• Aucun résultat trouvé

78

Stimulation of Type 1 parathyroid hormone receptor (PTH1R) mediates activation of multiple signaling pathways. As a paradigm, PTH-stimulated cAMP/PKA and IP3/Ca2+ signaling in kidney and bone is essential for its effects on serum calcium homeostasis and bone turnover, whereas MAPK signaling in osteoblasts may play an important role on osteoblastic proliferation and differentiation and thus on bone formation (128, 191, 205, 214). Cytoplasmic molecules such as -arrestin that interact with both PTH1R and multiple signaling effectors have been shown to regulate G proteins coupling/uncoupling and PTH1R fate/trafficking (245, 248, 251). In addition, -arrestin knock out mice have decreased bone mass and have an altered response to PTH anabolic stimulus on trabecular bone and an improved cortical response to anabolic PTH (249, 253). Our work now bridges these in vitro and in vivo findings by showing that -arrestin also mediates PTH-stimulated activation of MAPK ERK1/2 through c-Src and in turn that MAPKs activation is important for osteoblastic differentiation.

This discussion will now focus on the analysis and understanding of the somewhat divergent results obtained in HEK293 and osteoblastic cells.

First concerning PTH-induced MAPKs activation, both ERK1/2 and p38 were stimulated in osteoblastic (MC4) cells whereas in HEK293 cells and primary osteoblastic cells only ERK1/2 was activated. The activation profile was also different with a delayed activation of about one hour in MC4 cells and in contrary very rapid (after 5 min.) and transient in HEK293 cells. Consistent with our findings in HEK293 cells and primary osteoblastic cells, another study using osteoblastic (MC4) cells found that PTH-induced ERK1/2 activation was rapid (peak at 5 min) (129). In that study, however, ERK1/2 activation by PTH was apparently cAMP-dependent. In our hands, on another side, p38 activation in MC4 cells was cAMP/PKA-dependent (and delayed until 1 hour), whereas ERK1/2 activation was not. These differences could partly be explained by the presence or absence of B-Raf in MC4 cells and HEK293 cells respectively (129, 293). Moreover, the experimental conditions slightly differed [time of culture for MC4 cells was shorter (3-4 days) compared to ours (8-9 days)], suggesting that PTH1R-mediated signaling pathways

79

may be dependent on the cell type, duration and conditions of culture. Thus, in our culture conditions the delayed activation in MC4 cells could be explained by an indirect mechanism such as for instance an inhibition of phosphatase(s). The relevance of these findings with regard to physiological and pathophysiological processes in vivo should therefore be taken with caution. At best, these experiments show the potentiality of PTH and PTH1R to activate multiple signaling pathways, without necessarily indicating that theses events all occur in every tissue and/or conditions where the receptor is expressed.

Molecular mechanisms of ERK1/2 activation by various GPCRs have been extensively studied. In general, each GPCR is able to activate ERK1/2 through a variety of signaling pathways. Second messengers between the receptor and ERK1/2 are often similar, but their activation mechanisms and interactions with other molecules differ from each other. For example, c-Src is implicated in 2-adrenergic receptor (AR)-mediated ERK1/2 activation by binding to -arrestin (235), whereas c-Src directly binds to the 3-AR to induce ERK1/2 activation (228). It appears difficult therefore to predict the mechanisms of MAPKs activation through GPCRs based solely on their structural homology. In HEK293 cells we found that PTH-induced ERK1/2 activation was Gq/PKC-dependent, as also described in rat osteoblastic cells and opossum kidney cells (128, 206). Of note, in HEK293 cells B-Raf is absent (293), precluding direct activation by cAMP/PKA pathway and potentially explaining the predominant implication of Gq/PKC signaling for ERK1/2 activation in these cells (see figure 10 to see the signaling pathways). It is not described if B-Raf is present or absent in rat osteoblastic and opossum kidney cells (128, 206). On another side it has been shown that the Gq/PKC signaling pathway is implicated in PTH1R phosphorylation, subsequent -arrestin recruitment and PTH1R internalization (245).

Hence, in addition to directly activate ERK1/2, the Gq/PKC signaling pathway may trigger the cross talk between G proteins activation and -arrestin-dependent activation of ERK1/2.

