リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「The Regulation of Bone Metabolism and Disorders by Wnt Signaling」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

The Regulation of Bone Metabolism and Disorders by Wnt Signaling

Maeda, Kazuhiro Kobayashi, Yasuhiro Koide, Masanori Uehara, Shunsuke Okamoto, Masanori Ishihara, Akihiro Kayama, Tomohiro Saito, Mitsuhiro Marumo, Keishi 信州大学 DOI:31698687

2022.12.28

概要

Wnt, a secreted glycoprotein, has an approximate molecular weight of 40 kDa, and it is a cytokine involved in various biological phenomena including ontogeny, morphogenesis, carcinogenesis, and maintenance of stem cells. The Wnt signaling pathway can be classified into two main pathways: canonical and non-canonical. Of these, the canonical Wnt signaling pathway promotes osteogenesis. Sclerostin produced by osteocytes is an inhibitor of this pathway, thereby inhibiting osteogenesis. Recently, osteoporosis treatment using an anti-sclerostin therapy has been introduced. In this review, the basics of Wnt signaling, its role in bone metabolism and its involvement in skeletal disorders have been covered. Furthermore, the clinical significance and future scopes of Wnt signaling in osteoporosis, osteoarthritis, rheumatoid arthritis and neoplasia are discussed.

参考文献

1. Nusse, R.; van Ooyen, A.; Cox, D.; Fung, Y.K.; Varmus, H. Mode of proviral activation of a putative mammary oncogene (int-1) on mouse chromosome 15. Nature 1984, 307, 131–136. [CrossRef]

2. Sharma, R.P.; Chopra, V.L. Effect of the Wingless (wg1) mutation on wing and haltere development in Drosophila melanogaster. Dev. Biol. 1976, 48, 461–465. [CrossRef]

3. Rijsewijk, F.; Schuermann, M.; Wagenaar, E.; Parren, P.; Weigel, D.; Nusse, R. The Drosophila homolog of the mouse mammary oncogene int-1 is identical to the segment polarity gene wingless. Cell 1987, 50, 649–657. [CrossRef]

4. Gong, Y.; Slee, R.B.; Fukai, N.; Rawadi, G.; Roman-Roman, S.; Reginato, A.M.; Wang, H.; Cundy, T.; Glorieux, F.H.; Lev, D.; et al. LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development. Cell 2001, 107, 513–523. [CrossRef]

5. Nusse, R.; Clevers, H. Wnt/beta-Catenin Signaling, Disease, and Emerging Therapeutic Modalities. Cell 2017, 169, 985–999. [CrossRef] [PubMed]

6. Baron, R.; Kneissel, M. WNT signaling in bone homeostasis and disease: From human mutations to treatments. Nat. Med. 2013, 19, 179–192. [CrossRef] [PubMed]

7. Maeda, K.; Takahashi, N.; Kobayashi, Y. Roles of Wnt signals in bone resorption during physiological and pathological states. J. Mol. Med. 2013, 91, 15–23. [CrossRef] [PubMed]

8. Ke, J.; Xu, H.E.; Williams, B.O. Lipid modification in Wnt structure and function. Curr. Opin. Lipidol. 2013, 24, 129–133. [CrossRef] [PubMed]

9. Bartscherer, K.; Pelte, N.; Ingelfinger, D.; Boutros, M. Secretion of Wnt ligands requires Evi, a conserved transmembrane protein. Cell 2006, 125, 523–533. [CrossRef] [PubMed]

10. Banziger, C.; Soldini, D.; Schutt, C.; Zipperlen, P.; Hausmann, G.; Basler, K. Wntless, a conserved membrane protein dedicated to the secretion of Wnt proteins from signaling cells. Cell 2006, 125, 509–522. [CrossRef]

11. Karner, C.M.; Long, F. Wnt signaling and cellular metabolism in osteoblasts. Cell. Mol. Life Sci. 2017, 74, 1649–1657. [CrossRef] [PubMed]

12. Katoh, M. Multilayered prevention and treatment of chronic inflammation, organ fibrosis and cancer associated with canonical WNT/betacatenin signaling activation (Review). Int. J. Mol. Med. 2018, 42, 713–725. [PubMed]

13. Pai, S.G.; Carneiro, B.A.; Mota, J.M.; Costa, R.; Leite, C.A.; Barroso-Sousa, R.; Kaplan, J.B.; Chae, Y.K.; Giles, F.J. Wnt/beta-catenin pathway: Modulating anticancer immune response. J. Hematol. Oncol. 2017, 10, 101. [CrossRef] [PubMed]

14. Johnson, M.L.; Harnish, K.; Nusse, R.; Van Hul, W. LRP5 and Wnt signaling: A union made for bone. J. Bone Miner. Res. 2004, 19, 1749–1757. [CrossRef]

15. Endo, M.; Nishita, M.; Fujii, M.; Minami, Y. Insight into the role of Wnt5a-induced signaling in normal and cancer cells. Int. Rev. Cell Mol. Biol. 2015, 314, 117–148.

16. Cruciat, C.M.; Niehrs, C. Secreted and transmembrane wnt inhibitors and activators. Cold Spring Harb. Perspect. Biol. 2013, 5, a015081. [CrossRef]

17. Glinka, A.; Wu, W.; Delius, H.; Monaghan, A.P.; Blumenstock, C.; Niehrs, C. Dickkopf-1 is a member of a new family of secreted proteins and functions in head induction. Nature 1998, 391, 357–362. [CrossRef]

18. Balemans, W.; Ebeling, M.; Patel, N.; Van Hul, E.; Olson, P.; Dioszegi, M.; Lacza, C.; Wuyts, W.; Van Den Ende, J.; Willems, P.; et al. Increased bone density in sclerosteosis is due to the deficiency of a novel secreted protein (SOST). Hum. Mol. Genet. 2001, 10, 537–543. [CrossRef]

19. Brunkow, M.E.; Gardner, J.C.; Van Ness, J.; Paeper, B.W.; Kovacevich, B.R.; Proll, S.; Skonier, J.E.; Zhao, L.; Sabo, P.J.; Fu, Y.; et al. Bone dysplasia sclerosteosis results from loss of the SOST gene product, a novel cystine knot-containing protein. Am. J. Hum. Genet. 2001, 68, 577–589. [CrossRef]

20. Van Lierop, A.H.; Appelman-Dijkstra, N.M.; Papapoulos, S.E. Sclerostin deficiency in humans. Bone 2017, 96, 51–62. [CrossRef]

21. Sapir-Koren, R.; Livshits, G. Osteocyte control of bone remodeling: Is sclerostin a key molecular coordinator of the balanced bone resorption-formation cycles? Osteoporos. Int. 2014, 25, 2685–2700. [CrossRef] [PubMed]

22. Dallas, S.L.; Prideaux, M.; Bonewald, L.F. The osteocyte: An endocrine cell... and more. Endocr. Rev. 2013, 34, 658–690. [CrossRef] [PubMed]

23. O’Brien, C.A.; Nakashima, T.; Takayanagi, H. Osteocyte control of osteoclastogenesis. Bone 2013, 54, 258–263. [CrossRef] [PubMed]

24. Goldring, S.R. The osteocyte: Key player in regulating bone turnover. RMD Open 2015, 1, e000049. [CrossRef]

25. Leupin, O.; Piters, E.; Halleux, C.; Hu, S.; Kramer, I.; Morvan, F.; Bouwmeester, T.; Schirle, M.; Bueno-Lozano, M.; Fuentes, F.J.; et al. Bone overgrowth-associated mutations in the LRP4 gene impair sclerostin facilitator function. J. Biol. Chem. 2011, 286, 19489–19500. [CrossRef]

26. Kim, N.; Stiegler, A.L.; Cameron, T.O.; Hallock, P.T.; Gomez, A.M.; Huang, J.H.; Hubbard, S.R.; Dustin, M.L.; Burden, S.J. Lrp4 is a receptor for Agrin and forms a complex with MuSK. Cell 2008, 135, 334–342. [CrossRef]

27. Lara-Castillo, N.; Johnson, M.L. LRP receptor family member associated bone disease. Rev. Endocr. Metab. Disord. 2015, 16, 141–148. [CrossRef]

28. Van Wesenbeeck, L.; Cleiren, E.; Gram, J.; Beals, R.K.; Benichou, O.; Scopelliti, D.; Key, L.; Renton, T.; Bartels, C.; Gong, Y.; et al. Six novel missense mutations in the LDL receptor-related protein 5 (LRP5) gene in different conditions with an increased bone density. Am. J. Hum. Genet. 2003, 72, 763–771. [CrossRef]

29. Koide, M.; Kobayashi, Y. Regulatory mechanisms of sclerostin expression during bone remodeling. J. Bone Miner. Metab. 2019, 37, 9–17. [CrossRef]

30. Balemans, W.; Patel, N.; Ebeling, M.; Van Hul, E.; Wuyts, W.; Lacza, C.; Dioszegi, M.; Dikkers, F.G.; Hildering, P.; Willems, P.J.; et al. Identification of a 52 kb deletion downstream of the SOST gene in patients with van Buchem disease. J. Med. Genet. 2002, 39, 91–97. [CrossRef]

31. Li, Y.; Pawlik, B.; Elcioglu, N.; Aglan, M.; Kayserili, H.; Yigit, G.; Percin, F.; Goodman, F.; Nurnberg, G.; Cenani, A.; et al. LRP4 mutations alter Wnt/beta-catenin signaling and cause limb and kidney malformations in Cenani-Lenz syndrome. Am. J. Hum. Genet. 2010, 86, 696–706. [CrossRef] [PubMed]

32. Afzal, M.; Zaman, Q.; Kornak, U.; Mundlos, S.; Malik, S.; Flottmann, R. Novel splice mutation in LRP4 causes severe type of Cenani-Lenz syndactyly syndrome with oro-facial and skeletal symptoms. Eur. J. Med. Genet. 2017, 60, 421–425. [CrossRef] [PubMed]

