fbpx
Wikipedia

Bone marrow adipose tissue

Bone marrow adipose tissue (BMAT), sometimes referred to as marrow adipose tissue (MAT), is a type of fat deposit in bone marrow. It increases in states of low bone density -osteoporosis,[1][2] anorexia nervosa/caloric restriction,[3][4] skeletal unweighting such as that which occurs in space travel,[5][6] and anti-diabetes therapies.[7] BMAT decreases in anaemia, leukaemia, and hypertensive heart failure; in response to hormones such as oestrogen, leptin, and growth hormone; with exercise-induced weight loss or bariatric surgery; in response to chronic cold exposure; and in response to pharmacological agents such as bisphosphonates, teriparatide, and metformin.[8]

Bone marrow adipose tissue
Details
SystemMusculoskeletal (or locomotor)
Identifiers
Latinadipose ossium medulla
Anatomical terminology
[edit on Wikidata]
Bone marrow adipocytes are derived from mesenchymal stem cell (MSC) differentiation.

Anatomy edit

Bone marrow adipocytes (BMAds)[9] originate from mesenchymal stem cell (MSC) progenitors that also give rise to osteoblasts, among other cell types.[10] Thus, it is thought that BMAT results from preferential MSC differentiation into the adipocyte, rather than osteoblast, lineage in the setting of osteoporosis.[11] Since BMAT is increased in the setting of obesity[12][13][14] and is suppressed by endurance exercise,[15][12][16][17] or vibration,[18] it is likely that BMAT physiology, in the setting of mechanical input/exercise, approximates that of white adipose tissue (WAT).

Physiology edit

Exercise regulation edit

The first study to demonstrate exercise regulation of BMAT in rodents was published in 2014;[12] Now, exercise regulation of BMAT has been confirmed in a human,[19] adding clinical importance. Several studies demonstrated exercise reduction of BMAT which occurs along with an increase in bone quantity.[17][15][16][20] Since exercise increases bone quantity, reduces BMAT and increases expression of markers of fatty acid oxidation in bone, BMAT is thought to be providing needed fuel for exercise-induced bone formation or anabolism.[16] A notable exception occurs in the setting of caloric restriction: exercise suppression of BMAT does not yield an increase in bone formation and even appears to cause bone loss.[4][21][20] Indeed, energy availability appears to be a factor in the ability of exercise to regulate BMAT.[4] Another exception occurs in lipodystrophy, a condition with reduced overall adipose stores: exercise- induced anabolism is possible, even with minimal BMAT stores.[22]

Relationships to other types of fat edit

BMAT has been reported to have qualities of both white and brown fat.[23] However, more-recent functional and -omics studies have shown that BMAT is a unique adipose depot that is molecularly and functionally distinct to WAT or BAT.[24][25][26][27] Subcutaneous white fat contain excess energy, indicating a clear evolutionary advantage during times of scarcity. WAT is also the source of adipokines and inflammatory markers which have both positive (e.g., adiponectin)[28] and negative[29] effects on metabolic and cardiovascular endpoints. Visceral abdominal fat (VAT) is a distinct type of WAT that is "proportionally associated with negative metabolic and cardiovascular morbidity",[30] regenerates cortisol,[31] and recently has been tied to decreased bone formation[32][33] Both types of WAT substantially differ from brown adipose tissue (BAT) as by a group of proteins that help BAT's thermogenic role.[34] BMAT, by its "specific marrow location, and its adipocyte origin from at least LepR+ marrow MSC is separated from non-bone fat storage by larger expression of bone transcription factors",[35] and likely indicates a different fat phenotype.[36] Recently, BMAT was noted to "produce a greater proportion of adiponectin – an adipokine associated with improved metabolism – than WAT",[37] suggesting an endocrine function for this depot, akin, but different, from that of WAT.

Impact on bone health edit

BMAT increases in states of bone fragility. BMAT is thought to result from preferential MSC differentiation into an adipocyte, rather than osteoblast lineage in osteoporosis[11][20] based on the inverse relationship between bone and BMAT in bone-fragile osteoporotic states. An increase in BMAT is noted in osteoporosis clinical studies measured by MR spectroscopy.[38][39][40] Estrogen therapy in postmenopausal osteoporosis reduces BMAT.[41] Antiresorptive therapies like risedronate or zoledronate also decrease BMAT while increasing bone density, supporting an inverse relationship between bone quantity and BMAT. During aging, bone quantity declines[42][43] and fat redistributes from subcutaneous to ectopic sites such as bone marrow, muscle, and liver.[44] Aging is associated with lower osteogenic and greater adipogenic biasing of MSC.[45] This aging-related biasing of MSC away from osteoblast lineage may represent higher basal PPARγ expression[46] or decreased Wnt10b.[47][48][49] Thus, bone fragility, osteoporosis, and osteoporotic fractures are thought to be linked to mechanisms which promote BMAT accumulation.[citation needed]

