Skip to main content
Erschienen in: Calcified Tissue International 5/2007

01.05.2007

The New Field of Neuroskeletal Biology

verfasst von: M. S. Patel, F. Elefteriou

Erschienen in: Calcified Tissue International | Ausgabe 5/2007

Einloggen, um Zugang zu erhalten

Abstract

The fields of neuroscience and bone biology have recently converged following the discovery that bone remodeling is directly regulated by the brain. This work has defined bone remodeling as one of the cardinal physiological functions of the body, subject to homeostatic regulation and integrated with the other major physiological functions by the hypothalamus. Central to this discovery was the definition of the adipocyte-derived hormone leptin as a regulator of both arms of bone remodeling, formation and resorption, through its action on the ventromedial hypothalamus and subsequently via the sympathetic nervous system to osteoblasts. The characterization of the sympathetic nervous system as a regulator of bone remodeling has led to several large clinical studies demonstrating a substantial protective effect of ß-blockers, particularly ß1-blockers, on fracture risk. Studies in model organisms have reinforced the role of the central nervous system in the regulation of bone remodeling in vivo by the identification of several additional genes, namely cocaine and amphetamine regulated transcript (Cart), melanocortin 4 receptor (Mc4R), neuropeptide Y (NPY), Y2 receptor, cannabinoid receptor CB1 (Cnbr1), and the genes of the circadian clock. These genes have several common features, including high levels of expression in the hypothalamus and the ability to regulate other major physiological functions in addition to bone remodeling including energy homeostasis, body weight, and reproduction. We review the major pathways that define the new field of neuroskeletal biology and identify further avenues of inquiry.
Literatur
1.
Zurück zum Zitat Bonewald LF (2002) Osteocytes: a proposed multifunctional bone cell. J Musculoskelet Neuronal Interact 2:239–241PubMed Bonewald LF (2002) Osteocytes: a proposed multifunctional bone cell. J Musculoskelet Neuronal Interact 2:239–241PubMed
2.
3.
Zurück zum Zitat Cooper C, Melton LJI (1996) Magnitude and impact of osteoporosis and fractures. In: Marcus R, Feldman D, Kelsey J (eds), Osteoporosis. Academic Press, San Diego, pp 419–434 Cooper C, Melton LJI (1996) Magnitude and impact of osteoporosis and fractures. In: Marcus R, Feldman D, Kelsey J (eds), Osteoporosis. Academic Press, San Diego, pp 419–434
4.
Zurück zum Zitat Perkins R, Skirving AP (1987) Callus formation and the rate of healing of femoral fractures in patients with head injuries. J Bone Joint Surg Br 69:521–524PubMed Perkins R, Skirving AP (1987) Callus formation and the rate of healing of femoral fractures in patients with head injuries. J Bone Joint Surg Br 69:521–524PubMed
5.
Zurück zum Zitat Freehafer AA, Mast WA (1965) Lower extremity fractures in patients with spinal-cord injury. J Bone Joint Surg Am 47:683–694PubMed Freehafer AA, Mast WA (1965) Lower extremity fractures in patients with spinal-cord injury. J Bone Joint Surg Am 47:683–694PubMed
6.
Zurück zum Zitat Aro H (1985) Effect of nerve injury on fracture healing. Callus formation studied in the rat. Acta Orthop Scand 56:233–237PubMedCrossRef Aro H (1985) Effect of nerve injury on fracture healing. Callus formation studied in the rat. Acta Orthop Scand 56:233–237PubMedCrossRef
7.
Zurück zum Zitat Ramnemark A, Nyberg L, Lorentzon R, Englund U, Gustafson Y (1999) Progressive hemiosteoporosis on the paretic side and increased bone mineral density in the nonparetic arm the first year after severe stroke. Osteoporos Int 9:269–275PubMedCrossRef Ramnemark A, Nyberg L, Lorentzon R, Englund U, Gustafson Y (1999) Progressive hemiosteoporosis on the paretic side and increased bone mineral density in the nonparetic arm the first year after severe stroke. Osteoporos Int 9:269–275PubMedCrossRef
8.
Zurück zum Zitat Dauty M, Perrouin Verbe B, Maugars Y, Dubois C, Mathe JF (2000) Supralesional and sublesional bone mineral density in spinal cord-injured patients. Bone 27:305–309PubMedCrossRef Dauty M, Perrouin Verbe B, Maugars Y, Dubois C, Mathe JF (2000) Supralesional and sublesional bone mineral density in spinal cord-injured patients. Bone 27:305–309PubMedCrossRef
9.
Zurück zum Zitat Poole KE, Reeve J, Warburton EA (2002) Falls, fractures, and osteoporosis after stroke: time to think about protection? Stroke 33:1432–1436PubMedCrossRef Poole KE, Reeve J, Warburton EA (2002) Falls, fractures, and osteoporosis after stroke: time to think about protection? Stroke 33:1432–1436PubMedCrossRef
10.
Zurück zum Zitat Pearson J, Dancis J, Axelrod F, Grover N (1975) The sural nerve in familial dysautonomia. J Neuropathol Exp Neurol 34:413–424PubMed Pearson J, Dancis J, Axelrod F, Grover N (1975) The sural nerve in familial dysautonomia. J Neuropathol Exp Neurol 34:413–424PubMed
11.
Zurück zum Zitat Hukkanen M, Konttinen YT, Santavirta S, Paavolainen P, Gu XH, Terenghi G, Polak JM (1993) Rapid proliferation of calcitonin gene-related peptide-immunoreactive nerves during healing of rat tibial fracture suggests neural involvement in bone growth and remodelling. Neuroscience 54:969–979PubMedCrossRef Hukkanen M, Konttinen YT, Santavirta S, Paavolainen P, Gu XH, Terenghi G, Polak JM (1993) Rapid proliferation of calcitonin gene-related peptide-immunoreactive nerves during healing of rat tibial fracture suggests neural involvement in bone growth and remodelling. Neuroscience 54:969–979PubMedCrossRef
12.
Zurück zum Zitat Li J, Ahmad T, Spetea M, Ahmed M, Kreicbergs A (2001) Bone reinnervation after fracture: a study in the rat. J Bone Miner Res 16:1505–1510PubMedCrossRef Li J, Ahmad T, Spetea M, Ahmed M, Kreicbergs A (2001) Bone reinnervation after fracture: a study in the rat. J Bone Miner Res 16:1505–1510PubMedCrossRef
