Inviting an author to review:
Find an author and click ‘Invite to review selected article’ near their name.
Search for authorsSearch for similar articles
13
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      PIN1 is a new therapeutic target of craniosynostosis

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Gain-of-function mutations in fibroblast growth factor receptors (FGFRs) cause congenital skeletal anomalies, including craniosynostosis (CS), which is characterized by the premature closure of craniofacial sutures. Apert syndrome (AS) is one of the severest forms of CS, and the only treatment is surgical expansion of prematurely fused sutures in infants. Previously, we demonstrated that the prolyl isomerase peptidyl-prolyl cistrans isomerase interacting 1 (PIN1) plays a critical role in mediating FGFR signaling and that Pin1 +/− mice exhibit delayed closure of cranial sutures. In this study, using both genetic and pharmacological approaches, we tested whether PIN1 modulation could be used as a therapeutic regimen against AS. In the genetic approach, we crossbred Fgfr2 S252W/+ , a mouse model of AS, and Pin1 +/− mice. Downregulation of Pin1 gene dosage attenuated premature cranial suture closure and other phenotypes of AS in Fgfr2 S252W/+ mutant mice. In the pharmacological approach, we intraperitoneally administered juglone, a PIN1 enzyme inhibitor, to pregnant Fgfr2 S252W/+ mutant mice and found that this treatment successfully interrupted fetal development of AS phenotypes. Primary cultured osteoblasts from Fgfr2 S252W/+ mutant mice expressed high levels of FGFR2 downstream target genes, but this phenotype was attenuated by PIN1 inhibition. Post-translational stabilization and activation of Runt-related transcription factor 2 (RUNX2) in Fgfr2 S252W/+ osteoblasts were also attenuated by PIN1 inhibition. Based on these observations, we conclude that PIN1 enzyme activity is important for FGFR2-induced RUNX2 activation and craniofacial suture morphogenesis. Moreover, these findings highlight that juglone or other PIN1 inhibitors represent viable alternatives to surgical intervention for treatment of CS and other hyperostotic diseases.

          Related collections

          Most cited references72

          • Record: found
          • Abstract: found
          • Article: not found

          Efficient in vivo manipulation of mouse genomic sequences at the zygote stage.

          We describe a transgenic mouse line carrying the cre transgene under the control of the adenovirus EIIa promoter that targets expression of the Cre recombinase to the early mouse embryo. To assess the ability of this recombinase to excise loxP-flanked DNA sequences at early stages of development, we bred EIIa-cre transgenic mice to two different mouse lines carrying loxP-flanked target sequences: (i) a strain with a single gene-targeted neomycin resistance gene flanked by 1oxP sites and (ii) a transgenic line carrying multiple transgene copies with internal loxP sites. Mating either of these loxP-carrying mouse lines to EIIa-cre mice resulted in first generation progeny in which the loxP-flanked sequences had been efficiently deleted from all tissues tested, including the germ cells. Interbreeding of these first generation progeny resulted in efficient germ-line transmission of the deletion to subsequent generations. These results demonstrate a method by which loxP-flanked DNA sequences can be efficiently deleted in the early mouse embryo. Potential applications of this approach are discussed, including reduction of multicopy transgene loci to produce single-copy transgenic lines and introduction of a variety of subtle mutations into the line.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Tissue origins and interactions in the mammalian skull vault.

            During mammalian evolution, expansion of the cerebral hemispheres was accompanied by expansion of the frontal and parietal bones of the skull vault and deployment of the coronal (fronto-parietal) and sagittal (parietal-parietal) sutures as major growth centres. Using a transgenic mouse with a permanent neural crest cell lineage marker, Wnt1-Cre/R26R, we show that both sutures are formed at a neural crest-mesoderm interface: the frontal bones are neural crest-derived and the parietal bones mesodermal, with a tongue of neural crest between the two parietal bones. By detailed analysis of neural crest migration pathways using X-gal staining, and mesodermal tracing by DiI labelling, we show that the neural crest-mesodermal tissue juxtaposition that later forms the coronal suture is established at E9.5 as the caudal boundary of the frontonasal mesenchyme. As the cerebral hemispheres expand, they extend caudally, passing beneath the neural crest-mesodermal interface within the dermis, carrying with them a layer of neural crest cells that forms their meningeal covering. Exposure of embryos to retinoic acid at E10.0 reduces this meningeal neural crest and inhibits parietal ossification, suggesting that intramembranous ossification of this mesodermal bone requires interaction with neural crest-derived meninges, whereas ossification of the neural crest-derived frontal bone is autonomous. These observations provide new perspectives on skull evolution and on human genetic abnormalities of skull growth and ossification.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Regulation of osteoblast differentiation by Runx2.

              Runx2 protein is first detected in preosteoblasts, and the expression is upregulated in immature osteoblasts, but downregulated in mature osteoblasts. Runx2 is the first transcription factor required for determination of the osteoblast lineage, while Sp7 and canonical Wnt-signaling further direct the fate of mesenchymal cells to osteoblasts, blocking their differentiation into chondrocytes. Runx2 induces the differentiation of multipotent mesenchymal cells into immature osteoblasts, directing the formation of immature bone, but Runx2 inhibits osteoblast maturation and mature bone formation. Normally, the protein level of Runx2 in osteoblasts reduces during bone development, and osteoblasts acquire mature phenotypes, which are required for mature bone formation. Furthermore, Runx2 triggers the expression of major bone matrix genes during the early stages of osteoblast differentiation, but Runx2 is not essential for the maintenance of these gene expressions in mature osteoblasts.
                Bookmark

                Author and article information

                Journal
                Hum Mol Genet
                Hum. Mol. Genet
                hmg
                Human Molecular Genetics
                Oxford University Press
                0964-6906
                1460-2083
                15 November 2018
                11 July 2018
                11 July 2018
                : 27
                : 22
                : 3827-3839
                Affiliations
                [1 ]BK21 Program, Department of Molecular Genetics and Dental Pharmacology and Therapeutics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
                [2 ]Department of Periodontology, School of Dentistry, Seoul National University, Seoul, Republic of Korea
                [3 ]Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
                Author notes
                To whom correspondence should be addressed at: Department of Molecular Genetics and Dental Pharmacology, School of Dentistry, Seoul National University, Room 404, Building #86, 1 Kwanak-ro, Kwanak-gu, Seoul 088-26, Republic of Korea. Tel: +82 28802320; Fax: +82 27413103; Email: hmryoo@ 123456snu.ac.kr
                Article
                ddy252
                10.1093/hmg/ddy252
                6216213
                30007339
                80b3881d-ef5f-49c2-a725-a88355ba2773
                ©The Author(s) 2018. Published by Oxford University Press.

                This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License ( http://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@oup.com

                History
                : 16 January 2018
                : 23 May 2018
                : 5 July 2018
                Page count
                Pages: 13
                Funding
                Funded by: National Research Foundation of Korea 10.13039/501100003725
                Award ID: NRF-2014R1A2A2A01004865
                Award ID: NRF-2017R1A2B3011778
                Categories
                General Article

                Genetics
                Genetics

                Comments

                Comment on this article