Skip to main content
Erschienen in: Current Osteoporosis Reports 6/2019

20.11.2019 | Bone Marrow and Adipose Tissue (G Duque and B Lecka-Czernik, Section Editors)

Endogenous Glucocorticoid Signaling in the Regulation of Bone and Marrow Adiposity: Lessons from Metabolism and Cross Talk in Other Tissues

verfasst von: Anuj K. Sharma, Xingming Shi, Carlos M. Isales, Meghan E. McGee-Lawrence

Erschienen in: Current Osteoporosis Reports | Ausgabe 6/2019

Einloggen, um Zugang zu erhalten

Abstract

Purpose of Review

The development of adiposity in the bone marrow, known as marrow adipose tissue (MAT), is often associated with musculoskeletal frailty. Glucocorticoids, which are a key component of the biological response to stress, affect both bone and MAT. These molecules signal through receptors such as the glucocorticoid receptor (GR), but the role of the GR in regulation of MAT is not yet clear from previous studies. The purpose of this review is to establish and determine the role of GR-mediated signaling in marrow adiposity by comparing and contrasting what is known against other energy-storing tissues like adipose tissue, liver, and muscle, to provide better insight into the regulation of MAT during times of metabolic stress (e.g., dietary challenges, aging).

Recent Findings

GR-mediated glucocorticoid signaling is critical for proper storage and utilization of lipids in cells such as adipocytes and hepatocytes and proteolysis in muscle, impacting whole-body composition, energy utilization, and homeostasis through a complex network of tissue cross talk between these systems. Loss of GR signaling in bone promotes increased MAT and decreased bone mass.