33. Kiper, P.O.S.; Saito, H.; Gori, F.; Unger, S.; Hesse, E.; Yamana, K.; Kiviranta, R.; Solban, N.; Liu, J.; Brommage, R.; et al. Cortical-Bone Fragility—Insights from sFRP4 Deficiency in Pyle’s Disease. N. Engl. J. Med. 2016, 374, 2553–2562. [CrossRef] [PubMed]

34. Szenker-Ravi, E.; Altunoglu, U.; Leushacke, M.; Bosso-Lefevre, C.; Khatoo, M.; Tran, H.T.; Naert, T.; Noelanders, R.; Hajamohideen, A.; Beneteau, C.; et al. RSPO2 inhibition of RNF43 and ZNRF3 governs limb development independently of LGR4/5/6. Nature 2018, 557, 564–569. [CrossRef]

35. Grzeschik, K.H.; Bornholdt, D.; Oeffner, F.; Konig, A.; del Carmen Boente, M.; Enders, H.; Fritz, B.; Hertl, M.; Grasshoff, U.; Hofling, K.; et al. Deficiency of PORCN, a regulator of Wnt signaling, is associated with focal dermal hypoplasia. Nat. Genet. 2007, 39, 833–835. [CrossRef]

36. Laine, C.M.; Joeng, K.S.; Campeau, P.M.; Kiviranta, R.; Tarkkonen, K.; Grover, M.; Lu, J.T.; Pekkinen, M.; Wessman, M.; Heino, T.J.; et al. WNT1 mutations in early-onset osteoporosis and osteogenesis imperfecta. N. Engl. J. Med. 2013, 368, 1809–1816. [CrossRef]

37. Fahiminiya, S.; Majewski, J.; Mort, J.; Moffatt, P.; Glorieux, F.H.; Rauch, F. Mutations in WNT1 are a cause of osteogenesis imperfecta. J. Med. Genet. 2013, 50, 345–348. [CrossRef]

38. Keupp, K.; Beleggia, F.; Kayserili, H.; Barnes, A.M.; Steiner, M.; Semler, O.; Fischer, B.; Yigit, G.; Janda, C.Y.; Becker, J.; et al. Mutations in WNT1 cause different forms of bone fragility. Am. J. Hum. Genet. 2013, 92, 565–574. [CrossRef]

39. Pyott, S.M.; Tran, T.T.; Leistritz, D.F.; Pepin, M.G.; Mendelsohn, N.J.; Temme, R.T.; Fernandez, B.A.; Elsayed, S.M.; Elsobky, E.; Verma, I.; et al. WNT1 mutations in families affected by moderately severe and progressive recessive osteogenesis imperfecta. Am. J. Hum. Genet. 2013, 92, 590–597. [CrossRef]

40. Aldinger, K.A.; Mendelsohn, N.J.; Chung, B.H.; Zhang, W.; Cohn, D.H.; Fernandez, B.; Alkuraya, F.S.; Dobyns, W.B.; Curry, C.J. Variable brain phenotype primarily affects the brainstem and cerebellum in patients with osteogenesis imperfecta caused by recessive WNT1 mutations. J. Med. Genet. 2016, 53, 427–430. [CrossRef]

41. Kantaputra, P.N.; Sirirungruangsarn, Y.; Visrutaratna, P.; Petcharunpaisan, S.; Carlson, B.M.; Intachai, W.; Sudasna, J.; Kampuansai, J.; Dejkhamron, P. WNT1-associated osteogenesis imperfecta with atrophic frontal lobes and arachnoid cysts. J. Hum. Genet. 2019, 64, 291–296. [CrossRef] [PubMed]

42. Patton, M.A.; Afzal, A.R. Robinow syndrome. J. Med. Genet. 2002, 39, 305–310. [CrossRef] [PubMed]

43. Roifman, M.; Marcelis, C.L.; Paton, T.; Marshall, C.; Silver, R.; Lohr, J.L.; Yntema, H.G.; Venselaar, H.; Kayserili, H.; van Bon, B.; et al. De novo WNT5A-associated autosomal dominant Robinow syndrome suggests specificity of genotype and phenotype. Clin. Genet. 2015, 87, 34–41. [CrossRef] [PubMed]

44. Lintern, K.B.; Guidato, S.; Rowe, A.; Saldanha, J.W.; Itasaki, N. Characterization of wise protein and its molecular mechanism to interact with both Wnt and BMP signals. J. Biol. Chem. 2009, 284, 23159–23168. [CrossRef] [PubMed]

45. Hao, H.X.; Xie, Y.; Zhang, Y.; Charlat, O.; Oster, E.; Avello, M.; Lei, H.; Mickanin, C.; Liu, D.; Ruffner, H.; et al. ZNRF3 promotes Wnt receptor turnover in an R-spondin-sensitive manner. Nature 2012, 485, 195–200. [CrossRef] [PubMed]

46. Koo, B.K.; Spit, M.; Jordens, I.; Low, T.Y.; Stange, D.E.; van de Wetering, M.; van Es, J.H.; Mohammed, S.; Heck, A.J.; Maurice, M.M.; et al. Tumour suppressor RNF43 is a stem-cell E3 ligase that induces endocytosis of Wnt receptors. Nature 2012, 488, 665–669. [CrossRef] [PubMed]

47. De Lau, W.; Peng, W.C.; Gros, P.; Clevers, H. The R-spondin/Lgr5/Rnf43 module: Regulator of Wnt signal strength. Genes Dev. 2014, 28, 305–316. [CrossRef]

48. Bell, S.M.; Schreiner, C.M.; Wert, S.E.; Mucenski, M.L.; Scott, W.J.; Whitsett, J.A. R-spondin 2 is required for normal laryngeal-tracheal, lung and limb morphogenesis. Development 2008, 135, 1049–1058. [CrossRef]

49. Okamoto, K.; Nakashima, T.; Shinohara, M.; Negishi-Koga, T.; Komatsu, N.; Terashima, A.; Sawa, S.; Nitta, T.; Takayanagi, H. Osteoimmunology: The Conceptual Framework Unifying the Immune and Skeletal Systems. Physiol. Rev. 2017, 97, 1295–1349. [CrossRef]

50. Bennett, C.N.; Longo, K.A.; Wright, W.S.; Suva, L.J.; Lane, T.F.; Hankenson KDMacDougald, O.A. Regulation of osteoblastogenesis and bone mass by Wnt10b. Proc. Natl. Acad. Sci. USA 2005, 102, 3324–3329. [CrossRef]

51. Bennett, C.N.; Ouyang, H.; Ma, Y.L.; Zeng, Q.; Gerin, I.; Sousa, K.M.; Lane, T.F.; Krishnan, V.; Hankenson, K.D.; MacDougald, O.A. Wnt10b increases postnatal bone formation by enhancing osteoblast differentiation. J. Bone Miner. Res. 2007, 22, 1924–1932. [CrossRef] [PubMed]

52. Visweswaran, M.; Pohl, S.; Arfuso, F.; Newsholme, P.; Dilley, R.; Pervaiz, S.; Dharmarajan, A. Multi-lineage differentiation of mesenchymal stem cells—To Wnt, or not Wnt. Int. J. Biochem. Cell Biol. 2015, 68, 139–147. [CrossRef] [PubMed]

53. Nakanishi, R.; Akiyama, H.; Kimura, H.; Otsuki, B.; Shimizu, M.; Tsuboyama, T.; Nakamura, T. Osteoblast-targeted expression of Sfrp4 in mice results in low bone mass. J. Bone Miner. Res. 2008, 23, 271–277. [CrossRef] [PubMed]

54. Chang, M.K.; Kramer, I.; Huber, T.; Kinzel, B.; Guth-Gundel, S.; Leupin, O.; Kneissel, M. Disruption of Lrp4 function by genetic deletion or pharmacological blockade increases bone mass and serum sclerostin levels. Proc. Natl. Acad. Sci. USA 2014, 111, E5187–E5195. [CrossRef] [PubMed]

55. Xiong, L.; Jung, J.U.; Wu, H.; Xia, W.F.; Pan, J.X.; Shen, C.; Mei, L.; Xiong, W.C. Lrp4 in osteoblasts suppresses bone formation and promotes osteoclastogenesis and bone resorption. Proc. Natl. Acad. Sci. USA 2015, 112, 3487–3492. [CrossRef] [PubMed]

56. Shen, C.; Xiong, W.C.; Mei, L. LRP4 in neuromuscular junction and bone development and diseases. Bone 2015, 80, 101–108. [CrossRef] [PubMed]

57. Kato, M.; Patel, M.S.; Levasseur, R.; Lobov, I.; Chang, B.H.; Glass, D.A., II; Hartmann, C.; Li, L.; Hwang, T.H.; Brayton, C.F.; et al. Cbfa1-independent decrease in osteoblast proliferation, osteopenia, and persistent embryonic eye vascularization in mice deficient in Lrp5, a Wnt coreceptor. J. Cell Biol. 2002, 157, 303–314. [CrossRef] [PubMed]

58. Yadav, V.K.; Ryu, J.H.; Suda, N.; Tanaka, K.F.; Gingrich, J.A.; Schutz, G.; Glorieux, F.H.; Chiang, C.Y.; Zajac, J.D.; Insogna, K.L.; et al. Lrp5 controls bone formation by inhibiting serotonin synthesis in the duodenum. Cell 2008, 135, 825–837. [CrossRef]

59. Cui, Y.; Niziolek, P.J.; MacDonald, B.T.; Zylstra, C.R.; Alenina, N.; Robinson, D.R.; Zhong, Z.; Matthes, S.; Jacobsen, C.M.; Conlon, R.A.; et al. Lrp5 functions in bone to regulate bone mass. Nat. Med. 2011, 17, 684–691. [CrossRef]

60. Riddle, R.C.; Diegel, C.R.; Leslie, J.M.; Van Koevering, K.K.; Faugere, M.C.; Clemens, T.L.; Williams, B.O. Lrp5 and Lrp6 exert overlapping functions in osteoblasts during postnatal bone acquisition. PLoS ONE 2013, 8, e63323. [CrossRef]

