ASN's Mission

To create a world without kidney diseases, the ASN Alliance for Kidney Health elevates care by educating and informing, driving breakthroughs and innovation, and advocating for policies that create transformative changes in kidney medicine throughout the world.

learn more

Contact ASN

1401 H St, NW, Ste 900, Washington, DC 20005

email@asn-online.org

202-640-4660

The Latest on X

Kidney Week

Please note that you are viewing an archived section from 2022 and some content may be unavailable. To unlock all content for 2022, please visit the archives.

Abstract: TH-OR43

Analyzing Cell Type-Specific Dynamics of Metabolism in Kidney Repair

Session Information

Category: Acute Kidney Injury

  • 103 AKI: Mechanisms

Authors

  • Rabelink, Ton J., Leids Universitair Medisch Centrum, Leiden, Zuid-Holland, Netherlands
  • Wang, Gangqi, Leids Universitair Medisch Centrum, Leiden, Zuid-Holland, Netherlands
  • Heijs, Bram, Leids Universitair Medisch Centrum, Leiden, Zuid-Holland, Netherlands
  • Kostidis, Sarantos, Leids Universitair Medisch Centrum, Leiden, Zuid-Holland, Netherlands
  • Mahfouz, Ahmed, Leids Universitair Medisch Centrum, Leiden, Zuid-Holland, Netherlands
  • Rietjens, Rosalie, Leids Universitair Medisch Centrum, Leiden, Zuid-Holland, Netherlands
  • Bijkerk, Roel, Leids Universitair Medisch Centrum, Leiden, Zuid-Holland, Netherlands
  • Koudijs, Angela, Leids Universitair Medisch Centrum, Leiden, Zuid-Holland, Netherlands
  • van der Pluijm, Loïs, Leids Universitair Medisch Centrum, Leiden, Zuid-Holland, Netherlands
  • Van den Berg, Cathelijne W., Leids Universitair Medisch Centrum, Leiden, Zuid-Holland, Netherlands
  • Dumas, Sébastien J., Vlaams Instituut voor Biotechnologie KU Leuven Center for Cancer Biology, Leuven, Flemish Brabant, Belgium
  • Carmeliet, Peter, Vlaams Instituut voor Biotechnologie KU Leuven Center for Cancer Biology, Leuven, Flemish Brabant, Belgium
  • Giera, Martin, Leids Universitair Medisch Centrum, Leiden, Zuid-Holland, Netherlands
  • van den Berg, Bernard, Leids Universitair Medisch Centrum, Leiden, Zuid-Holland, Netherlands
Background

Conventional single-cell metabolomics approaches such as MALDI based mass spectrometry imaging (MALDI-MSI) generate biochemical snapshots, neglecting the inherent dynamic nature of metabolism. Here we describe a platform based on isotope tracing and MALDI-MSI that allows in-situ dynamic measurements of cell type-specific metabolism at single-cell resolution, and thus unravel cell metabolism within its tissue architecture.

Methods

We applied different 13C-isotope-labeled nutrients on vibratome slices of fresh mouse kidney. MALDI-MSI at single-cell resolution (i.e. pixel size of 5 × 5 μm2) was then applied to detect metabolites and lipids from the harvested tissues. Following MALDI-MSI analysis, post-MSI-analyzed sections were stained and subsequently imaged using multiplexed immunofluorescence (IF) microscopy for cell-type identification.

Results

We show that this method can map cell type-specific dynamic changes in the central carbon metabolism, as well as the contribution of different nutrients to energy metabolism in a complex heterogenous tissue architecture such as the kidney. Combined with multiplexed immunofluorescence staining, we can detect metabolic changes and nutrient partitioning in targeted cell types as demonstrated in a bilateral renal ischemia/reperfusion injury (bIRI) model. At baseline, we identified a marked heterogeneity in respect to TCA metabolite consumption and glycolysis in the outer stripe outer medullary proximal tubular segments (PT-S3) when compared to the cortical PT-S1/S2 segments. After bIRI, PT cells that failed to repair remained in a hyperglycolytic state. Meanwhile, PT cells with an apparent normal phenotype in the recovery phase still display a striking difference in tricarboxylic acid (TCA) cycle substrate use when compared to those in sham kidneys. As TCA metabolites serve biosynthesis as well as gene regulation this may be of relevance to the homeostatic capacity of the kidney microenvironment.

Conclusion

In sum, this method allows to achieve single-cell resolution in situ and hence interpret cell type-specific metabolic dynamics in the context of kidney structure and metabolism of neighboring cells.

Funding

  • Private Foundation Support