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

01-12-2019 | Magnetic Resonance Imaging | Bone Marrow and Adipose Tissue (G Duque and B Lecka-Czernik, Section Editors)

Bone Marrow Adipose Tissue Quantification by Imaging

Authors: Ebrahim Bani Hassan, Ali Ghasem-Zadeh, Mahdi Imani, Numan Kutaiba, David K. Wright, Tara Sepehrizadeh, Gustavo Duque

Published in: Current Osteoporosis Reports | Issue 6/2019

Login to get access

Abstract

Purpose of Review

The significance and roles of marrow adipose tissue (MAT) are increasingly known, and it is no more considered a passive fat storage but a tissue with significant paracrine and endocrine activities that can cause lipotoxicity and inflammation.

Recent Findings

Changes in the MAT volume and fatty acid composition appear to drive bone and hematopoietic marrow deterioration, and studying it may open new horizons to predict bone fragility and anemia development. MAT has the potential to negatively impact bone volume and strength through several mechanisms that are partially described by inflammaging and lipotoxicity terminology.

Summary

Evidence indicates paramount importance of MAT in age-associated decline of bone and red marrow structure and function. Currently, MAT measurement is being tested and validated by several techniques. However, purpose-specific adaptation of existing imaging technologies and, more importantly, development of new modalities to quantitatively measure MAT are yet to be done.
Literature
1.
go back to reference •• Bani Hassan E, Demontiero O, Vogrin S, Ng A, Duque G. Marrow adipose tissue in older men: association with visceral and subcutaneous fat, bone volume, metabolism, and inflammation. Calcif Tissue Int. 2018;103(2):164–74. https://doi.org/10.1007/s00223-018-0412-6This paper provides important evidences that MAT expansion in older men coincides with bone and hematopoietic marrow atrophy. Also, this reference provides evidences that most of the inflammatory cytokines produces by MAT are not released into systemic circulation, and may gain high local concentrations and severely impact bone and marrow health. Further, this paper is one of the limited number of publications that uses single energy CT scan for assesing bone, MAT and red marrow volumes. CrossRefPubMed •• Bani Hassan E, Demontiero O, Vogrin S, Ng A, Duque G. Marrow adipose tissue in older men: association with visceral and subcutaneous fat, bone volume, metabolism, and inflammation. Calcif Tissue Int. 2018;103(2):164–74. https://​doi.​org/​10.​1007/​s00223-018-0412-6This paper provides important evidences that MAT expansion in older men coincides with bone and hematopoietic marrow atrophy. Also, this reference provides evidences that most of the inflammatory cytokines produces by MAT are not released into systemic circulation, and may gain high local concentrations and severely impact bone and marrow health. Further, this paper is one of the limited number of publications that uses single energy CT scan for assesing bone, MAT and red marrow volumes. CrossRefPubMed
3.
go back to reference Schwartz RS, Shuman WP, Bradbury VL, Cain KC, Fellingham GW, Beard JC, et al. Body fat distribution in healthy young and older men. J Gerontol. 1990;45(6):M181–5.CrossRefPubMed Schwartz RS, Shuman WP, Bradbury VL, Cain KC, Fellingham GW, Beard JC, et al. Body fat distribution in healthy young and older men. J Gerontol. 1990;45(6):M181–5.CrossRefPubMed
4.
