Wishart, D. S. et al. NMR and metabolomics—a roadmap for the future. Metabolites 12, 678 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Alexandrov, T. Spatial metabolomics and imaging mass spectrometry in the age of artificial intelligence. Annu. Rev. Biomed. Data Sci. 3, 61–87 (2020).

    PubMed  PubMed Central  Google Scholar 

  6. Geier, B. et al. Spatial metabolomics of in situ host–microbe interactions at the micrometre scale. Nat. Microbiol. 5, 498–510 (2020).

    CAS  PubMed  Google Scholar 

  7. Esquenazi, E., Yang, Y.-L., Watrous, J., Gerwick, W. H. & Dorrestein, P. C. Imaging mass spectrometry of natural products. Nat. Prod. Rep. 26, 1521–1534 (2009).

    CAS  PubMed  Google Scholar 

  8. Vickerman, J. C. Molecular imaging and depth profiling by mass spectrometry—SIMS, MALDI or DESI. Analyst 136, 2199–2217 (2011).

    CAS  PubMed  Google Scholar 

  9. Pozebon, D., Scheffler, G. L., Dressler, V. L. & Nunes, M. A. G. Review of the applications of laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS) to the analysis of biological samples. J. Anal. At. Spectrom. 29, 2204–2228 (2014).

    CAS  Google Scholar 

  10. Heijs, B., Potthoff, A., Soltwisch, J. & Dreisewerd, K. MALDI-2 for the enhanced analysis of N-linked glycans by mass spectrometry imaging. Anal. Chem. 92, 13904–13911 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Stanback, A. E. et al. Regional N-glycan and lipid analysis from tissues using MALDI–mass spectrometry imaging. STAR Protoc. 2, 100304 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Sun, C. et al. Visualizing the spatial distribution and alteration of metabolites in continuously cropped Salvia miltiorrhiza Bge using MALDI–MSI. J. Pharm. Anal. 12, 719–724 (2022).

    PubMed  Google Scholar 

  13. Sogin, E. M. et al. Sugars dominate the seagrass rhizosphere. Nat. Ecol. Evol. 6, 866–877 (2022).

    PubMed  PubMed Central  Google Scholar 

  14. Bien, T., Koerfer, K., Schwenzfeier, J., Dreisewerd, K. & Soltwisch, J. Mass spectrometry imaging to explore molecular heterogeneity in cell culture. Proc. Natl Acad. Sci. USA 119, e2114365119 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Capolupo, L. et al. Sphingolipids control dermal fibroblast heterogeneity. Science 376, eabh1623 (2022).

    CAS  PubMed  Google Scholar 

  16. Bourceau, P., Michellod, D., Geier, B. & Liebeke, M. Spatial metabolomics shows contrasting phosphonolipid distributions in tissues of marine bivalves. PeerJ Anal. Chem. 4, e21 (2022).

    CAS  Google Scholar 

  17. Bowman, A. P. et al. Evaluation of lipid coverage and high spatial resolution MALDI-imaging capabilities of oversampling combined with laser post-ionisation. Anal. Bioanal. Chem. 412, 2277–2289 (2020).

    CAS  PubMed  Google Scholar 

  18. Rappez, L. et al. SpaceM reveals metabolic states of single cells. Nat. Methods 18, 799–805 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Wang, G. et al. Analyzing cell-type-specific dynamics of metabolism in kidney repair. Nat. Metab. 4, 1109–1118 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Zhu, X., Xu, T., Peng, C. & Wu, S. Advances in MALDI mass spectrometry imaging single cell and tissues. Front. Chem. 9, 782432 (2022).

    PubMed  PubMed Central  Google Scholar 

  21. Niehaus, M., Soltwisch, J., Belov, M. E. & Dreisewerd, K. Transmission-mode MALDI-2 mass spectrometry imaging of cells and tissues at subcellular resolution. Nat. Methods 16, 925–931 (2019).

    CAS  PubMed  Google Scholar 

  22. Kompauer, M., Heiles, S. & Spengler, B. Atmospheric pressure MALDI mass spectrometry imaging of tissues and cells at 1.4-μm lateral resolution. Nat. Methods 14, 90–96 (2017).