13.
Zurück zum Zitat Ducy P, Amling M, Takeda S, Priemel M, Schilling AF, Beil T, 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 T, 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
14.
Zurück zum Zitat Zigman JM, Elmquist JK (2003) From anorexia to obesity–the yin and yang of body weight control. Endocrinology 144:3749–3756PubMedCrossRef Zigman JM, Elmquist JK (2003) From anorexia to obesity–the yin and yang of body weight control. Endocrinology 144:3749–3756PubMedCrossRef
15.
Zurück zum Zitat Ahima RS, Saper CB, Flier JS, Elmquist JK (2000) Leptin regulation of neuroendocrine systems. Front Neuroendocrinol 21:263–307PubMedCrossRef Ahima RS, Saper CB, Flier JS, Elmquist JK (2000) Leptin regulation of neuroendocrine systems. Front Neuroendocrinol 21:263–307PubMedCrossRef
16.
Zurück zum Zitat Guidobono F, Pagani F, Sibilia V, Netti C, Lattuada N, Rapetti D, Mrak E, Villa I, Cavani F, Bertoni L, Palumbo C, Ferretti M, Marotti G, Rubinacci A (2006) Different skeletal regional response to continuous brain infusion of leptin in the rat. Peptides 27:1426–1433PubMedCrossRef Guidobono F, Pagani F, Sibilia V, Netti C, Lattuada N, Rapetti D, Mrak E, Villa I, Cavani F, Bertoni L, Palumbo C, Ferretti M, Marotti G, Rubinacci A (2006) Different skeletal regional response to continuous brain infusion of leptin in the rat. Peptides 27:1426–1433PubMedCrossRef
17.
Zurück zum Zitat Pogoda P, Egermann M, Schnell JC, Priemel M, Schilling AF, Alini M, Schinke T, Rueger JM, Schneider E, Clarke I, Amling M (2006) Leptin inhibits bone formation not only in rodents, but also in sheep. J Bone Miner Res 21:1591–1599PubMedCrossRef Pogoda P, Egermann M, Schnell JC, Priemel M, Schilling AF, Alini M, Schinke T, Rueger JM, Schneider E, Clarke I, Amling M (2006) Leptin inhibits bone formation not only in rodents, but also in sheep. J Bone Miner Res 21:1591–1599PubMedCrossRef
18.
Zurück zum Zitat Elefteriou F, Takeda S, Ebihara K, Magre J, Patano N, Kim CA, Ogawa Y, Liu X, Ware SM, Craigen WJ, Robert JJ, Vinson C, Nakao K, Capeau J, Karsenty G (2004) Serum leptin level is a regulator of bone mass. Proc Natl Acad Sci USA 101:3258–3263PubMedCrossRef Elefteriou F, Takeda S, Ebihara K, Magre J, Patano N, Kim CA, Ogawa Y, Liu X, Ware SM, Craigen WJ, Robert JJ, Vinson C, Nakao K, Capeau J, Karsenty G (2004) Serum leptin level is a regulator of bone mass. Proc Natl Acad Sci USA 101:3258–3263PubMedCrossRef
19.
Zurück zum Zitat Cock TA, Back J, Elefteriou F, Karsenty G, Kastner P, Chan S, Auwerx J (2004) Enhanced bone formation in lipodystrophic PPARγhyp/hyp mice relocates haematopoiesis to the spleen. EMBO Rep 5:1007–1012PubMedCrossRef Cock TA, Back J, Elefteriou F, Karsenty G, Kastner P, Chan S, Auwerx J (2004) Enhanced bone formation in lipodystrophic PPARγhyp/hyp mice relocates haematopoiesis to the spleen. EMBO Rep 5:1007–1012PubMedCrossRef
20.
Zurück zum Zitat 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
21.
Zurück zum Zitat Dhillon H, Zigman JM, Ye C, Lee CE, McGovern RA, Tang V, Kenny CD, Christiansen LM, White RD, Edelstein EA, Coppari R, Balthasar N, Cowley MA, Chua S Jr, Elmquist JK, Lowell BB (2006) Leptin directly activates SF1 neurons in the VMH, and this action by leptin is required for normal body-weight homeostasis. Neuron 49:191–203PubMedCrossRef Dhillon H, Zigman JM, Ye C, Lee CE, McGovern RA, Tang V, Kenny CD, Christiansen LM, White RD, Edelstein EA, Coppari R, Balthasar N, Cowley MA, Chua S Jr, Elmquist JK, Lowell BB (2006) Leptin directly activates SF1 neurons in the VMH, and this action by leptin is required for normal body-weight homeostasis. Neuron 49:191–203PubMedCrossRef
22.
Zurück zum Zitat Cornish J, Callon KE, Bava U, Lin C, Naot D, Hill BL, Grey AB, Broom N, Myers DE, Nicholson GC, Reid IR (2002) Leptin directly regulates bone cell function in vitro and reduces bone fragility in vivo. J Endocrinol 175:405–415PubMedCrossRef Cornish J, Callon KE, Bava U, Lin C, Naot D, Hill BL, Grey AB, Broom N, Myers DE, Nicholson GC, Reid IR (2002) Leptin directly regulates bone cell function in vitro and reduces bone fragility in vivo. J Endocrinol 175:405–415PubMedCrossRef
23.
Zurück zum Zitat 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
24.
Zurück zum Zitat Steppan CM, Crawford DT, Chidsey-Frink KL, Ke H, Swick AG (2000) Leptin is a potent stimulator of bone growth in ob/ob mice. Regul Pept 92:73–78PubMedCrossRef Steppan CM, Crawford DT, Chidsey-Frink KL, Ke H, Swick AG (2000) Leptin is a potent stimulator of bone growth in ob/ob mice. Regul Pept 92:73–78PubMedCrossRef
25.
Zurück zum Zitat 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 140:1630–1638PubMedCrossRef 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 140:1630–1638PubMedCrossRef
26.
Zurück zum Zitat 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
27.
Zurück zum Zitat Di Monaco M, Vallero F, Di Monaco R, Mautino F, Cavanna A (2003) Fat body mass, leptin and femur bone mineral density in hip-fractured women. J Endocrinol Invest 26:1180–1185PubMed Di Monaco M, Vallero F, Di Monaco R, Mautino F, Cavanna A (2003) Fat body mass, leptin and femur bone mineral density in hip-fractured women. J Endocrinol Invest 26:1180–1185PubMed
28.
Zurück zum Zitat 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
29.
Zurück zum Zitat Weiss LA, Barrett-Connor E, von Muhlen D, Clark P (2006) Leptin predicts BMD and bone resorption in older women but not older men: the Rancho Bernardo Study. J Bone Miner Res 21:758–764PubMedCrossRef Weiss LA, Barrett-Connor E, von Muhlen D, Clark P (2006) Leptin predicts BMD and bone resorption in older women but not older men: the Rancho Bernardo Study. J Bone Miner Res 21:758–764PubMedCrossRef