61. Weivoda, M.M.; Ruan, M.; Hachfeld, C.M.; Pederson, L.; Howe, A.; Davey, R.A.; Zajac, J.D.; Kobayashi, Y.; Williams, B.O.; Westendorf, J.J.; et al. Wnt Signaling Inhibits Osteoclast Differentiation by Activating Canonical and Noncanonical cAMP/PKA Pathways. J. Bone Miner. Res. 2016, 31, 65–75. [CrossRef] [PubMed]

62. Albers, J.; Keller, J.; Baranowsky, A.; Beil, F.T.; Catala-Lehnen, P.; Schulze, J.; Amling, M.; Schinke, T. Canonical Wnt signaling inhibits osteoclastogenesis independent of osteoprotegerin. J. Cell Biol. 2013, 200, 537–549. [CrossRef] [PubMed]

63. Albers, J.; Schulze, J.; Beil, F.T.; Gebauer, M.; Baranowsky, A.; Keller, J.; Marshall, R.P.; Wintges, K.; Friedrich, F.W.; Priemel, M.; et al. Control of bone formation by the serpentine receptor Frizzled-9. J. Cell Biol. 2011, 192, 1057–1072. [CrossRef] [PubMed]

64. Maeda, K.; Kobayashi, Y.; Udagawa, N.; Uehara, S.; Ishihara, A.; Mizoguchi, T.; Kikuchi, Y.; Takada, I.; Kato, S.; Kani, S.; et al. Wnt5a-Ror2 signaling between osteoblast-lineage cells and osteoclast precursors enhances osteoclastogenesis. Nat. Med. 2012, 18, 405–412. [CrossRef] [PubMed]

65. Uehara, S.; Udagawa, N.; Mukai, H.; Ishihara, A.; Maeda, K.; Yamashita, T.; Murakami, K.; Nishita, M.; Nakamura, T.; Kato, S.; et al. Protein kinase N3 promotes bone resorption by osteoclasts in response to Wnt5a-Ror2 signaling. Sci. Signal. 2017, 10. [CrossRef]

66. Li, X.; Ominsky, M.S.; Niu, Q.T.; Sun, N.; Daugherty, B.; D’Agostin, D.; Kurahara, C.; Gao, Y.; Cao, J.; Gong, J.; et al. Targeted deletion of the sclerostin gene in mice results in increased bone formation and bone strength. J. Bone Miner. Res. 2008, 23, 860–869. [CrossRef]

67. Haraguchi, R.; Kitazawa, R.; Mori, K.; Tachibana, R.; Kiyonari, H.; Imai, Y.; Abe, T.; Kitazawa, S. sFRP4-dependent Wnt signal modulation is critical for bone remodeling during postnatal development and age-related bone loss. Sci. Rep. 2016, 6, 25198. [CrossRef]

68. Morvan, F.; Boulukos, K.; Clement-Lacroix, P.; Roman-Roman, S.; Suc-Royer, I.; Vayssiere, B.; Ammann, P.; Martin, P.; Pinho, S.; Pognonec, P.; et al. Deletion of a single allele of the Dkk1 gene leads to an increase in bone formation and bone mass. J. Bone Miner. Res. 2006, 21, 934–945. [CrossRef]

69. Colditz, J.; Thiele, S.; Baschant, U.; Niehrs, C.; Bonewald, L.F.; Hofbauer, L.C.; Rauner, M. Postnatal Skeletal Deletion of Dickkopf-1 Increases Bone Formation and Bone Volume in Male and Female Mice, Despite Increased Sclerostin Expression. J. Bone Miner. Res. 2018, 33, 1698–1707. [CrossRef]

70. Knight, M.N.; Karuppaiah, K.; Lowe, M.; Mohanty, S.; Zondervan, R.L.; Bell, S.; Ahn, J.; Hankenson, K.D. R-spondin-2 is a Wnt agonist that regulates osteoblast activity and bone mass. Bone Res. 2018, 6, 24. [CrossRef]

71. Luo, J.; Zhou, W.; Zhou, X.; Li, D.; Weng, J.; Yi, Z.; Cho, S.G.; Li, C.; Yi, T.; Wu, X.; et al. Regulation of bone formation and remodeling by G-protein-coupled receptor 48. Development 2009, 136, 2747–2756. [CrossRef] [PubMed]

72. Sun, P.; Jia, K.; Zheng, C.; Zhu, X.; Li, J.; He, L.; Siwko, S.; Xue, F.; Liu, M.; Luo, J. Loss of Lgr4 inhibits differentiation, migration and apoptosis, and promotes proliferation in bone mesenchymal stem cells. J. Cell Physiol. 2019, 234, 10855–10867. [CrossRef] [PubMed]

73. Luo, J.; Yang, Z.; Ma, Y.; Yue, Z.; Lin, H.; Qu, G.; Huang, J.; Dai, W.; Li, C.; Zheng, C.; et al. LGR4 is a receptor for RANKL and negatively regulates osteoclast differentiation and bone resorption. Nat. Med. 2016, 22, 539–546. [CrossRef] [PubMed]

74. Zhong, Z.; Zylstra-Diegel, C.R.; Schumacher, C.A.; Baker, J.J.; Carpenter, A.C.; Rao, S.; Yao, W.; Guan, M.; Helms, J.A.; Lane, N.E.; et al. Wntless functions in mature osteoblasts to regulate bone mass. Proc. Natl. Acad. Sci. USA 2012, 109, E2197–E2204. [CrossRef] [PubMed]

75. Joeng, K.S.; Lee, Y.C.; Lim, J.; Chen, Y.; Jiang, M.M.; Munivez, E.; Ambrose, C.; Lee, B.H. Osteocyte-specific WNT1 regulates osteoblast function during bone homeostasis. J. Clin. Investig. 2017, 127, 2678–2688. [CrossRef]

76. Luther, J.; Yorgan, T.A.; Rolvien, T.; Ulsamer, L.; Koehne, T.; Liao, N.; Keller, D.; Vollersen, N.; Teufel, S.; Neven, M.; et al. Wnt1 is an Lrp5-independent bone-anabolic Wnt ligand. Sci. Transl. Med. 2018, 10. [CrossRef]

77. Yu, B.; Chang, J.; Liu, Y.; Li, J.; Kevork, K.; Al-Hezaimi, K.; Graves, D.T.; Park, N.H.; Wang, C.Y. Wnt4 signaling prevents skeletal aging and inflammation by inhibiting nuclear factor-kappaB. Nat. Med. 2014, 20, 1009–1017. [CrossRef]

78. Okamoto, M.; Udagawa, N.; Uehara, S.; Maeda, K.; Yamashita, T.; Nakamichi, Y.; Kato, H.; Saito, N.; Minami, Y.; Takahashi, N.; et al. Noncanonical Wnt5a enhances Wnt/beta-catenin signaling during osteoblastogenesis. Sci. Rep. 2014, 4, 4493. [CrossRef]

79. Chen, J.; Tu, X.; Esen, E.; Joeng, K.S.; Lin, C.; Arbeit, J.M.; Ruegg, M.A.; Hall, M.N.; Ma, L.; Long, F. WNT7B promotes bone formation in part through mTORC1. PLoS Genet. 2014, 10, e1004145. [CrossRef]

80. Moverare-Skrtic, S.; Henning, P.; Liu, X.; Nagano, K.; Saito, H.; Borjesson, A.E.; Sjogren, K.; Windahl, S.H.; Farman, H.; Kindlund, B.; et al. Osteoblast-derived WNT16 represses osteoclastogenesis and prevents cortical bone fragility fractures. Nat. Med. 2014, 20, 1279–1288. [CrossRef]

81. Glass, D.A., II; Bialek, P.; Ahn, J.D.; Starbuck, M.; Patel, M.S.; Clevers, H.; Taketo, M.M.; Long, F.; McMahon, A.P.; Lang, R.A.; et al. Canonical Wnt signaling in differentiated osteoblasts controls osteoclast differentiation. Dev. Cell 2005, 8, 751–764. [CrossRef] [PubMed]

82. Komori, T. Regulation of Proliferation, Differentiation and Functions of Osteoblasts by Runx2. Int. J. Mol. Sci. 2019, 20, 1694. [CrossRef] [PubMed]

83. Delgado-Calle, J.; Sanudo, C.; Bolado, A.; Fernandez, A.F.; Arozamena, J.; Pascual-Carra, M.A.; Rodriguez-Rey, J.C.; Fraga, M.F.; Bonewald, L.; Riancho, J.A. DNA methylation contributes to the regulation of sclerostin expression in human osteocytes. J. Bone Miner. Res. 2012, 27, 926–937. [CrossRef] [PubMed]

84. Leupin, O.; Kramer, I.; Collette, N.M.; Loots, G.G.; Natt, F.; Kneissel, M.; Keller, H. Control of the SOST bone enhancer by PTH using MEF2 transcription factors. J. Bone Miner. Res. 2007, 22, 1957–1967. [CrossRef]

85. O’Brien, C.A.; Plotkin, L.I.; Galli, C.; Goellner, J.J.; Gortazar, A.R.; Allen, M.R.; Robling, A.G.; Bouxsein, M.; Schipani, E.; Turner, C.H.; et al. Control of bone mass and remodeling by PTH receptor signaling in osteocytes. PLoS ONE 2008, 3, e2942. [CrossRef]

86. Drake, M.T.; Srinivasan, B.; Modder, U.I.; Peterson, J.M.; McCready, L.K.; Riggs, B.L.; Dwyer, D.; Stolina, M.; Kostenuik, P.; Khosla, S. Effects of parathyroid hormone treatment on circulating sclerostin levels in postmenopausal women. J. Clin. Endocrinol. Metab. 2010, 95, 5056–5062. [CrossRef]