    CAS  PubMed  Google Scholar 

  23. Yang, J. Y. et al. Primer on agar-based microbial imaging mass spectrometry. J. Bacteriol. 194, 6023–6028 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Yang, Y.-L., Xu, Y., Straight, P. & Dorrestein, P. C. Translating metabolic exchange with imaging mass spectrometry. Nat. Chem. Biol. 5, 885–887 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Si, T. et al. Characterization of Bacillus subtilis colony biofilms via mass spectrometry and fluorescence imaging. J. Proteome Res. 15, 1955–1962 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Feucherolles, M. & Frache, G. MALDI mass spectrometry imaging: a potential game-changer in a modern microbiology. Cells 11, 3900 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Dunham, S. J. B., Ellis, J. F., Li, B. & Sweedler, J. V. Mass spectrometry imaging of complex microbial communities. Acc. Chem. Res. 50, 96–104 (2017).

    CAS  PubMed  Google Scholar 

  28. Gahlmann, A. & Moerner, W. E. Exploring bacterial cell biology with single-molecule tracking and super-resolution imaging. Nat. Rev. Microbiol. 12, 9–22 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Hatzenpichler, R., Krukenberg, V., Spietz, R. L. & Jay, Z. J. Next-generation physiology approaches to study microbiome function at single cell level. Nat. Rev. Microbiol. 18, 241–256 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Amann, R. & Fuchs, B. M. Single-cell identification in microbial communities by improved fluorescence in situ hybridization techniques. Nat. Rev. Microbiol. 6, 339–348 (2008).

    CAS  PubMed  Google Scholar 

  31. Amann, R., Fuchs, B. M. & Behrens, S. The identification of microorganisms by fluorescence in situ hybridisation. Curr. Opin. Biotechnol. 12, 231–236 (2001).

    CAS  PubMed  Google Scholar 

  32. Barrero-Canosa, J., Moraru, C., Zeugner, L., Fuchs, B. M. & Amann, R. Direct-geneFISH: a simplified protocol for the simultaneous detection and quantification of genes and rRNA in microorganisms. Environ. Microbiol. 19, 70–82 (2017).

    CAS  PubMed  Google Scholar 

  33. Greuter, D., Loy, A., Horn, M. & Rattei, T. probeBase—an online resource for rRNA-targeted oligonucleotide probes and primers: new features 2016. Nucleic Acids Res. 44, D586–D589 (2015).

    PubMed  PubMed Central  Google Scholar 

  34. Smith, B. et al. Community analysis of bacteria colonizing intestinal tissue of neonates with necrotizing enterocolitis. BMC Microbiol. 11, 73 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Schimak, M. P., Toenshoff, E. R. & Bright, M. Simultaneous 16S and 18S rRNA fluorescence in situ hybridization (FISH) on LR White sections demonstrated in Vestimentifera (Siboglinidae) tubeworms. Acta Histochem. 114, 122–130 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Neugent, M. L., Gadhvi, J., Palmer, K. L., Zimmern, P. E. & De Nisco, N. J. Detection of tissue-resident bacteria in bladder biopsies by 16S rRNA fluorescence in situ hybridization. J Vis. Exp. 152, e60458 (2019).

    Google Scholar 

  37. Valm, A. M., Mark Welch, J. L. & Borisy, G. G. CLASI-FISH: principles of combinatorial labeling and spectral imaging. Syst. Appl. Microbiol. 35, 496–502 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Valm, A. M. et al. Systems-level analysis of microbial community organization through combinatorial labeling and spectral imaging. Proc. Natl Acad. Sci. USA 108, 4152–4157 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Kaltenpoth, M., Strupat, K. & Svatoš, A. Linking metabolite production to taxonomic identity in environmental samples by (MA)LDI–FISH. ISME J. 10, 527–531 (2016).