Summary

GR-mediated signaling in the liver, adipose tissue, and muscle is critical for whole-body energy and metabolic homeostasis, and both similarities and differences in GR-mediated GC signaling in MAT as compared with these tissues are readily apparent. It is clear that GC-induced pathways work together through these tissues to affect systemic biology, and understanding the role of bone in these patterns of tissue cross talk may lead to a better understanding of MAT-bone biology that improves treatment strategies for frailty-associated diseases.
Literatur
1.
Zurück zum Zitat Hawkes CP, Mostoufi-Moab S. Fat-bone interaction within the bone marrow milieu: impact on hematopoiesis and systemic energy metabolism. Bone. 2019;119:57–64.PubMedCrossRef Hawkes CP, Mostoufi-Moab S. Fat-bone interaction within the bone marrow milieu: impact on hematopoiesis and systemic energy metabolism. Bone. 2019;119:57–64.PubMedCrossRef
2.
Zurück zum Zitat Sulston RJ, Cawthorn WP. Bone marrow adipose tissue as an endocrine organ: close to the bone? Horm Mol Biol Clin Invest. 2016;28:21–38. Sulston RJ, Cawthorn WP. Bone marrow adipose tissue as an endocrine organ: close to the bone? Horm Mol Biol Clin Invest. 2016;28:21–38.
4.
Zurück zum Zitat Li J, Li J, Zuo B, Zhang L, Dai L, Zhang X. Osteoblast versus adipocyte: bone marrow microenvironment-guided epigenetic control. Case Rep Orthop Res. 2018;1:2–18.CrossRef Li J, Li J, Zuo B, Zhang L, Dai L, Zhang X. Osteoblast versus adipocyte: bone marrow microenvironment-guided epigenetic control. Case Rep Orthop Res. 2018;1:2–18.CrossRef
5.
Zurück zum Zitat Newton LA, Hanks JL, Davis M, Casazza K. The relationships among total body fat, bone mineral content and bone marrow adipose tissue in early-pubertal girls. Bonekey Rep. 2013;2:315.CrossRef Newton LA, Hanks JL, Davis M, Casazza K. The relationships among total body fat, bone mineral content and bone marrow adipose tissue in early-pubertal girls. Bonekey Rep. 2013;2:315.CrossRef
6.
Zurück zum Zitat Rharass T, and Lucas S (2018) Mechanisms in endocrinology: bone marrow adiposity and bone, a bad romance? Eur J Endocrinol Rharass T, and Lucas S (2018) Mechanisms in endocrinology: bone marrow adiposity and bone, a bad romance? Eur J Endocrinol
7.
Zurück zum Zitat Li T, Jiang S, Lu C, Yang W, Yang Z, Hu W, et al. Melatonin: another avenue for treating osteoporosis? J Pineal Res. 2019;66:e12548.PubMedCrossRef Li T, Jiang S, Lu C, Yang W, Yang Z, Hu W, et al. Melatonin: another avenue for treating osteoporosis? J Pineal Res. 2019;66:e12548.PubMedCrossRef
8.
Zurück zum Zitat Yang S, Liu H, Liu Y, Liu L, Zhang W, and En L (2019) Effect of adiponectin secreted from adipose-derived stem cells on bone-fat balance and bone defect healing. J Tissue Eng Regen Med Yang S, Liu H, Liu Y, Liu L, Zhang W, and En L (2019) Effect of adiponectin secreted from adipose-derived stem cells on bone-fat balance and bone defect healing. J Tissue Eng Regen Med
9.
Zurück zum Zitat Manolagas SC, O’Brien CA, Almeida M. The role of estrogen and androgen receptors in bone health and disease. Nat Rev Endocrinol. 2013;9:699–712.PubMedPubMedCentralCrossRef Manolagas SC, O’Brien CA, Almeida M. The role of estrogen and androgen receptors in bone health and disease. Nat Rev Endocrinol. 2013;9:699–712.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Zhang XZ, Kalu DN, Erbas B, Hopper JL, Seeman E. The effects of gonadectomy on bone size, mass, and volumetric density in growing rats are gender-, site-, and growth hormone-specific. J Bone Miner Res Off J Am Soc Bone Miner Res. 1999;14:802–9.CrossRef Zhang XZ, Kalu DN, Erbas B, Hopper JL, Seeman E. The effects of gonadectomy on bone size, mass, and volumetric density in growing rats are gender-, site-, and growth hormone-specific. J Bone Miner Res Off J Am Soc Bone Miner Res. 1999;14:802–9.CrossRef
11.
Zurück zum Zitat Bao T, Zeng L, Yang K, Li Y, Ren F, Zhang Y, et al. Can melatonin improve the osteopenia of perimenopausal and postmenopausal women? A meta-analysis. Int J Endocrinol. 2019;2019:5151678.PubMedPubMedCentralCrossRef Bao T, Zeng L, Yang K, Li Y, Ren F, Zhang Y, et al. Can melatonin improve the osteopenia of perimenopausal and postmenopausal women? A meta-analysis. Int J Endocrinol. 2019;2019:5151678.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Ikegame M, Hattori A, Tabata MJ, Kitamura KI, Tabuchi Y, Furusawa Y, et al. Melatonin is a potential drug for the prevention of bone loss during space flight. J Pineal Res. 2019;67:e12594.PubMedPubMedCentralCrossRef Ikegame M, Hattori A, Tabata MJ, Kitamura KI, Tabuchi Y, Furusawa Y, et al. Melatonin is a potential drug for the prevention of bone loss during space flight. J Pineal Res. 2019;67:e12594.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Fairfield H, Falank C, Harris E, Demambro V, McDonald M, Pettitt JA, et al. The skeletal cell-derived molecule sclerostin drives bone marrow adipogenesis. J Cell Physiol. 2018;233:1156–67.PubMedCrossRef Fairfield H, Falank C, Harris E, Demambro V, McDonald M, Pettitt JA, et al. The skeletal cell-derived molecule sclerostin drives bone marrow adipogenesis. J Cell Physiol. 2018;233:1156–67.PubMedCrossRef
