Skip to main content
Top
Published in: Journal of Bone and Mineral Metabolism 5/2015

01-09-2015 | Review Article

Roles of leptin in bone metabolism and bone diseases

Authors: Xu Xu Chen, Tianfu Yang

Published in: Journal of Bone and Mineral Metabolism | Issue 5/2015

Login to get access

Abstract

Adipose tissue has been more accepted as an active contributor to whole body homeostasis, rather than just a fat depot, since leptin, a 16 kDa protein, was discovered as the product of the obese gene in 1994. With more and more studies conducted on this hormone, it has been shown that there is a close relationship between adipose tissue and bone, which have important effects on each other. Bone is the source of many hormones, such as osteocalcin, that can affect energy metabolism and then the anabolism or catabolism of fat tissue. In contrast, the adipose tissue synthesizes and releases a series of adipokines, which are involved in bone metabolism through direct or indirect effects on bone formation and resorption. Interestingly, leptin, one of the most important cytokines derived from fat tissue, seems to account for the largest part of effects on bone, through direct or indirect involvement in bone remodeling and by playing a significant role in many bone diseases, such as osteoporosis, osteoarthritis, rheumatic arthritis, bone tumors and even fractures. In this review, we will discuss the progress in leptin research, particularly focusing on the roles of leptin in bone diseases.
Literature
1.
go back to reference Ducy P, Amling M, Takeda S, Priemel M, Schilling AF, Beil FT, Shen J, Vinson C, Rueger JM, Karsenty G (2000) Leptin inhibits bone formation through a hypothalamic relay: a central control of bone mass. Cell 100:197–207PubMedCrossRef Ducy P, Amling M, Takeda S, Priemel M, Schilling AF, Beil FT, Shen J, Vinson C, Rueger JM, Karsenty G (2000) Leptin inhibits bone formation through a hypothalamic relay: a central control of bone mass. Cell 100:197–207PubMedCrossRef
2.
go back to reference Iwaniec UT, Boghossian S, Lapke PD, Turner RT, Kalra SP (2007) Central leptin gene therapy corrects skeletal abnormalities in leptin-deficient ob/ob mice. Peptides 28:1012–1019PubMedCentralPubMedCrossRef Iwaniec UT, Boghossian S, Lapke PD, Turner RT, Kalra SP (2007) Central leptin gene therapy corrects skeletal abnormalities in leptin-deficient ob/ob mice. Peptides 28:1012–1019PubMedCentralPubMedCrossRef
3.
go back to reference Hamrick MW, Pennington C, Newton D, Xie D, Isales C (2004) Leptin deficiency produces contrasting phenotypes in bones of the limb and spine. Bone 34:376–383PubMedCrossRef Hamrick MW, Pennington C, Newton D, Xie D, Isales C (2004) Leptin deficiency produces contrasting phenotypes in bones of the limb and spine. Bone 34:376–383PubMedCrossRef
4.
go back to reference Parm AL, Jurimae J, Saar M, Parna K, Tillmann V, Maasalu K, Neissaar I, Jurimae T (2011) Plasma adipocytokine and ghrelin levels in relation to bone mineral density in prepubertal rhythmic gymnasts. J Bone Miner Metab 29:717–724PubMedCrossRef Parm AL, Jurimae J, Saar M, Parna K, Tillmann V, Maasalu K, Neissaar I, Jurimae T (2011) Plasma adipocytokine and ghrelin levels in relation to bone mineral density in prepubertal rhythmic gymnasts. J Bone Miner Metab 29:717–724PubMedCrossRef
5.
go back to reference Hamrick MW, Ferrari SL (2008) Leptin and the sympathetic connection of fat to bone. Osteoporos Int 19:905–912PubMedCrossRef Hamrick MW, Ferrari SL (2008) Leptin and the sympathetic connection of fat to bone. Osteoporos Int 19:905–912PubMedCrossRef
6.
go back to reference Tartaglia LA, Dembski M, Weng X, Deng N, Culpepper J, Devos R, Richards GJ, Campfield LA, Clark FT, Deeds J, Muir C, Sanker S, Moriarty A, Moore KJ, Smutko JS, Mays GG, Wool EA, Monroe CA, Tepper RI (1995) Identification and expression cloning of a leptin receptor, OB-R. Cell 83:1263–1271PubMedCrossRef Tartaglia LA, Dembski M, Weng X, Deng N, Culpepper J, Devos R, Richards GJ, Campfield LA, Clark FT, Deeds J, Muir C, Sanker S, Moriarty A, Moore KJ, Smutko JS, Mays GG, Wool EA, Monroe CA, Tepper RI (1995) Identification and expression cloning of a leptin receptor, OB-R. Cell 83:1263–1271PubMedCrossRef
7.
go back to reference Takeda S, Elefteriou F, Levasseur R, Liu X, Zhao L, Parker KL, Armstrong D, Ducy P, Karsenty G (2002) Leptin regulates bone formation via the sympathetic nervous system. Cell 111:305–317PubMedCrossRef Takeda S, Elefteriou F, Levasseur R, Liu X, Zhao L, Parker KL, Armstrong D, Ducy P, Karsenty G (2002) Leptin regulates bone formation via the sympathetic nervous system. Cell 111:305–317PubMedCrossRef
8.
go back to reference Elefteriou F, Ahn JD, Takeda S, Starbuck M, Yang X, Liu X, Kondo H, Richards WG, Bannon TW, Noda M, Clement K, Vaisse C, Karsenty G (2005) Leptin regulation of bone resorption by the sympathetic nervous system and CART. Nature 434:514–520PubMedCrossRef Elefteriou F, Ahn JD, Takeda S, Starbuck M, Yang X, Liu X, Kondo H, Richards WG, Bannon TW, Noda M, Clement K, Vaisse C, Karsenty G (2005) Leptin regulation of bone resorption by the sympathetic nervous system and CART. Nature 434:514–520PubMedCrossRef
9.
go back to reference Karsenty G (2006) Convergence between bone and energy homeostases: leptin regulation of bone mass. Cell Metab 4:341–348PubMedCrossRef Karsenty G (2006) Convergence between bone and energy homeostases: leptin regulation of bone mass. Cell Metab 4:341–348PubMedCrossRef
10.
go back to reference Schwetz V, Pieber T, Obermayer-Pietsch B (2012) The endocrine role of the skeleton: background and clinical evidence. Eur J Endocrinol 166:959–967PubMedCrossRef Schwetz V, Pieber T, Obermayer-Pietsch B (2012) The endocrine role of the skeleton: background and clinical evidence. Eur J Endocrinol 166:959–967PubMedCrossRef
11.
go back to reference Benovic JL, Bouvier M, Caron MG, Lefkowitz RJ (1988) Regulation of adenylyl cyclase-coupled beta-adrenergic receptors. Annu Rev Cell Biol 4:405–428PubMedCrossRef Benovic JL, Bouvier M, Caron MG, Lefkowitz RJ (1988) Regulation of adenylyl cyclase-coupled beta-adrenergic receptors. Annu Rev Cell Biol 4:405–428PubMedCrossRef
12.
go back to reference Fu L, Patel MS, Bradley A, Wagner EF, Karsenty G (2005) The molecular clock mediates leptin-regulated bone formation. Cell 122:803–815PubMedCrossRef Fu L, Patel MS, Bradley A, Wagner EF, Karsenty G (2005) The molecular clock mediates leptin-regulated bone formation. Cell 122:803–815PubMedCrossRef
13.
go back to reference Yang X, Matsuda K, Bialek P, Jacquot S, Masuoka HC, Schinke T, Li L, Brancorsini S, Sassone-Corsi P, Townes TM, Hanauer A, Karsenty G (2004) ATF4 is a substrate of RSK2 and an essential regulator of osteoblast biology; implication for Coffin–Lowry syndrome. Cell 117:387–398PubMedCrossRef Yang X, Matsuda K, Bialek P, Jacquot S, Masuoka HC, Schinke T, Li L, Brancorsini S, Sassone-Corsi P, Townes TM, Hanauer A, Karsenty G (2004) ATF4 is a substrate of RSK2 and an essential regulator of osteoblast biology; implication for Coffin–Lowry syndrome. Cell 117:387–398PubMedCrossRef
14.
go back to reference Teitelbaum SL, Ross FP (2003) Genetic regulation of osteoclast development and function. Nat Rev Genet 4:638–649PubMedCrossRef Teitelbaum SL, Ross FP (2003) Genetic regulation of osteoclast development and function. Nat Rev Genet 4:638–649PubMedCrossRef
15.