87. Baertschi, S.; Baur, N.; Lueders-Lefevre, V.; Voshol, J.; Keller, H. Class I and IIa histone deacetylases have opposite effects on sclerostin gene regulation. J. Biol. Chem. 2014, 289, 24995–25009. [CrossRef]

88. Wein, M.N.; Spatz, J.; Nishimori, S.; Doench, J.; Root, D.; Babij, P.; Nagano, K.; Baron, R.; Brooks, D.; Bouxsein, M.; et al. HDAC5 controls MEF2C-driven sclerostin expression in osteocytes. J. Bone Miner. Res. 2015, 30, 400–411. [CrossRef]

89. Stegen, S.; Stockmans, I.; Moermans, K.; Thienpont, B.; Maxwell, P.H.; Carmeliet, P.; Carmeliet, G. Osteocytic oxygen sensing controls bone mass through epigenetic regulation of sclerostin. Nat. Commun. 2018, 9, 2557. [CrossRef]

90. Wein, M.N.; Liang, Y.; Goransson, O.; Sundberg, T.B.; Wang, J.; Williams, E.A.; O’Meara, M.J.; Govea, N.; Beqo, B.; Nishimori, S.; et al. SIKs control osteocyte responses to parathyroid hormone. Nat. Commun. 2016, 7, 13176. [CrossRef]

91. Lombardi, M.S.; Gillieron, C.; Berkelaar, M.; Gabay, C. Salt-inducible kinases (SIK) inhibition reduces RANKL-induced osteoclastogenesis. PLoS ONE 2017, 12, e0185426. [CrossRef] [PubMed]

92. Taub, M. Salt Inducible Kinase Signaling Networks: Implications for Acute Kidney Injury and Therapeutic Potential. Int. J. Mol. Sci. 2019, 20, 3219. [CrossRef] [PubMed]

93. Bonnet, N.; Standley, K.N.; Bianchi, E.N.; Stadelmann, V.; Foti, M.; Conway, S.J.; Ferrari, S.L. The matricellular protein periostin is required for sost inhibition and the anabolic response to mechanical loading and physical activity. J. Biol. Chem. 2009, 284, 35939–35950. [CrossRef] [PubMed]

94. Bonnet, N.; Conway, S.J.; Ferrari, S.L. Regulation of beta catenin signaling and parathyroid hormone anabolic effects in bone by the matricellular protein periostin. Proc. Natl. Acad. Sci. USA 2012, 109, 15048–15053. [CrossRef]

95. Walker, E.C.; McGregor, N.E.; Poulton, I.J.; Solano, M.; Pompolo, S.; Fernandes, T.J.; Constable, M.J.; Nicholson, G.C.; Zhang, J.G.; Nicola, N.A.; et al. Oncostatin M promotes bone formation independently of resorption when signaling through leukemia inhibitory factor receptor in mice. J. Clin. Investig. 2010, 120, 582–592. [CrossRef]

96. Masuki, H.; Li, M.; Hasegawa, T.; Suzuki, R.; Ying, G.; Zhusheng, L.; Oda, K.; Yamamoto, T.; Kawanami, M.; Amizuka, N. Immunolocalization of DMP1 and sclerostin in the epiphyseal trabecule and diaphyseal cortical bone of osteoprotegerin deficient mice. BioMed Res. 2010, 31, 307–318. [CrossRef]

97. Koide, M.; Kobayashi, Y.; Yamashita, T.; Uehara, S.; Nakamura, M.; Hiraoka, B.Y.; Ozaki, Y.; Iimura, T.; Yasuda, H.; Takahashi, N.; et al. Bone Formation Is Coupled to Resorption Via Suppression of Sclerostin Expression by Osteoclasts. J. Bone Miner. Res. 2017, 32, 2074–2086. [CrossRef]

98. Kim, H.; Wrann, C.D.; Jedrychowski, M.; Vidoni, S.; Kitase, Y.; Nagano, K.; Zhou, C.; Chou, J.; Parkman, V.A.; Novick, S.J.; et al. Irisin Mediates Effects on Bone and Fat via alphaV Integrin Receptors. Cell 2018, 175, 1756–1768.e17. [CrossRef]

99. Rivadeneira, F.; Styrkarsdottir, U.; Estrada, K.; Halldorsson, B.V.; Hsu, Y.H.; Richards, J.B.; Zillikens, M.C.; Kavvoura, F.K.; Amin, N.; Aulchenko, Y.S.; et al. Twenty bone-mineral-density loci identified by large-scale meta-analysis of genome-wide association studies. Nat. Genet. 2009, 41, 1199–1206.

100. Zisimopoulou, P.; Evangelakou, P.; Tzartos, J.; Lazaridis, K.; Zouvelou, V.; Mantegazza, R.; Antozzi, C.; Andreetta, F.; Evoli, A.; Deymeer, F.; et al. A comprehensive analysis of the epidemiology and clinical characteristics of anti-LRP4 in myasthenia gravis. J. Autoimmun. 2014, 52, 139–145. [CrossRef]

101. Estrada, K.; Styrkarsdottir, U.; Evangelou, E.; Hsu, Y.H.; Duncan, E.L.; Ntzani, E.E.; Oei, L.; Albagha, O.M.; Amin, N.; Kemp, J.P.; et al. Genome-wide meta-analysis identifies 56 bone mineral density loci and reveals 14 loci associated with risk of fracture. Nat. Genet. 2012, 44, 491–501. [CrossRef]

102. Tu, X.; Delgado-Calle, J.; Condon, K.W.; Maycas, M.; Zhang, H.; Carlesso, N.; Taketo, M.M.; Burr, D.B.; Plotkin, L.I.; Bellido, T. Osteocytes mediate the anabolic actions of canonical Wnt/beta-catenin signaling in bone. Proc. Natl. Acad. Sci. USA 2015, 112, E478–E486. [CrossRef] [PubMed]

103. Styrkarsdottir, U.; Thorleifsson, G.; Gudjonsson, S.A.; Sigurdsson, A.; Center, J.R.; Lee, S.H.; Nguyen, T.V.; Kwok, T.C.Y.; Lee, J.S.W.; Ho, S.C.; et al. Sequence variants in the PTCH1 gene associate with spine bone mineral density and osteoporotic fractures. Nat. Commun. 2016, 7, 10129. [CrossRef] [PubMed]

104. Kemp, J.P.; Morris, J.A.; Medina-Gomez, C.; Forgetta, V.; Warrington, N.M.; Youlten, S.E.; Zheng, J.; Gregson, C.L.; Grundberg, E.; Trajanoska, K.; et al. Identification of 153 new loci associated with heel bone mineral density and functional involvement of GPC6 in osteoporosis. Nat. Genet. 2017, 49, 1468–1475. [CrossRef] [PubMed]

105. Trajanoska, K.; Morris, J.A.; Oei, L.; Zheng, H.F.; Evans, D.M.; Kiel, D.P.; Ohlsson, C.; Richards, J.B.; Rivadeneira, F.; consortium, G.G.; et al. Assessment of the genetic and clinical determinants of fracture risk: Genome wide association and mendelian randomisation study. BMJ 2018, 362, k3225. [CrossRef] [PubMed]

106. Shi, G.X.; Mao, W.W.; Zheng, X.F.; Jiang, L.S. The role of R-spondins and their receptors in bone metabolism. Prog. Biophys. Mol. Biol. 2016, 122, 93–100. [CrossRef]

107. Sharma, A.R.; Choi, B.S.; Park, J.M.; Lee, D.H.; Lee, J.E.; Kim, H.S.; Yoon, J.K.; Song, D.K.; Nam, J.S.; Lee, S.S. Rspo 1 promotes osteoblast differentiation via Wnt signaling pathway. Indian J. Biochem. Biophys. 2013, 50, 19–25.

108. Wang, H.; Brennan, T.A.; Russell, E.; Kim, J.H.; Egan, K.P.; Chen, Q.; Israelite, C.; Schultz, D.C.; Johnson, F.B.; Pignolo, R.J. R-Spondin 1 promotes vibration-induced bone formation in mouse models of osteoporosis. J. Mol. Med. 2013, 91, 1421–1429. [CrossRef]

109. Shi, G.X.; Zheng, X.F.; Zhu, C.; Li, B.; Wang, Y.R.; Jiang, S.D.; Jiang, L.S. Evidence of the Role of R-Spondin 1 and Its Receptor Lgr4 in the Transmission of Mechanical Stimuli to Biological Signals for Bone Formation. Int. J. Mol. Sci. 2017, 18, 564. [CrossRef]

110. Mahasarakham, C.P.A.; Ezura, Y.; Kawasaki, M.; Smriti, A.; Moriya, S.; Yamada, T.; Izu, Y.; Nifuji, A.; Nishimori, K.; Izumi, Y.; et al. BMP-2 Enhances Lgr4 Gene Expression in Osteoblastic Cells. J. Cell Physiol. 2016, 231, 887–895. [CrossRef]

111. Friedman, M.S.; Oyserman, S.M.; Hankenson, K.D. Wnt11 promotes osteoblast maturation and mineralization through R-spondin 2. J. Biol. Chem. 2009, 284, 14117–14125. [CrossRef] [PubMed]

112. Gunasekera, N.S.; Divito, J.K.; Kupper, T.S.; Huang, J.T.; Divito, S.J. Cross-Sectional Study Evaluating Skin, Hair, Nail, and Bone Disease in Patients with Focal Dermal Hypoplasia. Pediatr. Dermatol. 2017, 34, 197–198. [CrossRef] [PubMed]

113. Barrott, J.J.; Cash, G.M.; Smith, A.P.; Barrow, J.R.; Murtaugh, L.C. Deletion of mouse Porcn blocks Wnt ligand secretion and reveals an ectodermal etiology of human focal dermal hypoplasia/Goltz syndrome. Proc. Natl. Acad. Sci. USA 2011, 108, 12752–12757. [CrossRef] [PubMed]