14.
Zurück zum Zitat Tchernof A, Despres JP. Pathophysiology of human visceral obesity: an update. Physiol Rev. 2013;93:359–404.PubMedCrossRef Tchernof A, Despres JP. Pathophysiology of human visceral obesity: an update. Physiol Rev. 2013;93:359–404.PubMedCrossRef
15.
Zurück zum Zitat Bartelt A, Bruns OT, Reimer R, Hohenberg H, Ittrich H, Peldschus K, et al. Brown adipose tissue activity controls triglyceride clearance. Nat Med. 2011;17:200–5.PubMedCrossRef Bartelt A, Bruns OT, Reimer R, Hohenberg H, Ittrich H, Peldschus K, et al. Brown adipose tissue activity controls triglyceride clearance. Nat Med. 2011;17:200–5.PubMedCrossRef
16.
Zurück zum Zitat Devlin MJ, Rosen CJ. The bone-fat interface: basic and clinical implications of marrow adiposity. Lancet Diabetes Endocrinol. 2015;3:141–7.PubMedCrossRef Devlin MJ, Rosen CJ. The bone-fat interface: basic and clinical implications of marrow adiposity. Lancet Diabetes Endocrinol. 2015;3:141–7.PubMedCrossRef
17.
Zurück zum Zitat Hardouin P, Rharass T, Lucas S. Bone marrow adipose tissue: to be or not to be a typical adipose tissue? Front Endocrinol (Lausanne). 2016;7:85.CrossRef Hardouin P, Rharass T, Lucas S. Bone marrow adipose tissue: to be or not to be a typical adipose tissue? Front Endocrinol (Lausanne). 2016;7:85.CrossRef
18.
Zurück zum Zitat Ghali O, Al Rassy N, Hardouin P, Chauveau C. Increased bone marrow adiposity in a context of energy deficit: the tip of the iceberg? Front Endocrinol (Lausanne). 2016;7:125.CrossRef Ghali O, Al Rassy N, Hardouin P, Chauveau C. Increased bone marrow adiposity in a context of energy deficit: the tip of the iceberg? Front Endocrinol (Lausanne). 2016;7:125.CrossRef
19.
Zurück zum Zitat Scheller EL, Doucette CR, Learman BS, Cawthorn WP, Khandaker S, Schell B, et al. Region-specific variation in the properties of skeletal adipocytes reveals regulated and constitutive marrow adipose tissues. Nat Commun. 2015;6:7808.PubMedPubMedCentralCrossRef Scheller EL, Doucette CR, Learman BS, Cawthorn WP, Khandaker S, Schell B, et al. Region-specific variation in the properties of skeletal adipocytes reveals regulated and constitutive marrow adipose tissues. Nat Commun. 2015;6:7808.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Li Y, Meng Y, Yu X. The unique metabolic characteristics of bone marrow adipose tissue. Front Endocrinol (Lausanne). 2019;10:69.CrossRef Li Y, Meng Y, Yu X. The unique metabolic characteristics of bone marrow adipose tissue. Front Endocrinol (Lausanne). 2019;10:69.CrossRef
21.
Zurück zum Zitat Scheller EL, Cawthorn WP, Burr AA, Horowitz MC, MacDougald OA. Marrow adipose tissue: trimming the fat. Trends Endocrinol Metab. 2016;27:392–403.PubMedPubMedCentralCrossRef Scheller EL, Cawthorn WP, Burr AA, Horowitz MC, MacDougald OA. Marrow adipose tissue: trimming the fat. Trends Endocrinol Metab. 2016;27:392–403.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat • Cawthorn WP, Scheller EL, Parlee SD, Pham HA, Learman BS, Redshaw CM, et al. Expansion of bone marrow adipose tissue during caloric restriction is associated with increased circulating glucocorticoids and not with hypoleptinemia. Endocrinology. 2016;157:508–21 This study provided some of the first evidence that GC may drive MAT expansion during caloric restriction. PubMedCrossRef • Cawthorn WP, Scheller EL, Parlee SD, Pham HA, Learman BS, Redshaw CM, et al. Expansion of bone marrow adipose tissue during caloric restriction is associated with increased circulating glucocorticoids and not with hypoleptinemia. Endocrinology. 2016;157:508–21 This study provided some of the first evidence that GC may drive MAT expansion during caloric restriction. PubMedCrossRef
23.
Zurück zum Zitat Baek K, Bloomfield S. A. (2012) Blocking beta-adrenergic signaling attenuates reductions in circulating leptin, cancellous bone mass, and marrow adiposity seen with dietary energy restriction. J Appl Physiol. 1985;113:1792–801.CrossRef Baek K, Bloomfield S. A. (2012) Blocking beta-adrenergic signaling attenuates reductions in circulating leptin, cancellous bone mass, and marrow adiposity seen with dietary energy restriction. J Appl Physiol. 1985;113:1792–801.CrossRef
24.
Zurück zum Zitat Hooshmand S, Balakrishnan A, Clark RM, Owen KQ, Koo SI, Arjmandi BH. Dietary l-carnitine supplementation improves bone mineral density by suppressing bone turnover in aged ovariectomized rats. Phytomedicine. 2008;15:595–601.PubMedCrossRef Hooshmand S, Balakrishnan A, Clark RM, Owen KQ, Koo SI, Arjmandi BH. Dietary l-carnitine supplementation improves bone mineral density by suppressing bone turnover in aged ovariectomized rats. Phytomedicine. 2008;15:595–601.PubMedCrossRef
25.
