Skip to main content
Erschienen in: Current Osteoporosis Reports 4/2020

09.06.2020 | Muscle and Bone (A Bonetto and M Brotto, Section Editors)

Bone-Muscle Mutual Interactions

verfasst von: Nuria Lara-Castillo, Mark L. Johnson

Erschienen in: Current Osteoporosis Reports | Ausgabe 4/2020

Einloggen, um Zugang zu erhalten

Abstract

Purpose of Review

The purpose of this review is to describe the current state of our thinking regarding bone-muscle interactions beyond the mechanical perspective.

Recent Findings

Recent and prior evidence has begun to dissect many of the molecular mechanisms that bone and muscle use to communicate with each other and to modify each other’s function. Several signaling factors produced by muscle and bone have emerged as potential mediators of these biochemical/molecular interactions. These include muscle factors such as myostatin, Irisin, BAIBA, IL-6, and the IGF family and the bone factors FGF-23, Wnt1 and Wnt3a, PGE2, FGF9, RANKL, osteocalcin, and sclerostin.

Summary

The identification of these signaling molecules and their underlying mechanisms offers the very real and exciting possibility that new pharmaceutical approaches can be developed that will permit the simultaneous treatments of diseases that often occur in combination, such as osteoporosis and sarcopenia.
Literatur
1.
Zurück zum Zitat Gomarasca M, Banfi G, Lombardi G. Myokines: the endocrine coupling of skeletal muscle and bone. Adv Clin Chem. 2020;94:155–218.PubMedCrossRef Gomarasca M, Banfi G, Lombardi G. Myokines: the endocrine coupling of skeletal muscle and bone. Adv Clin Chem. 2020;94:155–218.PubMedCrossRef
2.
Zurück zum Zitat • Trajanoska K, et al. Genetics of bone and muscle interactions in humans. Curr Osteoporos Rep. 2019;17(2):86–95 High Importance: This manuscript describes key pleiotropic loci that have been identified by multivariate GWAS studies.PubMedPubMedCentralCrossRef • Trajanoska K, et al. Genetics of bone and muscle interactions in humans. Curr Osteoporos Rep. 2019;17(2):86–95 High Importance: This manuscript describes key pleiotropic loci that have been identified by multivariate GWAS studies.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Bonewald L. Use it or lose it to age: a review of bone and muscle communication. Bone. 2019;120:212–8.PubMedCrossRef Bonewald L. Use it or lose it to age: a review of bone and muscle communication. Bone. 2019;120:212–8.PubMedCrossRef
4.
Zurück zum Zitat Karsenty G, Mera P. Molecular bases of the crosstalk between bone and muscle. Bone. 2018;115:43–9.PubMedCrossRef Karsenty G, Mera P. Molecular bases of the crosstalk between bone and muscle. Bone. 2018;115:43–9.PubMedCrossRef
5.
Zurück zum Zitat Tagliaferri C, Wittrant Y, Davicco MJ, Walrand S, Coxam V. Muscle and bone, two interconnected tissues. Ageing Res Rev. 2015;21:55–70.PubMedCrossRef Tagliaferri C, Wittrant Y, Davicco MJ, Walrand S, Coxam V. Muscle and bone, two interconnected tissues. Ageing Res Rev. 2015;21:55–70.PubMedCrossRef
7.
Zurück zum Zitat Cianferotti L, Brandi ML. Muscle-bone interactions: basic and clinical aspects. Endocrine. 2014;45(2):165–77.PubMedCrossRef Cianferotti L, Brandi ML. Muscle-bone interactions: basic and clinical aspects. Endocrine. 2014;45(2):165–77.PubMedCrossRef
10.
Zurück zum Zitat Byrd HS, Spicer TE, Cierney G 3rd. Management of open tibial fractures. Plast Reconstr Surg. 1985;76(5):719–30.PubMedCrossRef Byrd HS, Spicer TE, Cierney G 3rd. Management of open tibial fractures. Plast Reconstr Surg. 1985;76(5):719–30.PubMedCrossRef
11.
Zurück zum Zitat Cierny G 3rd, Byrd HS, Jones RE. Primary versus delayed soft tissue coverage for severe open tibial fractures. A comparison of results. Clin Orthop Relat Res. 1983;178:54–63.CrossRef Cierny G 3rd, Byrd HS, Jones RE. Primary versus delayed soft tissue coverage for severe open tibial fractures. A comparison of results. Clin Orthop Relat Res. 1983;178:54–63.CrossRef
12.
Zurück zum Zitat Richards RR, Mahoney JL, Minas T. Influence of soft tissue coverage on the healing of cortical defects in canine diaphyseal bone. Ann Plast Surg. 1986;16(4):296–304.PubMedCrossRef Richards RR, Mahoney JL, Minas T. Influence of soft tissue coverage on the healing of cortical defects in canine diaphyseal bone. Ann Plast Surg. 1986;16(4):296–304.PubMedCrossRef
13.
Zurück zum Zitat Godina M. Early microsurgical reconstruction of complex trauma of the extremities. Plast Reconstr Surg. 1986;78(3):285–92.PubMedCrossRef Godina M. Early microsurgical reconstruction of complex trauma of the extremities. Plast Reconstr Surg. 1986;78(3):285–92.PubMedCrossRef
14.
Zurück zum Zitat Chan JK, et al. Soft-tissue reconstruction of open fractures of the lower limb: muscle versus fasciocutaneous flaps. Plast Reconstr Surg. 2012;130(2):284e–95e.PubMedPubMedCentralCrossRef Chan JK, et al. Soft-tissue reconstruction of open fractures of the lower limb: muscle versus fasciocutaneous flaps. Plast Reconstr Surg. 2012;130(2):284e–95e.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Cosman F, de Beur SJ, LeBoff M, Lewiecki EM, Tanner B, Randall S, et al. Clinician’s guide to prevention and treatment of osteoporosis. Osteoporos Int. 2014;25(10):2359–81.PubMedPubMedCentralCrossRef Cosman F, de Beur SJ, LeBoff M, Lewiecki EM, Tanner B, Randall S, et al. Clinician’s guide to prevention and treatment of osteoporosis. Osteoporos Int. 2014;25(10):2359–81.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Cruz-Jentoft AJ, Bahat G, Bauer J, Boirie Y, Bruyère O, Cederholm T, et al. Sarcopenia: revised European consensus on definition and diagnosis. Age Ageing. 2019;48(4):601.PubMedPubMedCentralCrossRef Cruz-Jentoft AJ, Bahat G, Bauer J, Boirie Y, Bruyère O, Cederholm T, et al. Sarcopenia: revised European consensus on definition and diagnosis. Age Ageing. 2019;48(4):601.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Cruz-Jentoft AJ, Baeyens JP, Bauer JM, Boirie Y, Cederholm T, Landi F, et al. Sarcopenia: European consensus on definition and diagnosis: report of the European Working Group on Sarcopenia in Older People. Age Ageing. 2010;39(4):412–23.PubMedPubMedCentralCrossRef Cruz-Jentoft AJ, Baeyens JP, Bauer JM, Boirie Y, Cederholm T, Landi F, et al. Sarcopenia: European consensus on definition and diagnosis: report of the European Working Group on Sarcopenia in Older People. Age Ageing. 2010;39(4):412–23.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Kull M, Kallikorm R, Lember M. Impact of a new sarco-osteopenia definition on health-related quality of life in a population-based cohort in Northern Europe. J Clin Densitom. 2012;15(1):32–8.PubMedCrossRef Kull M, Kallikorm R, Lember M. Impact of a new sarco-osteopenia definition on health-related quality of life in a population-based cohort in Northern Europe. J Clin Densitom. 2012;15(1):32–8.PubMedCrossRef
19.
Zurück zum Zitat Binkley N, Buehring B. Beyond FRAX: it’s time to consider “sarco-osteopenia”. J Clin Densitom. 2009;12(4):413–6.PubMedCrossRef Binkley N, Buehring B. Beyond FRAX: it’s time to consider “sarco-osteopenia”. J Clin Densitom. 2009;12(4):413–6.PubMedCrossRef
20.
Zurück zum Zitat Sepulveda-Loyola W, et al. The joint occurrence of osteoporosis and sarcopenia (osteosarcopenia): definitions and characteristics. J Am Med Dir Assoc. 2020;21(2):220–5.PubMedCrossRef Sepulveda-Loyola W, et al. The joint occurrence of osteoporosis and sarcopenia (osteosarcopenia): definitions and characteristics. J Am Med Dir Assoc. 2020;21(2):220–5.PubMedCrossRef
21.
Zurück zum Zitat Kirk B, Al Saedi A, Duque G. Osteosarcopenia: a case of geroscience. Aging Med (Milton). 2019;2(3):147–56.CrossRef Kirk B, Al Saedi A, Duque G. Osteosarcopenia: a case of geroscience. Aging Med (Milton). 2019;2(3):147–56.CrossRef
22.
