89
Views
0
CrossRef citations to date
0
Altmetric
Review

Hereditary multiple exostoses: are there new plausible treatment strategies?

Pages 385-391 | Received 25 Mar 2018, Accepted 29 May 2018, Published online: 07 Jun 2018

References

  • Jones KB. Glycobiology and the growth plate: current concepts in Multiple Hereditary Exostoses. J Pediatr Orthop. 2011;31:577–586.
  • Porter DE, Simpson AHRW. The neoplastic pathogenesis of solitary and multiple osteochondromas. J Pathol. 1999;188:119–125.
  • Stieber JR, Dormans JP. Manifestations of hereditary multiple exostoses. J Am Acad Orthop Surg. 2005;13:110–120.
  • Porter DE, Lonie L, Fraser M, et al. Severity of disease and risk in malignant change in hereditary multiple exostoses. J Bone Joint Surg Br. 2004;86:1041–1046.
  • Huegel J, Sgariglia F, Enomoto-Iwamoto M, et al. Heparan sulfate in skeletal development, growth, and pathology: the case of Hereditary Multiple exostoses. Dev Dyn. 2013;242:1021–1032.
  • Pacifici M. The pathogenic roles of heparan sulfate deficiency in hereditary multiple exostoses. Matrix Biol. 2018.
  • Cheung PK, McCormick C, Crawford BE, et al. Etiological point mutations in the hereditary multiple exostoses gene EXT1: a functional analysis of heparan sulfate polymerase activity. Am J Hum Genet. 2001;69:55–66.
  • Hecht JT, Hogue D, Strong LC, et al. Hereditary multiple exostosis and chondrosarcoma: linkage to chromosome 11 and loss of heterozygosity for EXT-linked markers on chromosome 11 and 8. Am J Hum Genet. 1995;56:1125–1131.
  • Anower-E-Khuda MF, Matsumoto K, Habuchi H, et al. Glycosaminoglycans in the blood of hereditary multiple exostoses patients: half reduction of heparan sulfate to chondroitin sulfate ratio and the possible diagnostic application. Glycobiology. 2013;23:865–876.
  • Bishop JR, Schuksz M, Esko JD. Heparan sulphate proteoglycans fine-tune mammalian physiology. Nature. 2007;446:1030–1037.
  • Billings PC, Pacifici M. Interactions of signaling proteins, growth factors and other proteins with heparan sulfate: mechanisms and mysteries. Connect Tissue Res. 2015;56:272–280.
  • Sarrazin S, Lamanna WC, Esko JD. Heparan sulfate proteoglycans. Cold Spring Harb Prospect Biol. 2011;3:a004952.
  • Mooij HL, BernelotMoens SJ, Gordts PL, et al. Ext1 heterozygosity causes a modest effect on postprandial lipid clearance in humans. J Lipid Res. 2015;56:665–673.
  • Goud AL, de Lange J, Scholtes VA, et al. Pain, physical and social functioning, and quality of life in individuals with multiple hereditary exostoses in The Netherlands: a national cohort study. J Bone Joint Surg Am. 2012;94:1013–1020.
  • Knudson AG. Hereditary cancer: two hits revisited. J Cancer Res Clin Oncol. 1996;122:135–140.
  • Bovee JV, Cleton-Jansen A-M, Wuyts W, et al. EXT-mutation analysis and loss of heterozygosity in sporadic and hereditary osteochondromas and secondary chondrosarcoma. Am J Hum Genet. 1999;65:689–698.
  • Hecht JT, Hayes E, Haynes R, et al. Differentiation-induced loss of heparan sulfate in human exostosis derived chondrocytes. Differentiation. 2005;73:212–221.
  • Jones KB, Piombo V, Searby C, et al. A mouse model of osteochondromagenesis from clonal inactivation of Ext1 in chondrocytes. Proc Natl Acad Sci USA. 2010;107:2054–2059.