Zurück zum Zitat Adamek G, Felix R, Guenther HL, Fleisch H. Fatty acid oxidation in bone tissue and bone cells in culture. Characterization and hormonal influences. Biochem J. 1987;248:129–37.PubMedPubMedCentralCrossRef Adamek G, Felix R, Guenther HL, Fleisch H. Fatty acid oxidation in bone tissue and bone cells in culture. Characterization and hormonal influences. Biochem J. 1987;248:129–37.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Frey JL, Li Z, Ellis JM, Zhang Q, Farber CR, Aja S, et al. Wnt-Lrp5 signaling regulates fatty acid metabolism in the osteoblast. Mol Cell Biol. 2015;35:1979–91.PubMedPubMedCentralCrossRef Frey JL, Li Z, Ellis JM, Zhang Q, Farber CR, Aja S, et al. Wnt-Lrp5 signaling regulates fatty acid metabolism in the osteoblast. Mol Cell Biol. 2015;35:1979–91.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Kim SP, Li Z, Zoch ML, Frey JL, Bowman CE, Kushwaha P, et al. Fatty acid oxidation by the osteoblast is required for normal bone acquisition in a sex- and diet-dependent manner. JCI Insight. 2017;2:92704.PubMedCrossRef Kim SP, Li Z, Zoch ML, Frey JL, Bowman CE, Kushwaha P, et al. Fatty acid oxidation by the osteoblast is required for normal bone acquisition in a sex- and diet-dependent manner. JCI Insight. 2017;2:92704.PubMedCrossRef
30.
Zurück zum Zitat McGee-Lawrence ME, Carpio LR, Schulze RJ, Pierce JL, McNiven MA, Farr JN, et al. Hdac3 deficiency increases marrow adiposity and induces lipid storage and glucocorticoid metabolism in osteochondroprogenitor cells. J Bone Miner Res Off J Am Soc Bone Miner Res. 2016;31:116–28.CrossRef McGee-Lawrence ME, Carpio LR, Schulze RJ, Pierce JL, McNiven MA, Farr JN, et al. Hdac3 deficiency increases marrow adiposity and induces lipid storage and glucocorticoid metabolism in osteochondroprogenitor cells. J Bone Miner Res Off J Am Soc Bone Miner Res. 2016;31:116–28.CrossRef
31.
Zurück zum Zitat Gunaratnam K, Vidal C, Gimble JM, Duque G. Mechanisms of palmitate-induced lipotoxicity in human osteoblasts. Endocrinology. 2014;155:108–16.PubMedCrossRef Gunaratnam K, Vidal C, Gimble JM, Duque G. Mechanisms of palmitate-induced lipotoxicity in human osteoblasts. Endocrinology. 2014;155:108–16.PubMedCrossRef
32.
Zurück zum Zitat Wei J, Ferron M, Clarke CJ, Hannun YA, Jiang H, Blaner WS, et al. Bone-specific insulin resistance disrupts whole-body glucose homeostasis via decreased osteocalcin activation. J Clin Invest. 2014;124:1–13.PubMedCrossRef Wei J, Ferron M, Clarke CJ, Hannun YA, Jiang H, Blaner WS, et al. Bone-specific insulin resistance disrupts whole-body glucose homeostasis via decreased osteocalcin activation. J Clin Invest. 2014;124:1–13.PubMedCrossRef
33.
Zurück zum Zitat Tommasini S, Nelson T, Faulkner-Filosa C, Rodeheffer MS, Rosen C, Lindskog D, and Horowitz M (2018) Selective deletion of marrow adipocytes leads to increased mesenchymal precursors, a shift in lineage allocation, and increased bone mass with improved bone biomechanics. In ASBMR Annual Meeting, American Society for Bone and Mineral Research, Montreal, Quebec, Canada Tommasini S, Nelson T, Faulkner-Filosa C, Rodeheffer MS, Rosen C, Lindskog D, and Horowitz M (2018) Selective deletion of marrow adipocytes leads to increased mesenchymal precursors, a shift in lineage allocation, and increased bone mass with improved bone biomechanics. In ASBMR Annual Meeting, American Society for Bone and Mineral Research, Montreal, Quebec, Canada
34.
Zurück zum Zitat Cao J, Ou G, Yang N, Ding K, Kream BE, Hamrick MW, et al. Impact of targeted PPARgamma disruption on bone remodeling. Mol Cell Endocrinol. 2015;410:27–34.PubMedPubMedCentralCrossRef Cao J, Ou G, Yang N, Ding K, Kream BE, Hamrick MW, et al. Impact of targeted PPARgamma disruption on bone remodeling. Mol Cell Endocrinol. 2015;410:27–34.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Bermeo S, Al Saedi A, Vidal C, Khalil M, Pang M, Troen BR, et al. Treatment with an inhibitor of fatty acid synthase attenuates bone loss in ovariectomized mice. Bone. 2019;122:114–22.PubMedCrossRef Bermeo S, Al Saedi A, Vidal C, Khalil M, Pang M, Troen BR, et al. Treatment with an inhibitor of fatty acid synthase attenuates bone loss in ovariectomized mice. Bone. 2019;122:114–22.PubMedCrossRef
36.
Zurück zum Zitat Duque G, Li W, Vidal C, Bermeo S, Rivas D, Henderson J. Pharmacological inhibition of PPARgamma increases osteoblastogenesis and bone mass in male C57BL/6 mice. J Bone Miner Res Off J Am Soc Bone Miner Res. 2013;28:639–48.CrossRef Duque G, Li W, Vidal C, Bermeo S, Rivas D, Henderson J. Pharmacological inhibition of PPARgamma increases osteoblastogenesis and bone mass in male C57BL/6 mice. J Bone Miner Res Off J Am Soc Bone Miner Res. 2013;28:639–48.CrossRef
37.
Zurück zum Zitat Rosario R, Baban B, Hamrick M, Isales C, Shi X. PPARγ in inflammation and aging. Denver: In ASBMR Annual Meeting, American Society for Bone and Mineral Research; 2017. Rosario R, Baban B, Hamrick M, Isales C, Shi X. PPARγ in inflammation and aging. Denver: In ASBMR Annual Meeting, American Society for Bone and Mineral Research; 2017.
38.
Zurück zum Zitat Rosario R, Ajith A, Ding K, Elsayed R, Su Y, Horuzsko A, et al. Complex role for PPARγ in bone, inflammation and immune function in aging animals. In: In ASBMR Annual Meeting. Montreal: American Society for Bone and Mineral Research; 2018. Rosario R, Ajith A, Ding K, Elsayed R, Su Y, Horuzsko A, et al. Complex role for PPARγ in bone, inflammation and immune function in aging animals. In: In ASBMR Annual Meeting. Montreal: American Society for Bone and Mineral Research; 2018.
42.
