De novo TRIM8 variants impair its protein localization to nuclear bodies and cause developmental delay, epilepsy, and focal segmental glomerulosclerosis

  • Patricia L Weng
  • Amar J Majmundar
  • Kamal Khan
  • Tze Y Lim
  • Shirlee Shril
  • Gina Jin
  • John Musgrove
  • Minxian Wang
  • Dina F Ahram
  • Vimla S Aggarwal
  • Louise E Bier
  • Erin L Heinzen
  • Ana C Onuchic-Whitford
  • Nina Mann
  • Florian Buerger
  • Ronen Schneider
  • Konstantin Deutsch
  • Thomas M Kitzler
  • Verena Klämbt
  • Amy Kolb
  • Youying Mao
  • Christelle Moufawad El Achkar
  • Adele Mitrotti
  • Jeremiah Martino
  • Bodo B Beck
  • Janine Altmüller
  • Marcus R Benz
  • Shoji Yano
  • Mohamad A Mikati
  • Talha Gunduz
  • Heidi Cope
  • Vandana Shashi
  • Howard Trachtman
  • Monica Bodria
  • Gianluca Caridi
  • Isabella Pisani
  • Enrico Fiaccadori
  • Asmaa S AbuMaziad
  • Julian A Martinez-Agosto
  • Ora Yadin
  • Jonathan Zuckerman
  • Arang Kim
  • Ulrike John-Kroegel
  • Amanda V Tyndall
  • Jillian S Parboosingh
  • A Micheil Innes
  • Agnieszka Bierzynska
  • Ania B Koziell
  • Mordi Muorah
  • Moin A Saleem
  • Julia Hoefele
  • Korbinian M Riedhammer
  • Ali G Gharavi
  • Vaidehi Jobanputra
  • Emma Pierce-Hoffman
  • Eleanor G Seaby
  • Anne O'Donnell-Luria
  • Heidi L Rehm
  • Shrikant Mane
  • Vivette D D'Agati
  • Martin R Pollak
  • Gian Marco Ghiggeri
  • Richard P Lifton
  • David B Goldstein
  • Erica E Davis
  • Friedhelm Hildebrandt
  • Simone Sanna-Cherchi
  • Undiagnosed Diseases Network

Abstract

Focal segmental glomerulosclerosis (FSGS) is the main pathology underlying steroid-resistant nephrotic syndrome (SRNS) and a leading cause of chronic kidney disease. Monogenic forms of pediatric SRNS are predominantly caused by recessive mutations, while the contribution of de novo variants (DNVs) to this trait is poorly understood. Using exome sequencing (ES) in a proband with FSGS/SRNS, developmental delay, and epilepsy, we discovered a nonsense DNV in TRIM8, which encodes the E3 ubiquitin ligase tripartite motif containing 8. To establish whether TRIM8 variants represent a cause of FSGS, we aggregated exome/genome-sequencing data for 2,501 pediatric FSGS/SRNS-affected individuals and 48,556 control subjects, detecting eight heterozygous TRIM8 truncating variants in affected subjects but none in control subjects (p = 3.28 × 10-11). In all six cases with available parental DNA, we demonstrated de novo inheritance (p = 2.21 × 10-15). Reverse phenotyping revealed neurodevelopmental disease in all eight families. We next analyzed ES from 9,067 individuals with epilepsy, yielding three additional families with truncating TRIM8 variants. Clinical review revealed FSGS in all. All TRIM8 variants cause protein truncation clustering within the last exon between residues 390 and 487 of the 551 amino acid protein, indicating a correlation between this syndrome and loss of the TRIM8 C-terminal region. Wild-type TRIM8 overexpressed in immortalized human podocytes and neuronal cells localized to nuclear bodies, while constructs harboring patient-specific variants mislocalized diffusely to the nucleoplasm. Co-localization studies demonstrated that Gemini and Cajal bodies frequently abut a TRIM8 nuclear body. Truncating TRIM8 DNVs cause a neuro-renal syndrome via aberrant TRIM8 localization, implicating nuclear bodies in FSGS and developmental brain disease.

Bibliographical data

Original languageEnglish
ISSN0002-9297
DOIs
Publication statusPublished - 04.02.2021
Externally publishedYes

Comment Deanary

Copyright © 2021 American Society of Human Genetics. Published by Elsevier Inc. All rights reserved.

PubMed 33508234