go back to reference Kristensen P, Judge ME, Thim L, Ribel U, Christjansen KN, Wulff BS, Clausen JT, Jensen PB, Madsen OD, Vrang N, Larsen PJ, Hastrup S (1998) Hypothalamic CART is a new anorectic peptide regulated by leptin. Nature 393:72–76PubMedCrossRef Kristensen P, Judge ME, Thim L, Ribel U, Christjansen KN, Wulff BS, Clausen JT, Jensen PB, Madsen OD, Vrang N, Larsen PJ, Hastrup S (1998) Hypothalamic CART is a new anorectic peptide regulated by leptin. Nature 393:72–76PubMedCrossRef
16.
go back to reference Karsenty G, Oury F (2010) The central regulation of bone mass, the first link between bone remodeling and energy metabolism. J Clin Endocrinol Metab 95:4795–4801PubMedCrossRef Karsenty G, Oury F (2010) The central regulation of bone mass, the first link between bone remodeling and energy metabolism. J Clin Endocrinol Metab 95:4795–4801PubMedCrossRef
17.
go back to reference Yadav VK, Oury F, Suda N, Liu ZW, Gao XB, Confavreux C, Klemenhagen KC, Tanaka KF, Gingrich JA, Guo XE, Tecott LH, Mann JJ, Hen R, Horvath TL, Karsenty G (2009) A serotonin-dependent mechanism explains the leptin regulation of bone mass, appetite, and energy expenditure. Cell 138:976–989PubMedCentralPubMedCrossRef Yadav VK, Oury F, Suda N, Liu ZW, Gao XB, Confavreux C, Klemenhagen KC, Tanaka KF, Gingrich JA, Guo XE, Tecott LH, Mann JJ, Hen R, Horvath TL, Karsenty G (2009) A serotonin-dependent mechanism explains the leptin regulation of bone mass, appetite, and energy expenditure. Cell 138:976–989PubMedCentralPubMedCrossRef
18.
go back to reference Hanada R, Teranishi H, Pearson JT, Kurokawa M, Hosoda H, Fukushima N, Fukue Y, Serino R, Fujihara H, Ueta Y, Ikawa M, Okabe M, Murakami N, Shirai M, Yoshimatsu H, Kangawa K, Kojima M (2004) Neuromedin U has a novel anorexigenic effect independent of the leptin signaling pathway. Nat Med 10:1067–1073PubMedCrossRef Hanada R, Teranishi H, Pearson JT, Kurokawa M, Hosoda H, Fukushima N, Fukue Y, Serino R, Fujihara H, Ueta Y, Ikawa M, Okabe M, Murakami N, Shirai M, Yoshimatsu H, Kangawa K, Kojima M (2004) Neuromedin U has a novel anorexigenic effect independent of the leptin signaling pathway. Nat Med 10:1067–1073PubMedCrossRef
19.
go back to reference Baldock PA, Allison S, McDonald MM, Sainsbury A, Enriquez RF, Little DG, Eisman JA, Gardiner EM, Herzog H (2006) Hypothalamic regulation of cortical bone mass: opposing activity of Y2 receptor and leptin pathways. J Bone Miner Res 21:1600–1607PubMedCrossRef Baldock PA, Allison S, McDonald MM, Sainsbury A, Enriquez RF, Little DG, Eisman JA, Gardiner EM, Herzog H (2006) Hypothalamic regulation of cortical bone mass: opposing activity of Y2 receptor and leptin pathways. J Bone Miner Res 21:1600–1607PubMedCrossRef
20.
go back to reference Baldock PA, Sainsbury A, Couzens M, Enriquez RF, Thomas GP, Gardiner EM, Herzog H (2002) Hypothalamic Y2 receptors regulate bone formation. J Clin Investig 109:915–921PubMedCentralPubMedCrossRef Baldock PA, Sainsbury A, Couzens M, Enriquez RF, Thomas GP, Gardiner EM, Herzog H (2002) Hypothalamic Y2 receptors regulate bone formation. J Clin Investig 109:915–921PubMedCentralPubMedCrossRef
21.
go back to reference Brighton PJ, Szekeres PG, Willars GB (2004) Neuromedin U and its receptors: structure, function, and physiological roles. Pharmacol Rev 56:231–248PubMedCrossRef Brighton PJ, Szekeres PG, Willars GB (2004) Neuromedin U and its receptors: structure, function, and physiological roles. Pharmacol Rev 56:231–248PubMedCrossRef
22.
go back to reference Sato S, Hanada R, Kimura A, Abe T, Matsumoto T, Iwasaki M, Inose H, Ida T, Mieda M, Takeuchi Y, Fukumoto S, Fujita T, Kato S, Kangawa K, Kojima M, Shinomiya K, Takeda S (2007) Central control of bone remodeling by neuromedin U. Nat Med 13:1234–1240PubMedCrossRef Sato S, Hanada R, Kimura A, Abe T, Matsumoto T, Iwasaki M, Inose H, Ida T, Mieda M, Takeuchi Y, Fukumoto S, Fujita T, Kato S, Kangawa K, Kojima M, Shinomiya K, Takeda S (2007) Central control of bone remodeling by neuromedin U. Nat Med 13:1234–1240PubMedCrossRef
23.
go back to reference Howard AD, Wang R, Pong SS, Mellin TN, Strack A et al (2000) Identification of receptors for neuromedin U and its role in feeding. Nature 406:70–74PubMedCrossRef Howard AD, Wang R, Pong SS, Mellin TN, Strack A et al (2000) Identification of receptors for neuromedin U and its role in feeding. Nature 406:70–74PubMedCrossRef
24.
go back to reference Wren AM, Small CJ, Abbott CR, Jethwa PH, Kennedy AR, Murphy KG, Stanley SA, Zollner AN, Ghatei MA, Bloom SR (2002) Hypothalamic actions of neuromedin U. Endocrinology 143:4227–4234PubMedCrossRef Wren AM, Small CJ, Abbott CR, Jethwa PH, Kennedy AR, Murphy KG, Stanley SA, Zollner AN, Ghatei MA, Bloom SR (2002) Hypothalamic actions of neuromedin U. Endocrinology 143:4227–4234PubMedCrossRef
25.
go back to reference Cirmanova V, Bayer M, Starka L, Zajickova K (2008) The effect of leptin on bone: an evolving concept of action. Physiol Res 57 Suppl 1:S143–S151PubMed Cirmanova V, Bayer M, Starka L, Zajickova K (2008) The effect of leptin on bone: an evolving concept of action. Physiol Res 57 Suppl 1:S143–S151PubMed
26.
go back to reference Lin S, Boey D, Herzog H (2004) NPY and Y receptors: lessons from transgenic and knockout models. Neuropeptides 38:189–200PubMedCrossRef Lin S, Boey D, Herzog H (2004) NPY and Y receptors: lessons from transgenic and knockout models. Neuropeptides 38:189–200PubMedCrossRef
27.
go back to reference Baldock PA, Lee NJ, Driessler F, Lin S, Allison S et al (2009) Neuropeptide Y knockout mice reveal a central role of NPY in the coordination of bone mass to body weight. PLoS ONE 4:e8415PubMedCentralPubMedCrossRef Baldock PA, Lee NJ, Driessler F, Lin S, Allison S et al (2009) Neuropeptide Y knockout mice reveal a central role of NPY in the coordination of bone mass to body weight. PLoS ONE 4:e8415PubMedCentralPubMedCrossRef
29.
go back to reference Teixeira L, Sousa DM, Nunes AF, Sousa MM, Herzog H, Lamghari M (2009) NPY revealed as a critical modulator of osteoblast function in vitro: new insights into the role of Y1 and Y2 receptors. J Cell Biochem 107:908–916PubMedCrossRef Teixeira L, Sousa DM, Nunes AF, Sousa MM, Herzog H, Lamghari M (2009) NPY revealed as a critical modulator of osteoblast function in vitro: new insights into the role of Y1 and Y2 receptors. J Cell Biochem 107:908–916PubMedCrossRef
30.
go back to reference Shi YC, Baldock PA (2012) Central and peripheral mechanisms of the NPY system in the regulation of bone and adipose tissue. Bone 50:430–436PubMedCrossRef Shi YC, Baldock PA (2012) Central and peripheral mechanisms of the NPY system in the regulation of bone and adipose tissue. Bone 50:430–436PubMedCrossRef
31.
go back to reference Stephens TW, Basinski M, Bristow PK, Bue-Valleskey JM, Burgett SG, Craft L, Hale J, Hoffmann J, Hsiung HM, Kriauciunas A et al (1995) The role of neuropeptide Y in the antiobesity action of the obese gene product. Nature 377:530–532PubMedCrossRef Stephens TW, Basinski M, Bristow PK, Bue-Valleskey JM, Burgett SG, Craft L, Hale J, Hoffmann J, Hsiung HM, Kriauciunas A et al (1995) The role of neuropeptide Y in the antiobesity action of the obese gene product. Nature 377:530–532PubMedCrossRef
32.