Zurück zum Zitat Vande Berg BC, Malghem J, Lecouvet FE, Devogelaer JP, Maldague B, Houssiau FA. Fat conversion of femoral marrow in glucocorticoid-treated patients: a cross-sectional and longitudinal study with magnetic resonance imaging. Arthritis Rheum. 1999;42:1405–11.PubMedCrossRef Vande Berg BC, Malghem J, Lecouvet FE, Devogelaer JP, Maldague B, Houssiau FA. Fat conversion of femoral marrow in glucocorticoid-treated patients: a cross-sectional and longitudinal study with magnetic resonance imaging. Arthritis Rheum. 1999;42:1405–11.PubMedCrossRef
43.
Zurück zum Zitat Esche J, Shi L, Hartmann MF, Schonau E, Wudy SA, Remer T. Glucocorticoids and body fat inversely associate with bone marrow density of the distal radius in healthy youths. J Clin Endocrinol Metab. 2019;104:2250–6.PubMedCrossRef Esche J, Shi L, Hartmann MF, Schonau E, Wudy SA, Remer T. Glucocorticoids and body fat inversely associate with bone marrow density of the distal radius in healthy youths. J Clin Endocrinol Metab. 2019;104:2250–6.PubMedCrossRef
44.
Zurück zum Zitat Canalis E, Mazziotti G, Giustina A, Bilezikian JP. Glucocorticoid-induced osteoporosis: pathophysiology and therapy. Osteoporos Int. 2007;18:1319–28.PubMedCrossRef Canalis E, Mazziotti G, Giustina A, Bilezikian JP. Glucocorticoid-induced osteoporosis: pathophysiology and therapy. Osteoporos Int. 2007;18:1319–28.PubMedCrossRef
45.
Zurück zum Zitat Hartmann K, Koenen M, Schauer S, Wittig-Blaich S, Ahmad M, Baschant U, et al. Molecular actions of glucocorticoids in cartilage and bone during health, disease, and steroid therapy. Physiol Rev. 2016;96:409–47.PubMedCrossRef Hartmann K, Koenen M, Schauer S, Wittig-Blaich S, Ahmad M, Baschant U, et al. Molecular actions of glucocorticoids in cartilage and bone during health, disease, and steroid therapy. Physiol Rev. 2016;96:409–47.PubMedCrossRef
46.
Zurück zum Zitat Zhao ZY, Lu FH, Xie Y, Fu YR, Bogdan A, Touitou Y. Cortisol secretion in the elderly. Influence of age, sex and cardiovascular disease in a Chinese population. Steroids. 2003;68:551–5.PubMedCrossRef Zhao ZY, Lu FH, Xie Y, Fu YR, Bogdan A, Touitou Y. Cortisol secretion in the elderly. Influence of age, sex and cardiovascular disease in a Chinese population. Steroids. 2003;68:551–5.PubMedCrossRef
47.
Zurück zum Zitat Johar H, Emeny RT, Bidlingmaier M, Reincke M, Thorand B, Peters A, et al. Blunted diurnal cortisol pattern is associated with frailty: a cross-sectional study of 745 participants aged 65 to 90 years. J Clin Endocrinol Metab. 2014;99:E464–8.PubMedCrossRef Johar H, Emeny RT, Bidlingmaier M, Reincke M, Thorand B, Peters A, et al. Blunted diurnal cortisol pattern is associated with frailty: a cross-sectional study of 745 participants aged 65 to 90 years. J Clin Endocrinol Metab. 2014;99:E464–8.PubMedCrossRef
48.
Zurück zum Zitat Reynolds RM, Dennison EM, Walker BR, Syddall HE, Wood PJ, Andrew R, et al. Cortisol secretion and rate of bone loss in a population-based cohort of elderly men and women. Calcif Tissue Int. 2005;77:134–8.PubMedCrossRef Reynolds RM, Dennison EM, Walker BR, Syddall HE, Wood PJ, Andrew R, et al. Cortisol secretion and rate of bone loss in a population-based cohort of elderly men and women. Calcif Tissue Int. 2005;77:134–8.PubMedCrossRef
49.
Zurück zum Zitat Henneicke H, Gasparini SJ, Brennan-Speranza TC, Zhou H, Seibel MJ. Glucocorticoids and bone: local effects and systemic implications. Trends Endocrinol Metab. 2014;25:197–211.PubMedCrossRef Henneicke H, Gasparini SJ, Brennan-Speranza TC, Zhou H, Seibel MJ. Glucocorticoids and bone: local effects and systemic implications. Trends Endocrinol Metab. 2014;25:197–211.PubMedCrossRef
50.
Zurück zum Zitat Hamrick MW, McGee-Lawrence ME, Frechette DM. Fatty infiltration of skeletal muscle: mechanisms and comparisons with bone marrow adiposity. Front Endocrinol (Lausanne). 2016;7:69.CrossRef Hamrick MW, McGee-Lawrence ME, Frechette DM. Fatty infiltration of skeletal muscle: mechanisms and comparisons with bone marrow adiposity. Front Endocrinol (Lausanne). 2016;7:69.CrossRef
51.
Zurück zum Zitat Bauerle KT, Hutson I, Scheller EL, Harris CA. Glucocorticoid receptor signaling is not required for in vivo adipogenesis. Endocrinology. 2018;159:2050–61.PubMedPubMedCentralCrossRef Bauerle KT, Hutson I, Scheller EL, Harris CA. Glucocorticoid receptor signaling is not required for in vivo adipogenesis. Endocrinology. 2018;159:2050–61.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Dalle H, Garcia M, Antoine B, Boehm V, Do TTH, Buyse M, et al. Adipocyte glucocorticoid receptor deficiency promotes adipose tissue expandability and improves the metabolic profile under corticosterone exposure. Diabetes. 2019;68:305–17.PubMedCrossRef Dalle H, Garcia M, Antoine B, Boehm V, Do TTH, Buyse M, et al. Adipocyte glucocorticoid receptor deficiency promotes adipose tissue expandability and improves the metabolic profile under corticosterone exposure. Diabetes. 2019;68:305–17.PubMedCrossRef
53.