    PubMed  Google Scholar 

  40. Geier, B. et al. Connecting structure and function from organisms to molecules in small-animal symbioses through chemo-histo-tomography. Proc. Natl Acad. Sci. USA 118, e2023773118 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Ponnudurai, R. et al. Metabolic and physiological interdependencies in the Bathymodiolus azoricus symbiosis. ISME J. 11, 463–477 (2017).

    CAS  PubMed  Google Scholar 

  42. Dreisewerd, K., Bien, T. & Soltwisch, J. in Mass Spectrometry Imaging of Small Molecules: Methods and Protocols (ed. Y.-J. Lee) 21–40 (Springer, 2022).

  43. Soltwisch, J. et al. Mass spectrometry imaging with laser-induced postionization. Science 348, 211–215 (2015).

    CAS  PubMed  Google Scholar 

  44. Ansorge, R. et al. Functional diversity enables multiple symbiont strains to coexist in deep-sea mussels. Nat. Microbiol. 4, 2487–2497 (2019).

    PubMed  Google Scholar 

  45. Fung, C. et al. High-resolution mapping reveals that microniches in the gastric glands control Helicobacter pylori colonization of the stomach. PLoS Biol. 17, e3000231 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Lackner, G., Peters, E. E., Helfrich, E. J. N. & Piel, J. Insights into the lifestyle of uncultured bacterial natural product factories associated with marine sponges. Proc. Natl Acad. Sci. USA 114, E347–E356 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Yang, H. et al. On-tissue derivatization of lipopolysaccharide for detection of Lipid A using MALDI-MSI. Anal. Chem. 92, 13667–13671 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Patel, E. et al. MALDI-MS imaging for the study of tissue pharmacodynamics and toxicodynamics. Bioanalysis 7, 91–101 (2015).

    CAS  PubMed  Google Scholar 

  49. Prideaux, B. et al. High-sensitivity MALDI–MRM–MS imaging of moxifloxacin distribution in tuberculosis-infected rabbit lungs and granulomatous lesions. Anal. Chem. 83, 2112–2118 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Daims, H., Brühl, A., Amann, R., Schleifer, K.-H. & Wagner, M. The domain-specific probe EUB338 is insufficient for the detection of all bacteria: development and evaluation of a more comprehensive probe set. Syst. Appl. Microbiol. 22, 434–444 (1999).

    CAS  PubMed  Google Scholar 

  51. Braun, P. et al. In-depth analysis of Bacillus anthracis 16S rRNA genes and transcripts reveals intra- and intergenomic diversity and facilitates anthrax detection. mSystems 7, e01361–01321 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Rath, C. M. et al. Molecular analysis of model gut microbiotas by imaging mass spectrometry and nanodesorption electrospray ionization reveals dietary metabolite transformations. Anal. Chem. 84, 9259–9267 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Piwosz, K., Mukherjee, I., Salcher, M. M., Grujčić, V. & Šimek, K. CARD-FISH in the sequencing era: opening a new universe of protistan ecology. Front. Microbiol. 12, 640066 (2021).

    PubMed  PubMed Central  Google Scholar 

  54. Morales, D. P. et al. Advances and challenges in fluorescence in situ hybridization for visualizing fungal endobacteria. Front. Microbiol. 13, 892227 (2022).

    PubMed  PubMed Central  Google Scholar 

  55. Raj, A., van den Bogaard, P., Rifkin, S. A., van Oudenaarden, A. & Tyagi, S. Imaging individual mRNA molecules using multiple singly labeled probes. Nat. Methods 5, 877–879 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Ticha, P. et al. A novel cryo-embedding method for in-depth analysis of craniofacial mini pig bone specimens. Sci. Rep. 10, 19510 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Hoffmann, F., Janussen, D., Dröse, W., Arp, G. & Reitner, J. Histological investigation of organisms with hard skeletons: a case study of siliceous sponges. Biotech. Histochem. 78, 191–199 (2003).

    CAS  PubMed  Google Scholar 

  58. Kompauer, M., Heiles, S. & Spengler, B. Autofocusing MALDI mass spectrometry imaging of tissue sections and 3D chemical topography of nonflat surfaces. Nat. Methods 14, 1156–1158 (2017).