80

In this study, we show more precisely that activation of Gq induces ERK1/2 phosphorylation by two different molecular mechanisms, one via the classical activation cascade of q-(PLC-/PKC/Raf1/MEK)-ERK1/2 and the other through the activation cascade of /c-Src-(Ras/Raf1/MEK)-ERK1/2 (see Figure 10). This latter shows the essential role of c-Src in ERK1/2 activation but also in stabilization of PTH1R internalization. Hence, activated c-Src binds to the PxxP motifs of PTH1R cytoplasmic tail through its SH3 domains and to -arrestin probably via its SH1 domain. Moreover, activation of transcription induced by this molecular complex shows that activated ERK1/2 migrate into the nucleus and does not only phosphorylate cytosolic substrates as generally described for GPCRs (234). Hence, ERK1/2 activation by Gq appears to be a general mechanism by which many GPCRs signals could eventually stimulate ERK1/2-dependent transcription. In contrast, the pathway involving the scaffolding of -arrestin2/c-Src/ to PTH1R polyproline motifs (PxxP) is quite unique to this receptor so far. Importantly, such mechanisms will target the signaling response towards its specific stimulus, in this case PTH.

Such targeting mechanisms could be crucial to determine the function of intracellular MAPK signaling. For instance, osteoblasts express both PTH1R and 2-adrenergic receptors (2-AR), yet stimulation by their respective agonists, PTH and noradrenalin (NA), has opposite effects on osteoblast functions (stimulation and inhibition of bone formation respectively) (71, 72, 94). Coupling the ERK1/2 activation cascade directly to the stimulated receptor could therefore copy the cellular response to its specific stimulus.

The purpose of using MC4 cells was to study molecular mechanisms of PTH-induced osteoblastic differentiation and mineralization. Here we confirm that PTH-stimulated cAMP is an important pathway to increase ALP activity and osteocalcin production (135-137, 195) which are both important for osteoblastic differentiation and mineralization. The pathway towards PTH-induced ALP activity was shown to be dependent on a definite signaling cascade beginning with cAMP production, then PKA and p38 activation, whereas for osteocalcin production, we showed that it was

81

dependent of cAMP, PKA and ERK1/2 without observing a definite activation cascade between these three molecules.

In these cells, we also show that PTH-induced p38 and ERK1/2 activation were delayed. This delayed activation could also be explained by an indirect mechanism such as for instance an inhibition of phosphatase(s). In fact, this response is different compared with a previous report in the same cells in which PTH induced a rapid activation of ERK1/2 that was found to be cAMP/B-Raf-dependent (129). The difference between our and this previous report is probably related to variation in culture conditions used to analyze PTH signaling. In the previous study, PTH was investigated in cells cultured for 3-4 days whereas in our analysis, PTH was added on cells cultured for 8-10 days. Thus, it seems that signaling pathways activated by PTH are dependent on the stage of differentiation.

Moreover, in our culture conditions, we observed that the basal activation of p38 increases with culture duration and it seems that PTH is able to induce its phosphorylation only during the differentiation phase and not during the proliferative phase. Therefore, p38 activation seems to be important to induce osteoblastic differentiation and PTH is able to increase its activity.

As a perspective and to investigate the relevance of the molecular mechanisms identified in vitro, the next step would be to test in vivo specific MAPKs inhibitors in mice treated with PTH and, in order to be more specific, to create an inducible knock out mice for p38 and ERK1/2 (tetracycline-on/-off system). These experiments would allow to investigate the role of MAPKs in PTH anabolic effects on bone.

Moreover to further elucidate the role of -arrestin in mediating PTH effects on bone, our group is currently developing osteoblasts targeted -arrestin1 and -arrestin2 knock out mice. All these models could be used to compare intermittent versus continuous PTH treatment in order to further delineate the role of -arrestin in PTH1R-mediated signaling and responses at the tissue level.