Zurück zum Zitat Fatima M, Brennan-Olsen SL, Duque G. Therapeutic approaches to osteosarcopenia: insights for the clinician. Ther Adv Musculoskelet Dis. 2019;11:1759720x19867009.PubMedPubMedCentralCrossRef Fatima M, Brennan-Olsen SL, Duque G. Therapeutic approaches to osteosarcopenia: insights for the clinician. Ther Adv Musculoskelet Dis. 2019;11:1759720x19867009.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Huo YR, Suriyaarachchi P, Gomez F, Curcio CL, Boersma D, Muir SW, et al. Phenotype of osteosarcopenia in older individuals with a history of falling. J Am Med Dir Assoc. 2015;16(4):290–5.PubMedCrossRef Huo YR, Suriyaarachchi P, Gomez F, Curcio CL, Boersma D, Muir SW, et al. Phenotype of osteosarcopenia in older individuals with a history of falling. J Am Med Dir Assoc. 2015;16(4):290–5.PubMedCrossRef
24.
Zurück zum Zitat Ho-Pham LT, Nguyen UD, Nguyen TV. Association between lean mass, fat mass, and bone mineral density: a meta-analysis. J Clin Endocrinol Metab. 2014;99(1):30–8.PubMedCrossRef Ho-Pham LT, Nguyen UD, Nguyen TV. Association between lean mass, fat mass, and bone mineral density: a meta-analysis. J Clin Endocrinol Metab. 2014;99(1):30–8.PubMedCrossRef
25.
Zurück zum Zitat Huh JH, Song MK, Park KH, Kim KJ, Kim JE, Rhee YM, et al. Gender-specific pleiotropic bone-muscle relationship in the elderly from a nationwide survey (KNHANES IV). Osteoporos Int. 2014;25(3):1053–61.PubMedCrossRef Huh JH, Song MK, Park KH, Kim KJ, Kim JE, Rhee YM, et al. Gender-specific pleiotropic bone-muscle relationship in the elderly from a nationwide survey (KNHANES IV). Osteoporos Int. 2014;25(3):1053–61.PubMedCrossRef
26•.
Zurück zum Zitat . Luo Y, Jiang K, He M. Association between grip strength and bone mineral density in general US population of NHANES 2013–2014. Arch Osteoporos. 2020;15(1):47 High Importance: This manuscript describes a US population study of hand grip strength and bone mineral density of the femoral neck and total lumbar spine. They found that grip strength can be associated with nonadjacent bones and grip strength of the dominant arm was highly correlated with BMD.PubMedCrossRef . Luo Y, Jiang K, He M. Association between grip strength and bone mineral density in general US population of NHANES 2013–2014. Arch Osteoporos. 2020;15(1):47 High Importance: This manuscript describes a US population study of hand grip strength and bone mineral density of the femoral neck and total lumbar spine. They found that grip strength can be associated with nonadjacent bones and grip strength of the dominant arm was highly correlated with BMD.PubMedCrossRef
27.
Zurück zum Zitat Locquet M, Beaudart C, Durieux N, Reginster JY, Bruyère O. Relationship between the changes over time of bone mass and muscle health in children and adults: a systematic review and meta-analysis. BMC Musculoskelet Disord. 2019;20(1):429.PubMedPubMedCentralCrossRef Locquet M, Beaudart C, Durieux N, Reginster JY, Bruyère O. Relationship between the changes over time of bone mass and muscle health in children and adults: a systematic review and meta-analysis. BMC Musculoskelet Disord. 2019;20(1):429.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Huang J, Hsu YH, Mo C, Abreu E, Kiel DP, Bonewald LF, et al. METTL21C is a potential pleiotropic gene for osteoporosis and sarcopenia acting through the modulation of the NF-kappaB signaling pathway. J Bone Miner Res. 2014;29(7):1531–40.PubMedCrossRef Huang J, Hsu YH, Mo C, Abreu E, Kiel DP, Bonewald LF, et al. METTL21C is a potential pleiotropic gene for osteoporosis and sarcopenia acting through the modulation of the NF-kappaB signaling pathway. J Bone Miner Res. 2014;29(7):1531–40.PubMedCrossRef
30.
Zurück zum Zitat Medina-Gomez C, Kemp JP, Dimou NL, Kreiner E, Chesi A, Zemel BS, et al. Bivariate genome-wide association meta-analysis of pediatric musculoskeletal traits reveals pleiotropic effects at the SREBF1/TOM1L2 locus. Nat Commun. 2017;8(1):121.PubMedPubMedCentralCrossRef Medina-Gomez C, Kemp JP, Dimou NL, Kreiner E, Chesi A, Zemel BS, et al. Bivariate genome-wide association meta-analysis of pediatric musculoskeletal traits reveals pleiotropic effects at the SREBF1/TOM1L2 locus. Nat Commun. 2017;8(1):121.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Kurosu H, Ogawa Y, Miyoshi M, Yamamoto M, Nandi A, Rosenblatt KP, et al. Regulation of fibroblast growth factor-23 signaling by klotho. J Biol Chem. 2006;281(10):6120–3.PubMedCrossRef Kurosu H, Ogawa Y, Miyoshi M, Yamamoto M, Nandi A, Rosenblatt KP, et al. Regulation of fibroblast growth factor-23 signaling by klotho. J Biol Chem. 2006;281(10):6120–3.PubMedCrossRef
32.
Zurück zum Zitat Gorski JP, Huffman NT, Vallejo J, Brotto L, Chittur SV, Breggia A, et al. Deletion of Mbtps1 (Pcsk8, S1p, Ski-1) gene in osteocytes stimulates soleus muscle regeneration and increased size and contractile force with age. J Biol Chem. 2016;291(9):4308–22.PubMedCrossRef Gorski JP, Huffman NT, Vallejo J, Brotto L, Chittur SV, Breggia A, et al. Deletion of Mbtps1 (Pcsk8, S1p, Ski-1) gene in osteocytes stimulates soleus muscle regeneration and increased size and contractile force with age. J Biol Chem. 2016;291(9):4308–22.PubMedCrossRef
33.
Zurück zum Zitat Gorski JP, Price JL. Bone muscle crosstalk targets muscle regeneration pathway regulated by core circadian transcriptional repressors DEC1 and DEC2. Bonekey Rep. 2016;5:850.PubMedPubMedCentralCrossRef Gorski JP, Price JL. Bone muscle crosstalk targets muscle regeneration pathway regulated by core circadian transcriptional repressors DEC1 and DEC2. Bonekey Rep. 2016;5:850.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Samsa WE, Vasanji A, Midura RJ, Kondratov RV. Deficiency of circadian clock protein BMAL1 in mice results in a low bone mass phenotype. Bone. 2016;84:194–203.PubMedPubMedCentralCrossRef Samsa WE, Vasanji A, Midura RJ, Kondratov RV. Deficiency of circadian clock protein BMAL1 in mice results in a low bone mass phenotype. Bone. 2016;84:194–203.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Fu L, Patel MS, Bradley A, Wagner EF, Karsenty G. The molecular clock mediates leptin-regulated bone formation. Cell. 2005;122(5):803–15.PubMedCrossRef Fu L, Patel MS, Bradley A, Wagner EF, Karsenty G. The molecular clock mediates leptin-regulated bone formation. Cell. 2005;122(5):803–15.PubMedCrossRef
36.
Zurück zum Zitat Andrews JL, Zhang X, McCarthy JJ, McDearmon EL, Hornberger TA, Russell B, et al. CLOCK and BMAL1 regulate MyoD and are necessary for maintenance of skeletal muscle phenotype and function. Proc Natl Acad Sci U S A. 2010;107(44):19090–5.PubMedPubMedCentralCrossRef Andrews JL, Zhang X, McCarthy JJ, McDearmon EL, Hornberger TA, Russell B, et al. CLOCK and BMAL1 regulate MyoD and are necessary for maintenance of skeletal muscle phenotype and function. Proc Natl Acad Sci U S A. 2010;107(44):19090–5.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Riley LA, Esser KA. The role of the molecular clock in skeletal muscle and what it is teaching us about muscle-bone crosstalk. Curr Osteoporos Rep. 2017;15(3):222–30.PubMedPubMedCentralCrossRef Riley LA, Esser KA. The role of the molecular clock in skeletal muscle and what it is teaching us about muscle-bone crosstalk. Curr Osteoporos Rep. 2017;15(3):222–30.PubMedPubMedCentralCrossRef
38•.
Zurück zum Zitat . Qin W, Dallas SL. Exosomes and extracellular RNA in muscle and bone aging and crosstalk. Curr Osteoporos Rep. 2019; High Importance: This review discusses the role of extracellular vesicles in bone-muscle crosstalk. . Qin W, Dallas SL. Exosomes and extracellular RNA in muscle and bone aging and crosstalk. Curr Osteoporos Rep. 2019; High Importance: This review discusses the role of extracellular vesicles in bone-muscle crosstalk.
39.
Zurück zum Zitat Ge M, Ke R, Cai T, Yang J, Mu X. Identification and proteomic analysis of osteoblast-derived exosomes. Biochem Biophys Res Commun. 2015;467(1):27–32.PubMedCrossRef Ge M, Ke R, Cai T, Yang J, Mu X. Identification and proteomic analysis of osteoblast-derived exosomes. Biochem Biophys Res Commun. 2015;467(1):27–32.PubMedCrossRef
40.
Zurück zum Zitat Cui Y, Luan J, Li H, Zhou X, Han J. Exosomes derived from mineralizing osteoblasts promote ST2 cell osteogenic differentiation by alteration of microRNA expression. FEBS Lett. 2016;590(1):185–92.PubMedCrossRef Cui Y, Luan J, Li H, Zhou X, Han J. Exosomes derived from mineralizing osteoblasts promote ST2 cell osteogenic differentiation by alteration of microRNA expression. FEBS Lett. 2016;590(1):185–92.PubMedCrossRef
41.