114. Liu, W.; Shaver, T.M.; Balasa, A.; Ljungberg, M.C.; Wang, X.; Wen, S.; Nguyen, H.; Van den Veyver, I.B. Deletion of Porcn in mice leads to multiple developmental defects and models human focal dermal hypoplasia (Goltz syndrome). PLoS ONE 2012, 7, e32331. [CrossRef] [PubMed]

115. Weivoda, M.M.; Ruan, M.; Pederson, L.; Hachfeld, C.; Davey, R.A.; Zajac, J.D.; Westendorf, J.J.; Khosla, S.; Oursler, M.J. Osteoclast TGF-beta Receptor Signaling Induces Wnt1 Secretion and Couples Bone Resorption to Bone Formation. J. Bone Miner. Res. 2016, 31, 76–85. [CrossRef]

116. Tasca, A.; Astleford, K.; Blixt, N.C.; Jensen, E.D.; Gopalakrishnan, R.; Mansky, K.C. SMAD1/5 signaling in osteoclasts regulates bone formation via coupling factors. PLoS ONE 2018, 13, e0203404. [CrossRef]

117. Weske, S.; Vaidya, M.; Reese, A.; von Wnuck Lipinski, K.; Keul, P.; Bayer, J.K.; Fischer, J.W.; Flogel, U.; Nelsen, J.; Epple, M.; et al. Targeting sphingosine-1-phosphate lyase as an anabolic therapy for bone loss. Nat. Med. 2018, 24, 667–678. [CrossRef]

118. Zhang, L.; Choi, H.J.; Estrada, K.; Leo, P.J.; Li, J.; Pei, Y.F.; Zhang, Y.; Lin, Y.; Shen, H.; Liu, Y.Z.; et al. Multistage genome-wide association meta-analyses identified two new loci for bone mineral density. Hum. Mol. Genet. 2014, 23, 1923–1933. [CrossRef]

119. Ozeki, N.; Mogi, M.; Hase, N.; Hiyama, T.; Yamaguchi, H.; Kawai, R.; Kondo, A.; Nakata, K. Wnt16 Signaling Is Required for IL-1beta-Induced Matrix Metalloproteinase-13-Regulated Proliferation of Human Stem Cell-Derived Osteoblastic Cells. Int. J. Mol. Sci. 2016, 17, 221. [CrossRef]

120. Chang, J.; Sonoyama, W.; Wang, Z.; Jin, Q.; Zhang, C.; Krebsbach, P.H.; Giannobile, W.; Shi, S.; Wang, C.Y. Noncanonical Wnt-4 signaling enhances bone regeneration of mesenchymal stem cells in craniofacial defects through activation of p38 MAPK. J. Biol. Chem. 2007, 282, 30938–30948. [CrossRef]

121. Santiago, F.; Oguma, J.; Brown, A.M.; Laurence, J. Noncanonical Wnt signaling promotes osteoclast differentiation and is facilitated by the human immunodeficiency virus protease inhibitor ritonavir. Biochem. Biophys. Res. Commun. 2012, 417, 223–230. [CrossRef] [PubMed]

122. Bellon, M.; Ko, N.L.; Lee, M.J.; Yao, Y.; Waldmann, T.A.; Trepel, J.B.; Nicot, C. Adult T-cell leukemia cells overexpress Wnt5a and promote osteoclast differentiation. Blood 2013, 121, 5045–5054. [CrossRef] [PubMed]

123. Lories, R.J.; Corr, M.; Lane, N.E. To Wnt or not to Wnt: The bone and joint health dilemma. Nat. Rev. Rheumatol. 2013, 9, 328–339. [CrossRef] [PubMed]

124. Uehara, S.; Udagawa, N.; Kobayashi, Y. Non-canonical Wnt signals regulate cytoskeletal remodeling in osteoclasts. Cell. Mol. Life Sci. 2018, 75, 3683–3692. [CrossRef]

125. Solling, A.S.K.; Harslof, T.; Langdahl, B. The clinical potential of romosozumab for the prevention of fractures in postmenopausal women with osteoporosis. Ther. Adv. Musculoskelet. Dis. 2018, 10, 105–115. [CrossRef]

126. Cosman, F.; Crittenden, D.B.; Adachi, J.D.; Binkley, N.; Czerwinski, E.; Ferrari, S.; Hofbauer, L.C.; Lau, E.; Lewiecki, E.M.; Miyauchi, A.; et al. Romosozumab Treatment in Postmenopausal Women with Osteoporosis. N. Engl. J. Med. 2016, 375, 1532–1543. [CrossRef]

127. Langdahl, B.L.; Libanati, C.; Crittenden, D.B.; Bolognese, M.A.; Brown, J.P.; Daizadeh, N.S.; Dokoupilova, E.; Engelke, K.; Finkelstein, J.S.; Genant, H.K.; et al. Romosozumab (sclerostin monoclonal antibody) versus teriparatide in postmenopausal women with osteoporosis transitioning from oral bisphosphonate therapy: A randomised, open-label, phase 3 trial. Lancet 2017, 390, 1585–1594. [CrossRef]

128. Saag, K.G.; Petersen, J.; Brandi, M.L.; Karaplis, A.C.; Lorentzon, M.; Thomas, T.; Maddox, J.; Fan, M.; Meisner, P.D.; Grauer, A. Romosozumab or Alendronate for Fracture Prevention in Women with Osteoporosis. N. Engl. J. Med. 2017, 377, 1417–1427. [CrossRef]

129. Lewiecki, E.M.; Blicharski, T.; Goemaere, S.; Lippuner, K.; Meisner, P.D.; Miller, P.D.; Miyauchi, A.; Maddox, J.; Chen, L.; Horlait, S. A Phase III Randomized Placebo-Controlled Trial to Evaluate Efficacy and Safety of Romosozumab in Men With Osteoporosis. J. Clin. Endocrinol. Metab. 2018, 103, 3183–3193. [CrossRef]

130. Gao, Y.; Huang, E.; Zhang, H.; Wang, J.; Wu, N.; Chen, X.; Wang, N.; Wen, S.; Nan, G.; Deng, F.; et al. Crosstalk between Wnt/beta-catenin and estrogen receptor signaling synergistically promotes osteogenic differentiation of mesenchymal progenitor cells. PLoS ONE 2013, 8, e82436. [CrossRef]

131. Kondoh, S.; Inoue, K.; Igarashi, K.; Sugizaki, H.; Shirode-Fukuda, Y.; Inoue, E.; Yu, T.; Takeuchi, J.K.; Kanno, J.; Bonewald, L.F.; et al. Estrogen receptor alpha in osteocytes regulates trabecular bone formation in female mice. Bone 2014, 60, 68–77. [CrossRef] [PubMed]

132. Roforth, M.M.; Fujita, K.; McGregor, U.I.; Kirmani, S.; McCready, L.K.; Peterson, J.M.; Drake, M.T.; Monroe, D.G.; Khosla, S. Effects of age on bone mRNA levels of sclerostin and other genes relevant to bone metabolism in humans. Bone 2014, 59, 1–6. [CrossRef] [PubMed]

133. Hay, E.; Bouaziz, W.; Funck-Brentano, T.; Cohen-Solal, M. Sclerostin and Bone Aging: A Mini-Review. Gerontology 2016, 62, 618–623. [CrossRef] [PubMed]

134. Weivoda, M.M.; Youssef, S.J.; Oursler, M.J. Sclerostin expression and functions beyond the osteocyte. Bone 2017, 96, 45–50. [CrossRef] [PubMed]

135. Mirza, F.S.; Padhi, I.D.; Raisz, L.G.; Lorenzo, J.A. Serum sclerostin levels negatively correlate with parathyroid hormone levels and free estrogen index in postmenopausal women. J. Clin. Endocrinol. Metab. 2010, 95, 1991–1997. [CrossRef] [PubMed]

136. Chung, Y.E.; Lee, S.H.; Lee, S.Y.; Kim, S.Y.; Kim, H.H.; Mirza, F.S.; Lee, S.K.; Lorenzo, J.A.; Kim, G.S.; Koh, J.M. Long-term treatment with raloxifene, but not bisphosphonates, reduces circulating sclerostin levels in postmenopausal women. Osteoporos. Int. 2012, 23, 1235–1243. [CrossRef]

137. Reid, I.R. Short-term and long-term effects of osteoporosis therapies. Nat. Rev. Endocrinol. 2015, 11, 418–428. [CrossRef]

138. MacNabb, C.; Patton, D.; Hayes, J.S. Sclerostin Antibody Therapy for the Treatment of Osteoporosis: Clinical Prospects and Challenges. J. Osteoporos. 2016, 2016, 6217286. [CrossRef]

139. Fukumoto, S.; Matsumoto, T. Recent advances in the management of osteoporosis. F1000Res. 2017, 6, 625. [CrossRef]

140. Appelman-Dijkstra, N.M.; Papapoulos, S.E. Clinical advantages and disadvantages of anabolic bone therapies targeting the WNT pathway. Nat. Rev. Endocrinol. 2018, 14, 605–623. [CrossRef]

141. van Geel, T.A.; van Helden, S.; Geusens, P.P.; Winkens, B.; Dinant, G.J. Clinical subsequent fractures cluster in time after first fractures. Ann. Rheum. Dis. 2009, 68, 99–102. [CrossRef] [PubMed]

142. Li, X.; Ominsky, M.S.; Warmington, K.S.; Morony, S.; Gong, J.; Cao, J.; Gao, Y.; Shalhoub, V.; Tipton, B.; Haldankar, R.; et al. Sclerostin antibody treatment increases bone formation, bone mass, and bone strength in a rat model of postmenopausal osteoporosis. J. Bone Miner. Res. 2009, 24, 578–588. [CrossRef] [PubMed]

143. Li, X.; Warmington, K.S.; Niu, Q.T.; Asuncion, F.J.; Barrero, M.; Grisanti, M.; Dwyer, D.; Stouch, B.; Thway, T.M.; Stolina, M.; et al. Inhibition of sclerostin by monoclonal antibody increases bone formation, bone mass, and bone strength in aged male rats. J. Bone Miner. Res. 2010, 25, 2647–2656. [CrossRef] [PubMed]