From our work, we can conclude that PTH has the property to activate multiple signaling pathways. This may therefore explain why, depending on the cell type and

82

localization in vivo, PTH may exert different, and sometimes opposite effects at the cellular level. Studies on shorter PTH analogs have depicted precise regions responsible for cAMP activation (aa 1-3), PKC stimulation (aa 29-32), and calcium mobilization (aa 3-7) (179, 180). Gardella et al. have shown that PTH analogs of only 11 or 14 amino acids are sufficient to activate PTH1R (294). Considering the complex receptor conformational changes required to bind -arrestin (295), it is however likely that very short PTH analogues have potential limitations with regard to triggering other signalling pathways, i.e. MAPKs. Our studies delineating the molecular mechanisms of PTH activity also show the complexity and intrications of PTH1Rc-mediated signaling pathways. Therefore, PTH appears to be more than a one-shot molecule and its effects on bone, as used for osteoporosis treatment, may encompass the variety of molecular events described here.

83

References

84

1. Lee, C.A., Einhorn, T. A. 2001. The bone organ system. In Osteoporosis. R.

Marcus, Feldman, D.,Kesley, J., editor. San Diego: Academic Press. 3-20.

2. Aubin, J.E., Turksen, K., and Heersche, J.N.M. 1993. Osteoblastic cell lineage.

In Cellular and molecular biology of bone. M. Noda, editor. New York: Academic Press. 1-45.

3. Aubin, J.E. 2000. Osteogenic cell differentiation. In Bone Engineering. J.E. Davis, editor. Toronton. 19-30.

4. Yamaguchi, A., Komori, T., and Suda, T. 2000. Regulation of osteoblast differentiation mediated by bone morphogenetic proteins, hedgehogs, and Cbfa1. Endocr Rev 21:393-411.

5. Nakashima, K., Zhou, X., Kunkel, G., Zhang, Z., Deng, J.M., Behringer, R.R., and de Crombrugghe, B. 2002. The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation. Cell 108:17-29.

6. Hu, H., Hilton, M.J., Tu, X., Yu, K., Ornitz, D.M., and Long, F. 2005. Sequential roles of Hedgehog and Wnt signaling in osteoblast development. Development 132:49-60.

7. Hill, T.P., Spater, D., Taketo, M.M., Birchmeier, W., and Hartmann, C. 2005.

Canonical Wnt/beta-catenin signaling prevents osteoblasts from differentiating into chondrocytes. Dev Cell 8:727-738.

8. Day, T.F., Guo, X., Garrett-Beal, L., and Yang, Y. 2005. Wnt/beta-catenin signaling in mesenchymal progenitors controls osteoblast and chondrocyte differentiation during vertebrate skeletogenesis. Dev Cell 8:739-750.

9. Ducy, P., Zhang, R., Geoffroy, V., Ridall, A.L., and Karsenty, G. 1997.

Osf2/Cbfa1: a transcriptional activator of osteoblast differentiation. Cell 89:747-754.

10. Komori, T., Yagi, H., Nomura, S., Yamaguchi, A., Sasaki, K., Deguchi, K., Shimizu, Y., Bronson, R.T., Gao, Y.H., Inada, M., et al. 1997. Targeted

85

disruption of Cbfa1 results in a complete lack of bone formation owing to maturational arrest of osteoblasts. Cell 89:755-764.

11. Hoshi, K., Komori, T., and Ozawa, H. 1999. Morphological characterization of skeletal cells in Cbfa1-deficient mice. Bone 25:639-651.

12. Fajas, L., Debril, M.B., and Auwerx, J. 2001. Peroxisome proliferator-activated receptor-gamma: from adipogenesis to carcinogenesis. J Mol Endocrinol 27:1-9.

13. Schilling, T., Noth, U., Klein-Hitpass, L., Jakob, F., and Schutze, N. 2007.

Plasticity in adipogenesis and osteogenesis of human mesenchymal stem cells.

Mol Cell Endocrinol 271:1-17.

14. Soroceanu, M.A., Miao, D., Bai, X.Y., Su, H., Goltzman, D., and Karaplis, A.C.

2004. Rosiglitazone impacts negatively on bone by promoting osteoblast/osteocyte apoptosis. J Endocrinol 183:203-216.

15. Rzonca, S.O., Suva, L.J., Gaddy, D., Montague, D.C., and Lecka-Czernik, B.

2004. Bone is a target for the antidiabetic compound rosiglitazone.

Endocrinology 145:401-406.