  • Matsumoto K, Irie F, Mackem S, et al. A mouse model of chondrocyte-specific somatic mutation reveals a role for Ext1 loss of heterozygosity in multiple hereditary exostoses. Proc Natl Acad Sci USA. 2010;107:10932–10937.
  • Huegel J, Mundy C, Sgariglia F, et al. Perichondrium phenotype and border function are regulated by Ext1 and heparan sulfate in developing long bones: a mechanism likely deranged in Hereditary Multiple exostoses. Dev Biol. 2013;377:100–112.
  • Sgariglia F, Candela ME, Huegel J, et al. Epiphyseal abnormalities, trabecular bone loss and articular chondrocyte hypertrophy develop in the long bones of postnatal Ext1-deficient mice. Bone. 2013;57:220–231.
  • Bovee JV. EXTra hit for mouse osteochondroma. Proc Natl Acad Sci USA. 2010;107:1813–1814.
  • Salazar VS, Gamer LW, Rosen V. BMP signaling in skeletal development, disease and repair. Nat Endocrinol. 2016;12:203–221.
  • Sinha R, Mundy C, Bechtold T, et al. Unsuspected osteochondroma-like outgrowths in the cranial base of Hereditary Multiple exostoses patients and modeling and treatment with a BMP antagonist in mice. PLoS Genetics. 2017;13:e1006742.
  • Ohkawara B, Iemura S, ten Dijke P, et al. Action range of BMP is defined by its N-terminal basic amino acid core. Curr Biol. 2002;12:205–209.
  • Ruppert R, Hoffmann E, Sebald W. Human bone morphogenetic protein 2 contains a heparin-binding site which modifies its biological activity. Eur J Biochem. 1996;237:295–302.
  • Inubushi T, Nazawa S, Matsumoto K, et al. Aberrant perichondrial BMP signaling mediates multiple osteochondromagenesis in mice. J Clin Inv Insight. 2017;2:e90049.
  • Yu PB, Deng DY, Lai CS, et al. BMP type I receptor inhibition reduces heterotopic ossification. Nat Med. 2008;14:1363–1369.
  • Boergermann JH, Kopf J, Yu PB, et al. Dorsomorphin and LDN-193189 inhibit BMP-mediated samd, p38 and Akt signaling in C2C12 cells. Int J Biochem Cell Biol. 2010;42:1802–1807.
  • Ao A, Hao J, Hopkins CR, et al. DMH1, a novel BMP small molecule inhibitor, increases cardiomyocyte progenitors and promotes cardiac differentiation in mouse embryonic stem cells. PLoS ONE. 2012;7:e41627.
  • Vogt J, Traynor R, Sapkota GP. The specificities of small molecule inhibitors of the TFGβ and BMP pathways. Cell Signal. 2011;23:1831–1842.
  • Bandyopadhyay A, Kubilus JK, Crochiere ML, et al. Identification of unique molecular subdomains in the perichondrium and periosteum and their role in regulating gene expression in the underlying chondrocytes. Dev Biol. 2008;321:162–174.
  • Colnot C, Lu C, Hu D, et al. Distinguishing the contributions of the perichondrium, cartilage, and vascular endothelium to skeletal development. Dev Biol. 2004;269:55–69.
  • Buckland RA, Collinson JM, Graham E, et al. Antagonistic effects of FGF4 on BMP induction of apoptosis and chondrogenesis in the chick limb bud. Mech Dev. 1998;71:143–150.
  • Yoon BS, Pogue R, Ovchinnikov DA, et al. BMPs regulate multiple aspects of growth plate chondrogenesis through opposing actions of FGF pathways. Development. 2006;133:4667–4678.
  • Matsushita T, Chan YY, Kawanami A, et al. Extracellular signal-regulated kinase 1 (ERK1) and ERK2 play essential roles in osteoblast differentiation and in supporting osteoclastogenesis. Mol Cell Biol. 2009;29:5843–5857.
  • Day TF, Guo X, Garrett-Beal L, et al. Wnt/β-catenin signaling in mesenchymal progenitors controls osteoblast and chondrocyte differentiation during vertebrate skeletogenesis. Dev Cell. 2005;8:739–750.