Maintenance of hematopoietic stem cells edit

BMAds secrete factors that promote HSC renewal in most bones.[50]

Hematopoietic cells (also known as blood cells) reside in the bone marrow along with BMAds. These hematopoietic cells are derived from hematopoietic stem cells (HSC) which give rise to diverse cells: cells of the blood, immune system, as well as cells that break down bone (osteoclasts). HSC renewal occurs in the marrow stem cell niche, a microenvironment that contains cells and secreted factors that promote appropriate renewal and differentiation of HSC. The study of the stem cell niche is relevant to the field of oncology in order to improve therapy for multiple hematologic cancers. As such cancers are often treated with bone marrow transplantation, there is interest in improving the renewal of HSC.[citation needed]

Measurement edit

In order to understand the physiology of BMAT, various analytic methods have been applied. BMAds are difficult to isolate and quantify because they are interspersed with bony and hematopoietic elements. Until recently, qualitative measurements of BMAT have relied on bone histology,[51][52] which is subject to site selection bias and cannot adequately quantify the volume of fat in the marrow. Nevertheless, histological techniques and fixation make possible visualization of BMAT, quantification of BMAd size, and BMAT's association with the surrounding endosteum, milieu of cells, and secreted factors.[53][54][55]

Recent advances in cell surface and intracellular marker identification and single-cell analyses led to greater resolution and high-throughput ex-vivo quantification. Flow cytometric quantification can be used to purify adipocytes from the stromal vascular fraction of most fat depots.[56] Early research with such machinery cited adipocytes as too large and fragile for cytometer-based purification, rendering them susceptible to lysis; however, recent advances have been made to mitigate this;[57] nevertheless, this methodology continues to pose technical challenges[58] and is inaccessible to much of the research community.

To improve quantification of BMAT, novel imaging techniques have been developed as a means to visualize and quantify BMAT. Although proton magnetic resonance spectroscopy (1H-MRS) has been used with success to quantify vertebral BMAT in humans,[59] it is difficult to employ in laboratory animals.[60] Magnetic resonance imaging (MRI) provides BMAT assessment in the vertebral skeleton[61] in conjunction with μCT-based marrow density measurements.[62] A volumetric method to identify, quantify, and localize BMAT in rodent bone has been recently developed, requiring osmium staining of bones and μCT imaging,[63] followed by advanced image analysis of osmium-bound lipid volume (in mm3) relative to bone volume.[12][16][15] This technique provides reproducible quantification and visualization of BMAT, enabling the ability to consistently quantify changes in BMAT with diet, exercise, and agents that constrain precursor lineage allocation. Although the osmium method is quantitatively precise, osmium is toxic and cannot be compared across batched experiments. Recently, researchers developed and validated[16] a 9.4T MRI scanner technique that allows localization and volumetric (3D) quantification that can be compared across experiments, as in.[4]

Several studies have also analysed BMAT function in vivo using positron emission tomography - computed tomography (PET-CT) combined with the tracer 18F-Fluorodeoxyglucose (FDG). This allows glucose uptake, a measure of metabolic activity, to be quantified in living organisms, including humans. Two recent studies found that, unlike brown adipose tissue, BMAT does not increase glucose uptake in response to cold exposure, demonstrating that BMAT is functionally distinct from BAT.[24][64] The full extent of BMAT's impact on systemic metabolic homeostasis remains to be determined.

Scientific societies edit

The International Bone Marrow Adiposity Society (BMAS) edit

Because of the increasing interest in BMAT from both researchers and clinicians, in 2018 The International Bone Marrow Adiposity Society (BMAS) was founded.[65] Work to build the society began in Lille, France in 2015, when the first International Meeting on Bone Marrow Adiposity (BMA2015) was held. The meeting was a great success and led to a second international meeting (BMA2016) in August 2016 held in Rotterdam, The Netherlands. Both meetings were a success in that they for the first time brought together scientists and physicians from different backgrounds (bone metabolism, cancer, obesity and diabetes) to share ideas and advance research into, and our understanding of, the patho/physiological role of BMAds.