go back to reference Allison SJ, Baldock PA, Herzog H (2007) The control of bone remodeling by neuropeptide Y receptors. Peptides 28:320–325PubMedCrossRef Allison SJ, Baldock PA, Herzog H (2007) The control of bone remodeling by neuropeptide Y receptors. Peptides 28:320–325PubMedCrossRef
33.
go back to reference Baldock PA, Sainsbury A, Allison S, Lin EJ, Couzens M, Boey D, Enriquez R, During M, Herzog H, Gardiner EM (2005) Hypothalamic control of bone formation: distinct actions of leptin and y2 receptor pathways. J Bone Miner Res 20:1851–1857PubMedCrossRef Baldock PA, Sainsbury A, Allison S, Lin EJ, Couzens M, Boey D, Enriquez R, During M, Herzog H, Gardiner EM (2005) Hypothalamic control of bone formation: distinct actions of leptin and y2 receptor pathways. J Bone Miner Res 20:1851–1857PubMedCrossRef
35.
go back to reference Mantzoros CS, Magkos F, Brinkoetter M, Sienkiewicz E, Dardeno TA, Kim SY, Hamnvik OP, Koniaris A (2011) Leptin in human physiology and pathophysiology. Am J Physiol Endocrinol Metab 301:E567–E584PubMedCentralPubMedCrossRef Mantzoros CS, Magkos F, Brinkoetter M, Sienkiewicz E, Dardeno TA, Kim SY, Hamnvik OP, Koniaris A (2011) Leptin in human physiology and pathophysiology. Am J Physiol Endocrinol Metab 301:E567–E584PubMedCentralPubMedCrossRef
36.
go back to reference Thomas T, Gori F, Khosla S, Jensen MD, Burguera B, Riggs BL (1999) Leptin acts on human marrow stromal cells to enhance differentiation to osteoblasts and to inhibit differentiation to adipocytes. Endocrinology 140:1630–1638PubMed Thomas T, Gori F, Khosla S, Jensen MD, Burguera B, Riggs BL (1999) Leptin acts on human marrow stromal cells to enhance differentiation to osteoblasts and to inhibit differentiation to adipocytes. Endocrinology 140:1630–1638PubMed
37.
go back to reference Hess R, Pino AM, Rios S, Fernandez M, Rodriguez JP (2005) High affinity leptin receptors are present in human mesenchymal stem cells (MSCs) derived from control and osteoporotic donors. J Cell Biochem 94:50–57PubMedCrossRef Hess R, Pino AM, Rios S, Fernandez M, Rodriguez JP (2005) High affinity leptin receptors are present in human mesenchymal stem cells (MSCs) derived from control and osteoporotic donors. J Cell Biochem 94:50–57PubMedCrossRef
38.
go back to reference Reseland JE, Syversen U, Bakke I, Qvigstad G, Eide LG, Hjertner O, Gordeladze JO, Drevon CA (2001) Leptin is expressed in and secreted from primary cultures of human osteoblasts and promotes bone mineralization. J Bone Miner Res 16:1426–1433PubMedCrossRef Reseland JE, Syversen U, Bakke I, Qvigstad G, Eide LG, Hjertner O, Gordeladze JO, Drevon CA (2001) Leptin is expressed in and secreted from primary cultures of human osteoblasts and promotes bone mineralization. J Bone Miner Res 16:1426–1433PubMedCrossRef
39.
go back to reference Astudillo P, Rios S, Pastenes L, Pino AM, Rodriguez JP (2008) Increased adipogenesis of osteoporotic human-mesenchymal stem cells (MSCs) characterizes by impaired leptin action. J Cell Biochem 103:1054–1065PubMedCrossRef Astudillo P, Rios S, Pastenes L, Pino AM, Rodriguez JP (2008) Increased adipogenesis of osteoporotic human-mesenchymal stem cells (MSCs) characterizes by impaired leptin action. J Cell Biochem 103:1054–1065PubMedCrossRef
40.
go back to reference Gordeladze JO, Drevon CA, Syversen U, Reseland JE (2002) Leptin stimulates human osteoblastic cell proliferation, de novo collagen synthesis, and mineralization: impact on differentiation markers, apoptosis, and osteoclastic signaling. J Cell Biochem 85:825–836PubMedCrossRef Gordeladze JO, Drevon CA, Syversen U, Reseland JE (2002) Leptin stimulates human osteoblastic cell proliferation, de novo collagen synthesis, and mineralization: impact on differentiation markers, apoptosis, and osteoclastic signaling. J Cell Biochem 85:825–836PubMedCrossRef
41.
go back to reference Holloway WR, Collier FM, Aitken CJ, Myers DE, Hodge JM, Malakellis M, Gough TJ, Collier GR, Nicholson GC (2002) Leptin inhibits osteoclast generation. J Bone Miner Res 17:200–209PubMedCrossRef Holloway WR, Collier FM, Aitken CJ, Myers DE, Hodge JM, Malakellis M, Gough TJ, Collier GR, Nicholson GC (2002) Leptin inhibits osteoclast generation. J Bone Miner Res 17:200–209PubMedCrossRef
42.
go back to reference Burguera B, Hofbauer LC, Thomas T, Gori F, Evans GL, Khosla S, Riggs BL, Turner RT (2001) Leptin reduces ovariectomy-induced bone loss in rats. Endocrinology 142:3546–3553PubMedCrossRef Burguera B, Hofbauer LC, Thomas T, Gori F, Evans GL, Khosla S, Riggs BL, Turner RT (2001) Leptin reduces ovariectomy-induced bone loss in rats. Endocrinology 142:3546–3553PubMedCrossRef
43.
go back to reference Martin A, de Vittoris R, David V, Moraes R, Begeot M, Lafage-Proust MH, Alexandre C, Vico L, Thomas T (2005) Leptin modulates both resorption and formation while preventing disuse-induced bone loss in tail-suspended female rats. Endocrinology 146:3652–3659PubMedCrossRef Martin A, de Vittoris R, David V, Moraes R, Begeot M, Lafage-Proust MH, Alexandre C, Vico L, Thomas T (2005) Leptin modulates both resorption and formation while preventing disuse-induced bone loss in tail-suspended female rats. Endocrinology 146:3652–3659PubMedCrossRef
44.
go back to reference Hamrick MW, Della-Fera MA, Choi YH, Pennington C, Hartzell D, Baile CA (2005) Leptin treatment induces loss of bone marrow adipocytes and increases bone formation in leptin-deficient ob/ob mice. J Bone Miner Res 20:994–1001PubMedCrossRef Hamrick MW, Della-Fera MA, Choi YH, Pennington C, Hartzell D, Baile CA (2005) Leptin treatment induces loss of bone marrow adipocytes and increases bone formation in leptin-deficient ob/ob mice. J Bone Miner Res 20:994–1001PubMedCrossRef
45.
go back to reference Turner RT, Kalra SP, Wong CP, Philbrick KA, Lindenmaier LB, Boghossian S, Iwaniec UT (2013) Peripheral leptin regulates bone formation. J Bone Miner Res 28:22–34PubMedCentralPubMedCrossRef Turner RT, Kalra SP, Wong CP, Philbrick KA, Lindenmaier LB, Boghossian S, Iwaniec UT (2013) Peripheral leptin regulates bone formation. J Bone Miner Res 28:22–34PubMedCentralPubMedCrossRef
46.
go back to reference Shi Y, Yadav VK, Suda N, Liu XS, Guo XE, Myers MG Jr, Karsenty G (2008) Dissociation of the neuronal regulation of bone mass and energy metabolism by leptin in vivo. Proc Natl Acad Sci USA 105:20529–20533PubMedCentralPubMedCrossRef Shi Y, Yadav VK, Suda N, Liu XS, Guo XE, Myers MG Jr, Karsenty G (2008) Dissociation of the neuronal regulation of bone mass and energy metabolism by leptin in vivo. Proc Natl Acad Sci USA 105:20529–20533PubMedCentralPubMedCrossRef
47.
go back to reference Figenschau Y, Knutsen G, Shahazeydi S, Johansen O, Sveinbjornsson B (2001) Human articular chondrocytes express functional leptin receptors. Biochem Biophys Res Commun 287:190–197PubMedCrossRef Figenschau Y, Knutsen G, Shahazeydi S, Johansen O, Sveinbjornsson B (2001) Human articular chondrocytes express functional leptin receptors. Biochem Biophys Res Commun 287:190–197PubMedCrossRef
48.