Zurück zum Zitat Deng L, Wang Y, Peng Y, Wu Y, Ding Y, Jiang Y, et al. Osteoblast-derived microvesicles: a novel mechanism for communication between osteoblasts and osteoclasts. Bone. 2015;79:37–42.PubMedCrossRef Deng L, Wang Y, Peng Y, Wu Y, Ding Y, Jiang Y, et al. Osteoblast-derived microvesicles: a novel mechanism for communication between osteoblasts and osteoclasts. Bone. 2015;79:37–42.PubMedCrossRef
42.
Zurück zum Zitat Huynh N, VonMoss L, Smith D, Rahman I, Felemban MF, Zuo J, et al. Characterization of regulatory extracellular vesicles from osteoclasts. J Dent Res. 2016;95(6):673–9.PubMedPubMedCentralCrossRef Huynh N, VonMoss L, Smith D, Rahman I, Felemban MF, Zuo J, et al. Characterization of regulatory extracellular vesicles from osteoclasts. J Dent Res. 2016;95(6):673–9.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Li D, Liu J, Guo B, Liang C, Dang L, Lu C, et al. Osteoclast-derived exosomal miR-214-3p inhibits osteoblastic bone formation. Nat Commun. 2016;7(1):10872.PubMedPubMedCentralCrossRef Li D, Liu J, Guo B, Liang C, Dang L, Lu C, et al. Osteoclast-derived exosomal miR-214-3p inhibits osteoblastic bone formation. Nat Commun. 2016;7(1):10872.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Sun W, Zhao C, Li Y, Wang L, Nie G, Peng J, et al. Osteoclast-derived microRNA-containing exosomes selectively inhibit osteoblast activity. Cell Discov. 2016;2:16015.PubMedPubMedCentralCrossRef Sun W, Zhao C, Li Y, Wang L, Nie G, Peng J, et al. Osteoclast-derived microRNA-containing exosomes selectively inhibit osteoblast activity. Cell Discov. 2016;2:16015.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Qin Y, Peng Y, Zhao W, Pan J, Ksiezak-Reding H, Cardozo C, et al. Myostatin inhibits osteoblastic differentiation by suppressing osteocyte-derived exosomal microRNA-218: a novel mechanism in muscle-bone communication. J Biol Chem. 2017;292(26):11021–33.PubMedPubMedCentralCrossRef Qin Y, Peng Y, Zhao W, Pan J, Ksiezak-Reding H, Cardozo C, et al. Myostatin inhibits osteoblastic differentiation by suppressing osteocyte-derived exosomal microRNA-218: a novel mechanism in muscle-bone communication. J Biol Chem. 2017;292(26):11021–33.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Veno PPM, Dusevich V, Bonewald L, Dallas S. Osteocytes release microvesicles that regulate osteoblast function. J Bone Miner Res. 2013;28(Suppl 1):S253. Veno PPM, Dusevich V, Bonewald L, Dallas S. Osteocytes release microvesicles that regulate osteoblast function. J Bone Miner Res. 2013;28(Suppl 1):S253.
47.
Zurück zum Zitat Sato M, Suzuki T, Kawano M, Tamura M. Circulating osteocyte-derived exosomes contain miRNAs which are enriched in exosomes from MLO-Y4 cells. Biomed Rep. 2017;6(2):223–31.PubMedCrossRef Sato M, Suzuki T, Kawano M, Tamura M. Circulating osteocyte-derived exosomes contain miRNAs which are enriched in exosomes from MLO-Y4 cells. Biomed Rep. 2017;6(2):223–31.PubMedCrossRef
48.
Zurück zum Zitat Lai X, Price C, Lu X(L), Wang L. Imaging and quantifying solute transport across periosteum: implications for muscle-bone crosstalk. Bone. 2014;66:82–9.PubMedPubMedCentralCrossRef Lai X, Price C, Lu X(L), Wang L. Imaging and quantifying solute transport across periosteum: implications for muscle-bone crosstalk. Bone. 2014;66:82–9.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Pedersen BK, Febbraio M. Muscle-derived interleukin-6--a possible link between skeletal muscle, adipose tissue, liver, and brain. Brain Behav Immun. 2005;19(5):371–6.PubMedCrossRef Pedersen BK, Febbraio M. Muscle-derived interleukin-6--a possible link between skeletal muscle, adipose tissue, liver, and brain. Brain Behav Immun. 2005;19(5):371–6.PubMedCrossRef
50.
Zurück zum Zitat Pedersen BK, et al. Role of myokines in exercise and metabolism. J Appl Physiol (1985). 2007;103(3):1093–8.CrossRef Pedersen BK, et al. Role of myokines in exercise and metabolism. J Appl Physiol (1985). 2007;103(3):1093–8.CrossRef
51.
Zurück zum Zitat Pedersen BK, Febbraio MA. Muscle as an endocrine organ: focus on muscle-derived interleukin-6. Physiol Rev. 2008;88(4):1379–406.PubMedCrossRef Pedersen BK, Febbraio MA. Muscle as an endocrine organ: focus on muscle-derived interleukin-6. Physiol Rev. 2008;88(4):1379–406.PubMedCrossRef
52.
Zurück zum Zitat McPherron AC, Lawler AM, Lee SJ. Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member. Nature. 1997;387(6628):83–90.PubMedCrossRef McPherron AC, Lawler AM, Lee SJ. Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member. Nature. 1997;387(6628):83–90.PubMedCrossRef
53.
Zurück zum Zitat Hamrick MW, McPherron AC, Lovejoy CO. Bone mineral content and density in the humerus of adult myostatin-deficient mice. Calcif Tissue Int. 2002;71(1):63–8.PubMedCrossRef Hamrick MW, McPherron AC, Lovejoy CO. Bone mineral content and density in the humerus of adult myostatin-deficient mice. Calcif Tissue Int. 2002;71(1):63–8.PubMedCrossRef
54.
Zurück zum Zitat Hamrick MW, Samaddar T, Pennington C, McCormick J. Increased muscle mass with myostatin deficiency improves gains in bone strength with exercise. J Bone Miner Res. 2006;21(3):477–83.PubMedCrossRef Hamrick MW, Samaddar T, Pennington C, McCormick J. Increased muscle mass with myostatin deficiency improves gains in bone strength with exercise. J Bone Miner Res. 2006;21(3):477–83.PubMedCrossRef
55.
Zurück zum Zitat Grobet L, Royo Martin LJ, Poncelet D, Pirottin D, Brouwers B, Riquet J, et al. A deletion in the bovine myostatin gene causes the double-muscled phenotype in cattle. Nat Genet. 1997;17(1):71–4.PubMedCrossRef Grobet L, Royo Martin LJ, Poncelet D, Pirottin D, Brouwers B, Riquet J, et al. A deletion in the bovine myostatin gene causes the double-muscled phenotype in cattle. Nat Genet. 1997;17(1):71–4.PubMedCrossRef
56.
Zurück zum Zitat Kambadur R, Sharma M, Smith TPL, Bass JJ. Mutations in myostatin (GDF8) in double-muscled Belgian Blue and Piedmontese cattle. Genome Res. 1997;7(9):910–6.PubMedCrossRef Kambadur R, Sharma M, Smith TPL, Bass JJ. Mutations in myostatin (GDF8) in double-muscled Belgian Blue and Piedmontese cattle. Genome Res. 1997;7(9):910–6.PubMedCrossRef
57.
Zurück zum Zitat Smith TP, et al. Myostatin maps to the interval containing the bovine mh locus. Mamm Genome. 1997;8(10):742–4.PubMedCrossRef Smith TP, et al. Myostatin maps to the interval containing the bovine mh locus. Mamm Genome. 1997;8(10):742–4.PubMedCrossRef
58.
Zurück zum Zitat Schuelke M, Wagner KR, Stolz LE, Hübner C, Riebel T, Kömen W, et al. Myostatin mutation associated with gross muscle hypertrophy in a child. N Engl J Med. 2004;350(26):2682–8.PubMedCrossRef Schuelke M, Wagner KR, Stolz LE, Hübner C, Riebel T, Kömen W, et al. Myostatin mutation associated with gross muscle hypertrophy in a child. N Engl J Med. 2004;350(26):2682–8.PubMedCrossRef
59.
Zurück zum Zitat Hamrick MW, Shi X, Zhang W, Pennington C, Thakore H, Haque M, et al. Loss of myostatin (GDF8) function increases osteogenic differentiation of bone marrow-derived mesenchymal stem cells but the osteogenic effect is ablated with unloading. Bone. 2007;40(6):1544–53.PubMedPubMedCentralCrossRef Hamrick MW, Shi X, Zhang W, Pennington C, Thakore H, Haque M, et al. Loss of myostatin (GDF8) function increases osteogenic differentiation of bone marrow-derived mesenchymal stem cells but the osteogenic effect is ablated with unloading. Bone. 2007;40(6):1544–53.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Dankbar B, Fennen M, Brunert D, Hayer S, Frank S, Wehmeyer C, et al. Myostatin is a direct regulator of osteoclast differentiation and its inhibition reduces inflammatory joint destruction in mice. Nat Med. 2015;21(9):1085–90.PubMedCrossRef Dankbar B, Fennen M, Brunert D, Hayer S, Frank S, Wehmeyer C, et al. Myostatin is a direct regulator of osteoclast differentiation and its inhibition reduces inflammatory joint destruction in mice. Nat Med. 2015;21(9):1085–90.PubMedCrossRef
61.
Zurück zum Zitat Chen Y-S, Guo Q, Guo LJ, Liu T, Wu XP, Lin ZY, et al. GDF8 inhibits bone formation and promotes bone resorption in mice. Clin Exp Pharmacol Physiol. 2017;44(4):500–8.PubMedCrossRef Chen Y-S, Guo Q, Guo LJ, Liu T, Wu XP, Lin ZY, et al. GDF8 inhibits bone formation and promotes bone resorption in mice. Clin Exp Pharmacol Physiol. 2017;44(4):500–8.PubMedCrossRef