30.
Zurück zum Zitat Chanprasertyothin S, Piaseu N, Chailurkit L, Rajatanavin R, Ongphiphadhanakul B (2005) Association of circulating leptin with bone mineral density in males and females. J Med Assoc Thai 88:655–659PubMed Chanprasertyothin S, Piaseu N, Chailurkit L, Rajatanavin R, Ongphiphadhanakul B (2005) Association of circulating leptin with bone mineral density in males and females. J Med Assoc Thai 88:655–659PubMed
31.
Zurück zum Zitat Blum M, Harris SS, Must A, Naumova EN, Phillips SM, Rand WM, Dawson-Hughes B (2003) Leptin, body composition and bone mineral density in premenopausal women. Calcif Tissue Int 73:27–32PubMedCrossRef Blum M, Harris SS, Must A, Naumova EN, Phillips SM, Rand WM, Dawson-Hughes B (2003) Leptin, body composition and bone mineral density in premenopausal women. Calcif Tissue Int 73:27–32PubMedCrossRef
32.
Zurück zum Zitat 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
33.
Zurück zum Zitat 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
34.
Zurück zum Zitat 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
35.
Zurück zum Zitat 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
36.
Zurück zum Zitat Garnett SP, Hogler W, Blades B, Baur LA, Peat J, Lee J, Cowell CT (2004) Relation between hormones and body composition, including bone, in prepubertal children. Am J Clin Nutr 80:966–972PubMed Garnett SP, Hogler W, Blades B, Baur LA, Peat J, Lee J, Cowell CT (2004) Relation between hormones and body composition, including bone, in prepubertal children. Am J Clin Nutr 80:966–972PubMed
37.
Zurück zum Zitat Roemmich JN, Clark PA, Mantzoros CS, Gurgol CM, Weltman A, Rogol AD (2003) Relationship of leptin to bone mineralization in children and adolescents. J Clin Endocrinol Metab 88:599–604PubMedCrossRef Roemmich JN, Clark PA, Mantzoros CS, Gurgol CM, Weltman A, Rogol AD (2003) Relationship of leptin to bone mineralization in children and adolescents. J Clin Endocrinol Metab 88:599–604PubMedCrossRef
38.
Zurück zum Zitat Oh KW, Lee WY, Rhee EJ, Baek KH, Yoon KH, Kang MI, Yun EJ, Park CY, Ihm SH, Choi MG, Yoo HJ, Park SW (2005) The relationship between serum resistin, leptin, adiponectin, ghrelin levels and bone mineral density in middle-aged men. Clin Endocrinol (Oxf) 63:131–138CrossRef Oh KW, Lee WY, Rhee EJ, Baek KH, Yoon KH, Kang MI, Yun EJ, Park CY, Ihm SH, Choi MG, Yoo HJ, Park SW (2005) The relationship between serum resistin, leptin, adiponectin, ghrelin levels and bone mineral density in middle-aged men. Clin Endocrinol (Oxf) 63:131–138CrossRef
39.
Zurück zum Zitat Papadopoulou F, Krassas GE, Kalothetou C, Koliakos G, Constantinidis TC (2004) Serum leptin values in relation to bone density and growth hormone-insulin like growth factors axis in healthy men. Arch Androl 50:97–103PubMedCrossRef Papadopoulou F, Krassas GE, Kalothetou C, Koliakos G, Constantinidis TC (2004) Serum leptin values in relation to bone density and growth hormone-insulin like growth factors axis in healthy men. Arch Androl 50:97–103PubMedCrossRef
40.
Zurück zum Zitat Zhong N, Wu XP, Xu ZR, Wang AH, Luo XH, Cao XZ, Xie H, Shan PF, Liao EY (2005) Relationship of serum leptin with age, body weight, body mass index, and bone mineral density in healthy mainland Chinese women. Clin Chim Acta 351:161–168PubMedCrossRef Zhong N, Wu XP, Xu ZR, Wang AH, Luo XH, Cao XZ, Xie H, Shan PF, Liao EY (2005) Relationship of serum leptin with age, body weight, body mass index, and bone mineral density in healthy mainland Chinese women. Clin Chim Acta 351:161–168PubMedCrossRef
41.
Zurück zum Zitat 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 23:87–91PubMed 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 23:87–91PubMed
42.
Zurück zum Zitat Rauch F, Blum WF, Klein K, Allolio B, Schonau E (1998) Does leptin have an effect on bone in adult women? Calcif Tissue Int 63:453–455PubMedCrossRef Rauch F, Blum WF, Klein K, Allolio B, Schonau E (1998) Does leptin have an effect on bone in adult women? Calcif Tissue Int 63:453–455PubMedCrossRef
43.
Zurück zum Zitat 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 (Oxf) 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 (Oxf) 55:341–347CrossRef
44.
Zurück zum Zitat Pasco JA, Henry MJ, Kotowicz MA, Collier GR, Ball MJ, Ugoni AM, Nicholson GC (2001) Serum leptin levels are associated with bone mass in nonobese women. J Clin Endocrinol Metab 86:1884–1887PubMedCrossRef Pasco JA, Henry MJ, Kotowicz MA, Collier GR, Ball MJ, Ugoni AM, Nicholson GC (2001) Serum leptin levels are associated with bone mass in nonobese women. J Clin Endocrinol Metab 86:1884–1887PubMedCrossRef
45.
Zurück zum Zitat Gibson WT, Farooqi IS, Moreau M, DePaoli AM, Lawrence E, O’Rahilly S, Trussell RA (2004) Congenital leptin deficiency due to homozygosity for the Delta133G mutation: report of another case and evaluation of response to four years of leptin therapy. J Clin Endocrinol Metab 89:4821–4826PubMedCrossRef Gibson WT, Farooqi IS, Moreau M, DePaoli AM, Lawrence E, O’Rahilly S, Trussell RA (2004) Congenital leptin deficiency due to homozygosity for the Delta133G mutation: report of another case and evaluation of response to four years of leptin therapy. J Clin Endocrinol Metab 89:4821–4826PubMedCrossRef
46.
Zurück zum Zitat 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
47.
Zurück zum Zitat Thorsell A, Heilig M (2002) Diverse functions of neuropeptide Y revealed using genetically modified animals. Neuropeptides 36:182–193PubMedCrossRef Thorsell A, Heilig M (2002) Diverse functions of neuropeptide Y revealed using genetically modified animals. Neuropeptides 36:182–193PubMedCrossRef
48.
