4.6 Article

Hyperactive Transforming Growth Factor-β1 Signaling Potentiates Skeletal Defects in a Neurofibromatosis Type 1 Mouse Model

Journal

JOURNAL OF BONE AND MINERAL RESEARCH
Volume 28, Issue 12, Pages 2476-2489

Publisher

WILEY
DOI: 10.1002/jbmr.1992

Keywords

TRANSFORMING GROWTH FACTOR-BETA; TGF-; SMAD; NEUROFIBROMATOSIS TYPE 1; FRACTURE NONUNION; OSTEOPOROSIS

Funding

  1. U.S. Department of Defense (DOD) [NF043032, NF073112]
  2. March of Dimes [YF08-246]
  3. Children's Tumor Foundation [2011-01-010]
  4. Indiana Clinical and Translational Sciences Institute PHS NCCR [5TL1RR025759-03]

Ask authors/readers for more resources

Dysregulated transforming growth factor beta (TGF-) signaling is associated with a spectrum of osseous defects as seen in Loeys-Dietz syndrome, Marfan syndrome, and Camurati-Engelmann disease. Intriguingly, neurofibromatosis type 1 (NF1) patients exhibit many of these characteristic skeletal features, including kyphoscoliosis, osteoporosis, tibial dysplasia, and pseudarthrosis; however, the molecular mechanisms mediating these phenotypes remain unclear. Here, we provide genetic and pharmacologic evidence that hyperactive TGF-1 signaling pivotally underpins osseous defects in Nf1(flox/-);Col2.3Cre mice, a model which closely recapitulates the skeletal abnormalities found in the human disease. Compared to controls, we show that serum TGF-1 levels are fivefold to sixfold increased both in Nf1(flox/-);Col2.3Cre mice and in a cohort of NF1 patients. Nf1-deficient osteoblasts, the principal source of TGF-1 in bone, overexpress TGF-1 in a gene dosage-dependent fashion. Moreover, Nf1-deficient osteoblasts and osteoclasts are hyperresponsive to TGF-1 stimulation, potentiating osteoclast bone resorptive activity while inhibiting osteoblast differentiation. These cellular phenotypes are further accompanied by p21-Ras-dependent hyperactivation of the canonical TGF-1-Smad pathway. Reexpression of the human, full-length neurofibromin guanosine triphosphatase (GTPase)-activating protein (GAP)-related domain (NF1 GRD) in primary Nf1-deficient osteoblast progenitors, attenuated TGF-1 expression levels and reduced Smad phosphorylation in response to TGF-1 stimulation. As an in vivo proof of principle, we demonstrate that administration of the TGF- receptor 1 (TRI) kinase inhibitor, SD-208, can rescue bone mass deficits and prevent tibial fracture nonunion in Nf1(flox/-);Col2.3Cre mice. In sum, these data demonstrate a pivotal role for hyperactive TGF-1 signaling in the pathogenesis of NF1-associated osteoporosis and pseudarthrosis, thus implicating the TGF- signaling pathway as a potential therapeutic target in the treatment of NF1 osseous defects that are refractory to current therapies. (c) 2013 American Society for Bone and Mineral Research.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available