Zurück zum Zitat • Mueller KM, Hartmann K, Kaltenecker D, Vettorazzi S, Bauer M, Mauser L, et al. Adipocyte glucocorticoid receptor deficiency attenuates aging- and HFD-induced obesity and impairs the feeding-fasting transition. Diabetes. 2017;66:272–86 This study demonstrates a key role for GR-mediated GC signaling in adipocyte lipolysis. PubMedCrossRef • Mueller KM, Hartmann K, Kaltenecker D, Vettorazzi S, Bauer M, Mauser L, et al. Adipocyte glucocorticoid receptor deficiency attenuates aging- and HFD-induced obesity and impairs the feeding-fasting transition. Diabetes. 2017;66:272–86 This study demonstrates a key role for GR-mediated GC signaling in adipocyte lipolysis. PubMedCrossRef
54.
Zurück zum Zitat Desarzens S, Faresse N. Adipocyte glucocorticoid receptor has a minor contribution in adipose tissue growth. J Endocrinol. 2016;230:1–11.PubMedCrossRef Desarzens S, Faresse N. Adipocyte glucocorticoid receptor has a minor contribution in adipose tissue growth. J Endocrinol. 2016;230:1–11.PubMedCrossRef
55.
Zurück zum Zitat Shen Y, Roh HC, Kumari M, Rosen ED. Adipocyte glucocorticoid receptor is important in lipolysis and insulin resistance due to exogenous steroids, but not insulin resistance caused by high fat feeding. Mol Metab. 2017;6:1150–60.PubMedPubMedCentralCrossRef Shen Y, Roh HC, Kumari M, Rosen ED. Adipocyte glucocorticoid receptor is important in lipolysis and insulin resistance due to exogenous steroids, but not insulin resistance caused by high fat feeding. Mol Metab. 2017;6:1150–60.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Bose SK, Hutson I, Harris CA. Hepatic glucocorticoid receptor plays a greater role than adipose gr in metabolic syndrome despite renal compensation. Endocrinology. 2016;157:4943–60.PubMedPubMedCentralCrossRef Bose SK, Hutson I, Harris CA. Hepatic glucocorticoid receptor plays a greater role than adipose gr in metabolic syndrome despite renal compensation. Endocrinology. 2016;157:4943–60.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Woods CP, Hazlehurst JM, Tomlinson JW. Glucocorticoids and non-alcoholic fatty liver disease. J Steroid Biochem Mol Biol. 2015;154:94–103.PubMedCrossRef Woods CP, Hazlehurst JM, Tomlinson JW. Glucocorticoids and non-alcoholic fatty liver disease. J Steroid Biochem Mol Biol. 2015;154:94–103.PubMedCrossRef
58.
Zurück zum Zitat John K, Marino JS, Sanchez ER, Hinds TD Jr. The glucocorticoid receptor: cause of or cure for obesity? Am J Physiol Endocrinol Metab. 2016;310:E249–57.PubMedCrossRef John K, Marino JS, Sanchez ER, Hinds TD Jr. The glucocorticoid receptor: cause of or cure for obesity? Am J Physiol Endocrinol Metab. 2016;310:E249–57.PubMedCrossRef
59.
Zurück zum Zitat Tuttle LJ, Sinacore DR, Mueller MJ. Intermuscular adipose tissue is muscle specific and associated with poor functional performance. J Aging Res. 2012;2012:172957.PubMedPubMedCentralCrossRef Tuttle LJ, Sinacore DR, Mueller MJ. Intermuscular adipose tissue is muscle specific and associated with poor functional performance. J Aging Res. 2012;2012:172957.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Choi SJ, Files DC, Zhang T, Wang ZM, Messi ML, Gregory H, et al. Intramyocellular lipid and impaired myofiber contraction in normal weight and obese older adults. J Gerontol A Biol Sci Med Sci. 2016;71:557–64.PubMedCrossRef Choi SJ, Files DC, Zhang T, Wang ZM, Messi ML, Gregory H, et al. Intramyocellular lipid and impaired myofiber contraction in normal weight and obese older adults. J Gerontol A Biol Sci Med Sci. 2016;71:557–64.PubMedCrossRef
61.
Zurück zum Zitat Reinders I, Murphy RA, Brouwer IA, Visser M, Launer L, Siggeirsdottir K, et al. Muscle quality and myosteatosis: novel associations with mortality risk: the Age, Gene/Environment Susceptibility (AGES)-Reykjavik study. Am J Epidemiol. 2016;183:53–60.PubMedCrossRef Reinders I, Murphy RA, Brouwer IA, Visser M, Launer L, Siggeirsdottir K, et al. Muscle quality and myosteatosis: novel associations with mortality risk: the Age, Gene/Environment Susceptibility (AGES)-Reykjavik study. Am J Epidemiol. 2016;183:53–60.PubMedCrossRef
62.
Zurück zum Zitat Khan IM, Perrard XY, Brunner G, Lui H, Sparks LM, Smith SR, et al. Intermuscular and perimuscular fat expansion in obesity correlates with skeletal muscle T cell and macrophage infiltration and insulin resistance. Int J Obes. 2015;39:1607–18.CrossRef Khan IM, Perrard XY, Brunner G, Lui H, Sparks LM, Smith SR, et al. Intermuscular and perimuscular fat expansion in obesity correlates with skeletal muscle T cell and macrophage infiltration and insulin resistance. Int J Obes. 2015;39:1607–18.CrossRef
63.
Zurück zum Zitat Addison O, Drummond MJ, LaStayo PC, Dibble LE, Wende AR, McClain DA, et al. Intramuscular fat and inflammation differ in older adults: the impact of frailty and inactivity. J Nutr Health Aging. 2014;18:532–8.PubMedPubMedCentralCrossRef Addison O, Drummond MJ, LaStayo PC, Dibble LE, Wende AR, McClain DA, et al. Intramuscular fat and inflammation differ in older adults: the impact of frailty and inactivity. J Nutr Health Aging. 2014;18:532–8.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Seelaender MC, Batista ML. Adipose tissue inflammation and cancer cachexia: the role of steroid hormones. Horm Mol Biol Clin Invest. 2014;17:5–12. Seelaender MC, Batista ML. Adipose tissue inflammation and cancer cachexia: the role of steroid hormones. Horm Mol Biol Clin Invest. 2014;17:5–12.
65.