144. Ominsky, M.S.; Boyce, R.W.; Li, X.; Ke, H.Z. Effects of sclerostin antibodies in animal models of osteoporosis. Bone 2017, 96, 63–75. [CrossRef]

145. Seefried, L.; Baumann, J.; Hemsley, S.; Hofmann, C.; Kunstmann, E.; Kiese, B.; Huang, Y.; Chivers, S.; Valentin, M.A.; Borah, B.; et al. Efficacy of anti-sclerostin monoclonal antibody BPS804 in adult patients with hypophosphatasia. J. Clin. Investig. 2017, 127, 2148–2158. [CrossRef]

146. Glorieux, F.H.; Devogelaer, J.P.; Durigova, M.; Goemaere, S.; Hemsley, S.; Jakob, F.; Junker, U.; Ruckle, J.; Seefried, L.; Winkle, P.J. BPS804 Anti-Sclerostin Antibody in Adults With Moderate Osteogenesis Imperfecta: Results of a Randomized Phase 2a Trial. J. Bone Miner. Res. 2017, 32, 1496–1504. [CrossRef]

147. New Therapy to Cost One-Third Less Than Other Bone-Building Agents Over Full Course of Therapy. Available online: https://www.amgen.com/media/news-releases/2019/04/evenity-romosozumabaqqg-nowavailable-in-the-united-states-for-the-treatment-of-osteoporosis-in-postmenopausal-women-at-highrisk-for-fracture/ (accessed on 29 October 2019).

148. van Lierop, A.H.; Moester, M.J.; Hamdy, N.A.; Papapoulos, S.E. Serum Dickkopf 1 levels in sclerostin deficiency. J. Clin. Endocrinol. Metab. 2014, 99, E252–E256. [CrossRef]

149. Stolina, M.; Dwyer, D.; Niu, Q.T.; Villasenor, K.S.; Kurimoto, P.; Grisanti, M.; Han, C.Y.; Liu, M.; Li, X.; Ominsky, M.S.; et al. Temporal changes in systemic and local expression of bone turnover markers during six months of sclerostin antibody administration to ovariectomized rats. Bone 2014, 67, 305–313. [CrossRef]

150. Witcher, P.C.; Miner, S.E.; Horan, D.J.; Bullock, W.A.; Lim, K.E.; Kang, K.S.; Adaniya, A.L.; Ross, R.D.; Loots, G.G.; Robling, A.G. Sclerostin neutralization unleashes the osteoanabolic effects of Dkk1 inhibition. JCI Insight. 2018, 3, 98673. [CrossRef]

151. Florio, M.; Gunasekaran, K.; Stolina, M.; Li, X.; Liu, L.; Tipton, B.; Salimi-Moosavi, H.; Asuncion, F.J.; Li, C.; Sun, B.; et al. A bispecific antibody targeting sclerostin and DKK-1 promotes bone mass accrual and fracture repair. Nat. Commun. 2016, 7, 11505. [CrossRef]

152. Usami, Y.; Gunawardena, A.T.; Iwamoto, M.; Enomoto-Iwamoto, M. Wnt signaling in cartilage development and diseases: Lessons from animal studies. Lab. Investig. 2016, 96, 186–196. [CrossRef] [PubMed]

153. Komori, T. Cell Death in Chondrocytes, Osteoblasts, and Osteocytes. Int. J. Mol. Sci. 2016, 17, 2045. [CrossRef] [PubMed]

154. Zhu, M.; Tang, D.; Wu, Q.; Hao, S.; Chen, M.; Xie, C.; Rosier, R.N.; O’Keefe, R.J.; Zuscik, M.; Chen, D. Activation of beta-catenin signaling in articular chondrocytes leads to osteoarthritis-like phenotype in adult beta-catenin conditional activation mice. J. Bone Miner. Res. 2009, 24, 12–21. [CrossRef] [PubMed]

155. Nakamura, Y.; Nawata, M.; Wakitani, S. Expression profiles and functional analyses of Wnt-related genes in human joint disorders. Am. J. Pathol. 2005, 167, 97–105. [CrossRef]

156. Zhang, Y.; Vasheghani, F.; Li, Y.H.; Blati, M.; Simeone, K.; Fahmi, H.; Lussier, B.; Roughley, P.; Lagares, D.; Pelletier, J.P.; et al. Cartilage-specific deletion of mTOR upregulates autophagy and protects mice from osteoarthritis. Ann. Rheum. Dis. 2015, 74, 1432–1440. [CrossRef] [PubMed]

157. Chan, B.Y.; Fuller, E.S.; Russell, A.K.; Smith, S.M.; Smith, M.M.; Jackson, M.T.; Cake, M.A.; Read, R.A.; Bateman, J.F.; Sambrook, P.N.; et al. Increased chondrocyte sclerostin may protect against cartilage degradation in osteoarthritis. Osteoarthr. Cartil. 2011, 19, 874–885. [CrossRef]

158. Bouaziz, W.; Funck-Brentano, T.; Lin, H.; Marty, C.; Ea, H.K.; Hay, E.; Cohen-Solal, M. Loss of sclerostin promotes osteoarthritis in mice via beta-catenin-dependent and -independent Wnt pathways. Arthritis Res. Ther. 2015, 17, 24. [CrossRef]

159. Deshmukh, V.; Hu, H.; Barroga, C.; Bossard, C.; Kc, S.; Dellamary, L.; Stewart, J.; Chiu, K.; Ibanez, M.; Pedraza, M.; et al. A small-molecule inhibitor of the Wnt pathway (SM04690) as a potential disease modifying agent for the treatment of osteoarthritis of the knee. Osteoarthr. Cartil. 2018, 26, 18–27. [CrossRef]

160. Lietman, C.; Wu, B.; Lechner, S.; Shinar, A.; Sehgal, M.; Rossomacha, E.; Datta, P.; Sharma, A.; Gandhi, R.; Kapoor, M.; et al. Inhibition of Wnt/beta-catenin signaling ameliorates osteoarthritis in a murine model of experimental osteoarthritis. JCI Insight 2018, 3, 96308. [CrossRef]

161. Tsukasaki, M.; Takayanagi, H. Osteoimmunology: Evolving concepts in bone-immune interactions in health and disease. Nat. Rev. Immunol. 2019, 19, 626–642. [CrossRef]

162. Swierkot, J.; Gruszecka, K.; Matuszewska, A.; Wiland, P. Assessment of the Effect of Methotrexate Therapy on Bone Metabolism in Patients with Rheumatoid Arthritis. Arch. Immunol. Ther. Exp. 2015, 63, 397–404. [CrossRef] [PubMed]

163. Diarra, D.; Stolina, M.; Polzer, K.; Zwerina, J.; Ominsky, M.S.; Dwyer, D.; Korb, A.; Smolen, J.; Hoffmann, M.; Scheinecker, C.; et al. Dickkopf-1 is a master regulator of joint remodeling. Nat. Med. 2007, 13, 156–163. [CrossRef] [PubMed]

164. Miao, C.G.; Yang, Y.Y.; He, X.; Li, X.F.; Huang, C.; Huang, Y.; Zhang, L.; Lv, X.W.; Jin, Y.; Li, J. Wnt signaling pathway in rheumatoid arthritis, with special emphasis on the different roles in synovial inflammation and bone remodeling. Cell Signal. 2013, 25, 2069–2078. [CrossRef] [PubMed]

165. Wehmeyer, C.; Frank, S.; Beckmann, D.; Bottcher, M.; Cromme, C.; Konig, U.; Fennen, M.; Held, A.; Paruzel, P.; Hartmann, C.; et al. Sclerostin inhibition promotes TNF-dependent inflammatory joint destruction. Sci. Transl. Med. 2016, 8, 330ra335. [CrossRef] [PubMed]

166. Chen, X.X.; Baum, W.; Dwyer, D.; Stock, M.; Schwabe, K.; Ke, H.Z.; Stolina, M.; Schett, G.; Bozec, A. Sclerostin inhibition reverses systemic, periarticular and local bone loss in arthritis. Ann. Rheum. Dis. 2013, 72, 1732–1736. [CrossRef] [PubMed]

167. Marenzana, M.; Vugler, A.; Moore, A.; Robinson, M. Effect of sclerostin-neutralising antibody on periarticular and systemic bone in a murine model of rheumatoid arthritis: A microCT study. Arthritis Res. Ther. 2013, 15, R125. [CrossRef]

168. Sen, M.; Lauterbach, K.; El-Gabalawy, H.; Firestein, G.S.; Corr, M.; Carson, D.A. Expression and function of wingless and frizzled homologs in rheumatoid arthritis. Proc. Natl. Acad. Sci. USA 2000, 97, 2791–2796. [CrossRef]

169. Sen, M.; Carson, D.A. Wnt signaling in rheumatoid synoviocyte activation. Mod. Rheumatol. 2002, 12, 5–9. [CrossRef]

170. Sen, M.; Chamorro, M.; Reifert, J.; Corr, M.; Carson, D.A. Blockade of Wnt-5A/frizzled 5 signaling inhibits rheumatoid synoviocyte activation. Arthritis Rheum. 2001, 44, 772–781. [CrossRef]

171. Kim, J.; Kim, J.; Kim, D.W.; Ha, Y.; Ihm, M.H.; Kim, H.; Song, K.; Lee, I. Wnt5a induces endothelial inflammation via beta-catenin-independent signaling. J. Immunol. 2010, 185, 1274–1282. [CrossRef]

172. Rauner, M.; Stein, N.; Winzer, M.; Goettsch, C.; Zwerina, J.; Schett, G.; Distler, J.H.; Albers, J.; Schulze, J.; Schinke, T.; et al. WNT5A is induced by inflammatory mediators in bone marrow stromal cells and regulates cytokine and chemokine production. J. Bone Miner. Res. 2012, 27, 575–585. [CrossRef] [PubMed]