62.
Zurück zum Zitat Campbell C, McMillan HJ, Mah JK, Tarnopolsky M, Selby K, McClure T, et al. Myostatin inhibitor ACE-031 treatment of ambulatory boys with Duchenne muscular dystrophy: results of a randomized, placebo-controlled clinical trial. Muscle Nerve. 2017;55(4):458–64.PubMedCrossRef Campbell C, McMillan HJ, Mah JK, Tarnopolsky M, Selby K, McClure T, et al. Myostatin inhibitor ACE-031 treatment of ambulatory boys with Duchenne muscular dystrophy: results of a randomized, placebo-controlled clinical trial. Muscle Nerve. 2017;55(4):458–64.PubMedCrossRef
63.
Zurück zum Zitat Long KK, O’Shea KM, Khairallah RJ, Howell K, Paushkin S, Chen KS, et al. Specific inhibition of myostatin activation is beneficial in mouse models of SMA therapy. Hum Mol Genet. 2019;28(7):1076–89.PubMedCrossRef Long KK, O’Shea KM, Khairallah RJ, Howell K, Paushkin S, Chen KS, et al. Specific inhibition of myostatin activation is beneficial in mouse models of SMA therapy. Hum Mol Genet. 2019;28(7):1076–89.PubMedCrossRef
64.
Zurück zum Zitat Boström P, Wu J, Jedrychowski MP, Korde A, Ye L, Lo JC, et al. A PGC1-α-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature. 2012;481(7382):463–8.PubMedPubMedCentralCrossRef Boström P, Wu J, Jedrychowski MP, Korde A, Ye L, Lo JC, et al. A PGC1-α-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature. 2012;481(7382):463–8.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Colaianni G, et al. Irisin enhances osteoblast differentiation in vitro. Int J Endocrinol. 2014;2014:–902186. Colaianni G, et al. Irisin enhances osteoblast differentiation in vitro. Int J Endocrinol. 2014;2014:–902186.
67.
Zurück zum Zitat Thomas T, Burguera B. Is leptin the link between fat and bone mass? J Bone Miner Res. 2002;17(9):1563–9.PubMedCrossRef Thomas T, Burguera B. Is leptin the link between fat and bone mass? J Bone Miner Res. 2002;17(9):1563–9.PubMedCrossRef
68.
Zurück zum Zitat Gimble JM, Nuttall ME. Bone and fat: old questions, new insights. Endocrine. 2004;23(2–3):183–8.PubMedCrossRef Gimble JM, Nuttall ME. Bone and fat: old questions, new insights. Endocrine. 2004;23(2–3):183–8.PubMedCrossRef
69.
Zurück zum Zitat Confavreux CB, Levine RL, Karsenty G. A paradigm of integrative physiology, the crosstalk between bone and energy metabolisms. Mol Cell Endocrinol. 2009;310(1–2):21–9.PubMedPubMedCentralCrossRef Confavreux CB, Levine RL, Karsenty G. A paradigm of integrative physiology, the crosstalk between bone and energy metabolisms. Mol Cell Endocrinol. 2009;310(1–2):21–9.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Colaianni G, Cuscito C, Mongelli T, Pignataro P, Buccoliero C, Liu P, et al. The myokine irisin increases cortical bone mass. Proc Natl Acad Sci U S A. 2015;112(39):12157–62.PubMedPubMedCentralCrossRef Colaianni G, Cuscito C, Mongelli T, Pignataro P, Buccoliero C, Liu P, et al. The myokine irisin increases cortical bone mass. Proc Natl Acad Sci U S A. 2015;112(39):12157–62.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Qiao X, Nie Y, Ma Y, Chen Y, Cheng R, Yin W, et al. Irisin promotes osteoblast proliferation and differentiation via activating the MAP kinase signaling pathways. Sci Rep. 2016;6:18732.PubMedCrossRef Qiao X, Nie Y, Ma Y, Chen Y, Cheng R, Yin W, et al. Irisin promotes osteoblast proliferation and differentiation via activating the MAP kinase signaling pathways. Sci Rep. 2016;6:18732.PubMedCrossRef
72.
Zurück zum Zitat Zhang J, Valverde P, Zhu X, Murray D, Wu Y, Yu L, et al. Exercise-induced irisin in bone and systemic irisin administration reveal new regulatory mechanisms of bone metabolism. Bone research. 2017;5:–16056. Zhang J, Valverde P, Zhu X, Murray D, Wu Y, Yu L, et al. Exercise-induced irisin in bone and systemic irisin administration reveal new regulatory mechanisms of bone metabolism. Bone research. 2017;5:–16056.
73.
Zurück zum Zitat Colaianni G, Mongelli T, Cuscito C, Pignataro P, Lippo L, Spiro G, et al. Irisin prevents and restores bone loss and muscle atrophy in hind-limb suspended mice. Sci Rep. 2017;7(1):2811.PubMedPubMedCentralCrossRef Colaianni G, Mongelli T, Cuscito C, Pignataro P, Lippo L, Spiro G, et al. Irisin prevents and restores bone loss and muscle atrophy in hind-limb suspended mice. Sci Rep. 2017;7(1):2811.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Kim H, et al. Irisin mediates effects on bone and fat via αV integrin receptors, 1756. Cell. 2018;175(7):–1768.e17. Kim H, et al. Irisin mediates effects on bone and fat via αV integrin receptors, 1756. Cell. 2018;175(7):–1768.e17.
75.
76.
Zurück zum Zitat Maisonneuve C, Igoudjil A, Begriche K, Lettéron P, Guimont MC, Bastin J, et al. Effects of zidovudine, stavudine and beta-aminoisobutyric acid on lipid homeostasis in mice: possible role in human fat wasting. Antivir Ther. 2004;9(5):801–10.PubMedCrossRef Maisonneuve C, Igoudjil A, Begriche K, Lettéron P, Guimont MC, Bastin J, et al. Effects of zidovudine, stavudine and beta-aminoisobutyric acid on lipid homeostasis in mice: possible role in human fat wasting. Antivir Ther. 2004;9(5):801–10.PubMedCrossRef
77.
Zurück zum Zitat Note R, Maisonneuve C, Lettéron P, Peytavin G, Djouadi F, Igoudjil A, et al. Mitochondrial and metabolic effects of nucleoside reverse transcriptase inhibitors (NRTIs) in mice receiving one of five single- and three dual-NRTI treatments. Antimicrob Agents Chemother. 2003;47(11):3384–92.PubMedPubMedCentralCrossRef Note R, Maisonneuve C, Lettéron P, Peytavin G, Djouadi F, Igoudjil A, et al. Mitochondrial and metabolic effects of nucleoside reverse transcriptase inhibitors (NRTIs) in mice receiving one of five single- and three dual-NRTI treatments. Antimicrob Agents Chemother. 2003;47(11):3384–92.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Igoudjil A, Abbey-Toby A, Begriche K, Grodet A, Chataigner K, Peytavin G, et al. High doses of stavudine induce fat wasting and mild liver damage without impairing mitochondrial respiration in mice. Antivir Ther. 2007;12(3):389–400.PubMedCrossRef Igoudjil A, Abbey-Toby A, Begriche K, Grodet A, Chataigner K, Peytavin G, et al. High doses of stavudine induce fat wasting and mild liver damage without impairing mitochondrial respiration in mice. Antivir Ther. 2007;12(3):389–400.PubMedCrossRef
79.
Zurück zum Zitat Begriche K, et al. Beta-aminoisobutyric acid prevents diet-induced obesity in mice with partial leptin deficiency. Obesity (Silver Spring, Md). 2008, 2053-2067;(16):9. Begriche K, et al. Beta-aminoisobutyric acid prevents diet-induced obesity in mice with partial leptin deficiency. Obesity (Silver Spring, Md). 2008, 2053-2067;(16):9.
80.
Zurück zum Zitat Begriche K, Massart J, Fromenty B. Effects of β-aminoisobutyric acid on leptin production and lipid homeostasis: mechanisms and possible relevance for the prevention of obesity. Fundam Clin Pharmacol. 2010;24(3):269–82.PubMedCrossRef Begriche K, Massart J, Fromenty B. Effects of β-aminoisobutyric acid on leptin production and lipid homeostasis: mechanisms and possible relevance for the prevention of obesity. Fundam Clin Pharmacol. 2010;24(3):269–82.PubMedCrossRef
81.
Zurück zum Zitat Calvo JA, et al. Muscle-specific expression of PPARgamma coactivator-1alpha improves exercise performance and increases peak oxygen uptake. J Appl Physiol (1985). 2008;104(5):1304–12.CrossRef Calvo JA, et al. Muscle-specific expression of PPARgamma coactivator-1alpha improves exercise performance and increases peak oxygen uptake. J Appl Physiol (1985). 2008;104(5):1304–12.CrossRef
82.