  • Hill TP, Spater D, Taketo MM, et al. Canonical Wnt/β-catenin signaling prevents osteoblasts from differentiating into chondrocytes. Dev Cell. 2005;8:727–738.
  • Zhou S, Xie Y, Tang J, et al. FGFR3 deficiency causes multiple chondroma-like lesions by upregulating hedgehog signaling. PLoS Genetics. 2015;11:e1005214.
  • Wehenkel M, Corr M, Guy CS, et al. Extracellular signal-regulated kinase signaling in CD4-expressing cells inhibits osteochondromas. Front Immunol. 2017;8:1–11.
  • Ornitz DM, Marie PJ. Fibroblast growth factor signaling in skeletal development and disease. Genes Dev. 2015;29:1463–1486.
  • Belov AA, Mohammadi M. Molecular mechanisms of fibroblast growth factor signaling in physiology and pathology. Cold Spring Harb Prospect Biol. 2013;5:1–24.
  • Li S, Christensen C, Kohler LB, et al. Agonists of fibroblast growth factor receptor induce neurite outgrowth and survival of cerebellar granule neurons. Develop Neurobiol. 2009;69:837–854.
  • Ge H, Tan L, Wu P, et al. Poly-L-ornithine promotes preferred differentiation of neural stem/progenitor cells via ERK signaling pathway. Sci Rep. 2015;5:15535.
  • Vortkamp A, Lee K, Lanske B, et al. Regulation of rate of cartilage differentiation by Indian hedgehog and PTH-related protein. Science. 1996;273:613–622.
  • Koyama E, Leatherman JL, Noji S, et al. Early chick limb cartilaginous elements possess polarizing activity and express Hedgehog-related morphogenetic factors. Dev Dynam. 1996;207(3):344–354.
  • Koziel L, Kunath M, Kelly OG, et al. Ext1-dependent heparan sulfate regulates the range of Ihh signaling during endochondral ossification. Dev Cell. 2004;6:801–813.
  • Koyama E, Young B, Nagayama M, et al. Conditional Kif3a ablation causes abnormal hedgehog signaling topography, growth plate dysfunction, and excessive bone and cartilage formation during mouse skeletogenesis. Development. 2007;134:2159–2169.
  • Xie J, Bartels CM, Barton SW, et al. Targeting hedgehog signaling in cancer: research and clinical developments. Onco Targets Ther. 2013;6:1425.
  • Mundy C, Bello A, Sgariglia F, et al. HhAntag, a hedgehog signaling antagonist, suppresses chondrogenesis and modulates canonical and non-canonical BMP signaling. J Cell Physiol. 2016;231:1033–1044.
  • Silapunt S, Chen L, Midgen MR. Hedgehog pathway inhibition in advanced basal cell carcinoma: latest evidence and clinical usefulness. Ther Adv Med Oncol. 2016;8:375–382.
  • Fux L, Ilan N, Sanderson RD, et al. Heparanase: busy at the cell surface. Trends Sci. 2009;34:511–519.
  • Vreys V, David G. Mammalian heparanase: what is the message? J Cell Mol Med. 2007;11:427–452.
  • Patel VN, Knox SM, Likar KM, et al. Heparanase cleavage of perlecan heparan sulfate modulates FGF10 activity during ex vivo submandibular gland branching morphogenesis. Development. 2007;134:4177–4186.
  • Trebicz-Geffen M, Robinson D, Evron Z, et al. The molecular and cellular basis of exostosis formation in hereditary multiple exostoses. Int J Exp Path. 2008;89:321–331.
  • Wang Y, Yang X, Yamagata S, et al. Involvement of Ext and heparanase in migration of mouse FBJ osteosarcoma cells. Mol Cell Biochem. 2013;373:63–72.
  • Vlodavsky I, Friedmann Y. Molecula properties and involvement of heparanase in cancer metastasis and angiogenesis. J Clin, Inv. 2001;108:341–447.