    CAS  PubMed  Google Scholar 

  59. Angerer, T. B., Bour, J., Biagi, J.-L., Moskovets, E. & Frache, G. Evaluation of 6 MALDI–matrices for 10 μm lipid imaging and on-tissue MSn with AP-MALDI-Orbitrap. J. Am. Soc. Mass Spectrom. 33, 760–771 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Leopold, J., Prabutzki, P., Engel, K. M. & Schiller, J. A five-year update on matrix compounds for MALDI–MS analysis of lipids. Biomolecules 13, 546 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Cerruti, C. D., Benabdellah, F., Laprévote, O., Touboul, D. & Brunelle, A. MALDI imaging and structural analysis of rat brain lipid negative ions with 9-aminoacridine matrix. Anal. Chem. 84, 2164–2171 (2012).

    CAS  PubMed  Google Scholar 

  62. Kaya, I., Jennische, E., Lange, S. & Malmberg, P. Dual polarity MALDI imaging mass spectrometry on the same pixel points reveals spatial lipid localizations at high-spatial resolutions in rat small intestine. Anal. Methods 10, 2428–2435 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Meisenbichler, C., Doppler, C., Bernhard, D. & Müller, T. Improved matrix coating for positive- and negative-ion-mode MALDI-TOF imaging of lipids in blood vessel tissues. Anal. Bioanal. Chem. 411, 3221–3227 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Shariatgorji, R. et al. Spatial visualization of comprehensive brain neurotransmitter systems and neuroactive substances by selective in situ chemical derivatization mass spectrometry imaging. Nat. Protoc. 16, 3298–3321 (2021).

    CAS  PubMed  Google Scholar 

  65. Iwama, T. et al. Development of an on-tissue derivatization method for MALDI mass spectrometry imaging of bioactive lipids containing phosphate monoester using Phos-tag. Anal. Chem. 93, 3867–3875 (2021).

    CAS  PubMed  Google Scholar 

  66. Harkin, C. et al. On-tissue chemical derivatization in mass spectrometry imaging. Mass Spectrom. Rev. 41, 662–694 (2022).

    CAS  PubMed  Google Scholar 

  67. Tressler, C. et al. Factorial design to optimize matrix spraying parameters for MALDI mass spectrometry imaging. J. Am. Soc. Mass Spectrom. 32, 2728–2737 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Ščupáková, K. et al. Cellular resolution in clinical MALDI mass spectrometry imaging: the latest advancements and current challenges. Clin. Chem. Lab. Med. 58, 914–929 (2020).

    PubMed  PubMed Central  Google Scholar 

  69. Schimak, M. P. et al. MiL-FISH: multilabeled oligonucleotides for fluorescence in situ hybridization improve visualization of bacterial cells. Appl. Environ. Microbiol. 82, 62–70 (2016).

    CAS  PubMed  Google Scholar 

  70. Amann, R. I., Krumholz, L. & Stahl, D. A. Fluorescent-oligonucleotide probing of whole cells for determinative, phylogenetic, and environmental studies in microbiology. J. Bacteriol. 172, 762–770 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Folberth, J., Begemann, K., Jöhren, O., Schwaninger, M. & Othman, A. MS(2) and LC libraries for untargeted metabolomics: Enhancing method development and identification confidence. J. Chromatogr. B 1145, 122105 (2020).

    CAS  Google Scholar 

  72. Chaleckis, R., Meister, I., Zhang, P. & Wheelock, C. E. Challenges, progress and promises of metabolite annotation for LC–MS-based metabolomics. Curr. Opin. Biotechnol. 55, 44–50 (2019).

    CAS  PubMed  Google Scholar 

  73. Palmer, A. et al. FDR-controlled metabolite annotation for high-resolution imaging mass spectrometry. Nat. Methods 14, 57–60 (2017).

    CAS  PubMed  Google Scholar 

  74. Spraggins, J. M. et al. High-performance molecular imaging with MALDI trapped ion-mobility time-of-flight (timsTOF) mass spectrometry. Anal. Chem. 91, 14552–14560 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Kim, Y. H. et al. In situ label-free visualization of orally dosed strictinin within mouse kidney by MALDI–MS imaging. J. Agric. Food Chem. 62, 9279–9285 (2014).