Zurück zum Zitat Roberts LD, Boström P, O’Sullivan JF, Schinzel RT, Lewis GD, Dejam A, et al. beta-Aminoisobutyric acid induces browning of white fat and hepatic beta-oxidation and is inversely correlated with cardiometabolic risk factors. Cell Metab. 2014;19(1):96–108.PubMedPubMedCentralCrossRef Roberts LD, Boström P, O’Sullivan JF, Schinzel RT, Lewis GD, Dejam A, et al. beta-Aminoisobutyric acid induces browning of white fat and hepatic beta-oxidation and is inversely correlated with cardiometabolic risk factors. Cell Metab. 2014;19(1):96–108.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Kitase Y, Vallejo JA, Gutheil W, Vemula H, Jähn K, Yi J, et al. beta-Aminoisobutyric acid, l-BAIBA, is a muscle-derived osteocyte survival factor. Cell Rep. 2018;22(6):1531–44.PubMedPubMedCentralCrossRef Kitase Y, Vallejo JA, Gutheil W, Vemula H, Jähn K, Yi J, et al. beta-Aminoisobutyric acid, l-BAIBA, is a muscle-derived osteocyte survival factor. Cell Rep. 2018;22(6):1531–44.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Shi CX, Zhao MX, Shu XD, Xiong XQ, Wang JJ, Gao XY, et al. beta-Aminoisobutyric acid attenuates hepatic endoplasmic reticulum stress and glucose/lipid metabolic disturbance in mice with type 2 diabetes. Sci Rep. 2016;6:21924.PubMedPubMedCentralCrossRef Shi CX, Zhao MX, Shu XD, Xiong XQ, Wang JJ, Gao XY, et al. beta-Aminoisobutyric acid attenuates hepatic endoplasmic reticulum stress and glucose/lipid metabolic disturbance in mice with type 2 diabetes. Sci Rep. 2016;6:21924.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Wang H, Qian J, Zhao X, Xing C, Sun B. beta-Aminoisobutyric acid ameliorates the renal fibrosis in mouse obstructed kidneys via inhibition of renal fibroblast activation and fibrosis. J Pharmacol Sci. 2017;133(4):203–13.PubMedCrossRef Wang H, Qian J, Zhao X, Xing C, Sun B. beta-Aminoisobutyric acid ameliorates the renal fibrosis in mouse obstructed kidneys via inhibition of renal fibroblast activation and fibrosis. J Pharmacol Sci. 2017;133(4):203–13.PubMedCrossRef
87.
Zurück zum Zitat Wang Z, Bian L, Mo C, Shen H, Zhao LJ, Su KJ, et al. Quantification of aminobutyric acids and their clinical applications as biomarkers for osteoporosis. Commun Biol. 2020;3(1):39.PubMedPubMedCentralCrossRef Wang Z, Bian L, Mo C, Shen H, Zhao LJ, Su KJ, et al. Quantification of aminobutyric acids and their clinical applications as biomarkers for osteoporosis. Commun Biol. 2020;3(1):39.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Ostrowski K, Rohde T, Zacho M, Asp S, Pedersen BK. Evidence that interleukin-6 is produced in human skeletal muscle during prolonged running. J Physiol. 1998;508(Pt 3):949–53.PubMedPubMedCentralCrossRef Ostrowski K, Rohde T, Zacho M, Asp S, Pedersen BK. Evidence that interleukin-6 is produced in human skeletal muscle during prolonged running. J Physiol. 1998;508(Pt 3):949–53.PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Nehlsen-Cannarella SL, et al. Carbohydrate and the cytokine response to 2.5 h of running. J Appl Physiol (1985). 1997;82(5):1662–7.CrossRef Nehlsen-Cannarella SL, et al. Carbohydrate and the cytokine response to 2.5 h of running. J Appl Physiol (1985). 1997;82(5):1662–7.CrossRef
90.
Zurück zum Zitat Steensberg A, van Hall G, Osada T, Sacchetti M, Saltin B, Pedersen BK. Production of interleukin-6 in contracting human skeletal muscles can account for the exercise-induced increase in plasma interleukin-6. J Physiol. 2000;529(Pt 1):237–42.PubMedPubMedCentralCrossRef Steensberg A, van Hall G, Osada T, Sacchetti M, Saltin B, Pedersen BK. Production of interleukin-6 in contracting human skeletal muscles can account for the exercise-induced increase in plasma interleukin-6. J Physiol. 2000;529(Pt 1):237–42.PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat Hiscock N, Chan MHS, Bisucci T, Darby IA, Febbraio MA. Skeletal myocytes are a source of interleukin-6 mRNA expression and protein release during contraction: evidence of fiber type specificity. FASEB J. 2004;18(9):992–4.PubMedCrossRef Hiscock N, Chan MHS, Bisucci T, Darby IA, Febbraio MA. Skeletal myocytes are a source of interleukin-6 mRNA expression and protein release during contraction: evidence of fiber type specificity. FASEB J. 2004;18(9):992–4.PubMedCrossRef
92.
Zurück zum Zitat Starkie RL, Angus DJ, Rolland J, Hargreaves M, Febbraio MA. Effect of prolonged, submaximal exercise and carbohydrate ingestion on monocyte intracellular cytokine production in humans. J Physiol. 2000;528(Pt 3):647–55.PubMedPubMedCentralCrossRef Starkie RL, Angus DJ, Rolland J, Hargreaves M, Febbraio MA. Effect of prolonged, submaximal exercise and carbohydrate ingestion on monocyte intracellular cytokine production in humans. J Physiol. 2000;528(Pt 3):647–55.PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Starkie RL, Rolland J, Angus DJ, Anderson MJ, Febbraio MA. Circulating monocytes are not the source of elevations in plasma IL-6 and TNF-alpha levels after prolonged running. Am J Phys Cell Phys. 2001;280(4):C769–74. Starkie RL, Rolland J, Angus DJ, Anderson MJ, Febbraio MA. Circulating monocytes are not the source of elevations in plasma IL-6 and TNF-alpha levels after prolonged running. Am J Phys Cell Phys. 2001;280(4):C769–74.
94.
Zurück zum Zitat Juffer P, Jaspers RT, Klein-Nulend J, Bakker AD. Mechanically loaded myotubes affect osteoclast formation. Calcif Tissue Int. 2014;94(3):319–26.PubMedCrossRef Juffer P, Jaspers RT, Klein-Nulend J, Bakker AD. Mechanically loaded myotubes affect osteoclast formation. Calcif Tissue Int. 2014;94(3):319–26.PubMedCrossRef
95.
Zurück zum Zitat Bakker AD, Kulkarni RN, Klein-Nulend J, Lems WF. IL-6 alters osteocyte signaling toward osteoblasts but not osteoclasts. J Dent Res. 2014;93(4):394–9.PubMedCrossRef Bakker AD, Kulkarni RN, Klein-Nulend J, Lems WF. IL-6 alters osteocyte signaling toward osteoblasts but not osteoclasts. J Dent Res. 2014;93(4):394–9.PubMedCrossRef
96.
Zurück zum Zitat McGregor NE, Murat M, Elango J, Poulton IJ, Walker EC, Crimeen-Irwin B, et al. IL-6 exhibits both cis- and trans-signaling in osteocytes and osteoblasts, but only trans-signaling promotes bone formation and osteoclastogenesis. J Biol Chem. 2019;294(19):7850–63.PubMedPubMedCentralCrossRef McGregor NE, Murat M, Elango J, Poulton IJ, Walker EC, Crimeen-Irwin B, et al. IL-6 exhibits both cis- and trans-signaling in osteocytes and osteoblasts, but only trans-signaling promotes bone formation and osteoclastogenesis. J Biol Chem. 2019;294(19):7850–63.PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Tresguerres FGF, Torres J, López-Quiles J, Hernández G, Vega JA, Tresguerres IF. The osteocyte: a multifunctional cell within the bone. Ann Anat. 2020;227:151422.PubMedCrossRef Tresguerres FGF, Torres J, López-Quiles J, Hernández G, Vega JA, Tresguerres IF. The osteocyte: a multifunctional cell within the bone. Ann Anat. 2020;227:151422.PubMedCrossRef
98.
Zurück zum Zitat Han Y, You X, Xing W, Zhang Z, Zou W. Paracrine and endocrine actions of bone-the functions of secretory proteins from osteoblasts, osteocytes, and osteoclasts. Bone Res. 2018;6:16.PubMedPubMedCentralCrossRef Han Y, You X, Xing W, Zhang Z, Zou W. Paracrine and endocrine actions of bone-the functions of secretory proteins from osteoblasts, osteocytes, and osteoclasts. Bone Res. 2018;6:16.PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Schaffler MB, Cheung WY, Majeska R, Kennedy O. Osteocytes: master orchestrators of bone. Calcif Tissue Int. 2014;94(1):5–24.PubMedCrossRef Schaffler MB, Cheung WY, Majeska R, Kennedy O. Osteocytes: master orchestrators of bone. Calcif Tissue Int. 2014;94(1):5–24.PubMedCrossRef
102.
103.
Zurück zum Zitat DiGirolamo DJ, Clemens TL, Kousteni S. The skeleton as an endocrine organ. Nat Rev Rheumatol. 2012;8(11):674–83.PubMedCrossRef DiGirolamo DJ, Clemens TL, Kousteni S. The skeleton as an endocrine organ. Nat Rev Rheumatol. 2012;8(11):674–83.PubMedCrossRef
105.
Zurück zum Zitat Yamashita T, Yoshioka M, Itoh N. Identification of a novel fibroblast growth factor, FGF-23, preferentially expressed in the ventrolateral thalamic nucleus of the brain. Biochem Biophys Res Commun. 2000;277(2):494–8.PubMedCrossRef Yamashita T, Yoshioka M, Itoh N. Identification of a novel fibroblast growth factor, FGF-23, preferentially expressed in the ventrolateral thalamic nucleus of the brain. Biochem Biophys Res Commun. 2000;277(2):494–8.PubMedCrossRef
106.