  • Huegel J, Enomoto-Iwamoto M, Sgariglia F, et al. Heparanase stimulates chondrogenesis and is up-regulated in human ectopic cartilage. A mechanism possibly involved in Hereditary Multiple exostoses. Am J Path. 2015;185:1676–1685.
  • Ritchie JP, Ramani VC, Ren Y, et al. SST0001, a chemically modified heparin, inhibits myeloma growth and angiogenesis via disruption of the heparanase/syndecan-1 shedding: a novel mechanism for stimulation of tumor growth and metastasis. Clin Cancer Res. 2011;17:1382–1393.
  • Cassinelli G, Lanzi C, Tortoreto M, et al. Antitumor efficacy of the heparanase inhibitor SST0001 alone or in combination with antiangiogenic agents in the treatment of human pediatric sarcoma models. Biochem Pharmacol. 2013;85:1424–1432.
  • Phan AQ, Pacifici M, Esko JD. Advances in the pathogenesis and possible treatments for multiple hereditary exostoses from the 2016 international MHE conference. Conn Tissue Res. 2018;59:85–98.
  • Wang Z, Hsieh PH, Xu Y, et al. Synthesis of 3-O-sulphated oligosaccharides to understand the relationship between structures and functions of heparan sulfate. J Am Chem Soc. 2017;139:5249–5256.
  • Miller RL, Wei W, Schworer R, et al. Composition, sequencing and ion mobility mass spectrometry of heparan sulfate-like octasaccahrides isomers differing in glucuronic and iduronic acid content. Eur J Mass Spectrometry. 2015;21:245–254.
  • Schultz V, Suflita M, Liu X, et al. Heparan sulfate domains required for fibroblast growth factor 1 and 2 signaling through fibroblast growth factor receptor 1c. J Biol Chem. 2017;292:2495–2509.
  • Evans RM, Mangelsdorf DJ. Nuclear receptors, RXR, and the big bang. Cell. 2014;157:255–266.
  • Sucov HM, Evans RM. Retinoic acid and retinoic acid receptors in development. Mol Neurobiol. 1995;10:169–184.
  • Hoffman LM, Garcha K, Karamboulas K, et al. BMP action in skeletogenesis involves attenuation of retinoid signaling. J Cell Biol. 2006;174:101–113.
  • Weston AD, Chandraratna RAS, Torchia J, et al. Requirement for RAR-mediated gene repression in skeletal progenitor differentiation. J Cell Biol. 2002;158:39–51.
  • Pacifici M, Cossu G, Molinaro M, et al. Vitamin A inhibits chondrogenesis but not myogenesis. Exp Cell Res. 1980;129:469–474.
  • Shimono K, Tung W-E, Macolino C, et al. Potent inhibition of heterotopic ossification by nuclear retinoic acid receptor-γ agonists. Nature Med. 2011;17:454–460.
  • Pacifici M. Retinoid roles and action in skeletal development and growth provide the rationale for an ongoing heterotopic ossification prevention trial. Bone. 2018;109:267–275.
  • Inubushi T, Lemire I, Irie F, et al. Palovarotene inhibits osteochondroma formation in a mouse model of multiple hereditary exostoses. J Bone Min Res. 2018;33:658–666.
  • Long C, Amoasii L, Mireault AA, et al. Postnatal genome editing partially restores dystrophin expression in a mous emodel of muscular dystrophy. Science. 2016;351:400–403.
  • Zuntini M, Pedrini E, Parra A, et al. Genetic models of osteochondroma onset and neoplastic progression: evidence for mechanisms alternative to EXT genes inactivation. Oncogene. 2010;29:3827–3834.
  • Ge X, Tsang K, He L, et al. NFAT restricts osteochondroma formation from entheseal progenitors. J Clin Inv Insight. 2016;1:e86254.
  • Nilsson O, Parker EA, Hedge A, et al. Gradients of bone morphogenetic protein-related gene expression across the growth plate. J Endocrinol. 2007;193:75–84.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.