go back to reference Kishida Y, Hirao M, Tamai N, Nampei A, Fujimoto T, Nakase T, Shimizu N, Yoshikawa H, Myoui A (2005) Leptin regulates chondrocyte differentiation and matrix maturation during endochondral ossification. Bone 37:607–621PubMedCrossRef Kishida Y, Hirao M, Tamai N, Nampei A, Fujimoto T, Nakase T, Shimizu N, Yoshikawa H, Myoui A (2005) Leptin regulates chondrocyte differentiation and matrix maturation during endochondral ossification. Bone 37:607–621PubMedCrossRef
49.
go back to reference Maor G, Rochwerger M, Segev Y, Phillip M (2002) Leptin acts as a growth factor on the chondrocytes of skeletal growth centers. J Bone Miner Res 17:1034–1043PubMedCrossRef Maor G, Rochwerger M, Segev Y, Phillip M (2002) Leptin acts as a growth factor on the chondrocytes of skeletal growth centers. J Bone Miner Res 17:1034–1043PubMedCrossRef
50.
go back to reference Hebebrand J, Muller TD, Holtkamp K, Herpertz-Dahlmann B (2007) The role of leptin in anorexia nervosa: clinical implications. Mol Psychiatry 12:23–35PubMedCrossRef Hebebrand J, Muller TD, Holtkamp K, Herpertz-Dahlmann B (2007) The role of leptin in anorexia nervosa: clinical implications. Mol Psychiatry 12:23–35PubMedCrossRef
51.
go back to reference Katsiki N, Mikhailidis DP, Gotzamani-Psarrakou A, Yovos JG, Karamitsos D (2011) Effect of various treatments on leptin, adiponectin, ghrelin and neuropeptide Y in patients with type 2 diabetes mellitus. Expert Opin Ther Targets 15:401–420PubMed Katsiki N, Mikhailidis DP, Gotzamani-Psarrakou A, Yovos JG, Karamitsos D (2011) Effect of various treatments on leptin, adiponectin, ghrelin and neuropeptide Y in patients with type 2 diabetes mellitus. Expert Opin Ther Targets 15:401–420PubMed
52.
53.
go back to reference Zhou R, Deng J, Zhang M, Zhou HD, Wang YJ (2011) Association between bone mineral density and the risk of Alzheimer’s disease. J Alzheimer’s Dis 24:101–108 Zhou R, Deng J, Zhang M, Zhou HD, Wang YJ (2011) Association between bone mineral density and the risk of Alzheimer’s disease. J Alzheimer’s Dis 24:101–108
55.
go back to reference Thomas T, Burguera B, Melton LJ 3rd, Atkinson EJ, O’Fallon WM, Riggs BL, Khosla S (2001) Role of serum leptin, insulin, and estrogen levels as potential mediators of the relationship between fat mass and bone mineral density in men versus women. Bone 29:114–120PubMedCrossRef Thomas T, Burguera B, Melton LJ 3rd, Atkinson EJ, O’Fallon WM, Riggs BL, Khosla S (2001) Role of serum leptin, insulin, and estrogen levels as potential mediators of the relationship between fat mass and bone mineral density in men versus women. Bone 29:114–120PubMedCrossRef
56.
go back to reference Yamauchi M, Sugimoto T, Yamaguchi T, Nakaoka D, Kanzawa M, Yano S, Ozuru R, Sugishita T, Chihara K (2001) Plasma leptin concentrations are associated with bone mineral density and the presence of vertebral fractures in postmenopausal women. Clin Endocrinol 55:341–347CrossRef Yamauchi M, Sugimoto T, Yamaguchi T, Nakaoka D, Kanzawa M, Yano S, Ozuru R, Sugishita T, Chihara K (2001) Plasma leptin concentrations are associated with bone mineral density and the presence of vertebral fractures in postmenopausal women. Clin Endocrinol 55:341–347CrossRef
57.
go back to reference Grigorie D, Neacsu E, Marinescu M, Popa O (2003) Circulating osteoprotegerin and leptin levels in postmenopausal women with and without osteoporosis. Roman J Int Med 41:409–415 Grigorie D, Neacsu E, Marinescu M, Popa O (2003) Circulating osteoprotegerin and leptin levels in postmenopausal women with and without osteoporosis. Roman J Int Med 41:409–415
58.
go back to reference Roux C, Arabi A, Porcher R, Garnero P (2003) Serum leptin as a determinant of bone resorption in healthy postmenopausal women. Bone 33:847–852PubMedCrossRef Roux C, Arabi A, Porcher R, Garnero P (2003) Serum leptin as a determinant of bone resorption in healthy postmenopausal women. Bone 33:847–852PubMedCrossRef
59.
go back to reference Sebastian-Ochoa A, Fernandez-Garcia D, Reyes-Garcia R, Mezquita-Raya P, Rozas-Moreno P, Alonso-Garcia G, Munoz-Torres M (2012) Adiponectin and leptin serum levels in osteoporotic postmenopausal women treated with raloxifene or alendronate. Menopause (New York, NY) 19:172–177CrossRef Sebastian-Ochoa A, Fernandez-Garcia D, Reyes-Garcia R, Mezquita-Raya P, Rozas-Moreno P, Alonso-Garcia G, Munoz-Torres M (2012) Adiponectin and leptin serum levels in osteoporotic postmenopausal women treated with raloxifene or alendronate. Menopause (New York, NY) 19:172–177CrossRef
60.
go back to reference Hipmair G, Bohler N, Maschek W, Soriguer F, Rojo-Martinez G, Schimetta W, Pichler R (2010) Serum leptin is correlated to high turnover in osteoporosis. Neuro Endocrinol Lett 31:155–160PubMed Hipmair G, Bohler N, Maschek W, Soriguer F, Rojo-Martinez G, Schimetta W, Pichler R (2010) Serum leptin is correlated to high turnover in osteoporosis. Neuro Endocrinol Lett 31:155–160PubMed
61.
go back to reference Di Carlo C, Tommaselli GA, Gargano V, Sammartino A, Bifulco G, Tauchmanova L, Colao A, Nappi C (2007) Effects of estrogen-progestin therapy on serum levels of RANKL, osteoprotegerin, osteocalcin, leptin, and ghrelin in postmenopausal women. Menopause (New York, NY) 14:38–44CrossRef Di Carlo C, Tommaselli GA, Gargano V, Sammartino A, Bifulco G, Tauchmanova L, Colao A, Nappi C (2007) Effects of estrogen-progestin therapy on serum levels of RANKL, osteoprotegerin, osteocalcin, leptin, and ghrelin in postmenopausal women. Menopause (New York, NY) 14:38–44CrossRef
62.
go back to reference Sato M, Takeda N, Sarui H, Takami R, Takami K, Hayashi M, Sasaki A, Kawachi S, Yoshino K, Yasuda K (2001) Association between serum leptin concentrations and bone mineral density, and biochemical markers of bone turnover in adult men. J Clin Endocrinol Metab 86:5273–5276PubMedCrossRef Sato M, Takeda N, Sarui H, Takami R, Takami K, Hayashi M, Sasaki A, Kawachi S, Yoshino K, Yasuda K (2001) Association between serum leptin concentrations and bone mineral density, and biochemical markers of bone turnover in adult men. J Clin Endocrinol Metab 86:5273–5276PubMedCrossRef
63.
go back to reference Ormarsdottir S, Ljunggren O, Mallmin H, Olofsson H, Blum WF, Loof L (2001) Inverse relationship between circulating levels of leptin and bone mineral density in chronic liver disease. J Gastroenterol Hepatol 16:1409–1414PubMedCrossRef Ormarsdottir S, Ljunggren O, Mallmin H, Olofsson H, Blum WF, Loof L (2001) Inverse relationship between circulating levels of leptin and bone mineral density in chronic liver disease. J Gastroenterol Hepatol 16:1409–1414PubMedCrossRef
64.
go back to reference Fontana L, Shew JL, Holloszy JO, Villareal DT (2005) Low bone mass in subjects on a long-term raw vegetarian diet. Arch Intern Med 165:684–689PubMedCrossRef Fontana L, Shew JL, Holloszy JO, Villareal DT (2005) Low bone mass in subjects on a long-term raw vegetarian diet. Arch Intern Med 165:684–689PubMedCrossRef
65.
go back to reference Pobeha P, Ukropec J, Skyba P, Ukropcova B, Joppa P, Kurdiova T, Javorsky M, Klimes I, Tkac I, Gasperikova D, Tkacova R (2011) Relationship between osteoporosis and adipose tissue leptin and osteoprotegerin in patients with chronic obstructive pulmonary disease. Bone 48:1008–1014PubMedCrossRef Pobeha P, Ukropec J, Skyba P, Ukropcova B, Joppa P, Kurdiova T, Javorsky M, Klimes I, Tkac I, Gasperikova D, Tkacova R (2011) Relationship between osteoporosis and adipose tissue leptin and osteoprotegerin in patients with chronic obstructive pulmonary disease. Bone 48:1008–1014PubMedCrossRef
66.