Zurück zum Zitat Stephens NA, Skipworth RJ, Macdonald AJ, Greig CA, Ross JA, Fearon KC. Intramyocellular lipid droplets increase with progression of cachexia in cancer patients. J Cachexia Sarcopenia Muscle. 2011;2:111–7.PubMedPubMedCentralCrossRef Stephens NA, Skipworth RJ, Macdonald AJ, Greig CA, Ross JA, Fearon KC. Intramyocellular lipid droplets increase with progression of cachexia in cancer patients. J Cachexia Sarcopenia Muscle. 2011;2:111–7.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Morgan SA, Gathercole LL, Simonet C, Hassan-Smith ZK, Bujalska I, Guest P, et al. Regulation of lipid metabolism by glucocorticoids and 11beta-HSD1 in skeletal muscle. Endocrinology. 2013;154:2374–84.PubMedCrossRef Morgan SA, Gathercole LL, Simonet C, Hassan-Smith ZK, Bujalska I, Guest P, et al. Regulation of lipid metabolism by glucocorticoids and 11beta-HSD1 in skeletal muscle. Endocrinology. 2013;154:2374–84.PubMedCrossRef
67.
Zurück zum Zitat Morgan SA, Hassan-Smith ZK, Doig CL, Sherlock M, Stewart PM, Lavery GG. Glucocorticoids and 11beta-HSD1 are major regulators of intramyocellular protein metabolism. J Endocrinol. 2016;229:277–86.PubMedPubMedCentralCrossRef Morgan SA, Hassan-Smith ZK, Doig CL, Sherlock M, Stewart PM, Lavery GG. Glucocorticoids and 11beta-HSD1 are major regulators of intramyocellular protein metabolism. J Endocrinol. 2016;229:277–86.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Chu W, Wei W, Han H, Gao Y, Liu K, Tian Y, et al. Muscle-specific downregulation of GR levels inhibits adipogenesis in porcine intramuscular adipocyte tissue. Sci Rep. 2017;7:510.PubMedPubMedCentralCrossRef Chu W, Wei W, Han H, Gao Y, Liu K, Tian Y, et al. Muscle-specific downregulation of GR levels inhibits adipogenesis in porcine intramuscular adipocyte tissue. Sci Rep. 2017;7:510.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Shimizu N, Maruyama T, Yoshikawa N, Matsumiya R, Ma Y, Ito N, et al. A muscle-liver-fat signalling axis is essential for central control of adaptive adipose remodelling. Nat Commun. 2015;6:6693.PubMedCrossRef Shimizu N, Maruyama T, Yoshikawa N, Matsumiya R, Ma Y, Ito N, et al. A muscle-liver-fat signalling axis is essential for central control of adaptive adipose remodelling. Nat Commun. 2015;6:6693.PubMedCrossRef
70.
Zurück zum Zitat de Theije CC, Schols A, Lamers WH, Ceelen JJM, van Gorp RH, Hermans JJR, et al. Glucocorticoid receptor signaling impairs protein turnover regulation in hypoxia-induced muscle atrophy in male mice. Endocrinology. 2018;159:519–34.PubMedCrossRef de Theije CC, Schols A, Lamers WH, Ceelen JJM, van Gorp RH, Hermans JJR, et al. Glucocorticoid receptor signaling impairs protein turnover regulation in hypoxia-induced muscle atrophy in male mice. Endocrinology. 2018;159:519–34.PubMedCrossRef
71.
Zurück zum Zitat Watson ML, Baehr LM, Reichardt HM, Tuckermann JP, Bodine SC, Furlow JD. A cell-autonomous role for the glucocorticoid receptor in skeletal muscle atrophy induced by systemic glucocorticoid exposure. Am J Physiol Endocrinol Metab. 2012;302:E1210–20.PubMedPubMedCentralCrossRef Watson ML, Baehr LM, Reichardt HM, Tuckermann JP, Bodine SC, Furlow JD. A cell-autonomous role for the glucocorticoid receptor in skeletal muscle atrophy induced by systemic glucocorticoid exposure. Am J Physiol Endocrinol Metab. 2012;302:E1210–20.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Hu Z, Wang H, Lee IH, Du J, Mitch WE. Endogenous glucocorticoids and impaired insulin signaling are both required to stimulate muscle wasting under pathophysiological conditions in mice. J Clin Invest. 2009;119:3059–69.PubMedPubMedCentral Hu Z, Wang H, Lee IH, Du J, Mitch WE. Endogenous glucocorticoids and impaired insulin signaling are both required to stimulate muscle wasting under pathophysiological conditions in mice. J Clin Invest. 2009;119:3059–69.PubMedPubMedCentral
73.
Zurück zum Zitat Faught E, Vijayan MM. Loss of the glucocorticoid receptor in zebrafish improves muscle glucose availability and increases growth. Am J Physiol Endocrinol Metab. 2019;316:E1093–104.PubMedCrossRefPubMedCentral Faught E, Vijayan MM. Loss of the glucocorticoid receptor in zebrafish improves muscle glucose availability and increases growth. Am J Physiol Endocrinol Metab. 2019;316:E1093–104.PubMedCrossRefPubMedCentral
74.
Zurück zum Zitat Kim HJ, Zhao H, Kitaura H, Bhattacharyya S, Brewer JA, Muglia LJ, et al. Glucocorticoids suppress bone formation via the osteoclast. J Clin Invest. 2006;116:2152–60.PubMedPubMedCentralCrossRef Kim HJ, Zhao H, Kitaura H, Bhattacharyya S, Brewer JA, Muglia LJ, et al. Glucocorticoids suppress bone formation via the osteoclast. J Clin Invest. 2006;116:2152–60.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Rauch A, Seitz S, Baschant U, Schilling AF, Illing A, Stride B, et al. Glucocorticoids suppress bone formation by attenuating osteoblast differentiation via the monomeric glucocorticoid receptor. Cell Metab. 2010;11:517–31.PubMedCrossRef Rauch A, Seitz S, Baschant U, Schilling AF, Illing A, Stride B, et al. Glucocorticoids suppress bone formation by attenuating osteoblast differentiation via the monomeric glucocorticoid receptor. Cell Metab. 2010;11:517–31.PubMedCrossRef