16. Akune, T., Ohba, S., Kamekura, S., Yamaguchi, M., Chung, U.I., Kubota, N., Terauchi, Y., Harada, Y., Azuma, Y., Nakamura, K., et al. 2004. PPARgamma insufficiency enhances osteogenesis through osteoblast formation from bone marrow progenitors. J Clin Invest 113:846-855.

17. Ali, A.A., Weinstein, R.S., Stewart, S.A., Parfitt, A.M., Manolagas, S.C., and Jilka, R.L. 2005. Rosiglitazone causes bone loss in mice by suppressing osteoblast differentiation and bone formation. Endocrinology 146:1226-1235.

18. 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. 2001. LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development. Cell 107:513-523.

19. Little, R.D., Carulli, J.P., Del Mastro, R.G., Dupuis, J., Osborne, M., Folz, C., Manning, S.P., Swain, P.M., Zhao, S.C., Eustace, B., et al. 2002. A mutation in the LDL receptor-related protein 5 gene results in the autosomal dominant high-bone-mass trait. Am J Hum Genet 70:11-19.

86

20. Boyden, L.M., Mao, J., Belsky, J., Mitzner, L., Farhi, A., Mitnick, M.A., Wu, D., Insogna, K., and Lifton, R.P. 2002. High bone density due to a mutation in LDL-receptor-related protein 5. N Engl J Med 346:1513-1521.

21. Semenov, M., Tamai, K., and He, X. 2005. SOST is a ligand for LRP5/LRP6 and a Wnt signaling inhibitor. J Biol Chem 280:26770-26775.

22. Tian, E., Zhan, F., Walker, R., Rasmussen, E., Ma, Y., Barlogie, B., and Shaughnessy, J.D., Jr. 2003. The role of the Wnt-signaling antagonist DKK1 in the development of osteolytic lesions in multiple myeloma. N Engl J Med 349:2483-2494.

23. Li, X., Liu, P., Liu, W., Maye, P., Zhang, J., Zhang, Y., Hurley, M., Guo, C., Boskey, A., Sun, L., et al. 2005. Dkk2 has a role in terminal osteoblast differentiation and mineralized matrix formation. Nat Genet 37:945-952.

24. Bodine, P.V., Zhao, W., Kharode, Y.P., Bex, F.J., Lambert, A.J., Goad, M.B., Gaur, T., Stein, G.S., Lian, J.B., and Komm, B.S. 2004. The Wnt antagonist secreted frizzled-related protein-1 is a negative regulator of trabecular bone formation in adult mice. Mol Endocrinol 18:1222-1237.

25. McCabe, L.R., Kockx, M., Lian, J., Stein, J., and Stein, G. 1995. Selective expression of fos- and jun-related genes during osteoblast proliferation and differentiation. Exp Cell Res 218:255-262.

26. Choi, J.Y., Lee, B.H., Song, K.B., Park, R.W., Kim, I.S., Sohn, K.Y., Jo, J.S., and Ryoo, H.M. 1996. Expression patterns of bone-related proteins during osteoblastic differentiation in MC3T3-E1 cells. J Cell Biochem 61:609-618.

27. Stein, G.S., Lian, J.B., Stein J.S., et al. 1996. Mechanisms regulating osteoblast proliferation and differentiation. In Principles of Bone Biology. J.P. Bilezikian, Raisz, L.G., Rodan, G.A., editor. San Diego, CA, USA: Academic Press. 69-86.

28. Ducy, P., Desbois, C., Boyce, B., Pinero, G., Story, B., Dunstan, C., Smith, E., Bonadio, J., Goldstein, S., Gundberg, C., et al. 1996. Increased bone formation in osteocalcin-deficient mice. Nature 382:448-452.

87

29. Lee, N.K., Sowa, H., Hinoi, E., Ferron, M., Ahn, J.D., Confavreux, C., Dacquin, R., Mee, P.J., McKee, M.D., Jung, D.Y., et al. 2007. Endocrine regulation of energy metabolism by the skeleton. Cell 130:456-469.

