PUBLICATION

Bi-allelic premature truncating variants in LTBP1 cause cutis laxa syndrome

Authors
Pottie, L., Adamo, C.S., Beyens, A., Lütke, S., Tapaneeyaphan, P., De Clercq, A., Salmon, P.L., De Rycke, R., Gezdirici, A., Gulec, E.Y., Khan, N., Urquhart, J.E., Newman, W.G., Metcalfe, K., Efthymiou, S., Maroofian, R., Anwar, N., Maqbool, S., Rahman, F., Altweijri, I., Alsaleh, M., Abdullah, S.M., Al-Owain, M., Hashem, M., Houlden, H., Alkuraya, F.S., Sips, P., Sengle, G., Callewaert, B.
ID
ZDB-PUB-210516-3
Date
2021
Source
American journal of human genetics   108(6): 1095-1114 (Journal)
Registered Authors
Callewaert, Bert, Sips, Patrick
Keywords
Danio rerio, LTBP1, transforming growth factor beta, collagen fibrillogenesis, craniosynostosis, cutis laxa syndrome, extracellular matrix, short stature, tissue mineral density
MeSH Terms
  • Adolescent
  • Alleles
  • Animals
  • Cells, Cultured
  • Child
  • Child, Preschool
  • Collagen/metabolism*
  • Cutis Laxa/etiology*
  • Cutis Laxa/pathology
  • Extracellular Matrix/metabolism
  • Female
  • Fibroblasts/metabolism
  • Fibroblasts/pathology
  • Genetic Variation*
  • Humans
  • Infant
  • Latent TGF-beta Binding Proteins/genetics*
  • Male
  • Pedigree
  • Skin/metabolism
  • Skin/pathology
  • Zebrafish
PubMed
33991472 Full text @ Am. J. Hum. Genet.
Abstract
Latent transforming growth factor β (TGFβ)-binding proteins (LTBPs) are microfibril-associated proteins essential for anchoring TGFβ in the extracellular matrix (ECM) as well as for correct assembly of ECM components. Variants in LTBP2, LTBP3, and LTBP4 have been identified in several autosomal recessive Mendelian disorders with skeletal abnormalities with or without impaired development of elastin-rich tissues. Thus far, the human phenotype associated with LTBP1 deficiency has remained enigmatic. In this study, we report homozygous premature truncating LTBP1 variants in eight affected individuals from four unrelated consanguineous families. Affected individuals present with connective tissue features (cutis laxa and inguinal hernia), craniofacial dysmorphology, variable heart defects, and prominent skeletal features (craniosynostosis, short stature, brachydactyly, and syndactyly). In vitro studies on proband-derived dermal fibroblasts indicate distinct molecular mechanisms depending on the position of the variant in LTBP1. C-terminal variants lead to an altered LTBP1 loosely anchored in the microfibrillar network and cause increased ECM deposition in cultured fibroblasts associated with excessive TGFβ growth factor activation and signaling. In contrast, N-terminal truncation results in a loss of LTBP1 that does not alter TGFβ levels or ECM assembly. In vivo validation with two independent zebrafish lines carrying mutations in ltbp1 induce abnormal collagen fibrillogenesis in skin and intervertebral ligaments and ectopic bone formation on the vertebrae. In addition, one of the mutant zebrafish lines shows voluminous and hypo-mineralized vertebrae. Overall, our findings in humans and zebrafish show that LTBP1 function is crucial for skin and bone ECM assembly and homeostasis.
Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping