MLK3 regulates bone development downstream of the faciogenital dysplasia protein FGD1 in mice.

TitleMLK3 regulates bone development downstream of the faciogenital dysplasia protein FGD1 in mice.
Publication TypeJournal Article
Year of Publication2011
AuthorsZou W, Greenblatt MB, Shim J-H, Kant S, Zhai B, Lotinun S, Brady N, Hu DZhang, Gygi SP, Baron R, Davis RJ, Jones D, Glimcher LH
JournalJ Clin Invest
Volume121
Issue11
Pagination4383-92
Date Published2011 Nov
ISSN1558-8238
KeywordsAnimals, Bone Development, cdc42 GTP-Binding Protein, Disease Models, Animal, Dwarfism, Enzyme Activation, Face, Female, Gene Knock-In Techniques, Genetic Diseases, X-Linked, Genitalia, Male, Guanine Nucleotide Exchange Factors, Hand Deformities, Congenital, Heart Defects, Congenital, Humans, Male, MAP Kinase Kinase Kinases, MAP Kinase Signaling System, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Mutant Strains, Mutation, Osteoblasts, p38 Mitogen-Activated Protein Kinases, Proteins
Abstract

Mutations in human FYVE, RhoGEF, and PH domain-containing 1 (FGD1) cause faciogenital dysplasia (FGDY; also known as Aarskog syndrome), an X-linked disorder that affects multiple skeletal structures. FGD1 encodes a guanine nucleotide exchange factor (GEF) that specifically activates the Rho GTPase CDC42. However, the mechanisms by which mutations in FGD1 affect skeletal development are unknown. Here, we describe what we believe to be a novel signaling pathway in osteoblasts initiated by FGD1 that involves the MAP3K mixed-lineage kinase 3 (MLK3). We observed that MLK3 functions downstream of FGD1 to regulate ERK and p38 MAPK, which in turn phosphorylate and activate the master regulator of osteoblast differentiation, Runx2. Mutations in FGD1 found in individuals with FGDY ablated its ability to activate MLK3. Consistent with our description of this pathway and the phenotype of patients with FGD1 mutations, mice with a targeted deletion of Mlk3 displayed multiple skeletal defects, including dental abnormalities, deficient calvarial mineralization, and reduced bone mass. Furthermore, mice with knockin of a mutant Mlk3 allele that is resistant to activation by FGD1/CDC42 displayed similar skeletal defects, demonstrating that activation of MLK3 specifically by FGD1/CDC42 is important for skeletal mineralization. Thus, our results provide a putative biochemical mechanism for the skeletal defects in human FGDY and suggest that modulating MAPK signaling may benefit these patients.

DOI10.1172/JCI59041
Alternate JournalJ Clin Invest
PubMed ID21965325
Grant ListK99AR055668 / AR / NIAMS NIH HHS / United States
/ HHMI / Howard Hughes Medical Institute / United States
HD55601 / HD / NICHD NIH HHS / United States
R01-CA065861 / CA / NCI NIH HHS / United States
Related Faculty: 
Matthew B. Greenblatt, M.D., Ph.D.

Pathology & Laboratory Medicine 1300 York Avenue New York, NY 10065 Phone: (212) 746-6464
Surgical Pathology: (212) 746-2700