173. Sato, A.; Kayama, H.; Shojima, K.; Matsumoto, S.; Koyama, H.; Minami, Y.; Nojima, S.; Morii, E.; Honda, H.; Takeda, K.; et al. The Wnt5a-Ror2 axis promotes the signaling circuit between interleukin-12 and interferon-gamma in colitis. Sci. Rep. 2015, 5, 10536. [CrossRef] [PubMed]

174. Miao, P.; Zhou, X.W.; Wang, P.; Zhao, R.; Chen, N.; Hu, C.Y.; Chen, X.H.; Qian, L.; Yu, Q.W.; Zhang, J.Y.; et al. Regulatory effect of anti-gp130 functional mAb on IL-6 mediated RANKL and Wnt5a expression through JAK-STAT3 signaling pathway in FLS. Oncotarget 2018, 9, 20366–20376. [CrossRef] [PubMed]

175. Kwon, Y.J.; Lee, S.W.; Park, Y.B.; Lee, S.K.; Park, M.C. Secreted frizzled-related protein 5 suppresses inflammatory response in rheumatoid arthritis fibroblast-like synoviocytes through down-regulation of c-Jun N-terminal kinase. Rheumatology 2014, 53, 1704–1711. [CrossRef]

176. Pukrop, T.; Klemm, F.; Hagemann, T.; Gradl, D.; Schulz, M.; Siemes, S.; Trumper, L.; Binder, C. Wnt 5a signaling is critical for macrophage-induced invasion of breast cancer cell lines. Proc. Natl. Acad. Sci. USA 2006, 103, 5454–5459. [CrossRef]

177. Enomoto, M.; Hayakawa, S.; Itsukushima, S.; Ren, D.Y.; Matsuo, M.; Tamada, K.; Oneyama, C.; Okada, M.; Takumi, T.; Nishita, M.; et al. Autonomous regulation of osteosarcoma cell invasiveness by Wnt5a/Ror2 signaling. Oncogene 2009, 28, 3197–3208. [CrossRef]

178. Smolen, J.S.; Landewe, R.; Bijlsma, J.; Burmester, G.; Chatzidionysiou, K.; Dougados, M.; Nam, J.; Ramiro, S.; Voshaar, M.; van Vollenhoven, R.; et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2016 update. Ann. Rheum. Dis. 2017, 76, 960–977. [CrossRef]

179. MacLauchlan, S.; Zuriaga, M.A.; Fuster, J.J.; Cuda, C.M.; Jonason, J.; Behzadi, F.; Duffen, J.P.; Haines, G.K., III; Aprahamian, T.; Perlman, H.; et al. Genetic deficiency of Wnt5a diminishes disease severity in a murine model of rheumatoid arthritis. Arthritis Res. Ther. 2017, 19, 166. [CrossRef]

180. Cao, W.; Niu, M.; Tong, Y.; Du, Y.; Lou, W.; Mao, Y.; Dou, Y.; Yuan, H.; Zhao, W. Depleting the carboxy-terminus of human Wnt5a attenuates collagen-induced arthritis in DBA/1 mice. Biochem. Biophys. Res. Commun. 2018, 504, 679–685. [CrossRef]

181. Takeuchi, T.; Tanaka, Y.; Soen, S.; Yamanaka, H.; Yoneda, T.; Tanaka, S.; Nitta, T.; Okubo, N.; Genant, H.K.; van der Heijde, D. Effects of the anti-RANKL antibody denosumab on joint structural damage in patients with rheumatoid arthritis treated with conventional synthetic disease-modifying antirheumatic drugs (DESIRABLE study): A randomised, double-blind, placebo-controlled phase 3 trial. Ann. Rheum. Dis. 2019, 78, 899–907.

182. Tanaka, S. RANKL is a therapeutic target of bone destruction in rheumatoid arthritis. F1000Res. 2019, 8. [CrossRef] [PubMed]

183. Tanaka, Y. Clinical immunity in bone and joints. J. Bone Miner. Metab. 2019, 37, 2–8. [CrossRef] [PubMed]

184. Fukuyo, S.; Yamaoka, K.; Sonomoto, K.; Oshita, K.; Okada, Y.; Saito, K.; Yoshida, Y.; Kanazawa, T.; Minami, Y.; Tanaka, Y. IL-6-accelerated calcification by induction of ROR2 in human adipose tissue-derived mesenchymal stem cells is STAT3 dependent. Rheumatology 2014, 53, 1282–1290. [CrossRef] [PubMed]

185. Sonomoto, K.; Yamaoka, K.; Oshita, K.; Fukuyo, S.; Zhang, X.; Nakano, K.; Okada, Y.; Tanaka, Y. Interleukin-1beta induces differentiation of human mesenchymal stem cells into osteoblasts via the Wnt-5a/receptor tyrosine kinase-like orphan receptor 2 pathway. Arthritis Rheum. 2012, 64, 3355–3363. [CrossRef] [PubMed]

186. Pridgeon, M.G.; Grohar, P.J.; Steensma, M.R.; Williams, B.O. Wnt Signaling in Ewing Sarcoma, Osteosarcoma, and Malignant Peripheral Nerve Sheath Tumors. Curr. Osteoporos. Rep. 2017, 15, 239–246. [CrossRef] [PubMed]

187. Kansara, M.; Tsang, M.; Kodjabachian, L.; Sims, N.A.; Trivett, M.K.; Ehrich, M.; Dobrovic, A.; Slavin, J.; Choong, P.F.; Simmons, P.J.; et al. Wnt inhibitory factor 1 is epigenetically silenced in human osteosarcoma, and targeted disruption accelerates osteosarcomagenesis in mice. J. Clin. Investig. 2009, 119, 837–851. [CrossRef] [PubMed]

188. Baker, E.K.; Taylor, S.; Gupte, A.; Chalk, A.M.; Bhattacharya, S.; Green, A.C.; Martin, T.J.; Strbenac, D.; Robinson, M.D.; Purton, L.E.; et al. Wnt inhibitory factor 1 (WIF1) is a marker of osteoblastic differentiation stage and is not silenced by DNA methylation in osteosarcoma. Bone 2015, 73, 223–232. [CrossRef] [PubMed]

189. Hoang, B.H.; Kubo, T.; Healey, J.H.; Yang, R.; Nathan, S.S.; Kolb, E.A.; Mazza, B.; Meyers, P.A.; Gorlick, R. Dickkopf 3 inhibits invasion and motility of Saos-2 osteosarcoma cells by modulating the Wnt-beta-catenin pathway. Cancer Res. 2004, 64, 2734–2739. [CrossRef]

190. Lin, C.H.; Guo, Y.; Ghaffar, S.; McQueen, P.; Pourmorady, J.; Christ, A.; Rooney, K.; Ji, T.; Eskander, R.; Zi, X.; et al. Dkk-3, a secreted wnt antagonist, suppresses tumorigenic potential and pulmonary metastasis in osteosarcoma. Sarcoma 2013, 2013, 147541. [CrossRef]

191. Techavichit, P.; Gao, Y.; Kurenbekova, L.; Shuck, R.; Donehower, L.A.; Yustein, J.T. Secreted Frizzled-Related Protein 2 (sFRP2) promotes osteosarcoma invasion and metastatic potential. BMC Cancer 2016, 16, 869. [CrossRef]

192. Danieau, G.; Morice, S.; Redini, F.; Verrecchia, F.; Royer, B.B. New Insights about the Wnt/beta-Catenin Signaling Pathway in Primary Bone Tumors and Their Microenvironment: A Promising Target to Develop Therapeutic Strategies? Int. J. Mol. Sci. 2019, 20, 3751. [CrossRef] [PubMed]

193. Delattre, O.; Zucman, J.; Plougastel, B.; Desmaze, C.; Melot, T.; Peter, M.; Kovar, H.; Joubert, I.; de Jong, P.; Rouleau, G.; et al. Gene fusion with an ETS DNA-binding domain caused by chromosome translocation in human tumours. Nature 1992, 359, 162–165. [CrossRef] [PubMed]

194. Delattre, O.; Zucman, J.; Melot, T.; Garau, X.S.; Zucker, J.M.; Lenoir, G.M.; Ambros, P.F.; Sheer, D.; Turc-Carel, C.; Triche, T.J.; et al. The Ewing family of tumors—A subgroup of small-round-cell tumors defined by specific chimeric transcripts. N. Engl. J. Med. 1994, 331, 294–299. [CrossRef] [PubMed]

195. Kauer, M.; Ban, J.; Kofler, R.; Walker, B.; Davis, S.; Meltzer, P.; Kovar, H. A molecular function map of Ewing’s sarcoma. PLoS ONE 2009, 4, e5415. [CrossRef]

196. Tanaka, M.; Yamazaki, Y.; Kanno, Y.; Igarashi, K.; Aisaki, K.; Kanno, J.; Nakamura, T. Ewing’s sarcoma precursors are highly enriched in embryonic osteochondrogenic progenitors. J. Clin. Investig. 2014, 124, 3061–3074. [CrossRef]

197. Crompton, J.G.; Ogura, K.; Bernthal, N.M.; Kawai, A.; Eilber, F.C. Local Control of Soft Tissue and Bone Sarcomas. J. Clin. Oncol. 2018, 36, 111–117. [CrossRef]

198. Shimozaki, S.; Yamamoto, N.; Domoto, T.; Nishida, H.; Hayashi, K.; Kimura, H.; Takeuchi, A.; Miwa, S.; Igarashi, K.; Kato, T.; et al. Efficacy of glycogen synthase kinase-3beta targeting against osteosarcoma via activation of beta-catenin. Oncotarget 2016, 7, 77038–77051. [CrossRef]

199. Wang, Z.; Smith, K.S.; Murphy, M.; Piloto, O.; Somervaille, T.C.; Cleary, M.L. Glycogen synthase kinase 3 in MLL leukaemia maintenance and targeted therapy. Nature 2008, 455, 1205–1209. [CrossRef]