 
Logo for The International Bone Marrow Adiposity Society

This success led to a network of researchers discussing the formation of a new society, focusing on bone marrow adiposity (BMA). This network worked together in 2016–2017 to lay the foundations for this society, which was then discussed further during the third international meeting held in Lausanne, Switzerland in 2017 (BMA2017). The statues were then signed at the fourth international meeting, held in 2018 again in Lille (BMA2018). As discussed in the following section, there have since been three further international meetings, held in Odense, Denmark in 2019 (BMA2019), virtually in 2020 (BMA2020), and in Athens, Greece in 2022 (BMA2022). The first BMAS Summer School was held virtually in the summer of 2021.

Since its foundation, BMAS working groups have published three position papers relating to nomenclature,[9] methodologies [66] and biobanking for BMA research.[67] These working groups remain active, with other working groups also focussing on clinical and translational issues, public engagement, and young researchers (Next Generation BMAS)

BMAS meetings edit

  • BMA2015 (Lille, France)[68]
  • BMA2016 (Rotterdam, Netherlands)[69]
  • BMA2017 (Lausanne, Switzerland)[70]
  • BMA2018 (Lille, France)[71]
  • BMA2019 (Odense, Denmark)
  • BMA2020 (virtual BMA meeting)[72]
  • BMA Summer School 2021 (virtual)[73]
  • BMA2022 (Athens, Greece)

American Society for Bone and Mineral Research edit

ASBMR has published hundreds of presentations and articles on BMAT featured in the ASBMR annual meetings, The Journal of Bone and Mineral Research ( JBMR) , JBMRPlus, and the Primer on the Metabolic Bone Diseases and Disorders of Mineral Metabolism.

Endocrine Society edit

Endocrine society features many presentations and articles on BMAT.

References edit

  This article incorporates text by Gabriel M. Pagnotti and Maya Styner available under the CC BY 4.0 license.

