#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

FGFR3 Deficiency Causes Multiple Chondroma-like Lesions by Upregulating Hedgehog Signaling


Benign cartilaginous tumors, especially osteochondromas and enchondromas, are the most common primary bone tumors in humans. Several hereditary diseases are characterized by the development of cartilaginous tumors, including hereditary multiple exostoses, metachondromatosis, and enchondromatosis, which are caused by mutations in genes such as exostosin 1 and 2, tyrosine protein phosphatase non-receptor type 11, parathyroid hormone receptor 1, and isocitrate dehydrogenase 1 and 2. The proteins encoded by these genes are directly or indirectly linked to fibroblast growth factor (FGF) signaling. In addition, osteochondroma was found in several members of a family with camptodactyly, tall stature, and hearing loss syndrome, a rare inherited disorder caused by a heterozygous missense mutation in Fgfr3. In this study, we found that Fgfr3 deficiency leads to the formation of cartilaginous tumors, including osteochondromas and enchondromas, likely due to dysregulated endochondral ossification in growth plates. We also show that cartilaginous tumorigenesis in Fgfr3-deficient mice results from excessive Indian hedgehog production mediated by the activation of mitogen-associated protein kinase signaling. Based on these results, we propose a model for cartilaginous tumor development in which FGFR3 functions as a tumor suppressor. Our findings also suggest that modulation of FGFR3 and related signaling pathways is a potential therapeutic strategy for treating benign cartilaginous tumors.


Vyšlo v časopise: FGFR3 Deficiency Causes Multiple Chondroma-like Lesions by Upregulating Hedgehog Signaling. PLoS Genet 11(6): e32767. doi:10.1371/journal.pgen.1005214
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1005214

Souhrn

Benign cartilaginous tumors, especially osteochondromas and enchondromas, are the most common primary bone tumors in humans. Several hereditary diseases are characterized by the development of cartilaginous tumors, including hereditary multiple exostoses, metachondromatosis, and enchondromatosis, which are caused by mutations in genes such as exostosin 1 and 2, tyrosine protein phosphatase non-receptor type 11, parathyroid hormone receptor 1, and isocitrate dehydrogenase 1 and 2. The proteins encoded by these genes are directly or indirectly linked to fibroblast growth factor (FGF) signaling. In addition, osteochondroma was found in several members of a family with camptodactyly, tall stature, and hearing loss syndrome, a rare inherited disorder caused by a heterozygous missense mutation in Fgfr3. In this study, we found that Fgfr3 deficiency leads to the formation of cartilaginous tumors, including osteochondromas and enchondromas, likely due to dysregulated endochondral ossification in growth plates. We also show that cartilaginous tumorigenesis in Fgfr3-deficient mice results from excessive Indian hedgehog production mediated by the activation of mitogen-associated protein kinase signaling. Based on these results, we propose a model for cartilaginous tumor development in which FGFR3 functions as a tumor suppressor. Our findings also suggest that modulation of FGFR3 and related signaling pathways is a potential therapeutic strategy for treating benign cartilaginous tumors.


Zdroje

1. Bovee JV, Hogendoorn PC, Wunder JS, Alman BA (2010) Cartilage tumours and bone development: molecular pathology and possible therapeutic targets. Nat Rev Cancer 10: 481–488. doi: 10.1038/nrc2869 20535132

2. Romeo S, Hogendoorn PC, Dei Tos AP (2009) Benign cartilaginous tumors of bone: from morphology to somatic and germ-line genetics. Adv Anat Pathol 16: 307–315. doi: 10.1097/PAP.0b013e3181b506a1 19700940

3. Garcia RA, Inwards CY, Unni KK (2011) Benign bone tumors—recent developments. Seminars in Diagnostic Pathology 28: 73–85. 21675379

4. Schwartz HS, Zimmerman N, Simon M, Wroble R, Millar E, et al. (1987) The malignant potential of enchondromatosis. The Journal of Bone & Joint Surgery 69: 269–274.