    CAS  PubMed  Google Scholar 

  76. Pirman, D. A., Reich, R. F., Kiss, A. A., Heeren, R. M. A. & Yost, R. A. Quantitative MALDI tandem mass spectrometric imaging of cocaine from brain tissue with a deuterated internal standard. Anal. Chem. 85, 1081–1089 (2013).

    CAS  PubMed  Google Scholar 

  77. Amann, R. I., Ludwig, W. & Schleifer, K. H. Phylogenetic identification and in situ detection of individual microbial cells without cultivation. Microbiol. Rev. 59, 143–169 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Janda, M. et al. Determination of abundant metabolite matrix adducts illuminates the dark metabolome of MALDI-mass spectrometry imaging datasets. Anal. Chem. 93, 8399–8407 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Protsyuk, I. et al. 3D molecular cartography using LC–MS facilitated by Optimus and ’ili software. Nat. Protoc. 13, 134–154 (2018).

    CAS  PubMed  Google Scholar 

  80. Garg, N. et al. Three-dimensional microbiome and metabolome cartography of a diseased human lung. Cell Host Microbe 22, 705–716.e704 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Shen, S. et al. Parallel, high-quality proteomic and targeted metabolomic quantification using laser capture microdissected tissues. Anal. Chem. 93, 8711–8718 (2021).

    CAS  PubMed  Google Scholar 

  82. Dilillo, M. et al. Mass spectrometry imaging, laser capture microdissection, and LC–MS/MS of the same tissue section. J. Proteome Res. 16, 2993–3001 (2017).

    CAS  PubMed  Google Scholar 

  83. Ščupáková, K., Dewez, F., Walch, A. K., Heeren, R. M. A. & Balluff, B. Morphometric cell classification for single-cell MALDI–mass spectrometry imaging. Angew. Chem. Int. Ed. 59, 17447–17450 (2020).

    Google Scholar 

  84. Prade, V. M. et al. De novo discovery of metabolic heterogeneity with immunophenotype-guided imaging mass spectrometry. Mol. Metabol. 36, 100953 (2020).

    CAS  Google Scholar 

  85. Blanc, L., Lenaerts, A., Dartois, V. & Prideaux, B. Visualization of mycobacterial biomarkers and tuberculosis drugs in infected tissue by MALDI–MS imaging. Anal. Chem. 90, 6275–6282 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Perry, W. J. et al. Staphylococcus aureus exhibits heterogeneous siderophore production within the vertebrate host. Proc. Natl Acad. Sci. USA 116, 21980–21982 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Tobias, F. & Hummon, A. B. Considerations for MALDI-based quantitative mass spectrometry imaging studies. J. Proteome Res. 19, 3620–3630 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Bakker, B. et al. Preparing ductal epithelial organoids for high-spatial-resolution molecular profiling using mass spectrometry imaging. Nat. Protoc. 17, 962–979 (2022).

    CAS  PubMed  Google Scholar 

  89. Paschke, C. et al. Mirion—a software package for automatic processing of mass spectrometric images. J. Am. Soc. Mass Spectrom. 24, 1296–1306 (2013).

    CAS  PubMed  Google Scholar 

  90. Bemis, K. D. et al. Cardinal: an R package for statistical analysis of mass spectrometry-based imaging experiments. Bioinformatics 31, 2418–2420 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Race, A. M., Styles, I. B. & Bunch, J. Inclusive sharing of mass spectrometry imaging data requires a converter for all. J. Proteomics 75, 5111–5112 (2012).

    CAS  PubMed  Google Scholar 

  92. Goracci, L. et al. Lipostar, a comprehensive platform-neutral cheminformatics tool for lipidomics. Anal. Chem. 89, 6257–6264 (2017).