go back to reference Odabasi E, Ozata M, Turan M, Bingol N, Yonem A, Cakir B, Kutlu M, Ozdemir IC (2000) Plasma leptin concentrations in postmenopausal women with osteoporosis. Eur J Endocrinol 142:170–173PubMedCrossRef Odabasi E, Ozata M, Turan M, Bingol N, Yonem A, Cakir B, Kutlu M, Ozdemir IC (2000) Plasma leptin concentrations in postmenopausal women with osteoporosis. Eur J Endocrinol 142:170–173PubMedCrossRef
67.
go back to reference Ruhl CE, Everhart JE (2002) Relationship of serum leptin concentration with bone mineral density in the United States population. J Bone Miner Res 17:1896–1903PubMedCrossRef Ruhl CE, Everhart JE (2002) Relationship of serum leptin concentration with bone mineral density in the United States population. J Bone Miner Res 17:1896–1903PubMedCrossRef
68.
go back to reference Hadji P, Bock K, Gotschalk M, Hars O, Backhus J, Emons G, Schulz KD (2003) The influence of serum leptin concentration on bone mass assessed by quantitative ultrasonometry in pre and postmenopausal women. Maturitas 44:141–148PubMedCrossRef Hadji P, Bock K, Gotschalk M, Hars O, Backhus J, Emons G, Schulz KD (2003) The influence of serum leptin concentration on bone mass assessed by quantitative ultrasonometry in pre and postmenopausal women. Maturitas 44:141–148PubMedCrossRef
69.
go back to reference Sahin G, Polat G, Baethis S, Milcan A, Baethdatoethlu O, Erdoethan C, Camdeviren H (2003) Body composition, bone mineral density, and circulating leptin levels in postmenopausal Turkish women. Rheumatol Int 3:87–91 Sahin G, Polat G, Baethis S, Milcan A, Baethdatoethlu O, Erdoethan C, Camdeviren H (2003) Body composition, bone mineral density, and circulating leptin levels in postmenopausal Turkish women. Rheumatol Int 3:87–91
70.
go back to reference Wu N, Wang QP, Li H, Wu XP, Sun ZQ, Luo XH (2010) Relationships between serum adiponectin, leptin concentrations and bone mineral density, and bone biochemical markers in Chinese women. Clin Chim Acta 411:771–775PubMedCrossRef Wu N, Wang QP, Li H, Wu XP, Sun ZQ, Luo XH (2010) Relationships between serum adiponectin, leptin concentrations and bone mineral density, and bone biochemical markers in Chinese women. Clin Chim Acta 411:771–775PubMedCrossRef
71.
go back to reference Gulhan I, Bilgili S, Gunaydin R, Gulhan S, Posaci C (2008) The effect of strontium ranelate on serum insulin like growth factor-1 and leptin levels in osteoporotic post-menopausal women: a prospective study. Arch Gynecol Obstet 278:437–441PubMedCrossRef Gulhan I, Bilgili S, Gunaydin R, Gulhan S, Posaci C (2008) The effect of strontium ranelate on serum insulin like growth factor-1 and leptin levels in osteoporotic post-menopausal women: a prospective study. Arch Gynecol Obstet 278:437–441PubMedCrossRef
72.
go back to reference Lajeunesse D, Pelletier JP, Martel-Pelletier J (2005) Osteoarthritis: a metabolic disease induced by local abnormal leptin activity? Curr Rheumatol Rep 7:79–81PubMedCrossRef Lajeunesse D, Pelletier JP, Martel-Pelletier J (2005) Osteoarthritis: a metabolic disease induced by local abnormal leptin activity? Curr Rheumatol Rep 7:79–81PubMedCrossRef
73.
go back to reference Aspden RM, Scheven BA, Hutchison JD (2001) Osteoarthritis as a systemic disorder including stromal cell differentiation and lipid metabolism. Lancet 357:1118–1120PubMedCrossRef Aspden RM, Scheven BA, Hutchison JD (2001) Osteoarthritis as a systemic disorder including stromal cell differentiation and lipid metabolism. Lancet 357:1118–1120PubMedCrossRef
74.
go back to reference Gegout PP, Francin PJ, Mainard D, Presle N (2008) Adipokines in osteoarthritis: friends or foes of cartilage homeostasis? Joint Bone Spine 75:669–671PubMedCrossRef Gegout PP, Francin PJ, Mainard D, Presle N (2008) Adipokines in osteoarthritis: friends or foes of cartilage homeostasis? Joint Bone Spine 75:669–671PubMedCrossRef
75.
go back to reference Cicuttini FM, Baker JR, Spector TD (1996) The association of obesity with osteoarthritis of the hand and knee in women: a twin study. J Rheumatol 23:1221–1226PubMed Cicuttini FM, Baker JR, Spector TD (1996) The association of obesity with osteoarthritis of the hand and knee in women: a twin study. J Rheumatol 23:1221–1226PubMed
76.
go back to reference Hu PF, Bao JP, Wu LD (2011) The emerging role of adipokines in osteoarthritis: a narrative review. Mol Biol Rep 38:873–878PubMedCrossRef Hu PF, Bao JP, Wu LD (2011) The emerging role of adipokines in osteoarthritis: a narrative review. Mol Biol Rep 38:873–878PubMedCrossRef
77.
go back to reference Gualillo O (2007) Further evidence for leptin involvement in cartilage homeostases. Osteoarthritis Cartilage 15:857–860PubMedCrossRef Gualillo O (2007) Further evidence for leptin involvement in cartilage homeostases. Osteoarthritis Cartilage 15:857–860PubMedCrossRef
78.
go back to reference Dumond H, Presle N, Terlain B, Mainard D, Loeuille D, Netter P, Pottie P (2003) Evidence for a key role of leptin in osteoarthritis. Arthritis Rheum 48:3118–3129PubMedCrossRef Dumond H, Presle N, Terlain B, Mainard D, Loeuille D, Netter P, Pottie P (2003) Evidence for a key role of leptin in osteoarthritis. Arthritis Rheum 48:3118–3129PubMedCrossRef
79.
go back to reference Luyten FP, Hascall VC, Nissley SP, Morales TI, Reddi AH (1988) Insulin-like growth factors maintain steady-state metabolism of proteoglycans in bovine articular cartilage explants. Arch Biochem Biophys 267:416–425PubMedCrossRef Luyten FP, Hascall VC, Nissley SP, Morales TI, Reddi AH (1988) Insulin-like growth factors maintain steady-state metabolism of proteoglycans in bovine articular cartilage explants. Arch Biochem Biophys 267:416–425PubMedCrossRef
80.
go back to reference Grimaud E, Heymann D, Redini F (2002) Recent advances in TGF-beta effects on chondrocyte metabolism. Potential therapeutic roles of TGF-beta in cartilage disorders. Cytokine Growth Factor Rev 13:241–257PubMedCrossRef Grimaud E, Heymann D, Redini F (2002) Recent advances in TGF-beta effects on chondrocyte metabolism. Potential therapeutic roles of TGF-beta in cartilage disorders. Cytokine Growth Factor Rev 13:241–257PubMedCrossRef
81.
go back to reference Simopoulou T, Malizos KN, Iliopoulos D, Stefanou N, Papatheodorou L, Ioannou M, Tsezou A (2007) Differential expression of leptin and leptin’s receptor isoform (Ob-Rb) mRNA between advanced and minimally affected osteoarthritic cartilage; effect on cartilage metabolism. Osteoarthritis Cartilage 15:872–883PubMedCrossRef Simopoulou T, Malizos KN, Iliopoulos D, Stefanou N, Papatheodorou L, Ioannou M, Tsezou A (2007) Differential expression of leptin and leptin’s receptor isoform (Ob-Rb) mRNA between advanced and minimally affected osteoarthritic cartilage; effect on cartilage metabolism. Osteoarthritis Cartilage 15:872–883PubMedCrossRef
82.
go back to reference Mutabaruka MS, Aoulad Aissa M, Delalandre A, Lavigne M, Lajeunesse D (2010) Local leptin production in osteoarthritis subchondral osteoblasts may be responsible for their abnormal phenotypic expression. Arthritis Res Ther 12:8CrossRef Mutabaruka MS, Aoulad Aissa M, Delalandre A, Lavigne M, Lajeunesse D (2010) Local leptin production in osteoarthritis subchondral osteoblasts may be responsible for their abnormal phenotypic expression. Arthritis Res Ther 12:8CrossRef
83.