5. Jennes I, Pedrini E, Zuntini M, Mordenti M, Balkassmi S, et al. (2009) Multiple osteochondromas: mutation update and description of the multiple osteochondromas mutation database (MOdb). Hum Mutat 30: 1620–1627. doi: 10.1002/humu.21123 19810120

6. Huegel J, Sgariglia F, Enomoto-Iwamoto M, Koyama E, Dormans JP, et al. (2013) Heparan sulfate in skeletal development, growth, and pathology: The case of hereditary multiple exostoses. Dev Dyn.

7. Zak BM, Schuksz M, Koyama E, Mundy C, Wells DE, et al. (2011) Compound heterozygous loss of Ext1 and Ext2 is sufficient for formation of multiple exostoses in mouse ribs and long bones. Bone 48: 979–987. doi: 10.1016/j.bone.2011.02.001 21310272

8. Jones KB, Piombo V, Searby C, Kurriger G, Yang B, et al. (2010) A mouse model of osteochondromagenesis from clonal inactivation of Ext1 in chondrocytes. Proc Natl Acad Sci U S A 107: 2054–2059. doi: 10.1073/pnas.0910875107 20080592

9. Matsumoto K, Irie F, Mackem S, Yamaguchi Y (2010) A mouse model of chondrocyte-specific somatic mutation reveals a role for Ext1 loss of heterozygosity in multiple hereditary exostoses. Proc Natl Acad Sci U S A 107: 10932–10937. doi: 10.1073/pnas.0914642107 20534475

10. Huegel J, Mundy C, Sgariglia F, Nygren P, Billings PC, et al. (2013) 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 377: 100–112. doi: 10.1016/j.ydbio.2013.02.008 23458899

11. Hopyan S, Gokgoz N, Poon R, Gensure RC, Yu C, et al. (2002) A mutant PTH/PTHrP type I receptor in enchondromatosis. Nat Genet 30: 306–310. 11850620

12. Tiet TD, Hopyan S, Nadesan P, Gokgoz N, Poon R, et al. (2006) Constitutive hedgehog signaling in chondrosarcoma up-regulates tumor cell proliferation. Am J Pathol 168: 321–330. 16400033

13. Rozeman LB, Hameetman L, Cleton-Jansen AM, Taminiau AH, Hogendoorn PC, et al. (2005) Absence of IHH and retention of PTHrP signalling in enchondromas and central chondrosarcomas. J Pathol 205: 476–482. 15685701

14. Amary MF, Bacsi K, Maggiani F, Damato S, Halai D, et al. (2011) IDH1 and IDH2 mutations are frequent events in central chondrosarcoma and central and periosteal chondromas but not in other mesenchymal tumours. J Pathol 224: 334–343. doi: 10.1002/path.2913 21598255

15. Amary MF, Damato S, Halai D, Eskandarpour M, Berisha F, et al. (2011) Ollier disease and Maffucci syndrome are caused by somatic mosaic mutations of IDH1 and IDH2. Nat Genet 43: 1262–1265. doi: 10.1038/ng.994 22057236

16. Pansuriya TC, van Eijk R, d'Adamo P, van Ruler MA, Kuijjer ML, et al. (2011) Somatic mosaic IDH1 and IDH2 mutations are associated with enchondroma and spindle cell hemangioma in Ollier disease and Maffucci syndrome. Nat Genet 43: 1256–1261. doi: 10.1038/ng.1004 22057234

17. Hirata M, Sasaki M, Cairns RA, Inoue S, Puviindran V, et al. (2015) MutantIDHis sufficient to initiate enchondromatosis in mice. Proceedings of the National Academy of Sciences 112: 2829–2834. doi: 10.1073/pnas.1424400112 25730874

18. Sobreira NL, Cirulli ET, Avramopoulos D, Wohler E, Oswald GL, et al. (2010) Whole-genome sequencing of a single proband together with linkage analysis identifies a Mendelian disease gene. PLoS Genet 6: e1000991. doi: 10.1371/journal.pgen.1000991 20577567

19. Bowen ME, Boyden ED, Holm IA, Campos-Xavier B, Bonafe L, et al. (2011) Loss-of-function mutations in PTPN11 cause metachondromatosis, but not Ollier disease or Maffucci syndrome. PLoS Genet 7: e1002050. doi: 10.1371/journal.pgen.1002050 21533187