Zurück zum Zitat Feng JQ, Ward LM, Liu S, Lu Y, Xie Y, Yuan B, et al. Loss of DMP1 causes rickets and osteomalacia and identifies a role for osteocytes in mineral metabolism. Nat Genet. 2006;38(11):1310–5.PubMedPubMedCentralCrossRef Feng JQ, Ward LM, Liu S, Lu Y, Xie Y, Yuan B, et al. Loss of DMP1 causes rickets and osteomalacia and identifies a role for osteocytes in mineral metabolism. Nat Genet. 2006;38(11):1310–5.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Gattineni J, Bates C, Twombley K, Dwarakanath V, Robinson ML, Goetz R, et al. FGF23 decreases renal NaPi-2a and NaPi-2c expression and induces hypophosphatemia in vivo predominantly via FGF receptor 1. Am J Physiol Ren Physiol. 2009;297(2):F282–91.CrossRef Gattineni J, Bates C, Twombley K, Dwarakanath V, Robinson ML, Goetz R, et al. FGF23 decreases renal NaPi-2a and NaPi-2c expression and induces hypophosphatemia in vivo predominantly via FGF receptor 1. Am J Physiol Ren Physiol. 2009;297(2):F282–91.CrossRef
108.
Zurück zum Zitat Shimada T, Kakitani M, Yamazaki Y, Hasegawa H, Takeuchi Y, Fujita T, et al. Targeted ablation of Fgf23 demonstrates an essential physiological role of FGF23 in phosphate and vitamin D metabolism. J Clin Invest. 2004;113(4):561–8.PubMedPubMedCentralCrossRef Shimada T, Kakitani M, Yamazaki Y, Hasegawa H, Takeuchi Y, Fujita T, et al. Targeted ablation of Fgf23 demonstrates an essential physiological role of FGF23 in phosphate and vitamin D metabolism. J Clin Invest. 2004;113(4):561–8.PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Urakawa I, Yamazaki Y, Shimada T, Iijima K, Hasegawa H, Okawa K, et al. Klotho converts canonical FGF receptor into a specific receptor for FGF23. Nature. 2006;444(7120):770–4.PubMedCrossRef Urakawa I, Yamazaki Y, Shimada T, Iijima K, Hasegawa H, Okawa K, et al. Klotho converts canonical FGF receptor into a specific receptor for FGF23. Nature. 2006;444(7120):770–4.PubMedCrossRef
110.
Zurück zum Zitat Richter B, Faul C. FGF23 actions on target tissues-with and without Klotho. Front Endocrinol (Lausanne). 2018;9:189.CrossRef Richter B, Faul C. FGF23 actions on target tissues-with and without Klotho. Front Endocrinol (Lausanne). 2018;9:189.CrossRef
111.
Zurück zum Zitat Faul C, Amaral AP, Oskouei B, Hu MC, Sloan A, Isakova T, et al. FGF23 induces left ventricular hypertrophy. J Clin Invest. 2011;121(11):4393–408.PubMedPubMedCentralCrossRef Faul C, Amaral AP, Oskouei B, Hu MC, Sloan A, Isakova T, et al. FGF23 induces left ventricular hypertrophy. J Clin Invest. 2011;121(11):4393–408.PubMedPubMedCentralCrossRef
112.
Zurück zum Zitat Touchberry CD, Green TM, Tchikrizov V, Mannix JE, Mao TF, Carney BW, et al. FGF23 is a novel regulator of intracellular calcium and cardiac contractility in addition to cardiac hypertrophy. Am J Physiol Endocrinol Metab. 2013;304(8):E863–73.PubMedPubMedCentralCrossRef Touchberry CD, Green TM, Tchikrizov V, Mannix JE, Mao TF, Carney BW, et al. FGF23 is a novel regulator of intracellular calcium and cardiac contractility in addition to cardiac hypertrophy. Am J Physiol Endocrinol Metab. 2013;304(8):E863–73.PubMedPubMedCentralCrossRef
113.
Zurück zum Zitat Kido S, Hashimoto Y, Segawa H, Tatsumi S, Miyamoto KI. Muscle atrophy in patients wirh ckd results from fgf23/klotho-mediated supression of insulin/igf-i signaling. Kidney Research and Clinical Practice. 2012;31(2):A44.CrossRef Kido S, Hashimoto Y, Segawa H, Tatsumi S, Miyamoto KI. Muscle atrophy in patients wirh ckd results from fgf23/klotho-mediated supression of insulin/igf-i signaling. Kidney Research and Clinical Practice. 2012;31(2):A44.CrossRef
114.
Zurück zum Zitat Avin KG, Vallejo JA, Chen NX, Wang K, Touchberry CD, Brotto M, et al. Fibroblast growth factor 23 does not directly influence skeletal muscle cell proliferation and differentiation or ex vivo muscle contractility. Am J Physiol Endocrinol Metab. 2018;315(4):E594–e604.PubMedPubMedCentralCrossRef Avin KG, Vallejo JA, Chen NX, Wang K, Touchberry CD, Brotto M, et al. Fibroblast growth factor 23 does not directly influence skeletal muscle cell proliferation and differentiation or ex vivo muscle contractility. Am J Physiol Endocrinol Metab. 2018;315(4):E594–e604.PubMedPubMedCentralCrossRef
115.
Zurück zum Zitat Wang K, le L, Chun BM, Tiede-Lewis LAM, Shiflett LA, Prideaux M, et al. A novel osteogenic cell line that differentiates into GFP-tagged osteocytes and forms mineral with a bone-like lacunocanalicular structure. J Bone Miner Res. 2019;34(6):979–95.PubMedCrossRef Wang K, le L, Chun BM, Tiede-Lewis LAM, Shiflett LA, Prideaux M, et al. A novel osteogenic cell line that differentiates into GFP-tagged osteocytes and forms mineral with a bone-like lacunocanalicular structure. J Bone Miner Res. 2019;34(6):979–95.PubMedCrossRef
116.
Zurück zum Zitat McCormick, L.A., et al., Role of FGF9 in promotion of early osteocyte differentiation and as a potent inducer of FGF23 expression in osteocytes. J Bone Mineral Res, 2016. American Society of Bone and Mineral Research (ASBMR Annual Meeting – Atlanta, GA 2016): p. Abstract 1122. McCormick, L.A., et al., Role of FGF9 in promotion of early osteocyte differentiation and as a potent inducer of FGF23 expression in osteocytes. J Bone Mineral Res, 2016. American Society of Bone and Mineral Research (ASBMR Annual Meeting – Atlanta, GA 2016): p. Abstract 1122.
117.
Zurück zum Zitat Oury F, Sumara G, Sumara O, Ferron M, Chang H, Smith CE, et al. Endocrine regulation of male fertility by the skeleton. Cell. 2011;144(5):796–809.PubMedPubMedCentralCrossRef Oury F, Sumara G, Sumara O, Ferron M, Chang H, Smith CE, et al. Endocrine regulation of male fertility by the skeleton. Cell. 2011;144(5):796–809.PubMedPubMedCentralCrossRef
118.
Zurück zum Zitat Chowdhury S, Schulz L, Palmisano B, Singh P, Berger JM, Yadav VK, et al. Muscle derived interleukin-6 increases exercise capacity by signaling in osteoblasts. J Clin Invest. 2020. Chowdhury S, Schulz L, Palmisano B, Singh P, Berger JM, Yadav VK, et al. Muscle derived interleukin-6 increases exercise capacity by signaling in osteoblasts. J Clin Invest. 2020.
119.
Zurück zum Zitat Dole NS, Mazur CM, Acevedo C, Lopez JP, Monteiro DA, Fowler TW, et al. Osteocyte-intrinsic TGF-beta signaling regulates bone quality through perilacunar/canalicular remodeling. Cell Rep. 2017;21(9):2585–96.PubMedPubMedCentralCrossRef Dole NS, Mazur CM, Acevedo C, Lopez JP, Monteiro DA, Fowler TW, et al. Osteocyte-intrinsic TGF-beta signaling regulates bone quality through perilacunar/canalicular remodeling. Cell Rep. 2017;21(9):2585–96.PubMedPubMedCentralCrossRef
120.
Zurück zum Zitat Waning DL, Mohammad KS, Reiken S, Xie W, Andersson DC, John S, et al. Excess TGF-beta mediates muscle weakness associated with bone metastases in mice. Nat Med. 2015;21(11):1262–71.PubMedPubMedCentralCrossRef Waning DL, Mohammad KS, Reiken S, Xie W, Andersson DC, John S, et al. Excess TGF-beta mediates muscle weakness associated with bone metastases in mice. Nat Med. 2015;21(11):1262–71.PubMedPubMedCentralCrossRef
121.
122.
Zurück zum Zitat Kim JA, Roh E, Hong SH, Lee YB, Kim NH, Yoo HJ, et al. Association of serum sclerostin levels with low skeletal muscle mass: the Korean Sarcopenic Obesity Study (KSOS). Bone. 2019;128:115053.PubMedCrossRef Kim JA, Roh E, Hong SH, Lee YB, Kim NH, Yoo HJ, et al. Association of serum sclerostin levels with low skeletal muscle mass: the Korean Sarcopenic Obesity Study (KSOS). Bone. 2019;128:115053.PubMedCrossRef
123.
Zurück zum Zitat Hesse E, et al. Sclerostin inhibition alleviates breast cancer-induced bone metastases and muscle weakness. JCI Insight. 2019;5. Hesse E, et al. Sclerostin inhibition alleviates breast cancer-induced bone metastases and muscle weakness. JCI Insight. 2019;5.
124.
Zurück zum Zitat Girardi F, Le Grand F. Wnt signaling in skeletal muscle development and regeneration. Prog Mol Biol Transl Sci. 2018;153:157–79.PubMedCrossRef Girardi F, Le Grand F. Wnt signaling in skeletal muscle development and regeneration. Prog Mol Biol Transl Sci. 2018;153:157–79.PubMedCrossRef
126.
Zurück zum Zitat Huang J, Romero-Suarez S, Lara N, Mo C, Kaja S, Brotto L, et al. Crosstalk between MLO-Y4 osteocytes and C2C12 muscle cells is mediated by the Wnt/beta-catenin pathway. JBMR Plus. 2017;1(2):86–100.PubMedPubMedCentralCrossRef Huang J, Romero-Suarez S, Lara N, Mo C, Kaja S, Brotto L, et al. Crosstalk between MLO-Y4 osteocytes and C2C12 muscle cells is mediated by the Wnt/beta-catenin pathway. JBMR Plus. 2017;1(2):86–100.PubMedPubMedCentralCrossRef