Zurück zum Zitat Bjurholm A, Kreicbergs A, Terenius L, Goldstein M, Schultzberg M (1988) Neuropeptide Y-, tyrosine hydroxylase- and vasoactive intestinal polypeptide-immunoreactive nerves in bone and surrounding tissues. J Auton Nerv Syst 25:119–125PubMedCrossRef Bjurholm A, Kreicbergs A, Terenius L, Goldstein M, Schultzberg M (1988) Neuropeptide Y-, tyrosine hydroxylase- and vasoactive intestinal polypeptide-immunoreactive nerves in bone and surrounding tissues. J Auton Nerv Syst 25:119–125PubMedCrossRef
49.
Zurück zum Zitat Hill EL, Elde R (1991) Distribution of CGRP-, VIP-, D beta H-, SP-, and NPY-immunoreactive nerves in the periosteum of the rat. Cell Tissue Res 264:469–480PubMedCrossRef Hill EL, Elde R (1991) Distribution of CGRP-, VIP-, D beta H-, SP-, and NPY-immunoreactive nerves in the periosteum of the rat. Cell Tissue Res 264:469–480PubMedCrossRef
50.
Zurück zum Zitat Baldock PA, Sainsbury A, Couzens M, Enriquez RF, Thomas GP, Gardiner EM, Herzog H (2002) Hypothalamic Y2 receptors regulate bone formation. J Clin Invest 109:915–921PubMedCrossRef Baldock PA, Sainsbury A, Couzens M, Enriquez RF, Thomas GP, Gardiner EM, Herzog H (2002) Hypothalamic Y2 receptors regulate bone formation. J Clin Invest 109:915–921PubMedCrossRef
51.
Zurück zum Zitat Sainsbury A, Schwarzer C, Couzens M, Herzog H (2002) Y2 receptor deletion attenuates the type 2 diabetic syndrome of ob/ob mice. Diabetes 51:3420–3427PubMedCrossRef Sainsbury A, Schwarzer C, Couzens M, Herzog H (2002) Y2 receptor deletion attenuates the type 2 diabetic syndrome of ob/ob mice. Diabetes 51:3420–3427PubMedCrossRef
52.
Zurück zum Zitat Wilding JP, Gilbey SG, Bailey CJ, Batt RA, Williams G, Ghatei MA, Bloom SR (1993) Increased neuropeptide-Y messenger ribonucleic acid (mRNA) and decreased neurotensin mRNA in the hypothalamus of the obese (ob/ob) mouse. Endocrinology 132:1939–1944PubMedCrossRef Wilding JP, Gilbey SG, Bailey CJ, Batt RA, Williams G, Ghatei MA, Bloom SR (1993) Increased neuropeptide-Y messenger ribonucleic acid (mRNA) and decreased neurotensin mRNA in the hypothalamus of the obese (ob/ob) mouse. Endocrinology 132:1939–1944PubMedCrossRef
53.
Zurück zum Zitat 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
54.
Zurück zum Zitat 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
55.
Zurück zum Zitat Sainsbury A, Baldock PA, Schwarzer C, Ueno N, Enriquez RF, Couzens M, Inui A, Herzog H, Gardiner EM (2003) Synergistic effects of Y2 and Y4 receptors on adiposity and bone mass revealed in double knockout mice. Mol Cell Biol 23:5225–5233PubMedCrossRef Sainsbury A, Baldock PA, Schwarzer C, Ueno N, Enriquez RF, Couzens M, Inui A, Herzog H, Gardiner EM (2003) Synergistic effects of Y2 and Y4 receptors on adiposity and bone mass revealed in double knockout mice. Mol Cell Biol 23:5225–5233PubMedCrossRef
56.
Zurück zum Zitat Baldock PA, Allison S, Sainsbury A, Enriquez RF, Gardiner EM, Herzog H, Eisman JA (2006) Hypothalamic neuropeptide Y exerts a negative effect on cortical bone formation. In: Abstracts of the 28th annual meeting of the American Society for Bone and Mineral Research September 2006. JBMR, vol 21 (suppl 1). Abstract no S65, p 1246 Baldock PA, Allison S, Sainsbury A, Enriquez RF, Gardiner EM, Herzog H, Eisman JA (2006) Hypothalamic neuropeptide Y exerts a negative effect on cortical bone formation. In: Abstracts of the 28th annual meeting of the American Society for Bone and Mineral Research September 2006. JBMR, vol 21 (suppl 1). Abstract no S65, p 1246
57.
Zurück zum Zitat Elefteriou F, Takeda S, Liu X, Armstrong D, Karsenty G (2003) Monosodium glutamate-sensitive hypothalamic neurons contribute to the control of bone mass. Endocrinology 144:3842–3847PubMedCrossRef Elefteriou F, Takeda S, Liu X, Armstrong D, Karsenty G (2003) Monosodium glutamate-sensitive hypothalamic neurons contribute to the control of bone mass. Endocrinology 144:3842–3847PubMedCrossRef
58.
Zurück zum Zitat Lutz B (2002) Molecular biology of cannabinoid receptors. Prostaglandins Leukot Essent Fatty Acids 66:123–142PubMedCrossRef Lutz B (2002) Molecular biology of cannabinoid receptors. Prostaglandins Leukot Essent Fatty Acids 66:123–142PubMedCrossRef
59.
Zurück zum Zitat Idris AI, van’t Hof RJ, Greig IR, Ridge SA, Baker D, Ross RA, Ralston SH (2005) Regulation of bone mass, bone loss and osteoclast activity by cannabinoid receptors. Nat Med 11:774–779PubMedCrossRef Idris AI, van’t Hof RJ, Greig IR, Ridge SA, Baker D, Ross RA, Ralston SH (2005) Regulation of bone mass, bone loss and osteoclast activity by cannabinoid receptors. Nat Med 11:774–779PubMedCrossRef
60.
Zurück zum Zitat Tam J, Ofek O, Fride E, Ledent C, Gabet Y, Muller R, Zimmer A, Mackie K, Mechoulam R, Shohami E, Bab I (2006) Involvement of neuronal cannabinoid receptor CB1 in regulation of bone mass and bone remodeling. Mol Pharmacol 70:786–792PubMedCrossRef Tam J, Ofek O, Fride E, Ledent C, Gabet Y, Muller R, Zimmer A, Mackie K, Mechoulam R, Shohami E, Bab I (2006) Involvement of neuronal cannabinoid receptor CB1 in regulation of bone mass and bone remodeling. Mol Pharmacol 70:786–792PubMedCrossRef
61.
Zurück zum Zitat Ravinet Trillou C, Delgorge C, Menet C, Arnone M, Soubrie P (2004) CB1 cannabinoid receptor knockout in mice leads to leanness, resistance to diet-induced obesity and enhanced leptin sensitivity. Int J Obes Relat Metab Disord 28:640–648PubMedCrossRef Ravinet Trillou C, Delgorge C, Menet C, Arnone M, Soubrie P (2004) CB1 cannabinoid receptor knockout in mice leads to leanness, resistance to diet-induced obesity and enhanced leptin sensitivity. Int J Obes Relat Metab Disord 28:640–648PubMedCrossRef
62.
Zurück zum Zitat Ishac EJ, Jiang L, Lake KD, Varga K, Abood ME, Kunos G (1996) Inhibition of exocytotic noradrenaline release by presynaptic cannabinoid CB1 receptors on peripheral sympathetic nerves. Br J Pharmacol 118:2023–2028PubMed Ishac EJ, Jiang L, Lake KD, Varga K, Abood ME, Kunos G (1996) Inhibition of exocytotic noradrenaline release by presynaptic cannabinoid CB1 receptors on peripheral sympathetic nerves. Br J Pharmacol 118:2023–2028PubMed