20. Grossmann KS, Rosario M, Birchmeier C, Birchmeier W (2010) The tyrosine phosphatase Shp2 in development and cancer. Adv Cancer Res 106: 53–89. doi: 10.1016/S0065-230X(10)06002-1 20399956

21. Bauler TJ, Kamiya N, Lapinski PE, Langewisch E, Mishina Y, et al. (2011) Development of severe skeletal defects in induced SHP-2-deficient adult mice: a model of skeletal malformation in humans with SHP-2 mutations. Dis Model Mech 4: 228–239. doi: 10.1242/dmm.006130 21068439

22. Lapinski PE, Meyer MF, Feng GS, Kamiya N, King PD (2013) Deletion of SHP-2 in mesenchymal stem cells causes growth retardation, limb and chest deformity, and calvarial defects in mice. Dis Model Mech 6: 1448–1458. doi: 10.1242/dmm.012849 24077964

23. Yang W, Wang J, Moore DC, Liang H, Dooner M, et al. (2013) Ptpn11 deletion in a novel progenitor causes metachondromatosis by inducing hedgehog signalling. Nature.

24. Bowen ME, Ayturk UM, Kurek KC, Yang W, Warman ML (2014) SHP2 Regulates Chondrocyte Terminal Differentiation, Growth Plate Architecture and Skeletal Cell Fates. PLoS Genet 10: e1004364. doi: 10.1371/journal.pgen.1004364 24875294

25. Kronenberg HM (2006) PTHrP and skeletal development. Annals of the New York Academy of Sciences 1068: 1–13. 16831900

26. Sgariglia F, Candela ME, Huegel J, Jacenko O, Koyama E, et al. (2013) Epiphyseal abnormalities, trabecular bone loss and articular chondrocyte hypertrophy develop in the long bones of postnatal Ext1-deficient mice. Bone 57: 220–231. doi: 10.1016/j.bone.2013.08.012 23958822

27. Foldynova-Trantirkova S, Wilcox WR, Krejci P (2012) Sixteen years and counting: the current understanding of fibroblast growth factor receptor 3 (FGFR3) signaling in skeletal dysplasias. Hum Mutat 33: 29–41. doi: 10.1002/humu.21636 22045636

28. Chen L, Adar R, Yang X, Monsonego EO, Li C, et al. (1999) Gly369Cys mutation in mouse FGFR3 causes achondroplasia by affecting both chondrogenesis and osteogenesis. Journal of Clinical Investigation 104: 1517–1525. 10587515

29. Chen L, Li C, Qiao W, Xu X, Deng C (2001) A Ser365→ Cys mutation of fibroblast growth factor receptor 3 in mouse downregulates Ihh/PTHrP signals and causes severe achondroplasia. Human molecular genetics 10: 457–465. 11181569

30. Iwata T, Chen L, Li C, Ovchinnikov DA, Behringer RR, et al. (2000) A neonatal lethal mutation in FGFR3 uncouples proliferation and differentiation of growth plate chondrocytes in embryos. Hum Mol Genet 9: 1603–1613. 10861287

31. Toydemir RM, Brassington AE, Bayrak-Toydemir P, Krakowiak PA, Jorde LB, et al. (2006) A novel mutation in FGFR3 causes camptodactyly, tall stature, and hearing loss (CATSHL) syndrome. Am J Hum Genet 79: 935–941. 17033969

32. Makrythanasis P, Temtamy S, Aglan MS, Otaify GA, Hamamy H, et al. (2014) A novel homozygous mutation in FGFR3 causes tall stature, severe lateral tibial deviation, scoliosis, hearing impairment, camptodactyly and arachnodactyly. Hum Mutat.