30. Goldring, M.B., and Goldring, S.R. 1990. Skeletal tissue response to cytokines.

Clin Orthop Relat Res:245-278.

31. Tondravi, M.M., McKercher, S.R., Anderson, K., Erdmann, J.M., Quiroz, M., Maki, R., and Teitelbaum, S.L. 1997. Osteopetrosis in mice lacking haematopoietic transcription factor PU.1. Nature 386:81-84.

32. Cui, W., Cuartas, E., Ke, J., Zhang, Q., Einarsson, H.B., Sedgwick, J.D., Li, J., and Vignery, A. 2007. CD200 and its receptor, CD200R, modulate bone mass via the differentiation of osteoclasts. Proc Natl Acad Sci U S A 104:14436-14441.

33. Lacey, D.L., Timms, E., Tan, H.L., Kelley, M.J., Dunstan, C.R., Burgess, T., Elliott, R., Colombero, A., Elliott, G., Scully, S., et al. 1998. Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell 93:165-176.

34. Yasuda, H., Shima, N., Nakagawa, N., Yamaguchi, K., Kinosaki, M., Mochizuki, S., Tomoyasu, A., Yano, K., Goto, M., Murakami, A., et al. 1998.

Osteoclast differentiation factor is a ligand for osteoprotegerin/osteoclastogenesis-inhibitory factor and is identical to TRANCE/RANKL. Proc Natl Acad Sci U S A 95:3597-3602.

35. Burgess, T.L., Qian, Y., Kaufman, S., Ring, B.D., Van, G., Capparelli, C., Kelley, M., Hsu, H., Boyle, W.J., Dunstan, C.R., et al. 1999. The ligand for osteoprotegerin (OPGL) directly activates mature osteoclasts. J Cell Biol 145:527-538.

36. Li, Y.P., Chen, W., Liang, Y., Li, E., and Stashenko, P. 1999. Atp6i-deficient mice exhibit severe osteopetrosis due to loss of osteoclast-mediated extracellular acidification. Nat Genet 23:447-451.

37. Simonet, W.S., Lacey, D.L., Dunstan, C.R., Kelley, M., Chang, M.S., Luthy, R., Nguyen, H.Q., Wooden, S., Bennett, L., Boone, T., et al. 1997. Osteoprotegerin:

88

a novel secreted protein involved in the regulation of bone density. Cell 89:309-319.

38. Choi, Y., Woo, K.M., Ko, S.H., Lee, Y.J., Park, S.J., Kim, H.M., and Kwon, B.S.

2001. Osteoclastogenesis is enhanced by activated B cells but suppressed by activated CD8(+) T cells. Eur J Immunol 31:2179-2188.

39. Grcevic, D., Lee, S.K., Marusic, A., and Lorenzo, J.A. 2000. Depletion of CD4 and CD8 T lymphocytes in mice in vivo enhances 1,25-dihydroxyvitamin D3-stimulated osteoclast-like cell formation in vitro by a mechanism that is dependent on prostaglandin synthesis. J Immunol 165:4231-4238.

40. John, V., Hock, J.M., Short, L.L., Glasebrook, A.L., and Galvin, R.J. 1996. A role for CD8+ T lymphocytes in osteoclast differentiation in vitro. Endocrinology 137:2457-2463.

41. Li, Y., Toraldo, G., Li, A., Yang, X., Zhang, H., Qian, W.P., and Weitzmann, M.N. 2007. B cells and T cells are critical for the preservation of bone homeostasis and attainment of peak bone mass in vivo. Blood 109:3839-3848.

42. Parfitt, A.M. 1995. Problems in the application of in vitro systems to the study of human bone remodeling. Calcif Tissue Int 56 Suppl 1:S5-7.

43. Parfitt, A. 1993. Physiology and pharmacology of bone. Heidelberg: Springer.

1-65.

44. Frost, H.M. 1969. Tetracycline-based histological analysis of bone remodeling.

Calcif Tissue Res 3:211-237.

45. Burr, D.B., Martin, R.B., Schaffler, M.B., and Radin, E.L. 1985. Bone remodeling in response to in vivo fatigue microdamage. J Biomech 18:189-200.