    CAS  PubMed  Google Scholar 

  93. Tortorella, S. et al. LipostarMSI: comprehensive, vendor-neutral software for visualization, data analysis, and automated molecular identification in mass spectrometry imaging. J. Am. Soc. Mass Spectrom. 31, 155–163 (2020).

    CAS  PubMed  Google Scholar 

  94. Treu, A. & Römpp, A. Matrix ions as internal standard for high mass accuracy matrix-assisted laser desorption/ionization mass spectrometry imaging. Rapid Commun. Mass Spectrom. 35, e9110 (2021).

    CAS  PubMed  Google Scholar 

  95. Sládková, K., Houška, J. & Havel, J. Laser desorption ionization of red phosphorus clusters and their use for mass calibration in time-of-flight mass spectrometry. Rapid Commun. Mass Spectrom. 23, 3114–3118 (2009).

    PubMed  Google Scholar 

  96. He, J., Mo, D., Chen, J. & Luo, L. Combined whole-mount fluorescence in situ hybridization and antibody staining in zebrafish embryos and larvae. Nat. Protoc. 15, 3361–3379 (2020).

    CAS  PubMed  Google Scholar 

  97. Richardson, D. S. et al. Tissue clearing. Nat. Rev. Methods Primers 1, 84 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Schramm, T. et al. imzML—a common data format for the flexible exchange and processing of mass spectrometry imaging data. J. Proteomics 75, 5106–5110 (2012).

    CAS  PubMed  Google Scholar 

  99. Sumner, L. W. et al. Proposed minimum reporting standards for chemical analysis Chemical Analysis Working Group (CAWG) Metabolomics Standards Initiative (MSI). Metabolomics 3, 211–221 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Bik, L. et al. In vivo dermal delivery of bleomycin with electronic pneumatic injection: drug visualization and quantification with mass spectrometry. Expert Opin. Drug Deliv. 19, 213–219 (2022).

    CAS  PubMed  Google Scholar 

  101. Cuypers, E. et al. ‘On the spot’ digital pathology of breast cancer based on single-cell mass spectrometry imaging. Anal. Chem. 94, 6180–6190 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Duperron, S. et al. A dual symbiosis shared by two mussel species, Bathymodiolus azoricus and Bathymodiolus puteoserpentis (Bivalvia: Mytilidae), from hydrothermal vents along the northern Mid-Atlantic Ridge. Environ. Microbiol. 8, 1441–1447 (2006).

    CAS  PubMed  Google Scholar 

  103. Zielinski, F. U. et al. Widespread occurrence of an intranuclear bacterial parasite in vent and seep bathymodiolin mussels. Environ. Microbiol. 11, 1150–1167 (2009).

    CAS  PubMed  Google Scholar 

  104. Raskin, L., Stromley, J. M., Rittmann, B. E. & Stahl, D. A. Group-specific 16S rRNA hybridization probes to describe natural communities of methanogens. Appl. Environ. Microbiol. 60, 1232–1240 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Manz, W., Amann, R., Ludwig, W., Wagner, M. & Schleifer, K.-H. Phylogenetic oligodeoxynucleotide probes for the major subclasses of proteobacteria: problems and solutions. Syst. Appl. Microbiol. 15, 593–600 (1992).

    Google Scholar 

  106. Hayasaka, T., Goto-Inoue, N., Masaki, N., Ikegami, K. & Setou, M. Application of 2,5-dihydroxyacetophenone with sublimation provides efficient ionization of lipid species by atmospheric pressure matrix-assisted laser desorption/ionization imaging mass spectrometry. Surf. Interface Anal. 46, 1219–1222 (2014).

    CAS  Google Scholar 

  107. Bien, T., Hambleton, E. A., Dreisewerd, K. & Soltwisch, J. Molecular insights into symbiosis—mapping sterols in a marine flatworm-algae-system using high spatial resolution MALDI-2–MS imaging with ion mobility separation. Anal. Bioanal. Chem. 413, 2767–2777 (2021).

    CAS  PubMed  Google Scholar 

  108. Ellis, S. R., Soltwisch, J., Paine, M. R. L., Dreisewerd, K. & Heeren, R. M. A. Laser post-ionisation combined with a high resolving power orbitrap mass spectrometer for enhanced MALDI–MS imaging of lipids. Chem. Commun. 53, 7246–7249 (2017).