33. Deng C, Wynshaw-Boris A, Zhou F, Kuo A, Leder P (1996) Fibroblast growth factor receptor 3 is a negative regulator of bone growth. Cell 84: 911–921. 8601314

34. Colvin JS, Bohne BA, Harding GW, McEwen DG, Ornitz DM (1996) Skeletal overgrowth and deafness in mice lacking fibroblast growth factor receptor 3. Nat Genet 12: 390–397. 8630492

35. Liu Z, Xu J, Colvin JS, Ornitz DM (2002) Coordination of chondrogenesis and osteogenesis by fibroblast growth factor 18. Genes Dev 16: 859–869. 11937493

36. Goetz R, Mohammadi M (2013) Exploring mechanisms of FGF signalling through the lens of structural biology. Nat Rev Mol Cell Biol 14: 166–180. doi: 10.1038/nrm3528 23403721

37. Krejci P, Bryja V, Pachernik J, Hampl A, Pogue R, et al. (2004) FGF2 inhibits proliferation and alters the cartilage-like phenotype of RCS cells. Experimental cell research 297: 152–164. 15194433

38. Krejci P, Masri B, Salazar L, Farrington-Rock C, Prats H, et al. (2007) Bisindolylmaleimide I suppresses fibroblast growth factor-mediated activation of Erk MAP kinase in chondrocytes by preventing Shp2 association with the Frs2 and Gab1 adaptor proteins. Journal of biological chemistry 282: 2929–2936. 17145761

39. Legeai-Mallet L, Benoist-Lasselin C, Delezoide AL, Munnich A, Bonaventure J (1998) Fibroblast Growth Factor Receptor 3 Mutations Promote Apoptosis but Do Not Alter Chondrocyte Proliferation in Thanatophoric Dysplasia. Journal of Biological Chemistry 273: 13007–13014. 9582336

40. L'Hote CG, Knowles MA (2005) Cell responses to FGFR3 signalling: growth, differentiation and apoptosis. Exp Cell Res 304: 417–431. 15748888

41. Harada D, Yamanaka Y, Ueda K, Nishimura R, Morishima T, et al. (2007) Sustained phosphorylation of mutated FGFR3 is a crucial feature of genetic dwarfism and induces apoptosis in the ATDC5 chondrogenic cell line via PLCgamma-activated STAT1. Bone 41: 273–281. 17561467

42. Krejci P, Prochazkova J, Smutny J, Chlebova K, Lin P, et al. (2010) FGFR3 signaling induces a reversible senescence phenotype in chondrocytes similar to oncogene-induced premature senescence. Bone 47: 102–110. doi: 10.1016/j.bone.2010.03.021 20362703

43. Amizuka N, Davidson D, Liu H, Valverde-Franco G, Chai S, et al. (2004) Signalling by fibroblast growth factor receptor 3 and parathyroid hormone-related peptide coordinate cartilage and bone development. Bone 34: 13–25. 14751559

44. Michigami T (2013) Regulatory mechanisms for the development of growth plate cartilage. Cell Mol Life Sci 70: 4213–4221. doi: 10.1007/s00018-013-1346-9 23640571

45. Long F, Ornitz DM (2013) Development of the endochondral skeleton. Cold Spring Harb Perspect Biol 5: a008334. doi: 10.1101/cshperspect.a008334 23284041

46. Naski MC, Wang Q, Xu J, Ornitz DM (1996) Graded activation of fibroblast growth factor receptor 3 by mutations causing achondroplasia and thanatophoric dysplasia. Nat Genet 13: 233–237. 8640234

47. Li C, Chen L, Iwata T, Kitagawa M, Fu X-Y, et al. (1999) A Lys644Glu substitution in fibroblast growth factor receptor 3 (FGFR3) causes dwarfism in mice by activation of STATs and ink4 cell cycle inhibitors. Human molecular genetics 8: 35–44. 9887329

48. Kim HK, Feng GS, Chen D, King PD, Kamiya N (2013) Targeted disruption of Shp2 in chondrocytes leads to metachondromatosis with multiple cartilaginous protrusions. J Bone Miner Res.

49. Xie J, Bartels CM, Barton SW, Gu D (2013) Targeting hedgehog signaling in cancer: research and clinical developments. OncoTargets and therapy 6: 1425. doi: 10.2147/OTT.S34678 24143114

50. Rudin CM, Hann CL, Laterra J, Yauch RL, Callahan CA, et al. (2009) Treatment of medulloblastoma with hedgehog pathway inhibitor GDC-0449. New England Journal of Medicine 361: 1173–1178. doi: 10.1056/NEJMoa0902903 19726761

51. Krejci P (2014) The paradox of FGFR3 signaling in skeletal dysplasia: Why chondrocytes growth arrest while other cells over proliferate. Mutation Research/Reviews in Mutation Research 759: 40–48. doi: 10.1016/j.mrrev.2013.11.001 24295726