46. Noble, B. 2003. Bone microdamage and cell apoptosis. Eur Cell Mater 6:46-55;

discussion 55.

47. Parfitt, A.M., Mundy, G.R., Roodman, G.D., Hughes, D.E., and Boyce, B.F.

1996. A new model for the regulation of bone resorption, with particular reference to the effects of bisphosphonates. J Bone Miner Res 11:150-159.

89

48. Parfitt, A.M. 1998. Osteoclast precursors as leukocytes: importance of the area code. Bone 23:491-494.

49. Parfitt, A.M. 2002. Targeted and nontargeted bone remodeling: relationship to basic multicellular unit origination and progression. Bone 30:5-7.

50. Riggs, B.L., and Parfitt, A.M. 2005. Drugs used to treat osteoporosis: the critical need for a uniform nomenclature based on their action on bone remodeling. J Bone Miner Res 20:177-184.

51. Zaidi, M. 2007. Skeletal remodeling in health and disease. Nat Med 13:791-801.

52. Rosen, C.J., and Donahue, L.R. 1998. Insulin-like growth factors and bone: the osteoporosis connection revisited. Proc Soc Exp Biol Med 219:1-7.

53. Bonewald, L.F., and Dallas, S.L. 1994. Role of active and latent transforming growth factor beta in bone formation. J Cell Biochem 55:350-357.

54. Sakou, T. 1998. Bone morphogenetic proteins: from basic studies to clinical approaches. Bone 22:591-603.

55. Kingsley, D.M. 1994. What do BMPs do in mammals? Clues from the mouse short-ear mutation. Trends Genet 10:16-21.

56. Zhang, M., Xuan, S., Bouxsein, M.L., von Stechow, D., Akeno, N., Faugere, M.C., Malluche, H., Zhao, G., Rosen, C.J., Efstratiadis, A., et al. 2002.

Osteoblast-specific knockout of the insulin-like growth factor (IGF) receptor gene reveals an essential role of IGF signaling in bone matrix mineralization. J Biol Chem 277:44005-44012.

57. Fiedler, J., Etzel, N., and Brenner, R.E. 2004. To go or not to go: Migration of human mesenchymal progenitor cells stimulated by isoforms of PDGF. J Cell Biochem 93:990-998.

58. Valta, M.P., Hentunen, T., Qu, Q., Valve, E.M., Harjula, A., Seppanen, J.A., Vaananen, H.K., and Harkonen, P.L. 2006. Regulation of osteoblast differentiation: a novel function for fibroblast growth factor 8. Endocrinology 147:2171-2182.

90

59. Zhao, C., Irie, N., Takada, Y., Shimoda, K., Miyamoto, T., Nishiwaki, T., Suda, T., and Matsuo, K. 2006. Bidirectional ephrinB2-EphB4 signaling controls bone homeostasis. Cell Metab 4:111-121.

60. Gardiner, E.M., Baldock, P.A., Thomas, G.P., Sims, N.A., Henderson, N.K., Hollis, B., White, C.P., Sunn, K.L., Morrison, N.A., Walsh, W.R., et al. 2000.

Increased formation and decreased resorption of bone in mice with elevated vitamin D receptor in mature cells of the osteoblastic lineage. Faseb J 14:1908-1916.

61. Hock, J.M., and Gera, I. 1992. Effects of continuous and intermittent administration and inhibition of resorption on the anabolic response of bone to parathyroid hormone. J Bone Miner Res 7:65-72.

62. Tam, C.S., Heersche, J.N., Murray, T.M., and Parsons, J.A. 1982. Parathyroid hormone stimulates the bone apposition rate independently of its resorptive action: differential effects of intermittent and continuous administration.

Endocrinology 110:506-512.

63. Ongphiphadhanakul, B., Piaseu, N., Chailurkit, L., and Rajatanavin, R. 1998.

Suppression of bone resorption in early postmenopausal women by intranasal salmon calcitonin in relation to dosage and basal bone turnover. Calcif Tissue Int 62:379-382.