go back to reference Anandacoomarasamy A, Giuffre BM, Leibman S, Caterson ID, Smith GS, Fransen M, Sambrook PN, March LM (2009) Delayed gadolinium-enhanced magnetic resonance imaging of cartilage: clinical associations in obese adults. J Rheumatol 36:1056–1062PubMedCrossRef Anandacoomarasamy A, Giuffre BM, Leibman S, Caterson ID, Smith GS, Fransen M, Sambrook PN, March LM (2009) Delayed gadolinium-enhanced magnetic resonance imaging of cartilage: clinical associations in obese adults. J Rheumatol 36:1056–1062PubMedCrossRef
84.
go back to reference Stannus OP, Jones G, Quinn SJ, Cicuttini FM, Dore D, Ding C (2010) The association between leptin, interleukin-6, and hip radiographic osteoarthritis in older people: a cross-sectional study. Arthritis Res Ther 12:19CrossRef Stannus OP, Jones G, Quinn SJ, Cicuttini FM, Dore D, Ding C (2010) The association between leptin, interleukin-6, and hip radiographic osteoarthritis in older people: a cross-sectional study. Arthritis Res Ther 12:19CrossRef
85.
go back to reference Ku JH, Lee CK, Joo BS, An BM, Choi SH, Wang TH, Cho HL (2009) Correlation of synovial fluid leptin concentrations with the severity of osteoarthritis. Clin Rheumatol 28:1431–1435PubMedCrossRef Ku JH, Lee CK, Joo BS, An BM, Choi SH, Wang TH, Cho HL (2009) Correlation of synovial fluid leptin concentrations with the severity of osteoarthritis. Clin Rheumatol 28:1431–1435PubMedCrossRef
86.
go back to reference Koskinen A, Vuolteenaho K, Nieminen R, Moilanen T, Moilanen E (2011) Leptin enhances MMP-1, MMP-3 and MMP-13 production in human osteoarthritic cartilage and correlates with MMP-1 and MMP-3 in synovial fluid from OA patients. Clin Exp Rheumatol 29:57–64PubMed Koskinen A, Vuolteenaho K, Nieminen R, Moilanen T, Moilanen E (2011) Leptin enhances MMP-1, MMP-3 and MMP-13 production in human osteoarthritic cartilage and correlates with MMP-1 and MMP-3 in synovial fluid from OA patients. Clin Exp Rheumatol 29:57–64PubMed
87.
go back to reference Qin J, Shi D, Dai J, Zhu L, Tsezou A, Jiang Q (2010) Association of the leptin gene with knee osteoarthritis susceptibility in a Han Chinese population: a case–control study. J Hum Genet 55:704–706PubMedCrossRef Qin J, Shi D, Dai J, Zhu L, Tsezou A, Jiang Q (2010) Association of the leptin gene with knee osteoarthritis susceptibility in a Han Chinese population: a case–control study. J Hum Genet 55:704–706PubMedCrossRef
88.
go back to reference Iliopoulos D, Malizos KN, Tsezou A (2007) Epigenetic regulation of leptin affects MMP-13 expression in osteoarthritic chondrocytes: possible molecular target for osteoarthritis therapeutic intervention. Ann Rheum Dis 66:1616–1621PubMedCentralPubMedCrossRef Iliopoulos D, Malizos KN, Tsezou A (2007) Epigenetic regulation of leptin affects MMP-13 expression in osteoarthritic chondrocytes: possible molecular target for osteoarthritis therapeutic intervention. Ann Rheum Dis 66:1616–1621PubMedCentralPubMedCrossRef
89.
go back to reference Vuolteenaho K, Koskinen A, Kukkonen M, Nieminen R, Paivarinta U, Moilanen T, Moilanen E (2009) Leptin enhances synthesis of proinflammatory mediators in human osteoarthritic cartilage—mediator role of NO in leptin-induced PGE2, IL-6, and IL-8 production. Mediators Inflamm 345838:13 Vuolteenaho K, Koskinen A, Kukkonen M, Nieminen R, Paivarinta U, Moilanen T, Moilanen E (2009) Leptin enhances synthesis of proinflammatory mediators in human osteoarthritic cartilage—mediator role of NO in leptin-induced PGE2, IL-6, and IL-8 production. Mediators Inflamm 345838:13
92.
go back to reference Gomez R, Conde J, Scotece M, Gomez-Reino JJ, Lago F, Gualillo O (2011) What’s new in our understanding of the role of adipokines in rheumatic diseases? Nat Rev Rheumatol 7:528–536PubMedCrossRef Gomez R, Conde J, Scotece M, Gomez-Reino JJ, Lago F, Gualillo O (2011) What’s new in our understanding of the role of adipokines in rheumatic diseases? Nat Rev Rheumatol 7:528–536PubMedCrossRef
93.
go back to reference Matarese G, Procaccini C, De Rosa V, Horvath TL, La Cava A (2010) Regulatory T cells in obesity: the leptin connection. Trends Mol Med 16:247–256PubMedCrossRef Matarese G, Procaccini C, De Rosa V, Horvath TL, La Cava A (2010) Regulatory T cells in obesity: the leptin connection. Trends Mol Med 16:247–256PubMedCrossRef
94.
go back to reference Notley CA, Ehrenstein MR (2010) The yin and yang of regulatory T cells and inflammation in RA. Nat Rev Rheumatol 6:572–577PubMedCrossRef Notley CA, Ehrenstein MR (2010) The yin and yang of regulatory T cells and inflammation in RA. Nat Rev Rheumatol 6:572–577PubMedCrossRef
95.
go back to reference Bokarewa M, Bokarew D, Hultgren O, Tarkowski A (2003) Leptin consumption in the inflamed joints of patients with rheumatoid arthritis. Ann Rheum Dis 62:952–956PubMedCentralPubMedCrossRef Bokarewa M, Bokarew D, Hultgren O, Tarkowski A (2003) Leptin consumption in the inflamed joints of patients with rheumatoid arthritis. Ann Rheum Dis 62:952–956PubMedCentralPubMedCrossRef
96.
go back to reference Gunaydin R, Kaya T, Atay A, Olmez N, Hur A, Koseoglu M (2006) Serum leptin levels in rheumatoid arthritis and relationship with disease activity. South Med J 99:1078–1083PubMedCrossRef Gunaydin R, Kaya T, Atay A, Olmez N, Hur A, Koseoglu M (2006) Serum leptin levels in rheumatoid arthritis and relationship with disease activity. South Med J 99:1078–1083PubMedCrossRef
97.
go back to reference Otero M, Lago R, Gomez R, Lago F, Dieguez C, Gomez-Reino JJ, Gualillo O (2006) Changes in plasma levels of fat-derived hormones adiponectin, leptin, resistin and visfatin in patients with rheumatoid arthritis. Ann Rheum Dis 65:1198–1201PubMedCentralPubMedCrossRef Otero M, Lago R, Gomez R, Lago F, Dieguez C, Gomez-Reino JJ, Gualillo O (2006) Changes in plasma levels of fat-derived hormones adiponectin, leptin, resistin and visfatin in patients with rheumatoid arthritis. Ann Rheum Dis 65:1198–1201PubMedCentralPubMedCrossRef
98.
go back to reference Ibrahim SM, Hamdy MS, Amer N (2008) Plasma and synovial fluid adipocytokines in patients with rheumatoid arthritis and osteoarthritis. Egypt J Immunol 15:159–170PubMed Ibrahim SM, Hamdy MS, Amer N (2008) Plasma and synovial fluid adipocytokines in patients with rheumatoid arthritis and osteoarthritis. Egypt J Immunol 15:159–170PubMed
99.
go back to reference Derdemezis CS, Filippatos TD, Voulgari PV, Tselepis AD, Drosos AA, Kiortsis DN (2009) Effects of a 6-month infliximab treatment on plasma levels of leptin and adiponectin in patients with rheumatoid arthritis. Fundam Clin Pharmacol 23:595–600PubMedCrossRef Derdemezis CS, Filippatos TD, Voulgari PV, Tselepis AD, Drosos AA, Kiortsis DN (2009) Effects of a 6-month infliximab treatment on plasma levels of leptin and adiponectin in patients with rheumatoid arthritis. Fundam Clin Pharmacol 23:595–600PubMedCrossRef
100.
go back to reference Rho YH, Solus J, Sokka T, Oeser A, Chung CP, Gebretsadik T, Shintani A, Pincus T, Stein CM (2009) Adipocytokines are associated with radiographic joint damage in rheumatoid arthritis. Arthritis Rheum 60:1906–1914PubMedCentralPubMedCrossRef Rho YH, Solus J, Sokka T, Oeser A, Chung CP, Gebretsadik T, Shintani A, Pincus T, Stein CM (2009) Adipocytokines are associated with radiographic joint damage in rheumatoid arthritis. Arthritis Rheum 60:1906–1914PubMedCentralPubMedCrossRef
101.