76.
Zurück zum Zitat Rapp AE, Hachemi Y, Kemmler J, Koenen M, Tuckermann J, Ignatius A. Induced global deletion of glucocorticoid receptor impairs fracture healing. FASEB J: official publication of the Federation of American Societies for Experimental Biology. 2018;32:2235–45.CrossRef Rapp AE, Hachemi Y, Kemmler J, Koenen M, Tuckermann J, Ignatius A. Induced global deletion of glucocorticoid receptor impairs fracture healing. FASEB J: official publication of the Federation of American Societies for Experimental Biology. 2018;32:2235–45.CrossRef
77.
Zurück zum Zitat •• Pierce JL, Ding KH, Xu J, Sharma AK, Yu K, Del Mazo Arbona N, Rodriguez-Santos Z, Bernard P, Bollag WB, Johnson MH, Hamrick MW, Begun DL, Shi XM, Isales CM, and McGee-Lawrence ME (2019) The glucocorticoid receptor in osteoprogenitors regulates bone mass and marrow fat. J Endocrinol. This study was the first to show that loss of the GR in bone promoted increased MAT deposition. •• Pierce JL, Ding KH, Xu J, Sharma AK, Yu K, Del Mazo Arbona N, Rodriguez-Santos Z, Bernard P, Bollag WB, Johnson MH, Hamrick MW, Begun DL, Shi XM, Isales CM, and McGee-Lawrence ME (2019) The glucocorticoid receptor in osteoprogenitors regulates bone mass and marrow fat. J Endocrinol. This study was the first to show that loss of the GR in bone promoted increased MAT deposition.
78.
Zurück zum Zitat Arriza JL, Weinberger C, Cerelli G, Glaser TM, Handelin BL, Housman DE, et al. Cloning of human mineralocorticoid receptor complementary DNA: structural and functional kinship with the glucocorticoid receptor. Science. 1987;237:268–75.PubMedCrossRef Arriza JL, Weinberger C, Cerelli G, Glaser TM, Handelin BL, Housman DE, et al. Cloning of human mineralocorticoid receptor complementary DNA: structural and functional kinship with the glucocorticoid receptor. Science. 1987;237:268–75.PubMedCrossRef
79.
Zurück zum Zitat Fumoto T, Ishii KA, Ito M, Berger S, Schutz G, Ikeda K. Mineralocorticoid receptor function in bone metabolism and its role in glucocorticoid-induced osteopenia. Biochem Biophys Res Commun. 2014;447:407–12.PubMedCrossRef Fumoto T, Ishii KA, Ito M, Berger S, Schutz G, Ikeda K. Mineralocorticoid receptor function in bone metabolism and its role in glucocorticoid-induced osteopenia. Biochem Biophys Res Commun. 2014;447:407–12.PubMedCrossRef
80.
Zurück zum Zitat Hinds TD Jr, Ramakrishnan S, Cash HA, Stechschulte LA, Heinrich G, Najjar SM, et al. Discovery of glucocorticoid receptor-beta in mice with a role in metabolism. Mol Endocrinol. 2010;24:1715–27.PubMedPubMedCentralCrossRef Hinds TD Jr, Ramakrishnan S, Cash HA, Stechschulte LA, Heinrich G, Najjar SM, et al. Discovery of glucocorticoid receptor-beta in mice with a role in metabolism. Mol Endocrinol. 2010;24:1715–27.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Oakley RH, Cidlowski JA. The biology of the glucocorticoid receptor: new signaling mechanisms in health and disease. J Allergy Clin Immunol. 2013;132:1033–44.PubMedPubMedCentralCrossRef Oakley RH, Cidlowski JA. The biology of the glucocorticoid receptor: new signaling mechanisms in health and disease. J Allergy Clin Immunol. 2013;132:1033–44.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Beavan S, Horner A, Bord S, Ireland D, Compston J. Colocalization of glucocorticoid and mineralocorticoid receptors in human bone. J Bone Miner Res Off J Am Soc Bone Miner Res. 2001;16:1496–504.CrossRef Beavan S, Horner A, Bord S, Ireland D, Compston J. Colocalization of glucocorticoid and mineralocorticoid receptors in human bone. J Bone Miner Res Off J Am Soc Bone Miner Res. 2001;16:1496–504.CrossRef
Metadaten
Titel
Endogenous Glucocorticoid Signaling in the Regulation of Bone and Marrow Adiposity: Lessons from Metabolism and Cross Talk in Other Tissues
verfasst von
Anuj K. Sharma
Xingming Shi
Carlos M. Isales
Meghan E. McGee-Lawrence
Publikationsdatum
20.11.2019
Verlag
Springer US
Erschienen in
Current Osteoporosis Reports / Ausgabe 6/2019
Print ISSN: 1544-1873
Elektronische ISSN: 1544-2241
DOI
https://doi.org/10.1007/s11914-019-00554-6

Weitere Artikel der Ausgabe 6/2019

Current Osteoporosis Reports 6/2019 Zur Ausgabe

Quality of Care in Osteoporosis (S Silverman and J Curtis, Section Editors)

Pragmatic Clinical Trials in Osteoporosis

Skeletal Biology and Regulation (M Forwood and A Robling, Section Editors)

Monocyte Chemoattractant Protein-1 (MCP-1/CCL2) Drives Activation of Bone Remodelling and Skeletal Metastasis

Osteocytes (J Klein-Nulend, Section editor)

Aging and Mechanoadaptive Responsiveness of Bone

Bone Marrow and Adipose Tissue (G Duque and B Lecka-Czernik, Section Editors)

Bone Marrow Adipose Tissue Quantification by Imaging

Quality of Care in Osteoporosis (S Silverman and J Curtis, Section Editors)

Bone Health TeleECHO: a Force Multiplier to Improve the Care of Skeletal Diseases in Underserved Communities

Bone Marrow and Adipose Tissue (G Duque and B Lecka-Czernik, Section Editors)

Insulin Signaling in Bone Marrow Adipocytes

Arthropedia

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

Update Orthopädie und Unfallchirurgie

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