64. van der Wiel, H.E., Lips, P., Nauta, J., Kwakkel, G., Hazenberg, G., Netelenbos, J.C., and van der Vijgh, W.J. 1993. Intranasal calcitonin suppresses increased bone resorption during short-term immobilization: a double-blind study of the effects of intranasal calcitonin on biochemical parameters of bone turnover. J Bone Miner Res 8:1459-1465.

65. Syed, F., and Khosla, S. 2005. Mechanisms of sex steroid effects on bone.

Biochem Biophys Res Commun 328:688-696.

66. Kawaguchi, H., Pilbeam, C.C., and Raisz, L.G. 1994. Anabolic effects of 3,3',5-triiodothyronine and triiodothyroacetic acid in cultured neonatal mouse parietal bones. Endocrinology 135:971-976.

91

67. Sun, L., Peng, Y., Sharrow, A.C., Iqbal, J., Zhang, Z., Papachristou, D.J., Zaidi, S., Zhu, L.L., Yaroslavskiy, B.B., Zhou, H., et al. 2006. FSH directly regulates bone mass. Cell 125:247-260.

68. Ducy, P., Amling, M., Takeda, S., Priemel, M., Schilling, A.F., Beil, F.T., Shen, J., Vinson, C., Rueger, J.M., and Karsenty, G. 2000. Leptin inhibits bone formation through a hypothalamic relay: a central control of bone mass. Cell 100:197-207.

69. Friedman, J.M., and Halaas, J.L. 1998. Leptin and the regulation of body weight in mammals. Nature 395:763-770.

70. Spiegelman, B.M., and Flier, J.S. 1996. Adipogenesis and obesity: rounding out the big picture. Cell 87:377-389.

71. Takeda, S., Elefteriou, F., Levasseur, R., Liu, X., Zhao, L., Parker, K.L., Armstrong, D., Ducy, P., and Karsenty, G. 2002. Leptin regulates bone formation via the sympathetic nervous system. Cell 111:305-317.

72. Elefteriou, F., Ahn, J.D., Takeda, S., Starbuck, M., Yang, X., Liu, X., Kondo, H., Richards, W.G., Bannon, T.W., Noda, M., et al. 2005. Leptin regulation of bone resorption by the sympathetic nervous system and CART. Nature 434:514-520.

73. Baldock, P.A., Sainsbury, A., Couzens, M., Enriquez, R.F., Thomas, G.P., Gardiner, E.M., and Herzog, H. 2002. Hypothalamic Y2 receptors regulate bone formation. J Clin Invest 109:915-921.

74. Baldock, P.A., Allison, S., McDonald, M.M., Sainsbury, A., Enriquez, R.F., Little, D.G., Eisman, J.A., Gardiner, E.M., and Herzog, H. 2006. Hypothalamic regulation of cortical bone mass: opposing activity of Y2 receptor and leptin pathways. J Bone Miner Res 21:1600-1607.

75. Lundberg, P., Allison, S.J., Lee, N.J., Baldock, P.A., Brouard, N., Rost, S., Enriquez, R.F., Sainsbury, A., Lamghari, M., Simmons, P., et al. 2007. Greater bone formation of Y2 knockout mice is associated with increased osteoprogenitor numbers and altered Y1 receptor expression. J Biol Chem 282:19082-19091.

92

76. Sato, S., Hanada, R., Kimura, A., Abe, T., Matsumoto, T., Iwasaki, M., Inose, H., Ida, T., Mieda, M., Takeuchi, Y., et al. 2007. Central control of bone remodeling by neuromedin U. Nat Med 13:1234-1240.

77. Raisz, L.G. 1997. The osteoporosis revolution. Ann Intern Med 126:458-462.

78. Riggs, B.L., Khosla, S., and Melton, L.J., 3rd. 1998. A unitary model for involutional osteoporosis: estrogen deficiency causes both type I and type II osteoporosis in postmenopausal women and contributes to bone loss in aging

78. Riggs, B.L., Khosla, S., and Melton, L.J., 3rd. 1998. A unitary model for involutional osteoporosis: estrogen deficiency causes both type I and type II osteoporosis in postmenopausal women and contributes to bone loss in aging