63.
Zurück zum Zitat Ofek O, Karsak M, Leclerc N, Fogel M, Frenkel B, Wright K, Tam J, Attar-Namdar M, Kram V, Shohami E, Mechoulam R, Zimmer A, Bab I (2006) Peripheral cannabinoid receptor, CB2, regulates bone mass. Proc Natl Acad Sci USA 103:696–701PubMedCrossRef Ofek O, Karsak M, Leclerc N, Fogel M, Frenkel B, Wright K, Tam J, Attar-Namdar M, Kram V, Shohami E, Mechoulam R, Zimmer A, Bab I (2006) Peripheral cannabinoid receptor, CB2, regulates bone mass. Proc Natl Acad Sci USA 103:696–701PubMedCrossRef
64.
Zurück zum Zitat Tam J, Alexandrovich A, Di Marzo V, Petrosino S, Trembovler V, Zimmer A, Ledent C, Mackie K, Mechoulam R, Shohami E, Bab I (2006) CB1, but not CB2 cannabinoid receptor mediates stimulation of bone formation induced by traumatic brain injury. Abstracts of the 28th Annual Meeting of the American Society for Bone and Mineral Research September 2006. JBMR, vol 21 (suppl 1). Abstract no 1032, p S10 Tam J, Alexandrovich A, Di Marzo V, Petrosino S, Trembovler V, Zimmer A, Ledent C, Mackie K, Mechoulam R, Shohami E, Bab I (2006) CB1, but not CB2 cannabinoid receptor mediates stimulation of bone formation induced by traumatic brain injury. Abstracts of the 28th Annual Meeting of the American Society for Bone and Mineral Research September 2006. JBMR, vol 21 (suppl 1). Abstract no 1032, p S10
65.
Zurück zum Zitat Devoto M, Shimoya K, Caminis J, Ott J, Tenenhouse A, Whyte MP, Sereda L, Hall S, Considine E, Williams CJ, Tromp G, Kuivaniemi H, Ala-Kokko L, Prockop DJ, Spotila LD (1998) First-stage autosomal genome screen in extended pedigrees suggests genes predisposing to low bone mineral density on chromosomes 1p, 2p and 4q. Eur J Hum Genet 6:151–157PubMedCrossRef Devoto M, Shimoya K, Caminis J, Ott J, Tenenhouse A, Whyte MP, Sereda L, Hall S, Considine E, Williams CJ, Tromp G, Kuivaniemi H, Ala-Kokko L, Prockop DJ, Spotila LD (1998) First-stage autosomal genome screen in extended pedigrees suggests genes predisposing to low bone mineral density on chromosomes 1p, 2p and 4q. Eur J Hum Genet 6:151–157PubMedCrossRef
66.
Zurück zum Zitat Karsak M, Cohen-Solal M, Freudenberg J, Ostertag A, Morieux C, Kornak U, Essig J, Erxlebe E, Bab I, Kubisch C, de Vernejoul MC, Zimmer A (2005) Cannabinoid receptor type 2 gene is associated with human osteoporosis. Hum Mol Genet 14:3389–3396PubMedCrossRef Karsak M, Cohen-Solal M, Freudenberg J, Ostertag A, Morieux C, Kornak U, Essig J, Erxlebe E, Bab I, Kubisch C, de Vernejoul MC, Zimmer A (2005) Cannabinoid receptor type 2 gene is associated with human osteoporosis. Hum Mol Genet 14:3389–3396PubMedCrossRef
67.
Zurück zum Zitat Douglass J, McKinzie AA, Couceyro P (1995) PCR differential display identifies a rat brain mRNA that is transcriptionally regulated by cocaine and amphetamine. J Neurosci 15:2471–2481PubMed Douglass J, McKinzie AA, Couceyro P (1995) PCR differential display identifies a rat brain mRNA that is transcriptionally regulated by cocaine and amphetamine. J Neurosci 15:2471–2481PubMed
68.
Zurück zum Zitat 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
69.
Zurück zum Zitat 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
70.
Zurück zum Zitat Ahn JD, Dubern B, Lubrano-Berthelier C, Clement K, Karsenty G (2006) Cart overexpression is the only identifiable cause of high bone mass in melanocortin 4 receptor deficiency. Endocrinology 147:3196–3202PubMedCrossRef Ahn JD, Dubern B, Lubrano-Berthelier C, Clement K, Karsenty G (2006) Cart overexpression is the only identifiable cause of high bone mass in melanocortin 4 receptor deficiency. Endocrinology 147:3196–3202PubMedCrossRef
71.
Zurück zum Zitat Orwoll B, Bouxsein ML, Marks DL, Cone RD, Klein RF (2004) Increased bone mass and strength in melanocortin-4 receptor-deficient mice. In: Orthopaedic Research Society/American Academy of Orthopaedic Surgeons Presentations 2003, 71st Annual Meeting of the AAOS. San Francisco, CA Orwoll B, Bouxsein ML, Marks DL, Cone RD, Klein RF (2004) Increased bone mass and strength in melanocortin-4 receptor-deficient mice. In: Orthopaedic Research Society/American Academy of Orthopaedic Surgeons Presentations 2003, 71st Annual Meeting of the AAOS. San Francisco, CA
72.
Zurück zum Zitat Aguirre J, Buttery L, O’Shaughnessy M, Afzal F, Fernandez de Marticorena I, Hukkanen M, Huang P, MacIntyre I, Polak J (2001) Endothelial nitric oxide synthase gene-deficient mice demonstrate marked retardation in postnatal bone formation, reduced bone volume, and defects in osteoblast maturation and activity. Am J Pathol 158:247–257PubMed Aguirre J, Buttery L, O’Shaughnessy M, Afzal F, Fernandez de Marticorena I, Hukkanen M, Huang P, MacIntyre I, Polak J (2001) Endothelial nitric oxide synthase gene-deficient mice demonstrate marked retardation in postnatal bone formation, reduced bone volume, and defects in osteoblast maturation and activity. Am J Pathol 158:247–257PubMed
73.
Zurück zum Zitat Armour KE, Armour KJ, Gallagher ME, Godecke A, Helfrich MH, Reid DM, Ralston SH (2001) Defective bone formation and anabolic response to exogenous estrogen in mice with targeted disruption of endothelial nitric oxide synthase. Endocrinology 142:760–766PubMedCrossRef Armour KE, Armour KJ, Gallagher ME, Godecke A, Helfrich MH, Reid DM, Ralston SH (2001) Defective bone formation and anabolic response to exogenous estrogen in mice with targeted disruption of endothelial nitric oxide synthase. Endocrinology 142:760–766PubMedCrossRef
74.