go back to reference • Elbaz A, Wu X, Rivas D, Gimble JM, Duque G. Inhibition of fatty acid biosynthesis prevents adipocyte lipotoxicity on human osteoblasts in vitro. J Cell Mol Med. 2010;14(4):982–91. https://doi.org/10.1111/j.1582-4934.2009.00751.xThis paper provides first evidences that toxic effects of MAT-derived fatty acids on osteoblastss can be prevented by inhibiting the biosynthesis of such fatty acids, establishing a possible causality association. CrossRefPubMed • Elbaz A, Wu X, Rivas D, Gimble JM, Duque G. Inhibition of fatty acid biosynthesis prevents adipocyte lipotoxicity on human osteoblasts in vitro. J Cell Mol Med. 2010;14(4):982–91. https://​doi.​org/​10.​1111/​j.​1582-4934.​2009.​00751.​xThis paper provides first evidences that toxic effects of MAT-derived fatty acids on osteoblastss can be prevented by inhibiting the biosynthesis of such fatty acids, establishing a possible causality association. CrossRefPubMed
7.
go back to reference Kirkland JL, Tchkonia T, Pirtskhalava T, Han J, Karagiannides I. Adipogenesis and aging: does aging make fat go MAD? Exp Gerontol. 2002;37(6):757–67.CrossRefPubMed Kirkland JL, Tchkonia T, Pirtskhalava T, Han J, Karagiannides I. Adipogenesis and aging: does aging make fat go MAD? Exp Gerontol. 2002;37(6):757–67.CrossRefPubMed
13.
go back to reference •• Bani Hassan E, Alderghaffar M, Wauquier F, Coxam V, Demontiero O, Vogrin S, et al. The effects of dietary fatty acids on bone, hematopoietic marrow and marrow adipose tissue in a murine model of senile osteoporosis. Aging (Albany NY). 2019;11(18):7938-7947. doi: 10.18632/aging.102299. This recent publication provides important evidences that the negative asciation netween MAT vs bone and hematopoietic marrow seen in men (Ref 1), also occurs in a progeria mouse model. Importantly, it provides evidences that abbrogating lipotoxicity by provision of omega 3 fatty acids may prevent age-associated loss of bone and red marrow atrophy in this model. •• Bani Hassan E, Alderghaffar M, Wauquier F, Coxam V, Demontiero O, Vogrin S, et al. The effects of dietary fatty acids on bone, hematopoietic marrow and marrow adipose tissue in a murine model of senile osteoporosis. Aging (Albany NY). 2019;11(18):7938-7947. doi: 10.18632/aging.102299.  This recent publication provides important evidences that the negative asciation netween MAT vs bone and hematopoietic marrow seen in men (Ref 1), also occurs in a progeria mouse model. Importantly, it provides evidences that abbrogating lipotoxicity by provision of omega 3 fatty acids may prevent age-associated loss of bone and red marrow atrophy in this model.
33.
go back to reference Malkov S, Cawthon PM, Peters KW, Cauley JA, Murphy RA, Visser M, et al. Hip fractures risk in older men and women associated with DXA-derived measures of thigh subcutaneous fat thickness, cross-sectional muscle area, and muscle density. J Bone Miner Res. 2015;30(8):1414–21. https://doi.org/10.1002/jbmr.2469.CrossRefPubMed Malkov S, Cawthon PM, Peters KW, Cauley JA, Murphy RA, Visser M, et al. Hip fractures risk in older men and women associated with DXA-derived measures of thigh subcutaneous fat thickness, cross-sectional muscle area, and muscle density. J Bone Miner Res. 2015;30(8):1414–21. https://​doi.​org/​10.​1002/​jbmr.​2469.CrossRefPubMed
34.
36.
go back to reference Meunier P, Aaron J, Edouard C, Vignon G. Osteoporosis and the replacement of cell populations of the marrow by adipose tissue. A quantitative study of 84 iliac bone biopsies. Clin Orthop Relat Res. 1971;80:147–54.CrossRefPubMed Meunier P, Aaron J, Edouard C, Vignon G. Osteoporosis and the replacement of cell populations of the marrow by adipose tissue. A quantitative study of 84 iliac bone biopsies. Clin Orthop Relat Res. 1971;80:147–54.CrossRefPubMed
37.
go back to reference Rosen CJ, Ackert-Bicknell C, Rodriguez JP, Pino AM. Marrow fat and the bone microenvironment: developmental, functional, and pathological implications. Crit Rev Eukaryot Gene Expr. 2009;19(2):109–24.CrossRefPubMedPubMedCentral Rosen CJ, Ackert-Bicknell C, Rodriguez JP, Pino AM. Marrow fat and the bone microenvironment: developmental, functional, and pathological implications. Crit Rev Eukaryot Gene Expr. 2009;19(2):109–24.CrossRefPubMedPubMedCentral