go back to reference Seven A, Guzel S, Aslan M, Hamuryudan V (2009) Serum and synovial fluid leptin levels and markers of inflammation in rheumatoid arthritis. Rheumatol Int 29:743–747PubMedCrossRef Seven A, Guzel S, Aslan M, Hamuryudan V (2009) Serum and synovial fluid leptin levels and markers of inflammation in rheumatoid arthritis. Rheumatol Int 29:743–747PubMedCrossRef
102.
go back to reference Olama SM, Senna MK, Elarman M (2012) Synovial/serum leptin ratio in rheumatoid arthritis: the association with activity and erosion. Rheumatol Int 32:683–690PubMedCrossRef Olama SM, Senna MK, Elarman M (2012) Synovial/serum leptin ratio in rheumatoid arthritis: the association with activity and erosion. Rheumatol Int 32:683–690PubMedCrossRef
103.
go back to reference Lee SW, Park MC, Park YB, Lee SK (2007) Measurement of the serum leptin level could assist disease activity monitoring in rheumatoid arthritis. Rheumatol Int 27:537–540PubMedCrossRef Lee SW, Park MC, Park YB, Lee SK (2007) Measurement of the serum leptin level could assist disease activity monitoring in rheumatoid arthritis. Rheumatol Int 27:537–540PubMedCrossRef
104.
go back to reference Targonska-Stepniak B, Majdan M, Dryglewska M (2008) Leptin serum levels in rheumatoid arthritis patients: relation to disease duration and activity. Rheumatol Int 28:585–591PubMedCrossRef Targonska-Stepniak B, Majdan M, Dryglewska M (2008) Leptin serum levels in rheumatoid arthritis patients: relation to disease duration and activity. Rheumatol Int 28:585–591PubMedCrossRef
105.
go back to reference Popa C, Netea MG, Radstake TR, van Riel PL, Barrera P, van der Meer JW (2005) Markers of inflammation are negatively correlated with serum leptin in rheumatoid arthritis. Ann Rheum Dis 64:1195–1198PubMedCentralPubMedCrossRef Popa C, Netea MG, Radstake TR, van Riel PL, Barrera P, van der Meer JW (2005) Markers of inflammation are negatively correlated with serum leptin in rheumatoid arthritis. Ann Rheum Dis 64:1195–1198PubMedCentralPubMedCrossRef
106.
go back to reference Anders HJ, Rihl M, Heufelder A, Loch O, Schattenkirchner M (1999) Leptin serum levels are not correlated with disease activity in patients with rheumatoid arthritis. Metab Clin Exp 48:745–748PubMedCrossRef Anders HJ, Rihl M, Heufelder A, Loch O, Schattenkirchner M (1999) Leptin serum levels are not correlated with disease activity in patients with rheumatoid arthritis. Metab Clin Exp 48:745–748PubMedCrossRef
107.
go back to reference Nishiya K, Nishiyama M, Chang A, Shinto A, Hashimoto K (2002) Serum leptin levels in patients with rheumatoid arthritis are correlated with body mass index. Rinsho Byori 50:524–527PubMed Nishiya K, Nishiyama M, Chang A, Shinto A, Hashimoto K (2002) Serum leptin levels in patients with rheumatoid arthritis are correlated with body mass index. Rinsho Byori 50:524–527PubMed
108.
go back to reference Hizmetli S, Kisa M, Gokalp N, Bakici MZ (2007) Are plasma and synovial fluid leptin levels correlated with disease activity in rheumatoid arthritis? Rheumatol Int 27:335–338PubMedCrossRef Hizmetli S, Kisa M, Gokalp N, Bakici MZ (2007) Are plasma and synovial fluid leptin levels correlated with disease activity in rheumatoid arthritis? Rheumatol Int 27:335–338PubMedCrossRef
109.
go back to reference Popa C, Netea MG, de Graaf J, van den Hoogen FH, Radstake TR, Toenhake-Dijkstra H, van Riel PL, van der Meer JW, Stalenhoef AF, Barrera P (2009) Circulating leptin and adiponectin concentrations during tumor necrosis factor blockade in patients with active rheumatoid arthritis. J Rheumatol 36:724–730PubMedCrossRef Popa C, Netea MG, de Graaf J, van den Hoogen FH, Radstake TR, Toenhake-Dijkstra H, van Riel PL, van der Meer JW, Stalenhoef AF, Barrera P (2009) Circulating leptin and adiponectin concentrations during tumor necrosis factor blockade in patients with active rheumatoid arthritis. J Rheumatol 36:724–730PubMedCrossRef
110.
go back to reference Garcia-Bermudez M, Gonzalez-Juanatey C, Rodriguez-Rodriguez L, Vazquez-Rodriguez TR, Miranda-Filloy JA, Fernandez-Gutierrez B, Llorca J, Martin J, Gonzalez-Gay MA (2011) Lack of association between LEP rs2167270 (19 G>A) polymorphism and disease susceptibility and cardiovascular disease in patients with rheumatoid arthritis. Clin Exp Rheumatol 29:293–298PubMed Garcia-Bermudez M, Gonzalez-Juanatey C, Rodriguez-Rodriguez L, Vazquez-Rodriguez TR, Miranda-Filloy JA, Fernandez-Gutierrez B, Llorca J, Martin J, Gonzalez-Gay MA (2011) Lack of association between LEP rs2167270 (19 G>A) polymorphism and disease susceptibility and cardiovascular disease in patients with rheumatoid arthritis. Clin Exp Rheumatol 29:293–298PubMed
111.
go back to reference Harle P, Sarzi-Puttini P, Cutolo M, Straub RH (2006) No change of serum levels of leptin and adiponectin during anti-tumour necrosis factor antibody treatment with adalimumab in patients with rheumatoid arthritis. Ann Rheum Dis 65:970–971PubMedCentralPubMedCrossRef Harle P, Sarzi-Puttini P, Cutolo M, Straub RH (2006) No change of serum levels of leptin and adiponectin during anti-tumour necrosis factor antibody treatment with adalimumab in patients with rheumatoid arthritis. Ann Rheum Dis 65:970–971PubMedCentralPubMedCrossRef
112.
go back to reference Gonzalez-Gay MA, Garcia-Unzueta MT, Berja A, Gonzalez-Juanatey C, Miranda-Filloy JA, Vazquez-Rodriguez TR, de Matias JM, Martin J, Dessein PH, Llorca J (2009) Anti-TNF-alpha therapy does not modulate leptin in patients with severe rheumatoid arthritis. Clin Exp Rheumatol 27:222–228PubMed Gonzalez-Gay MA, Garcia-Unzueta MT, Berja A, Gonzalez-Juanatey C, Miranda-Filloy JA, Vazquez-Rodriguez TR, de Matias JM, Martin J, Dessein PH, Llorca J (2009) Anti-TNF-alpha therapy does not modulate leptin in patients with severe rheumatoid arthritis. Clin Exp Rheumatol 27:222–228PubMed
113.
go back to reference de Punder YM, Fransen J, Kievit W, Houtman PM, Visser H, van de Laar MA, van Riel PL (2012) The prevalence of clinical remission in RA patients treated with anti-TNF: results from the Dutch Rheumatoid Arthritis Monitoring (DREAM) registry. Rheumatology (Oxford, England) 51:1610–1617CrossRef de Punder YM, Fransen J, Kievit W, Houtman PM, Visser H, van de Laar MA, van Riel PL (2012) The prevalence of clinical remission in RA patients treated with anti-TNF: results from the Dutch Rheumatoid Arthritis Monitoring (DREAM) registry. Rheumatology (Oxford, England) 51:1610–1617CrossRef
114.
go back to reference Schett G, Kiechl S, Bonora E, Redlich K, Woloszczuk W, Oberhollenzer F, Jocher J, Dorizzi R, Muggeo M, Smolen J, Willeit J (2004) Serum leptin level and the risk of nontraumatic fracture. Am J Med 117:952–956PubMedCrossRef Schett G, Kiechl S, Bonora E, Redlich K, Woloszczuk W, Oberhollenzer F, Jocher J, Dorizzi R, Muggeo M, Smolen J, Willeit J (2004) Serum leptin level and the risk of nontraumatic fracture. Am J Med 117:952–956PubMedCrossRef
115.