200. Xiao, H.; Jensen, P.E.; Chen, X. Elimination of Osteosarcoma by Necroptosis with Graphene Oxide-Associated Anti-HER2 Antibodies. Int. J. Mol. Sci. 2019, 20, 4360. [CrossRef]

201. Abe, M.; Harada, T.; Matsumoto, T. Concise review: Defining and targeting myeloma stem cell-like cells. Stem. Cells 2014, 32, 1067–1073. [CrossRef]

202. Marino, S.; Roodman, G.D. Multiple Myeloma and Bone: The Fatal Interaction. Cold Spring Harb. Perspect. Med. 2018, 8. [CrossRef] [PubMed]

203. Eda, H.; Santo, L.; Wein, M.N.; Hu, D.Z.; Cirstea, D.D.; Nemani, N.; Tai, Y.T.; Raines, S.E.; Kuhstoss, S.A.; Munshi, N.C.; et al. Regulation of Sclerostin Expression in Multiple Myeloma by Dkk-1: A Potential Therapeutic Strategy for Myeloma Bone Disease. J. Bone Miner. Res. 2016, 31, 1225–1234. [CrossRef] [PubMed]

204. Delgado-Calle, J.; Anderson, J.; Cregor, M.D.; Condon, K.W.; Kuhstoss, S.A.; Plotkin, L.I.; Bellido, T.; Roodman, G.D. Genetic deletion of Sost or pharmacological inhibition of sclerostin prevent multiple myeloma-induced bone disease without affecting tumor growth. Leukemia 2017, 31, 2686–2694. [CrossRef] [PubMed]

205. McDonald, M.M.; Reagan, M.R.; Youlten, S.E.; Mohanty, S.T.; Seckinger, A.; Terry, R.L.; Pettitt, J.A.; Simic, M.K.; Cheng, T.L.; Morse, A.; et al. Inhibiting the osteocyte-specific protein sclerostin increases bone mass and fracture resistance in multiple myeloma. Blood 2017, 129, 3452–3464. [CrossRef] [PubMed]

206. Zhuang, X.; Zhang, H.; Li, X.; Li, X.; Cong, M.; Peng, F.; Yu, J.; Zhang, X.; Yang, Q.; Hu, G. Differential effects on lung and bone metastasis of breast cancer by Wnt signalling inhibitor DKK1. Nat. Cell Biol. 2017, 19, 1274–1285. [CrossRef]

207. D’Oronzo, S.; Coleman, R.; Brown, J.; Silvestris, F. Metastatic bone disease: Pathogenesis and therapeutic options: Up-date on bone metastasis management. J. Bone Oncol. 2019, 15. [CrossRef]

208. Robinson, D.; Van Allen, E.M.; Wu, Y.M.; Schultz, N.; Lonigro, R.J.; Mosquera, J.M.; Montgomery, B.; Taplin, M.E.; Pritchard, C.C.; Attard, G.; et al. Integrative clinical genomics of advanced prostate cancer. Cell 2015, 161, 1215–1228. [CrossRef]

209. Bailey, P.; Chang, D.K.; Nones, K.; Johns, A.L.; Patch, A.M.; Gingras, M.C.; Miller, D.K.; Christ, A.N.; Bruxner, T.J.; Quinn, M.C.; et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 2016, 531, 47–52. [CrossRef]

210. Longerich, T.; Endris, V.; Neumann, O.; Rempel, E.; Kirchner, M.; Abadi, Z.; Uhrig, S.; Kriegsmann, M.; Weiss, K.H.; Breuhahn, K.; et al. RSPO2 gene rearrangement: A powerful driver of beta-catenin activation in liver tumours. Gut 2019, 68, 1287–1296. [CrossRef]

211. Tammela, T.; Sanchez-Rivera, F.J.; Cetinbas, N.M.; Wu, K.; Joshi, N.S.; Helenius, K.; Park, Y.; Azimi, R.; Kerper, N.R.; Wesselhoeft, R.A.; et al. A Wnt-producing niche drives proliferative potential and progression in lung adenocarcinoma. Nature 2017, 545, 355–359. [CrossRef]

212. Katoh, M.; Katoh, M. Molecular genetics and targeted therapy of WNT-related human diseases (Review). Int. J. Mol. Med. 2017, 40, 587–606. [CrossRef] [PubMed]

213. Madan, B.; Ke, Z.; Harmston, N.; Ho, S.Y.; Frois, A.O.; Alam, J.; Jeyaraj, D.A.; Pendharkar, V.; Ghosh, K.; Virshup, I.H.; et al. Wnt addiction of genetically defined cancers reversed by PORCN inhibition. Oncogene 2016, 35, 2197–2207. [CrossRef] [PubMed]

214. Liu, J.; Pan, S.; Hsieh, M.H.; Ng, N.; Sun, F.; Wang, T.; Kasibhatla, S.; Schuller, A.G.; Li, A.G.; Cheng, D.; et al. Targeting Wnt-driven cancer through the inhibition of Porcupine by LGK974. Proc. Natl. Acad. Sci. USA 2013, 110, 20224–20229. [CrossRef]

215. Le, P.N.; McDermott, J.D.; Jimeno, A. Targeting the Wnt pathway in human cancers: Therapeutic targeting with a focus on OMP-54F28. Pharmacol. Ther. 2015, 146, 1–11. [CrossRef] [PubMed]

216. Jimeno, A.; Gordon, M.; Chugh, R.; Messersmith, W.; Mendelson, D.; Dupont, J.; Stagg, R.; Kapoun, A.M.; Xu, L.; Uttamsingh, S.; et al. A First-in-Human Phase I Study of the Anticancer Stem Cell Agent Ipafricept (OMP-54F28), a Decoy Receptor for Wnt Ligands, in Patients with Advanced Solid Tumors. Clin. Cancer Res. 2017, 23, 7490–7497. [CrossRef]

217. Giraudet, A.L.; Cassier, P.A.; Iwao-Fukukawa, C.; Garin, G.; Badel, J.N.; Kryza, D.; Chabaud, S.; Gilles-Afchain, L.; Clapisson, G.; Desuzinges, C.; et al. A first-in-human study investigating biodistribution, safety and recommended dose of a new radiolabeled MAb targeting FZD10 in metastatic synovial sarcoma patients. BMC Cancer 2018, 18, 646. [CrossRef]

218. Gurney, A.; Axelrod, F.; Bond, C.J.; Cain, J.; Chartier, C.; Donigan, L.; Fischer, M.; Chaudhari, A.; Ji, M.; Kapoun, A.M.; et al. Wnt pathway inhibition via the targeting of Frizzled receptors results in decreased growth and tumorigenicity of human tumors. Proc. Natl. Acad. Sci. USA 2012, 109, 11717–11722. [CrossRef]

219. Lenz, H.J.; Kahn, M. Safely targeting cancer stem cells via selective catenin coactivator antagonism. Cancer Sci. 2014, 105, 1087–1092. [CrossRef]

220. Manegold, P.; Lai, K.K.Y.; Wu, Y.; Teo, J.L.; Lenz, H.J.; Genyk, Y.S.; Pandol, S.J.; Wu, K.; Lin, D.P.; Chen, Y.; et al. Differentiation Therapy Targeting the beta-Catenin/CBP Interaction in Pancreatic Cancer. Cancers 2018, 10, 95. [CrossRef]

221. Yu, J.; Chen, L.; Cui, B.; Widhopf, G.F., II; Shen, Z.; Wu, R.; Zhang, L.; Zhang, S.; Briggs, S.P.; Kipps, T.J. Wnt5a induces ROR1/ROR2 heterooligomerization to enhance leukemia chemotaxis and proliferation. J. Clin. Investig. 2016, 126, 585–598. [CrossRef]

222. Choi, M.Y.; Widhopf, G.F., II; Ghia, E.M.; Kidwell, R.L.; Hasan, M.K.; Yu, J.; Rassenti, L.Z.; Chen, L.; Chen, Y.; Pittman, E.; et al. Phase I Trial: Cirmtuzumab Inhibits ROR1 Signaling and Stemness Signatures in Patients with Chronic Lymphocytic Leukemia. Cell Stem Cell 2018, 22, 951–959.e3. [CrossRef] [PubMed]

223. Funck-Brentano, T.; Nilsson, K.H.; Brommage, R.; Henning, P.; Lerner, U.H.; Koskela, A.; Tuukkanen, J.; Cohen-Solal, M.; Moverare-Skrtic, S.; Ohlsson, C. Porcupine inhibitors impair trabecular and cortical bone mass and strength in mice. J. Endocrinol. 2018, 238, 13–23. [CrossRef] [PubMed]

224. Madan, B.; McDonald, M.J.; Foxa, G.E.; Diegel, C.R.; Williams, B.O.; Virshup, D.M. Bone loss from Wnt inhibition mitigated by concurrent alendronate therapy. Bone Res. 2018, 6, 17. [CrossRef] [PubMed]

225. Van Bezooijen, R.L.; Deruiter, M.C.; Vilain, N.; Monteiro, R.M.; Visser, A.; van der Wee-Pals, L.; van Munsteren, C.J.; Hogendoorn, P.C.; Aguet, M.; Mummery, C.L.; et al. SOST expression is restricted to the great arteries during embryonic and neonatal cardiovascular development. Dev. Dyn. 2007, 236, 606–612. [CrossRef] [PubMed]

226. Wang, X.R.; Yuan, L.; Zhang, J.J.; Hao, L.; Wang, D.G. Serum sclerostin values are associated with abdominal aortic calcification and predict cardiovascular events in patients with chronic kidney disease stages 3-5D. Nephrology 2017, 22, 286–292. [CrossRef]

227. Sato, M.; Hanafusa, N.; Kawaguchi, H.; Tsuchiya, K.; Nitta, K. A Prospective Cohort Study Showing No Association Between Serum Sclerostin Level and Mortality in Maintenance Hemodialysis Patients. Kidney Blood Press. Res. 2018, 43, 1023–1033. [CrossRef]

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る