Your browser doesn't support javascript.
loading
Dominant negative variants in KIF5B cause osteogenesis imperfecta via down regulation of mTOR signaling.
Marom, Ronit; Zhang, Bo; Washington, Megan E; Song, I-Wen; Burrage, Lindsay C; Rossi, Vittoria C; Berrier, Ava S; Lindsey, Anika; Lesinski, Jacob; Nonet, Michael L; Chen, Jian; Baldridge, Dustin; Silverman, Gary A; Sutton, V Reid; Rosenfeld, Jill A; Tran, Alyssa A; Hicks, M John; Murdock, David R; Dai, Hongzheng; Weis, MaryAnn; Jhangiani, Shalini N; Muzny, Donna M; Gibbs, Richard A; Caswell, Richard; Pottinger, Carrie; Cilliers, Deirdre; Stals, Karen; Eyre, David; Krakow, Deborah; Schedl, Tim; Pak, Stephen C; Lee, Brendan H.
Afiliação
  • Marom R; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America.
  • Zhang B; Texas Children's Hospital, Houston, Texas, United States of America.
  • Washington ME; Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri, United States of America.
  • Song IW; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America.
  • Burrage LC; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America.
  • Rossi VC; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America.
  • Berrier AS; Texas Children's Hospital, Houston, Texas, United States of America.
  • Lindsey A; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America.
  • Lesinski J; Texas Children's Hospital, Houston, Texas, United States of America.
  • Nonet ML; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America.
  • Chen J; Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri, United States of America.
  • Baldridge D; Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri, United States of America.
  • Silverman GA; Department of Neuroscience, Washington University School of Medicine, St Louis, Missouri, United States of America.
  • Sutton VR; Department of Genetics, Washington University School of Medicine, St Louis, Missouri, United States of America.
  • Rosenfeld JA; Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri, United States of America.
  • Tran AA; Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri, United States of America.
  • Hicks MJ; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America.
  • Murdock DR; Texas Children's Hospital, Houston, Texas, United States of America.
  • Dai H; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America.
  • Weis M; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America.
  • Jhangiani SN; Texas Children's Hospital, Houston, Texas, United States of America.
  • Muzny DM; Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America.
  • Gibbs RA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America.
  • Caswell R; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America.
  • Pottinger C; Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America.
  • Cilliers D; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America.
  • Stals K; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America.
  • Eyre D; Exeter Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom.
  • Krakow D; All Wales Medical Genomics Service, Wrexham Maelor Hospital, Wrexham, UK.
  • Schedl T; Oxford Centre for Genomic Medicine, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom.
  • Pak SC; Exeter Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom.
PLoS Genet ; 19(11): e1011005, 2023 Nov.
Article em En | MEDLINE | ID: mdl-37934770
ABSTRACT

BACKGROUND:

Kinesin motor proteins transport intracellular cargo, including mRNA, proteins, and organelles. Pathogenic variants in kinesin-related genes have been implicated in neurodevelopmental disorders and skeletal dysplasias. We identified de novo, heterozygous variants in KIF5B, encoding a kinesin-1 subunit, in four individuals with osteogenesis imperfecta. The variants cluster within the highly conserved kinesin motor domain and are predicted to interfere with nucleotide binding, although the mechanistic consequences on cell signaling and function are unknown.

METHODS:

To understand the in vivo genetic mechanism of KIF5B variants, we modeled the p.Thr87Ile variant that was found in two patients in the C. elegans ortholog, unc-116, at the corresponding position (Thr90Ile) by CRISPR/Cas9 editing and performed functional analysis. Next, we studied the cellular and molecular consequences of the recurrent p.Thr87Ile variant by microscopy, RNA and protein analysis in NIH3T3 cells, primary human fibroblasts and bone biopsy.

RESULTS:

C. elegans heterozygous for the unc-116 Thr90Ile variant displayed abnormal body length and motility phenotypes that were suppressed by additional copies of the wild type allele, consistent with a dominant negative mechanism. Time-lapse imaging of GFP-tagged mitochondria showed defective mitochondria transport in unc-116 Thr90Ile neurons providing strong evidence for disrupted kinesin motor function. Microscopy studies in human cells showed dilated endoplasmic reticulum, multiple intracellular vacuoles, and abnormal distribution of the Golgi complex, supporting an intracellular trafficking defect. RNA sequencing, proteomic analysis, and bone immunohistochemistry demonstrated down regulation of the mTOR signaling pathway that was partially rescued with leucine supplementation in patient cells.

CONCLUSION:

We report dominant negative variants in the KIF5B kinesin motor domain in individuals with osteogenesis imperfecta. This study expands the spectrum of kinesin-related disorders and identifies dysregulated signaling targets for KIF5B in skeletal development.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Osteogênese Imperfeita / Cinesinas Limite: Animals / Humans Idioma: En Revista: PLoS Genet Assunto da revista: GENETICA Ano de publicação: 2023 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Osteogênese Imperfeita / Cinesinas Limite: Animals / Humans Idioma: En Revista: PLoS Genet Assunto da revista: GENETICA Ano de publicação: 2023 Tipo de documento: Article País de afiliação: Estados Unidos