Zurück zum Zitat van’t Hof RJ, Ralston SH (1997) Cytokine-induced nitric oxide inhibits bone resorption by inducing apoptosis of osteoclast progenitors and suppressing osteoclast activity. J Bone Miner Res 12:1797–1804PubMedCrossRef van’t Hof RJ, Ralston SH (1997) Cytokine-induced nitric oxide inhibits bone resorption by inducing apoptosis of osteoclast progenitors and suppressing osteoclast activity. J Bone Miner Res 12:1797–1804PubMedCrossRef
75.
Zurück zum Zitat van’t Hof RJ, Armour KJ, Smith LM, Armour KE, Wei XQ, Liew FY, Ralston SH (2000) Requirement of the inducible nitric oxide synthase pathway for IL-1-induced osteoclastic bone resorption. Proc Natl Acad Sci USA 97:7993–7998PubMedCrossRef van’t Hof RJ, Armour KJ, Smith LM, Armour KE, Wei XQ, Liew FY, Ralston SH (2000) Requirement of the inducible nitric oxide synthase pathway for IL-1-induced osteoclastic bone resorption. Proc Natl Acad Sci USA 97:7993–7998PubMedCrossRef
76.
Zurück zum Zitat Lowik CW, Nibbering PH, van de Ruit M, Papapoulos SE (1994) Inducible production of nitric oxide in osteoblast-like cells and in fetal mouse bone explants is associated with suppression of osteoclastic bone resorption. J Clin Invest 93:1465–1472PubMed Lowik CW, Nibbering PH, van de Ruit M, Papapoulos SE (1994) Inducible production of nitric oxide in osteoblast-like cells and in fetal mouse bone explants is associated with suppression of osteoclastic bone resorption. J Clin Invest 93:1465–1472PubMed
77.
Zurück zum Zitat van’t Hof RJ, Macphee J, Libouban H, Helfrich MH, Ralston SH (2004) Regulation of bone mass and bone turnover by neuronal nitric oxide synthase. Endocrinology 145:5068–5074PubMedCrossRef van’t Hof RJ, Macphee J, Libouban H, Helfrich MH, Ralston SH (2004) Regulation of bone mass and bone turnover by neuronal nitric oxide synthase. Endocrinology 145:5068–5074PubMedCrossRef
78.
Zurück zum Zitat Perkins MN, Rothwell NJ, Stock MJ, Stone TW (1981) Activation of brown adipose tissue thermogenesis by the ventromedial hypothalamus. Nature 289:401–402PubMedCrossRef Perkins MN, Rothwell NJ, Stock MJ, Stone TW (1981) Activation of brown adipose tissue thermogenesis by the ventromedial hypothalamus. Nature 289:401–402PubMedCrossRef
79.
Zurück zum Zitat Satoh N, Ogawa Y, Katsuura G, Numata Y, Tsuji T, Hayase M, Ebihara K, Masuzaki H, Hosoda K, Yoshimasa Y, Nakao K (1999) Sympathetic activation of leptin via the ventromedial hypothalamus: leptin-induced increase in catecholamine secretion. Diabetes 48:1787–1793PubMedCrossRef Satoh N, Ogawa Y, Katsuura G, Numata Y, Tsuji T, Hayase M, Ebihara K, Masuzaki H, Hosoda K, Yoshimasa Y, Nakao K (1999) Sympathetic activation of leptin via the ventromedial hypothalamus: leptin-induced increase in catecholamine secretion. Diabetes 48:1787–1793PubMedCrossRef
80.
Zurück zum Zitat Elefteriou F (2005) Neuronal signaling and the regulation of bone remodeling. Cell Mol Life Sci 62:2339–2349PubMedCrossRef Elefteriou F (2005) Neuronal signaling and the regulation of bone remodeling. Cell Mol Life Sci 62:2339–2349PubMedCrossRef
81.
Zurück zum Zitat Bonnet N, Benhamou CL, Brunet-Imbault B, Arlettaz A, Horcajada MN, Richard O, Vico L, Collomp K, Courteix D (2005) Severe bone alterations under beta2 agonist treatments: bone mass, microarchitecture and strength analyses in female rats. Bone 37:622–633PubMedCrossRef Bonnet N, Benhamou CL, Brunet-Imbault B, Arlettaz A, Horcajada MN, Richard O, Vico L, Collomp K, Courteix D (2005) Severe bone alterations under beta2 agonist treatments: bone mass, microarchitecture and strength analyses in female rats. Bone 37:622–633PubMedCrossRef
82.
Zurück zum Zitat Minkowitz B, Boskey AL, Lane JM, Pearlman HS, Vigorita VJ (1991) Effects of propranolol on bone metabolism in the rat. J Orthop Res 9:869–875PubMedCrossRef Minkowitz B, Boskey AL, Lane JM, Pearlman HS, Vigorita VJ (1991) Effects of propranolol on bone metabolism in the rat. J Orthop Res 9:869–875PubMedCrossRef
83.
Zurück zum Zitat Pierroz D, Baldock P, Bouxsein ML, Ferrari S (2006) Low cortical bone mass in mice lacking beta 1 and beta 2 adrenergic receptors is associated with low bone formation and circulating IGF-1. Abstracts of the 28th Annual Meeting of the American Society for Bone and Mineral Research September 2006. JBMR, vol 21 (suppl 1). Abstract no 1091, p S26 Pierroz D, Baldock P, Bouxsein ML, Ferrari S (2006) Low cortical bone mass in mice lacking beta 1 and beta 2 adrenergic receptors is associated with low bone formation and circulating IGF-1. Abstracts of the 28th Annual Meeting of the American Society for Bone and Mineral Research September 2006. JBMR, vol 21 (suppl 1). Abstract no 1091, p S26
84.
Zurück zum Zitat Wiens M, Etminan M, Gill SS, Takkouche B (2006) Effects of antihypertensive drug treatments on fracture outcomes: a meta-analysis of observational studies. J Intern Med 260:350–362PubMedCrossRef Wiens M, Etminan M, Gill SS, Takkouche B (2006) Effects of antihypertensive drug treatments on fracture outcomes: a meta-analysis of observational studies. J Intern Med 260:350–362PubMedCrossRef
85.
Zurück zum Zitat Rejnmark L, Vestergaard P, Mosekilde L (2006) Treatment with beta-blockers, ACE inhibitors, and calcium-channel blockers is associated with a reduced fracture risk: a nationwide case-control study. J Hypertens 24:581–589PubMedCrossRef Rejnmark L, Vestergaard P, Mosekilde L (2006) Treatment with beta-blockers, ACE inhibitors, and calcium-channel blockers is associated with a reduced fracture risk: a nationwide case-control study. J Hypertens 24:581–589PubMedCrossRef
86.
Zurück zum Zitat Schlienger RG, Kraenzlin ME, Jick SS, Meier CR (2004) Use of beta-blockers and risk of fractures. JAMA 292:1326–1332PubMedCrossRef Schlienger RG, Kraenzlin ME, Jick SS, Meier CR (2004) Use of beta-blockers and risk of fractures. JAMA 292:1326–1332PubMedCrossRef
87.