go back to reference Barbour KE, Zmuda JM, Boudreau R, Strotmeyer ES, Horwitz MJ, Evans RW, Kanaya AM, Harris TB, Bauer DC, Cauley JA (2011) Adipokines and the risk of fracture in older adults. J Bone Miner Res 26:1568–1576PubMedCentralPubMedCrossRef Barbour KE, Zmuda JM, Boudreau R, Strotmeyer ES, Horwitz MJ, Evans RW, Kanaya AM, Harris TB, Bauer DC, Cauley JA (2011) Adipokines and the risk of fracture in older adults. J Bone Miner Res 26:1568–1576PubMedCentralPubMedCrossRef
116.
go back to reference Wang L, Tang X, Zhang H, Yuan J, Ding H, Wei Y (2011) Elevated leptin expression in rat model of traumatic spinal cord injury and femoral fracture. J Spinal Cord Med 34:501–509PubMedCentralPubMedCrossRef Wang L, Tang X, Zhang H, Yuan J, Ding H, Wei Y (2011) Elevated leptin expression in rat model of traumatic spinal cord injury and femoral fracture. J Spinal Cord Med 34:501–509PubMedCentralPubMedCrossRef
117.
go back to reference Wang L, Yuan JS, Zhang HX, Ding H, Tang XG, Wei YZ (2011) Effect of leptin on bone metabolism in rat model of traumatic brain injury and femoral fracture. Chin J Traumatol 14:7–13PubMed Wang L, Yuan JS, Zhang HX, Ding H, Tang XG, Wei YZ (2011) Effect of leptin on bone metabolism in rat model of traumatic brain injury and femoral fracture. Chin J Traumatol 14:7–13PubMed
118.
go back to reference Park MC, Lee SW, Choi ST, Park YB, Lee SK (2007) Serum leptin levels correlate with interleukin-6 levels and disease activity in patients with ankylosing spondylitis. Scand J Rheumatol 36:101–106PubMedCrossRef Park MC, Lee SW, Choi ST, Park YB, Lee SK (2007) Serum leptin levels correlate with interleukin-6 levels and disease activity in patients with ankylosing spondylitis. Scand J Rheumatol 36:101–106PubMedCrossRef
119.
go back to reference Park MC, Chung SJ, Park YB, Lee SK (2009) Pro-inflammatory effect of leptin on peripheral blood mononuclear cells of patients with ankylosing spondylitis. Joint Bone Spine 76:170–175PubMedCrossRef Park MC, Chung SJ, Park YB, Lee SK (2009) Pro-inflammatory effect of leptin on peripheral blood mononuclear cells of patients with ankylosing spondylitis. Joint Bone Spine 76:170–175PubMedCrossRef
120.
go back to reference Sari I, Demir T, Kozaci LD, Akar S, Kavak T, Birlik M, Onen F, Akkoc N (2007) Body composition, insulin, and leptin levels in patients with ankylosing spondylitis. Clin Rheumatol 26:1427–1432PubMedCrossRef Sari I, Demir T, Kozaci LD, Akar S, Kavak T, Birlik M, Onen F, Akkoc N (2007) Body composition, insulin, and leptin levels in patients with ankylosing spondylitis. Clin Rheumatol 26:1427–1432PubMedCrossRef
121.
go back to reference Kim KJ, Kim JY, Park SJ, Yoon H, Yoon CH, Kim WU, Cho CS (2012) Serum leptin levels are associated with the presence of syndesmophytes in male patients with ankylosing spondylitis. Clin Rheumatol 31:1231–1238PubMedCrossRef Kim KJ, Kim JY, Park SJ, Yoon H, Yoon CH, Kim WU, Cho CS (2012) Serum leptin levels are associated with the presence of syndesmophytes in male patients with ankylosing spondylitis. Clin Rheumatol 31:1231–1238PubMedCrossRef
122.
go back to reference Kushlinskii NE, Solov’ev YN, Babkina IV, Abbasova SG, Kostanyan IA, Lipkin VM, Trapeznikov NN (2000) Leptin and apoptosis inhibitor soluble Fas antigen in the serum of patients with osteosarcoma and neuroectodermal bone tumors. Bull Exp Biol Med 129:496–498PubMedCrossRef Kushlinskii NE, Solov’ev YN, Babkina IV, Abbasova SG, Kostanyan IA, Lipkin VM, Trapeznikov NN (2000) Leptin and apoptosis inhibitor soluble Fas antigen in the serum of patients with osteosarcoma and neuroectodermal bone tumors. Bull Exp Biol Med 129:496–498PubMedCrossRef
123.
go back to reference Yang SN, Chen HT, Tsou HK, Huang CY, Yang WH, Su CM, Fong YC, Tseng WP, Tang CH (2009) Leptin enhances cell migration in human chondrosarcoma cells through OBRl leptin receptor. Carcinogenesis 30:566–574PubMedCrossRef Yang SN, Chen HT, Tsou HK, Huang CY, Yang WH, Su CM, Fong YC, Tseng WP, Tang CH (2009) Leptin enhances cell migration in human chondrosarcoma cells through OBRl leptin receptor. Carcinogenesis 30:566–574PubMedCrossRef
124.
go back to reference Wu YP, Chen WS, Xu SJ, Zhang N (2010) Osteoporosis as a potential contributor to the bone metastases. Med Hypotheses 75:514–516PubMedCrossRef Wu YP, Chen WS, Xu SJ, Zhang N (2010) Osteoporosis as a potential contributor to the bone metastases. Med Hypotheses 75:514–516PubMedCrossRef
126.
go back to reference Burwell RG, Dangerfield PH, Moulton A, Anderson SI (2008) Etiologic theories of idiopathic scoliosis: autonomic nervous system and the leptin-sympathetic nervous system concept for the pathogenesis of adolescent idiopathic scoliosis. Stud Health Technol Inform 140:197–207PubMed Burwell RG, Dangerfield PH, Moulton A, Anderson SI (2008) Etiologic theories of idiopathic scoliosis: autonomic nervous system and the leptin-sympathetic nervous system concept for the pathogenesis of adolescent idiopathic scoliosis. Stud Health Technol Inform 140:197–207PubMed
127.
go back to reference Qiu Y, Sun X, Qiu X, Li W, Zhu Z, Zhu F, Wang B, Yu Y, Qian B (2007) Decreased circulating leptin level and its association with body and bone mass in girls with adolescent idiopathic scoliosis. Spine 32:2703–2710PubMedCrossRef Qiu Y, Sun X, Qiu X, Li W, Zhu Z, Zhu F, Wang B, Yu Y, Qian B (2007) Decreased circulating leptin level and its association with body and bone mass in girls with adolescent idiopathic scoliosis. Spine 32:2703–2710PubMedCrossRef
128.
go back to reference Liu Z, Tam EM, Sun GQ, Lam TP, Zhu ZZ, Sun X, Lee KM, Ng TB, Qiu Y, Cheng JC, Yeung HY (2012) Abnormal leptin bioavailability in adolescent idiopathic scoliosis: an important new finding. Spine 37:599–604PubMedCrossRef Liu Z, Tam EM, Sun GQ, Lam TP, Zhu ZZ, Sun X, Lee KM, Ng TB, Qiu Y, Cheng JC, Yeung HY (2012) Abnormal leptin bioavailability in adolescent idiopathic scoliosis: an important new finding. Spine 37:599–604PubMedCrossRef
129.
go back to reference Tsuyama N (1984) Ossification of the posterior longitudinal ligament of the spine. Clin Orthop Relat Res 184:71–84PubMed Tsuyama N (1984) Ossification of the posterior longitudinal ligament of the spine. Clin Orthop Relat Res 184:71–84PubMed
130.
go back to reference Ikeda Y, Nakajima A, Aiba A, Koda M, Okawa A, Takahashi K, Yamazaki M (2011) Association between serum leptin and bone metabolic markers, and the development of heterotopic ossification of the spinal ligament in female patients with ossification of the posterior longitudinal ligament. Eur Spine J 20:1450–1458PubMedCentralPubMedCrossRef Ikeda Y, Nakajima A, Aiba A, Koda M, Okawa A, Takahashi K, Yamazaki M (2011) Association between serum leptin and bone metabolic markers, and the development of heterotopic ossification of the spinal ligament in female patients with ossification of the posterior longitudinal ligament. Eur Spine J 20:1450–1458PubMedCentralPubMedCrossRef
Metadata
Title
Roles of leptin in bone metabolism and bone diseases
Authors
Xu Xu Chen
Tianfu Yang
Publication date
01-09-2015
Publisher
Springer Japan
Published in
Journal of Bone and Mineral Metabolism / Issue 5/2015
Print ISSN: 0914-8779
Electronic ISSN: 1435-5604
DOI
https://doi.org/10.1007/s00774-014-0569-7

Other articles of this Issue 5/2015

Journal of Bone and Mineral Metabolism 5/2015 Go to the issue
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discuss last year's major advances in heart failure and cardiomyopathies.