41.
go back to reference Pelegrino Bastos Maues NH, Fattori Alves AF, Menegatti Pavan AL, Marrone Ribeiro S, Yamashita S, Petean Trindade A, et al. Abdomen-pelvis computed tomography protocol optimization: an image quality and dose assessment. Radiat Prot Dosim. 2019;184(1):66–72. https://doi.org/10.1093/rpd/ncy181.CrossRef Pelegrino Bastos Maues NH, Fattori Alves AF, Menegatti Pavan AL, Marrone Ribeiro S, Yamashita S, Petean Trindade A, et al. Abdomen-pelvis computed tomography protocol optimization: an image quality and dose assessment. Radiat Prot Dosim. 2019;184(1):66–72. https://​doi.​org/​10.​1093/​rpd/​ncy181.CrossRef
43.
go back to reference Chowdhury B, Sjöström L, Alpsten M, Kostanty J, Kvist H, Löfgren R. A multicompartment body composition technique based on computerized tomography. Int J Obes Relat Metab Disord. 1994;18(4):219–34.PubMed Chowdhury B, Sjöström L, Alpsten M, Kostanty J, Kvist H, Löfgren R. A multicompartment body composition technique based on computerized tomography. Int J Obes Relat Metab Disord. 1994;18(4):219–34.PubMed
48.
go back to reference Lareida A, Beckmann F, Schrott-Fischer A, Glueckert R, Freysinger W, Müller B. High-resolution X-ray tomography of the human inner ear: synchrotron radiation-based study of nerve fibre bundles, membranes and ganglion cells. J Microsc. 2009;234(1):95–102.CrossRefPubMed Lareida A, Beckmann F, Schrott-Fischer A, Glueckert R, Freysinger W, Müller B. High-resolution X-ray tomography of the human inner ear: synchrotron radiation-based study of nerve fibre bundles, membranes and ganglion cells. J Microsc. 2009;234(1):95–102.CrossRefPubMed
50.
go back to reference Hardouin P, Marie PJ, Rosen CJ. New insights into bone marrow adipocytes: report from the first European meeting on bone marrow adiposity (BMA 2015). Bone. 2016;93:212–5.CrossRefPubMed Hardouin P, Marie PJ, Rosen CJ. New insights into bone marrow adipocytes: report from the first European meeting on bone marrow adiposity (BMA 2015). Bone. 2016;93:212–5.CrossRefPubMed
51.
go back to reference Doucette CR, Horowitz MC, Berry R, MacDougald OA, Anunciado-Koza R, Koza RA, et al. A high fat diet increases bone marrow adipose tissue (MAT) but does not alter trabecular or cortical bone mass in C57BL/6J mice. J Cell Physiol. 2015;230(9):2032–7.CrossRefPubMedPubMedCentral Doucette CR, Horowitz MC, Berry R, MacDougald OA, Anunciado-Koza R, Koza RA, et al. A high fat diet increases bone marrow adipose tissue (MAT) but does not alter trabecular or cortical bone mass in C57BL/6J mice. J Cell Physiol. 2015;230(9):2032–7.CrossRefPubMedPubMedCentral
52.
go back to reference Cawthorn WP, Scheller EL, Learman BS, Parlee SD, Simon BR, Mori H, et al. Bone marrow adipose tissue is an endocrine organ that contributes to increased circulating adiponectin during caloric restriction. Cell Metab. 2014;20(2):368–75.CrossRefPubMedPubMedCentral Cawthorn WP, Scheller EL, Learman BS, Parlee SD, Simon BR, Mori H, et al. Bone marrow adipose tissue is an endocrine organ that contributes to increased circulating adiponectin during caloric restriction. Cell Metab. 2014;20(2):368–75.CrossRefPubMedPubMedCentral
53.
go back to reference Simon BR, Learman BS, Parlee SD, Scheller EL, Mori H, Cawthorn WP, et al. Sweet taste receptor deficient mice have decreased adiposity and increased bone mass. PLoS One. 2014;9(1):e86454.CrossRefPubMedPubMedCentral Simon BR, Learman BS, Parlee SD, Scheller EL, Mori H, Cawthorn WP, et al. Sweet taste receptor deficient mice have decreased adiposity and increased bone mass. PLoS One. 2014;9(1):e86454.CrossRefPubMedPubMedCentral
54.