Zurück zum Zitat Levasseur R, Marcelli C, Sabatier JP, Dargent-Molina P, Breart G (2005) Beta-blocker use, bone mineral density, and fracture risk in older women: results from the Epidemiologie de l’Osteoporose prospective study. J Am Geriatr Soc 53:550–552PubMedCrossRef Levasseur R, Marcelli C, Sabatier JP, Dargent-Molina P, Breart G (2005) Beta-blocker use, bone mineral density, and fracture risk in older women: results from the Epidemiologie de l’Osteoporose prospective study. J Am Geriatr Soc 53:550–552PubMedCrossRef
88.
Zurück zum Zitat 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
89.
Zurück zum Zitat Reid IR, Lucas J, Wattie D, Horne A, Bolland M, Gamble GD, Davidson JS, Grey AB (2005) Effects of a beta-blocker on bone turnover in normal postmenopausal women: a randomized controlled trial. J Clin Endocrinol Metab 90:5212–5216PubMedCrossRef Reid IR, Lucas J, Wattie D, Horne A, Bolland M, Gamble GD, Davidson JS, Grey AB (2005) Effects of a beta-blocker on bone turnover in normal postmenopausal women: a randomized controlled trial. J Clin Endocrinol Metab 90:5212–5216PubMedCrossRef
90.
Zurück zum Zitat Denes A, Boldogkoi Z, Uhereczky G, Hornyak A, Rusvai M, Palkovits M, Kovacs KJ (2005) Central autonomic control of the bone marrow: multisynaptic tract tracing by recombinant pseudorabies virus. Neuroscience 134:947–963PubMedCrossRef Denes A, Boldogkoi Z, Uhereczky G, Hornyak A, Rusvai M, Palkovits M, Kovacs KJ (2005) Central autonomic control of the bone marrow: multisynaptic tract tracing by recombinant pseudorabies virus. Neuroscience 134:947–963PubMedCrossRef
91.
Zurück zum Zitat Mauer AM (1965) Diurnal variation of proliferative activity in the human bone marrow. Blood 26:1–7PubMed Mauer AM (1965) Diurnal variation of proliferative activity in the human bone marrow. Blood 26:1–7PubMed
92.
Zurück zum Zitat Simmons DJ, Nichols G Jr (1966) Diurnal periodicity in the metabolic activity of bone tissue. Am J Physiol 210:411–418PubMed Simmons DJ, Nichols G Jr (1966) Diurnal periodicity in the metabolic activity of bone tissue. Am J Physiol 210:411–418PubMed
93.
Zurück zum Zitat Schlemmer A, Hassager C, Jensen SB, Christiansen C (1992) Marked diurnal variation in urinary excretion of pyridinium cross-links in premenopausal women. J Clin Endocrinol Metab 74:476–480PubMedCrossRef Schlemmer A, Hassager C, Jensen SB, Christiansen C (1992) Marked diurnal variation in urinary excretion of pyridinium cross-links in premenopausal women. J Clin Endocrinol Metab 74:476–480PubMedCrossRef
94.
Zurück zum Zitat Srivastava AK, Bhattacharyya S, Li X, Mohan S, Baylink DJ (2001) Circadian and longitudinal variation of serum C-telopeptide, osteocalcin, and skeletal alkaline phosphatase in C3H/HeJ mice. Bone 29:361–367PubMedCrossRef Srivastava AK, Bhattacharyya S, Li X, Mohan S, Baylink DJ (2001) Circadian and longitudinal variation of serum C-telopeptide, osteocalcin, and skeletal alkaline phosphatase in C3H/HeJ mice. Bone 29:361–367PubMedCrossRef
95.
Zurück zum Zitat Ladlow JF, Hoffmann WE, Breur GJ, Richardson DC, Allen MJ (2002) Biological variability in serum and urinary indices of bone formation and resorption in dogs. Calcif Tissue Int 70:186–193PubMedCrossRef Ladlow JF, Hoffmann WE, Breur GJ, Richardson DC, Allen MJ (2002) Biological variability in serum and urinary indices of bone formation and resorption in dogs. Calcif Tissue Int 70:186–193PubMedCrossRef
96.
Zurück zum Zitat 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
97.
Zurück zum Zitat Reppert SM, Weaver DR (2002) Coordination of circadian timing in mammals. Nature 418:935–941PubMedCrossRef Reppert SM, Weaver DR (2002) Coordination of circadian timing in mammals. Nature 418:935–941PubMedCrossRef
98.
Zurück zum Zitat Rosen CJ, Morrison A, Zhou H, Storm D, Hunter SJ, Musgrave K, Chen T, Wei W, Holick MF (1994) Elderly women in northern New England exhibit seasonal changes in bone mineral density and calciotropic hormones. Bone Miner 25:83–92PubMedCrossRef Rosen CJ, Morrison A, Zhou H, Storm D, Hunter SJ, Musgrave K, Chen T, Wei W, Holick MF (1994) Elderly women in northern New England exhibit seasonal changes in bone mineral density and calciotropic hormones. Bone Miner 25:83–92PubMedCrossRef
99.
Zurück zum Zitat Dawson-Hughes B, Dallal GE, Krall EA, Harris S, Sokoll LJ, Falconer G (1991) Effect of vitamin D supplementation on wintertime and overall bone loss in healthy postmenopausal women. Ann Intern Med 115:505–512PubMed Dawson-Hughes B, Dallal GE, Krall EA, Harris S, Sokoll LJ, Falconer G (1991) Effect of vitamin D supplementation on wintertime and overall bone loss in healthy postmenopausal women. Ann Intern Med 115:505–512PubMed
100.
Zurück zum Zitat Ko CH, Takahashi JS (2006) Molecular components of the mammalian circadian clock. Hum Mol Genet 15(special issue 2):R271–R277PubMedCrossRef Ko CH, Takahashi JS (2006) Molecular components of the mammalian circadian clock. Hum Mol Genet 15(special issue 2):R271–R277PubMedCrossRef
101.
Zurück zum Zitat Damiola F, Le Minh N, Preitner N, Kornmann B, Fleury-Olela F, Schibler U (2000) Restricted feeding uncouples circadian oscillators in peripheral tissues from the central pacemaker in the suprachiasmatic nucleus. Genes Dev 14:2950–2961PubMedCrossRef Damiola F, Le Minh N, Preitner N, Kornmann B, Fleury-Olela F, Schibler U (2000) Restricted feeding uncouples circadian oscillators in peripheral tissues from the central pacemaker in the suprachiasmatic nucleus. Genes Dev 14:2950–2961PubMedCrossRef
Metadaten
Titel
The New Field of Neuroskeletal Biology
verfasst von
M. S. Patel
F. Elefteriou
Publikationsdatum
01.05.2007
Verlag
Springer-Verlag
Erschienen in
Calcified Tissue International / Ausgabe 5/2007
Print ISSN: 0171-967X
Elektronische ISSN: 1432-0827
DOI
https://doi.org/10.1007/s00223-007-9015-3