127.
Zurück zum Zitat Kamel MA, Picconi JL, Lara-Castillo N, Johnson ML. Activation of beta-catenin signaling in MLO-Y4 osteocytic cells versus 2T3 osteoblastic cells by fluid flow shear stress and PGE2: implications for the study of mechanosensation in bone. Bone. 2010;47(5):872–81.PubMedPubMedCentralCrossRef Kamel MA, Picconi JL, Lara-Castillo N, Johnson ML. Activation of beta-catenin signaling in MLO-Y4 osteocytic cells versus 2T3 osteoblastic cells by fluid flow shear stress and PGE2: implications for the study of mechanosensation in bone. Bone. 2010;47(5):872–81.PubMedPubMedCentralCrossRef
128.
Zurück zum Zitat Mo C, Romero-Suarez S, Bonewald L, Johnson M, Brotto M. Prostaglandin E2: from clinical applications to its potential role in bone- muscle crosstalk and myogenic differentiation. Recent Pat Biotechnol. 2012;6(3):223–9.PubMedPubMedCentralCrossRef Mo C, Romero-Suarez S, Bonewald L, Johnson M, Brotto M. Prostaglandin E2: from clinical applications to its potential role in bone- muscle crosstalk and myogenic differentiation. Recent Pat Biotechnol. 2012;6(3):223–9.PubMedPubMedCentralCrossRef
129.
Zurück zum Zitat Mo C, Zhao R, Vallejo J, Igwe O, Bonewald L, Wetmore L, et al. Prostaglandin E2 promotes proliferation of skeletal muscle myoblasts via EP4 receptor activation. Cell Cycle. 2015;14(10):1507–16.PubMedPubMedCentralCrossRef Mo C, Zhao R, Vallejo J, Igwe O, Bonewald L, Wetmore L, et al. Prostaglandin E2 promotes proliferation of skeletal muscle myoblasts via EP4 receptor activation. Cell Cycle. 2015;14(10):1507–16.PubMedPubMedCentralCrossRef
130.
Zurück zum Zitat Bothwell W, Verburg M, Wynalda M, Daniels EG, Fitzpatrick FA. A radioimmunoassay for the unstable pulmonary metabolites of prostaglandin E1 and E2: an indirect index of their in vivo disposition and pharmacokinetics. J Pharmacol Exp Ther. 1982;220(2):229–35.PubMed Bothwell W, Verburg M, Wynalda M, Daniels EG, Fitzpatrick FA. A radioimmunoassay for the unstable pulmonary metabolites of prostaglandin E1 and E2: an indirect index of their in vivo disposition and pharmacokinetics. J Pharmacol Exp Ther. 1982;220(2):229–35.PubMed
131.
Zurück zum Zitat Xiong J, Onal M, Jilka RL, Weinstein RS, Manolagas SC, O’Brien CA. Matrix-embedded cells control osteoclast formation. Nat Med. 2011;17(10):1235–41.PubMedPubMedCentralCrossRef Xiong J, Onal M, Jilka RL, Weinstein RS, Manolagas SC, O’Brien CA. Matrix-embedded cells control osteoclast formation. Nat Med. 2011;17(10):1235–41.PubMedPubMedCentralCrossRef
132.
Zurück zum Zitat Nakashima T, Hayashi M, Fukunaga T, Kurata K, Oh-hora M, Feng JQ, et al. Evidence for osteocyte regulation of bone homeostasis through RANKL expression. Nat Med. 2011;17(10):1231–4.PubMedCrossRef Nakashima T, Hayashi M, Fukunaga T, Kurata K, Oh-hora M, Feng JQ, et al. Evidence for osteocyte regulation of bone homeostasis through RANKL expression. Nat Med. 2011;17(10):1231–4.PubMedCrossRef
133.
Zurück zum Zitat Wijenayaka AR, Kogawa M, Lim HP, Bonewald LF, Findlay DM, Atkins GJ. Sclerostin stimulates osteocyte support of osteoclast activity by a RANKL-dependent pathway. PLoS One. 2011;6(10):e25900.PubMedPubMedCentralCrossRef Wijenayaka AR, Kogawa M, Lim HP, Bonewald LF, Findlay DM, Atkins GJ. Sclerostin stimulates osteocyte support of osteoclast activity by a RANKL-dependent pathway. PLoS One. 2011;6(10):e25900.PubMedPubMedCentralCrossRef
134.
Zurück zum Zitat Kong YY, Yoshida H, Sarosi I, Tan HL, Timms E, Capparelli C, et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature. 1999;397(6717):315–23.PubMedCrossRef Kong YY, Yoshida H, Sarosi I, Tan HL, Timms E, Capparelli C, et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature. 1999;397(6717):315–23.PubMedCrossRef
135.
Zurück zum Zitat Bonnet N, Bourgoin L, Biver E, Douni E, Ferrari S. RANKL inhibition improves muscle strength and insulin sensitivity and restores bone mass. J Clin Invest. 2019;129(8):3214–23.PubMedPubMedCentralCrossRef Bonnet N, Bourgoin L, Biver E, Douni E, Ferrari S. RANKL inhibition improves muscle strength and insulin sensitivity and restores bone mass. J Clin Invest. 2019;129(8):3214–23.PubMedPubMedCentralCrossRef
136.
Zurück zum Zitat Hamoudi D, Bouredji Z, Marcadet L, Yagita H, Landry LB, Argaw A, et al. Muscle weakness and selective muscle atrophy in osteoprotegerin-deficient mice. Hum Mol Genet. 2020;29(3):483–94.PubMedCrossRef Hamoudi D, Bouredji Z, Marcadet L, Yagita H, Landry LB, Argaw A, et al. Muscle weakness and selective muscle atrophy in osteoprotegerin-deficient mice. Hum Mol Genet. 2020;29(3):483–94.PubMedCrossRef
137.
Zurück zum Zitat Boulanger Piette A, Hamoudi D, Marcadet L, Morin F, Argaw A, Ward L, et al. Targeting the muscle-bone unit: filling two needs with one deed in the treatment of Duchenne muscular dystrophy. Curr Osteoporos Rep. 2018;16(5):541–53.PubMedCrossRef Boulanger Piette A, Hamoudi D, Marcadet L, Morin F, Argaw A, Ward L, et al. Targeting the muscle-bone unit: filling two needs with one deed in the treatment of Duchenne muscular dystrophy. Curr Osteoporos Rep. 2018;16(5):541–53.PubMedCrossRef
138.
Zurück zum Zitat Hamoudi D, Marcadet L, Piette Boulanger A, Yagita H, Bouredji Z, Argaw A, et al. An anti-RANKL treatment reduces muscle inflammation and dysfunction and strengthens bone in dystrophic mice. Hum Mol Genet. 2019;28(18):3101–12.PubMedCrossRef Hamoudi D, Marcadet L, Piette Boulanger A, Yagita H, Bouredji Z, Argaw A, et al. An anti-RANKL treatment reduces muscle inflammation and dysfunction and strengthens bone in dystrophic mice. Hum Mol Genet. 2019;28(18):3101–12.PubMedCrossRef
139.
Zurück zum Zitat Brun J, Berthou F, Trajkovski M, Maechler P, Foti M, Bonnet N. Bone regulates browning and energy metabolism through mature osteoblast/osteocyte PPARgamma expression. Diabetes. 2017;66(10):2541–54.PubMedCrossRef Brun J, Berthou F, Trajkovski M, Maechler P, Foti M, Bonnet N. Bone regulates browning and energy metabolism through mature osteoblast/osteocyte PPARgamma expression. Diabetes. 2017;66(10):2541–54.PubMedCrossRef
140.
Zurück zum Zitat Mera P, Ferron M, Mosialou I. Regulation of energy metabolism by bone-derived hormones. Cold Spring Harb Perspect Med. 2018:8(6). Mera P, Ferron M, Mosialou I. Regulation of energy metabolism by bone-derived hormones. Cold Spring Harb Perspect Med. 2018:8(6).
141.
Zurück zum Zitat Picca A, Calvani R, Manes-Gravina E, Spaziani L, Landi F, Bernabei R, et al. Bone-muscle crosstalk: unraveling new therapeutic targets for osteoporosis. Curr Pharm Des. 2017;23(41):6256–63.PubMedCrossRef Picca A, Calvani R, Manes-Gravina E, Spaziani L, Landi F, Bernabei R, et al. Bone-muscle crosstalk: unraveling new therapeutic targets for osteoporosis. Curr Pharm Des. 2017;23(41):6256–63.PubMedCrossRef
142.
Zurück zum Zitat Maurel DB, Jahn K, Lara-Castillo N. Muscle-bone crosstalk: emerging opportunities for novel therapeutic approaches to treat musculoskeletal pathologies. Biomedicines. 2017:5(4). Maurel DB, Jahn K, Lara-Castillo N. Muscle-bone crosstalk: emerging opportunities for novel therapeutic approaches to treat musculoskeletal pathologies. Biomedicines. 2017:5(4).
143.
Zurück zum Zitat Compston J. Emerging therapeutic concepts for muscle and bone preservation/building. Bone. 2015;80:150–6.PubMedCrossRef Compston J. Emerging therapeutic concepts for muscle and bone preservation/building. Bone. 2015;80:150–6.PubMedCrossRef
144.
Zurück zum Zitat Girgis CM. Integrated therapies for osteoporosis and sarcopenia: from signaling pathways to clinical trials. Calcif Tissue Int. 2015;96(3):243–55.PubMedCrossRef Girgis CM. Integrated therapies for osteoporosis and sarcopenia: from signaling pathways to clinical trials. Calcif Tissue Int. 2015;96(3):243–55.PubMedCrossRef
Metadaten
Titel
Bone-Muscle Mutual Interactions
verfasst von
Nuria Lara-Castillo
Mark L. Johnson
Publikationsdatum
09.06.2020
Verlag
Springer US
Erschienen in
Current Osteoporosis Reports / Ausgabe 4/2020
Print ISSN: 1544-1873
Elektronische ISSN: 1544-2241
DOI
https://doi.org/10.1007/s11914-020-00602-6

