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 2020 and some content may be unavailable. To unlock all content for 2020, please visit the archives.

Abstract: TH-OR22

Cell-Autonomous Expression of Membrane Transport Proteins in Mammalian Distal Nephron

Session Information

Category: Fluid, Electrolyte, and Acid-Base Disorders

  • 901 Fluid, Electrolyte, and Acid-Base Disorders: Basic

Authors

  • Thomson, Martin N., Department of Functional Anatomy, Charité - Universitätsmedizin Berlin, Berlin, Berlin, Germany
  • Mutig, Kerim, Department of Functional Anatomy, Charité - Universitätsmedizin Berlin, Berlin, Berlin, Germany
  • Bachmann, Sebastian, Department of Functional Anatomy, Charité - Universitätsmedizin Berlin, Berlin, Berlin, Germany
Background


Reabsorption of NaCl in kidney thick ascending limb (TAL) via NKCC2 involves the action of luminal (ROMK) and basolateral (Kir4.1/Kir5.1 multimers) potassium channels, a basolateral calcium sensing receptor (CaSR), and the claudin (Cldn) family of proteins. Morphological heterogeneity of TAL cells has been reported, as well as mosaic expression of ROMK and Kir4.1. We hypothesized that this variability between TAL cells extends to other aspects of their function.

Methods


We studied TAL EM morphology, zonal and cell-autonomous heterogeneity of the transport proteins at steady state in mice, rats and humans, and under stimulation by vasopressin (AVP; V2R agonist dDAVP for 72 h) using AVP-deficient Brattleboro rats. NKCC2, phosphorylated (p) NKCC2, ROMK, Kir4.1, CaSR, Cld-10 and Cld-16 signals were analyzed by immunofluorescence, in situ hybridization (ISH), EM and Western blot (WB).

Results


Between cortex and medullary kidney zones, TAL morphological cell heterogeneity was observed, but not at a cell-to-cell level within each zone. NKCC2 was continuously expressed in all TAL cells, while pNKCC2 signals were heterogeneous, increasing from inner stripe of outer medulla to cortex and varying between cells of each zone. ROMK and Kir4.1 protein expression showed conspicuous heterogeneity in a mutually exclusive pattern, with stronger pNKCC2 expression in the ROMK-negative, Kir4.1-positive cell type. CaSR and Cldn-16 signals were moderate to absent in ROMK-positive cells, but intensified in ROMK-negative cells, while Cld-10 was strongly expressed only in ROMK-positive cells. ISH revealed no cell heterogeneity of ROMK mRNA. In Brattleboro rats, 72h dDAVP increased the number of ROMK- and Kir4.1-positive cells and the overlap of both signals in mTAL, and stimulated NKCC2 phosphorylation. Cldn-10 expression was induced in the outer stripe of outer medulla. Morphological alterations in the TAL included proliferation of microvilli. WB showed 2.2-fold increase of ROMK and 1.3-fold increase of Kir4.1 abundance, but down-regulation of CaSR to 60%.

Conclusion


These results demonstrate mosaic expression of ROMK, Kir4.1, CaSR, Cld-10, and Cld-16 in TAL, which correlate with cell-heterogeneous levels of NKCC2 activation.

Funding

  • Government Support - Non-U.S.