Weitere Artikel der Ausgabe 5/2007

Calcified Tissue International 5/2007 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

GLP-1-Agonisten können Fortschreiten diabetischer Retinopathie begünstigen

24.05.2024 Diabetische Retinopathie Nachrichten

Möglicherweise hängt es von der Art der Diabetesmedikamente ab, wie hoch das Risiko der Betroffenen ist, dass sich sehkraftgefährdende Komplikationen verschlimmern. Das lassen retrospektive Daten vermuten.

Mehr Lebenszeit mit Abemaciclib bei fortgeschrittenem Brustkrebs?

24.05.2024 Mammakarzinom Nachrichten

In der MONARCHE-3-Studie lebten Frauen mit fortgeschrittenem Hormonrezeptor-positivem, HER2-negativem Brustkrebs länger, wenn sie zusätzlich zu einem nicht steroidalen Aromatasehemmer mit Abemaciclib behandelt wurden; allerdings verfehlte der numerische Zugewinn die statistische Signifikanz.

ADT zur Radiatio nach Prostatektomie: Wenn, dann wohl länger

24.05.2024 Prostatakarzinom Nachrichten

Welchen Nutzen es trägt, wenn die Strahlentherapie nach radikaler Prostatektomie um eine Androgendeprivation ergänzt wird, hat die RADICALS-HD-Studie untersucht. Nun liegen die Ergebnisse vor. Sie sprechen für länger dauernden Hormonentzug.

Wie der Klimawandel gefährliche Pilzinfektionen begünstigt

24.05.2024 Candida-Mykosen Nachrichten

Dass sich invasive Pilzinfektionen in letzter Zeit weltweit häufen, liegt wahrscheinlich auch am Klimawandel. Ausbrüche mit dem Hefepilz Candida auris stellen eine zunehmende Gefahr für Immungeschwächte dar – auch in Deutschland.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.