    CAS  Google Scholar 

  109. Zhou, Q. et al. A caged in-source laser-cleavable MALDI matrix with high vacuum stability for extended MALDI–MS imaging. Angew. Chem. Int. Ed. 62, e202217047 (2023).

    CAS  Google Scholar 

  110. Lukowski, J. K. et al. Storage conditions of human kidney tissue sections affect spatial lipidomics analysis reproducibility. J. Am. Soc. Mass Spectrom. 31, 2538–2546 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Stoecker, K., Dorninger, C., Daims, H. & Wagner, M. Double labeling of oligonucleotide probes for fluorescence in situ hybridization (DOPE-FISH) improves signal intensity and increases rRNA accessibility. Appl. Environ. Microbiol. 76, 922–926 (2010).

    CAS  PubMed  Google Scholar 

  112. Pernthaler, A., Pernthaler, J. & Amann, R. Fluorescence in situ hybridization and catalyzed reporter deposition for the identification of marine bacteria. Appl. Environ. Microbiol. 68, 3094–3101 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Teixeira, H., Sousa, A. L. & Azevedo, A. S. in Fluorescence In-Situ Hybridization (FISH) for Microbial Cells: Methods and Concepts (eds N. F. Azevedo & C. Almeida) 35–50 (Springer, 2021).

  114. Hugenholtz, P., Tyson, G. W. & Blackall, L. L. in Gene Probes: Principles and Protocols (eds M. A. de Muro & Ralph Rapley) 29–42 (Humana Press, 2002).

Download references

Acknowledgements

We acknowledge M. Sadowski (MPI Bremen) and J. Beckmann (MPI Bremen) for technical assistance with FISH and MSI, and Bruker Daltonics for access to timsTOF fleX instrumentation. P.B., B.G. and M.L. thank the Max Planck Society for funding. J.S. and K.D. are grateful for funding from Deutsche Forschungsgemeinschaft (DFG): DR 416/12-1 and SO976/41, SO976/5-1 and DR416/13-1, and CRC TRR332 (Z1). B.G. thanks the Human Frontier in Science Program for postdoctoral funding via a long-term fellowship (LT0015/2022-L).

Author information

Authors and Affiliations

Authors

Contributions

P.B., B.G., T.B. and V.S. recorded MSI data. V.S., J.S., T.B. and K.D. assisted in the interpretation of results and writing the manuscript. T.B., V.S. and P.B. conducted the protocol validation experiments. P.B., B.G. and M.L. conceived and designed the study. P.B., B.G. and M.L. wrote the manuscript.

Corresponding author

Correspondence to Manuel Liebeke.

Ethics declarations

Competing interests

T.B. is an employee of Bruker Daltonics GmbH & Co. KG (Bremen). All other authors declare no competing interests.

Peer review

Peer review information

Nature Protocols thanks Laura Sanchez 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.

Related links

Key references using this protocol

Geier, B. et al. Nat. Microbiol. 5, 498–510 (2020): https://doi.org/10.1038/s41564-019-0664-6

Geier, B. et al. Proc. Natl Acad. Sci. USA 118, e2023773118 (2021): https://doi.org/10.1073/pnas.2023773118

Bien, T. et al. Proc. Natl Acad. Sci. USA 119, e2114365119 (2022): https://doi.org/10.1073/pnas.2114365119

Niehaus, M. et al. Nat. Methods 16, 925–931 (2019): https://doi.org/10.1038/s41592-019-0536-2

Supplementary information

Supplementary Information

Supplementary Methods 1–4, Tables 1–13.

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

Bourceau, P., Geier, B., Suerdieck, V. et al. Visualization of metabolites and microbes at high spatial resolution using MALDI mass spectrometry imaging and in situ fluorescence labeling. Nat Protoc 18, 3050–3079 (2023). https://doi.org/10.1038/s41596-023-00864-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-023-00864-1

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

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