go back to reference Lecka-Czernik B, Stechschulte LA, Czernik PJ, Sherman SB, Huang S, Krings A. Marrow adipose tissue: skeletal location, sexual dimorphism, and response to sex steroid deficiency. Front Endocrinol. 2017;8:188.CrossRef Lecka-Czernik B, Stechschulte LA, Czernik PJ, Sherman SB, Huang S, Krings A. Marrow adipose tissue: skeletal location, sexual dimorphism, and response to sex steroid deficiency. Front Endocrinol. 2017;8:188.CrossRef
55.
go back to reference Styner M, Thompson WR, Galior K, Uzer G, Wu X, Kadari S, et al. Bone marrow fat accumulation accelerated by high fat diet is suppressed by exercise. Bone. 2014;64:39–46.CrossRefPubMedPubMedCentral Styner M, Thompson WR, Galior K, Uzer G, Wu X, Kadari S, et al. Bone marrow fat accumulation accelerated by high fat diet is suppressed by exercise. Bone. 2014;64:39–46.CrossRefPubMedPubMedCentral
56.
go back to reference Styner M, Pagnotti GM, McGrath C, Wu X, Sen B, Uzer G, et al. Exercise decreases marrow adipose tissue through ß-oxidation in obese running mice. J Bone Miner Res. 2017;32(8):1692–702.CrossRefPubMed Styner M, Pagnotti GM, McGrath C, Wu X, Sen B, Uzer G, et al. Exercise decreases marrow adipose tissue through ß-oxidation in obese running mice. J Bone Miner Res. 2017;32(8):1692–702.CrossRefPubMed
57.
go back to reference Sulston RJ, Learman BS, Zhang B, Scheller EL, Parlee SD, Simon BR, et al. Increased circulating adiponectin in response to thiazolidinediones: investigating the role of bone marrow adipose tissue. Front Endocrinol. 2016;7:128.CrossRef Sulston RJ, Learman BS, Zhang B, Scheller EL, Parlee SD, Simon BR, et al. Increased circulating adiponectin in response to thiazolidinediones: investigating the role of bone marrow adipose tissue. Front Endocrinol. 2016;7:128.CrossRef
58.
go back to reference 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(2):1156–67.CrossRefPubMed 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(2):1156–67.CrossRefPubMed
59.
go back to reference Pagnotti GM, Styner M. Exercise regulation of marrow adipose tissue. Front Endocrinol. 2016;7:94.CrossRef Pagnotti GM, Styner M. Exercise regulation of marrow adipose tissue. Front Endocrinol. 2016;7:94.CrossRef
60.
go back to reference Johnson JT, Hansen MS, Wu I, Healy LJ, Johnson CR, Jones GM, et al. Virtual histology of transgenic mouse embryos for high-throughput phenotyping. PLoS Genet. 2006;2(4):e61.CrossRefPubMedPubMedCentral Johnson JT, Hansen MS, Wu I, Healy LJ, Johnson CR, Jones GM, et al. Virtual histology of transgenic mouse embryos for high-throughput phenotyping. PLoS Genet. 2006;2(4):e61.CrossRefPubMedPubMedCentral
61.
go back to reference Bentley MD, Jorgensen SM, Lerman LO, Ritman EL, Romero JC. Visualization of three-dimensional nephron structure with microcomputed tomography. Anat Rec Adv Integr Anat Evol Biol. 2007;290(3):277–83.CrossRef Bentley MD, Jorgensen SM, Lerman LO, Ritman EL, Romero JC. Visualization of three-dimensional nephron structure with microcomputed tomography. Anat Rec Adv Integr Anat Evol Biol. 2007;290(3):277–83.CrossRef
62.
go back to reference Metscher BD. X-ray microtomographic imaging of intact vertebrate embryos. Cold Spring Harb Protoc. 2011;2011(12) pdb. prot067033.CrossRefPubMed Metscher BD. X-ray microtomographic imaging of intact vertebrate embryos. Cold Spring Harb Protoc. 2011;2011(12) pdb. prot067033.CrossRefPubMed
63.
go back to reference Litzlbauer HD, Neuhaeuser C, Moell A, Greschus S, Breithecker A, Franke FE, et al. Three-dimensional imaging and morphometric analysis of alveolar tissue from microfocal X-ray-computed tomography. Am J Phys Lung Cell Mol Phys. 2006;291(3):L535–L45. Litzlbauer HD, Neuhaeuser C, Moell A, Greschus S, Breithecker A, Franke FE, et al. Three-dimensional imaging and morphometric analysis of alveolar tissue from microfocal X-ray-computed tomography. Am J Phys Lung Cell Mol Phys. 2006;291(3):L535–L45.
64.