52. Ho L, Stojanovski A, Whetstone H, Wei QX, Mau E, et al. (2009) Gli2 and p53 cooperate to regulate IGFBP-3- mediated chondrocyte apoptosis in the progression from benign to malignant cartilage tumors. Cancer Cell 16: 126–136. doi: 10.1016/j.ccr.2009.05.013 19647223

53. Jones KB, Pacifici M, Hilton MJ (2014) Multiple Hereditary Exostoses (MHE): Elucidating the Pathogenesis of a Rare Skeletal Disorder Through Interdisciplinary Research. Connective tissue research: 1–26.

54. De Bari C, Dell'Accio F, Luyten FP (2001) Human periosteum-derived cells maintain phenotypic stability and chondrogenic potential throughout expansion regardless of donor age. Arthritis Rheum 44: 85–95. 11212180

55. Ito Y, Fitzsimmons JS, Sanyal A, Mello MA, Mukherjee N, et al. (2001) Localization of chondrocyte precursors in periosteum. Osteoarthritis Cartilage 9: 215–223. 11300744

56. Superti-Furga A, Spranger J, Nishimura G (2012) Enchondromatosis revisited: new classification with molecular basis. Am J Med Genet C Semin Med Genet 160C: 154–164. doi: 10.1002/ajmg.c.31331 22791316

57. Zelzer E, Mamluk R, Ferrara N, Johnson RS, Schipani E, et al. (2004) VEGFA is necessary for chondrocyte survival during bone development. Development 131: 2161–2171. 15073147

58. Sebastian A, Matsushita T, Kawanami A, Mackem S, Landreth GE, et al. (2011) Genetic inactivation of ERK1 and ERK2 in chondrocytes promotes bone growth and enlarges the spinal canal. J Orthop Res 29: 375–379. doi: 10.1002/jor.21262 20922792

59. Matsushita T, Chan YY, Kawanami A, Balmes G, Landreth GE, et al. (2009) Extracellular signal-regulated kinase 1 (ERK1) and ERK2 play essential roles in osteoblast differentiation and in supporting osteoclastogenesis. Molecular and cellular biology 29: 5843–5857. doi: 10.1128/MCB.01549-08 19737917

60. Murakami S, Balmes G, McKinney S, Zhang Z, Givol D, et al. (2004) Constitutive activation of MEK1 in chondrocytes causes Stat1-independent achondroplasia-like dwarfism and rescues the Fgfr3-deficient mouse phenotype. Genes Dev 18: 290–305. 14871928

61. Maeda Y, Nakamura E, Nguyen MT, Suva LJ, Swain FL, et al. (2007) Indian Hedgehog produced by postnatal chondrocytes is essential for maintaining a growth plate and trabecular bone. Proc Natl Acad Sci U S A 104: 6382–6387. 17409191

62. Brechbiel JL, Ng JM, Curran T (2011) PTHrP treatment fails to rescue bone defects caused by Hedgehog pathway inhibition in young mice. Toxicologic pathology 39: 478–485. doi: 10.1177/0192623311399788 21411723

63. Su N, Xu X, Li C, He Q, Zhao L, et al. (2010) Generation of Fgfr3 conditional knockout mice. International journal of biological sciences 6: 327. 20582225

64. Zhu M, Chen M, Lichtler A, O'Keefe R, Chen D (2008) Tamoxifen-inducible Cre-recombination in articular chondrocytes of adult Col2a1-CreERT2 transgenic mice. Osteoarthritis and Cartilage 16: 129–130. 17888690

65. Yang G, Sun Q, Teng Y, Li F, Weng T, et al. (2008) PTEN deficiency causes dyschondroplasia in mice by enhanced hypoxia-inducible factor 1alpha signaling and endoplasmic reticulum stress. Development 135: 3587–3597. doi: 10.1242/dev.028118 18832389

66. Arango NA, Lovell-Badge R, Behringer RR (1999) Targeted mutagenesis of the endogenous mouse Mis gene promoter: in vivo definition of genetic pathways of vertebrate sexual development. Cell 99: 409–419. 10571183

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2015 Číslo 6
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#