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 2021 and some content may be unavailable. To unlock all content for 2021, please visit the archives.

Abstract: PO1090

Mechanistic Importance of Reduced KLHL3 and CUL3 Expression in CUL3-Δ9-Mediated Familial Hyperkalemic Hypertension

Session Information

Category: Fluid, Electrolyte, and Acid-Base Disorders

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

Authors

  • Maeoka, Yujiro, Oregon Health & Science University, Portland, Oregon, United States
  • Ferdaus, Mohammed Zubaerul, Oregon Health & Science University, Portland, Oregon, United States
  • Sharma, Avika, Oregon Health & Science University, Portland, Oregon, United States
  • Cornelius, Ryan J., Oregon Health & Science University, Portland, Oregon, United States
  • Su, Xiao-Tong, Oregon Health & Science University, Portland, Oregon, United States
  • Robertson, Joshua A., Oregon Health & Science University, Portland, Oregon, United States
  • Gurley, Susan B., Oregon Health & Science University, Portland, Oregon, United States
  • Ellison, David H., Oregon Health & Science University, Portland, Oregon, United States
  • McCormick, James A., Oregon Health & Science University, Portland, Oregon, United States
Background

Mutations in the ubiquitin ligase scaffold protein Cullin 3 (CUL3) cause the disease familial hyperkalemic hypertension (FHHt). In the kidney, mutant CUL3 (CUL3-Δ9) cannot interact with COP9 signalosome subunit JAB1 that negatively regulates CUL3 activity. This leads to CUL3-Δ9 autodegradation, and increased abundance of With-No-Lysine [K] Kinase 4 (WNK4), which inappropriately activates the downstream kinase SPAK, which then phosphorylates and hyperactivates the Na+-Cl cotransporter (NCC). We showed lower that CUL3 alone does not increase WNK4, so the precise mechanism by which CUL3-Δ9 causes FHHt is unclear. We hypothesized CUL3-Δ9 degrades Kelch-like 3 (KLHL3), the CUL3 substrate adaptor for WNK4; thus reduced abundance of KLHL3 combined with reduced CUL3 are mechanistically important in CUL3-Δ9-mediated FHHt.

Methods

We studied Cul3 KO (Cul3–/–) mice, Cul3 KO mice expressing CUL3-Δ9 (Cul3–/–/Δ9), Cul3 heterozygotes expressing CUL3-Δ9 (Cul3+/–/Δ9), compound Cul3 and Klhl3 heterozygotes (Cul3+/–Klhl3+/–), and Jab1 KO (Jab1–/–) mice. All mouse lines were inducible and renal tubule-specific.

Results

CUL3-Δ9 did not promote degradation of CUL3 targets that accumulate in Cul3–/– kidney: WNK4, cyclin E, or NQO1 (a surrogate for the CUL3 substrate Nrf2). In Cul3–/–/Δ9 mice, CUL3-Δ9 prevented KLHL3 accumulation seen in Cul3–/– kidney and promoted KLHL3 degradation in Cul3+/–/Δ9 mice. Higher NQO1 and lower cyclin E abundances were observed in Cul3+/–/Δ9 mice compared to control mice. Cul3+/–Klhl3+/– mice displayed increased WNK4-SPAK activation and phospho-NCC abundance, and FHHt-like phenotype with increased plasma [K+] and salt-sensitive blood pressure. Similarly, reduced CUL3 and KLHL3 abundances and increased abundances of WNK4 and phsopho-NCC were observed in Jab1–/– mice.

Conclusion

Together, these data provide evidence for a mechanism of reduced KLHL3 and reduced CUL3 in CUL3-Δ9-mediated FHHt. CUL3-Δ9 potently degrades KLHL3, but also exerts modest effects on other CUL3 targets, raising the possibility of unidentified renal phenotypes in the human disease.

Funding

  • NIDDK Support