go back to reference Ritman EL. Molecular imaging in small animals—roles for micro-CT. J Cell Biochem. 2002;87(S39):116–24.CrossRef Ritman EL. Molecular imaging in small animals—roles for micro-CT. J Cell Biochem. 2002;87(S39):116–24.CrossRef
65.
go back to reference Henning AL, Jiang MX, Yalcin HC, Butcher JT. Quantitative three-dimensional imaging of live avian embryonic morphogenesis via micro-computed tomography. Dev Dyn. 2011;240(8):1949–57.CrossRefPubMedPubMedCentral Henning AL, Jiang MX, Yalcin HC, Butcher JT. Quantitative three-dimensional imaging of live avian embryonic morphogenesis via micro-computed tomography. Dev Dyn. 2011;240(8):1949–57.CrossRefPubMedPubMedCentral
66.
go back to reference Scheller EL, Troiano N, VanHoutan JN, Bouxsein MA, Fretz JA, Xi Y, et al. Use of osmium tetroxide staining with microcomputerized tomography to visualize and quantify bone marrow adipose tissue in vivo. Methods Enzymol. Elsevier. 2014;537:123–39.CrossRefPubMedPubMedCentral Scheller EL, Troiano N, VanHoutan JN, Bouxsein MA, Fretz JA, Xi Y, et al. Use of osmium tetroxide staining with microcomputerized tomography to visualize and quantify bone marrow adipose tissue in vivo. Methods Enzymol. Elsevier. 2014;537:123–39.CrossRefPubMedPubMedCentral
67.
go back to reference Scheller EL, Khoury B, Moller KL, Wee NK, Khandaker S, Kozloff KM, et al. Changes in skeletal integrity and marrow adiposity during high-fat diet and after weight loss. Front Endocrinol. 2016;7:102.CrossRef Scheller EL, Khoury B, Moller KL, Wee NK, Khandaker S, Kozloff KM, et al. Changes in skeletal integrity and marrow adiposity during high-fat diet and after weight loss. Front Endocrinol. 2016;7:102.CrossRef
68.
69.
go back to reference de Crespigny A, Bou-Reslan H, Nishimura MC, Phillips H, Carano RA, D’Arceuil HE. 3D micro-CT imaging of the postmortem brain. J Neurosci Methods. 2008;171(2):207–13.CrossRefPubMedPubMedCentral de Crespigny A, Bou-Reslan H, Nishimura MC, Phillips H, Carano RA, D’Arceuil HE. 3D micro-CT imaging of the postmortem brain. J Neurosci Methods. 2008;171(2):207–13.CrossRefPubMedPubMedCentral
70.
go back to reference Hardouin P, Rharass T, Lucas S. Bone marrow adipose tissue: to be or not to be a typical adipose tissue? Front Endocrinol. 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. 2016;7:85.CrossRef
71.
go back to reference 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.CrossRefPubMedPubMedCentral 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.CrossRefPubMedPubMedCentral
72.
go back to reference Alecci M, Collins CM, Smith MB, Jezzard P. Radio frequency magnetic field mapping of a 3 Tesla birdcage coil: experimental and theoretical dependence on sample properties. Magn Reson Med. 2001;46(2):379–85.CrossRefPubMed Alecci M, Collins CM, Smith MB, Jezzard P. Radio frequency magnetic field mapping of a 3 Tesla birdcage coil: experimental and theoretical dependence on sample properties. Magn Reson Med. 2001;46(2):379–85.CrossRefPubMed
74.
go back to reference Porter BA, Shields AF, Olson DO. Magnetic resonance imaging of bone marrow disorders. Radiol Clin N Am. 1986;24(2):269–89.PubMed Porter BA, Shields AF, Olson DO. Magnetic resonance imaging of bone marrow disorders. Radiol Clin N Am. 1986;24(2):269–89.PubMed
75.
go back to reference Baum T, Yap SP, Karampinos DC, Nardo L, Kuo D, Burghardt AJ, et al. Does vertebral bone marrow fat content correlate with abdominal adipose tissue, lumbar spine bone mineral density, and blood biomarkers in women with type 2 diabetes mellitus? J Magn Reson Imaging. 2012;35(1):117–24. https://doi.org/10.1002/jmri.22757.CrossRefPubMed Baum T, Yap SP, Karampinos DC, Nardo L, Kuo D, Burghardt AJ, et al. Does vertebral bone marrow fat content correlate with abdominal adipose tissue, lumbar spine bone mineral density, and blood biomarkers in women with type 2 diabetes mellitus? J Magn Reson Imaging. 2012;35(1):117–24. https://​doi.​org/​10.​1002/​jmri.​22757.CrossRefPubMed