Weitere Artikel der Ausgabe 4/2020

Current Osteoporosis Reports 4/2020 Zur Ausgabe

Bone and Diabetes (A Schwartz and P Vestergaard, Section Editors)

Update on the Acute Effects of Glucose, Insulin, and Incretins on Bone Turnover In Vivo

Rare Bone Diseases (CB Langman and E Shore, Section Editors)

Stem Cell Therapy as a Treatment for Osteogenesis Imperfecta

Bone and Diabetes (A Schwartz and P Vestergaard, Section Editors)

The Impact of Exercise on Bone Health in Type 2 Diabetes Mellitus—a Systematic Review

Arthropedia

Grundlagenwissen der Arthroskopie und Gelenkchirurgie. Erweitert durch Fallbeispiele, Videos und Abbildungen. 
» Jetzt entdecken

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Arthroskopie kann Knieprothese nicht hinauszögern

25.04.2024 Gonarthrose Nachrichten

Ein arthroskopischer Eingriff bei Kniearthrose macht im Hinblick darauf, ob und wann ein Gelenkersatz fällig wird, offenbar keinen Unterschied.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Ärztliche Empathie hilft gegen Rückenschmerzen

23.04.2024 Leitsymptom Rückenschmerzen Nachrichten

Personen mit chronischen Rückenschmerzen, die von einfühlsamen Ärzten und Ärztinnen betreut werden, berichten über weniger Beschwerden und eine bessere Lebensqualität.

Update Orthopädie und Unfallchirurgie

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