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: SA-PO566

OXGR1 Is a Candidate Disease Gene for Human Calcium Oxalate Nephrolithiasis

Session Information

  • Genetic Diseases: Diagnosis
    November 05, 2022 | Location: Exhibit Hall, Orange County Convention Center‚ West Building
    Abstract Time: 10:00 AM - 12:00 PM

Category: Genetic Diseases of the Kidneys

  • 1102 Genetic Diseases of the Kidneys: Non-Cystic

Authors

  • Majmundar, Amar J., Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
  • Widmeier, Eugen, Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
  • Heneghan, John F., Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States
  • Daga, Ankana, Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
  • Hugo, Hannah, Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
  • Amar, Ali, Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, United States
  • Jobst-Schwan, Tilman, Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, United States
  • Nelson, Caleb, Department of Urology, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts, United States
  • Halbritter, Jan, Division of Nephrology, Department of Internal Medicine, University of Leipzig, Leipzig, Germany
  • Sayer, John Andrew, Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom
  • Fathy, Hanan, Pediatric Nephrology Unit, Alexandria University, Alexandria, Egypt
  • Baum, Michelle Amy, Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
  • Shril, Shirlee, Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
  • Mane, Shrikant M., Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, United States
  • Alper, Seth L., Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States
  • Hildebrandt, Friedhelm, Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
Background

Nephrolithiasis (NL) affects 1 in 11 individuals worldwide. Causative genetic variants are detected in 11-28% of NL and/or associated nephrocalcinosis (NC) (Halbritter JASN 2015; Braun cJASN 2016; Daga KI 2018). OXGR1 encodes 2-oxoglutarate receptor 1, which is expressed in collecting duct Type B intercalated cells. OXGR1 mediates cellular Ca2+ uptake in response to α-ketoglutarate (AKG) (He Nature 2004), a renally excreted metabolite derived from the NL treatment citrate (Krebs Biochem J 1938; Coe Nat Rev Neph 2016). Genetic inactivation in mice of the Oxgr1 effector Pendrin leads to hypercalciuria, a risk factor for NL/NC (Tokonami JCI 2013; Amlal AJPCP 2010; Barone NDT 2016).

Methods

Exome and targeted sequencing of the OXGR1 locus was performed in a worldwide NL/NC cohort. Putatively deleterious rare OXGR1 variants were functionally characterized.

Results

A heterozygous missense OXGR1 variant (c.371T>G, p.L124R) co-segregated with calcium oxalate NL/NC in an autosomal dominant inheritance pattern within a multi-generational family with five affected individuals. Strong amino acid conservation in orthologues and paralogues, severe in silico prediction scores (SIFT, PolyPhen2.0, CADD), and extreme rarity in exome/genome population databases suggested the variant was deleterious. Interrogation of the OXGR1 locus in 1107 NL/NC families identified five additional dominant alleles (p.Ser56Profs*7, p.Tyr93His, p.Cys217Arg, p.Ser233Arg, p.Ser287Phe) in five families with calcium oxalate NL/NC. All were rare variants (<5 alleles in ExAC, <10 alleles in gnomAD) with multiple severe prediction scores. Rare, potentially deleterious OXGR1 variants were enriched in NL/NC subjects relative to ExAC controls (0.54% versus 0.16%; Χ2=7.117, p=0.0076). Wildtype OXGR1-expressing Xenopus oocytes exhibited AKG-responsive Ca2+ uptake. Four of five NL/NC-associated missense variants revealed impaired AKG-dependent Ca2+ uptake at pH 5 and/or 7.4, demonstrating loss-of-function.

Conclusion

Rare, dominant OXGR1 variants are a candidate etiology of NL/NC, suggesting a novel mechanism for human NL/NC disease.

Funding

  • NIDDK Support