78.
go back to reference Moulopoulos LA, Dimopoulos MA. Magnetic resonance imaging of the bone marrow in hematologic malignancies. Blood. 1997;90(6):2127–47.CrossRefPubMed Moulopoulos LA, Dimopoulos MA. Magnetic resonance imaging of the bone marrow in hematologic malignancies. Blood. 1997;90(6):2127–47.CrossRefPubMed
79.
go back to reference • Karampinos DC, Ruschke S, Dieckmeyer M, Diefenbach M, Franz D, Gersing AS, et al. Quantitative MRI and spectroscopy of bone marrow. J Magn Reson Imaging. 2018;47(2):332–53. https://doi.org/10.1002/jmri.25769This paper is one of the most recent and important publicaitons that discusses the quantitiative MRI approach towards MAT imaging. CrossRefPubMed • Karampinos DC, Ruschke S, Dieckmeyer M, Diefenbach M, Franz D, Gersing AS, et al. Quantitative MRI and spectroscopy of bone marrow. J Magn Reson Imaging. 2018;47(2):332–53. https://​doi.​org/​10.​1002/​jmri.​25769This paper is one of the most recent and important publicaitons that discusses the quantitiative MRI approach towards MAT imaging. CrossRefPubMed
85.
go back to reference Schick F, Bongers H, Jung WI, Skalej M, Lutz O, Claussen CD. Volume-selective proton MRS in vertebral bodies. Magn Reson Med. 1992;26(2):207–17.CrossRefPubMed Schick F, Bongers H, Jung WI, Skalej M, Lutz O, Claussen CD. Volume-selective proton MRS in vertebral bodies. Magn Reson Med. 1992;26(2):207–17.CrossRefPubMed
91.
98.
go back to reference Al Saedi A, Bani Hassan E, Duque G. The diagnostic role of fat in osteosarcopenia. J Lab Precis Med 2019;4:7.CrossRef Al Saedi A, Bani Hassan E, Duque G. The diagnostic role of fat in osteosarcopenia. J Lab Precis Med 2019;4:7.CrossRef
99.
go back to reference •• Duque G, Li W, Adams M, Xu S, Phipps R. Effects of risedronate on bone marrow adipocytes in postmenopausal women. Osteoporos Int. 2011;22(5):1547–53. https://doi.org/10.1007/s00198-010-1353-8Bisphosphonate therapy decreases MAT volume about 10 times more than it increases bone volume; which is the very much ignored aspect of how MAT impacts bone health. Interestingly, increased bone volumes only explain a small fraction of fracture risk decline that may be predicted by MAT decline—if reliable and affordable measurement tools are developed. CrossRefPubMed •• Duque G, Li W, Adams M, Xu S, Phipps R. Effects of risedronate on bone marrow adipocytes in postmenopausal women. Osteoporos Int. 2011;22(5):1547–53. https://​doi.​org/​10.​1007/​s00198-010-1353-8Bisphosphonate therapy decreases MAT volume about 10 times more than it increases bone volume; which is the very much ignored aspect of how MAT impacts bone health. Interestingly, increased bone volumes only explain a small fraction of fracture risk decline that may be predicted by MAT decline—if reliable and affordable measurement tools are developed. CrossRefPubMed
Metadata
Title
Bone Marrow Adipose Tissue Quantification by Imaging
Authors
Ebrahim Bani Hassan
Ali Ghasem-Zadeh
Mahdi Imani
Numan Kutaiba
David K. Wright
Tara Sepehrizadeh
Gustavo Duque
Publication date
01-12-2019
Publisher
Springer US
Published in
Current Osteoporosis Reports / Issue 6/2019
Print ISSN: 1544-1873
Electronic ISSN: 1544-2241
DOI
https://doi.org/10.1007/s11914-019-00539-5

Other articles of this Issue 6/2019

Current Osteoporosis Reports 6/2019 Go to the issue

Osteocytes (J Klein-Nulend, Section editor)

Aging and Mechanoadaptive Responsiveness of Bone

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

Quality Improvement Initiatives in Fragility Fracture Care and Prevention

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

Effects of Diabetes on Bone Material Properties

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

Insulin Signaling in Bone Marrow Adipocytes