  1. ^ Cohen A, Dempster DW, Stein EM, Nickolas TL, Zhou H, McMahon DJ, et al. (August 2012). "Increased marrow adiposity in premenopausal women with idiopathic osteoporosis". The Journal of Clinical Endocrinology and Metabolism. 97 (8): 2782–2791. doi:10.1210/jc.2012-1477. PMC 3410269. PMID 22701013.
  2. ^ Meunier P, Aaron J, Edouard C, Vignon G (October 1971). "Osteoporosis and the replacement of cell populations of the marrow by adipose tissue. A quantitative study of 84 iliac bone biopsies". Clinical Orthopaedics and Related Research. 80: 147–154. doi:10.1097/00003086-197110000-00021. PMID 5133320.
  3. ^ Fazeli PK, Horowitz MC, MacDougald OA, Scheller EL, Rodeheffer MS, Rosen CJ, Klibanski A (March 2013). "Marrow fat and bone--new perspectives". The Journal of Clinical Endocrinology and Metabolism. 98 (3): 935–945. doi:10.1210/jc.2012-3634. PMC 3590487. PMID 23393168.
  4. ^ a b c d McGrath C, Sankaran JS, Misaghian-Xanthos N, Sen B, Xie Z, Styner MA, et al. (January 2020). "Exercise Degrades Bone in Caloric Restriction, Despite Suppression of Marrow Adipose Tissue (MAT)". Journal of Bone and Mineral Research. 35 (1): 106–115. doi:10.1002/jbmr.3872. PMC 6980282. PMID 31509274.
  5. ^ Ahdjoudj S, Lasmoles F, Holy X, Zerath E, Marie PJ (April 2002). "Transforming growth factor beta2 inhibits adipocyte differentiation induced by skeletal unloading in rat bone marrow stroma". Journal of Bone and Mineral Research. 17 (4): 668–677. doi:10.1359/jbmr.2002.17.4.668. PMID 11918224. S2CID 23060094.
  6. ^ Wronski TJ, Morey ER (1982-01-01). "Skeletal abnormalities in rats induced by simulated weightlessness". Metabolic Bone Disease & Related Research. 4 (1): 69–75. doi:10.1016/0221-8747(82)90011-X. PMID 7121257.
  7. ^ Rubin MR, Manavalan JS, Agarwal S, McMahon DJ, Nino A, Fitzpatrick LA, Bilezikian JP (October 2014). "Effects of rosiglitazone vs metformin on circulating osteoclast and osteogenic precursor cells in postmenopausal women with type 2 diabetes mellitus". The Journal of Clinical Endocrinology and Metabolism. 99 (10): E1933–E1942. doi:10.1210/jc.2013-3666. PMID 24905061.
  8. ^ Cawthorn W (2020). "Bone Marrow Adipose Tissue". Encyclopedia of Bone Biology. Oxford, UK: Academic Press. pp. 156–177. doi:10.1016/B978-0-12-801238-3.11207-3. ISBN 978-0-12-814082-6. S2CID 213762507.
  9. ^ a b Bravenboer N, Bredella MA, Chauveau C, Corsi A, Douni E, Ferris WF, et al. (2020). "Standardised Nomenclature, Abbreviations, and Units for the Study of Bone Marrow Adiposity: Report of the Nomenclature Working Group of the International Bone Marrow Adiposity Society". Frontiers in Endocrinology. 10: 923. doi:10.3389/fendo.2019.00923. PMC 6993042. PMID 32038486.
  10. ^ Muruganandan S, Roman AA, Sinal CJ (January 2009). "Adipocyte differentiation of bone marrow-derived mesenchymal stem cells: cross talk with the osteoblastogenic program". Cellular and Molecular Life Sciences. 66 (2): 236–253. doi:10.1007/s00018-008-8429-z. PMID 18854943. S2CID 5558912.
  11. ^ a b Paccou J, Hardouin P, Cotten A, Penel G, Cortet B (October 2015). "The Role of Bone Marrow Fat in Skeletal Health: Usefulness and Perspectives for Clinicians". The Journal of Clinical Endocrinology and Metabolism. 100 (10): 3613–3621. doi:10.1210/jc.2015-2338. PMID 26244490.
  12. ^ a b c d Styner M, Thompson WR, Galior K, Uzer G, Wu X, Kadari S, et al. (July 2014). "Bone marrow fat accumulation accelerated by high fat diet is suppressed by exercise". Bone. 64: 39–46. doi:10.1016/j.bone.2014.03.044. PMC 4041820. PMID 24709686.
  13. ^ Scheller EL, Khoury B, Moller KL, Wee NK, Khandaker S, Kozloff KM, et al. (2016). "Changes in Skeletal Integrity and Marrow Adiposity during High-Fat Diet and after Weight Loss". Frontiers in Endocrinology. 7: 102. doi:10.3389/fendo.2016.00102. PMC 4961699. PMID 27512386.
  14. ^ Doucette CR, Horowitz MC, Berry R, MacDougald OA, Anunciado-Koza R, Koza RA, Rosen CJ (September 2015). "A High Fat Diet Increases Bone Marrow Adipose Tissue (MAT) But Does Not Alter Trabecular or Cortical Bone Mass in C57BL/6J Mice". Journal of Cellular Physiology. 230 (9): 2032–2037. doi:10.1002/jcp.24954. PMC 4580244. PMID 25663195.
  15. ^ a b c Styner M, Pagnotti GM, Galior K, Wu X, Thompson WR, Uzer G, et al. (August 2015). "Exercise Regulation of Marrow Fat in the Setting of PPARγ Agonist Treatment in Female C57BL/6 Mice". Endocrinology. 156 (8): 2753–2761. doi:10.1210/en.2015-1213. PMC 4511140. PMID 26052898.
  16. ^ a b c d e Styner M, Pagnotti GM, McGrath C, Wu X, Sen B, Uzer G, et al. (August 2017). "Exercise Decreases Marrow Adipose Tissue Through ß-Oxidation in Obese Running Mice". Journal of Bone and Mineral Research. 32 (8): 1692–1702. doi:10.1002/jbmr.3159. PMC 5550355. PMID 28436105.
  17. ^ a b Pagnotti GM, Styner M, Uzer G, Patel VS, Wright LE, Ness KK, et al. (June 2019). "Combating osteoporosis and obesity with exercise: leveraging cell mechanosensitivity". Nature Reviews. Endocrinology. 15 (6): 339–355. doi:10.1038/s41574-019-0170-1. PMC 6520125. PMID 30814687.
  18. ^ Luu YK, Pessin JE, Judex S, Rubin J, Rubin CT (April 2009). "Mechanical Signals As a Non-Invasive Means to Influence Mesenchymal Stem Cell Fate, Promoting Bone and Suppressing the Fat Phenotype". BoneKEy Osteovision. 6 (4): 132–149. doi:10.1138/20090371. PMC 3255555. PMID 22241295.
  19. ^ Belavy DL, Quittner MJ, Ridgers ND, Shiekh A, Rantalainen T, Trudel G (April 2018). "Specific Modulation of Vertebral Marrow Adipose Tissue by Physical Activity". Journal of Bone and Mineral Research. 33 (4): 651–657. doi:10.1002/jbmr.3357. hdl:10536/DRO/DU:30106029. PMID 29336053.
  20. ^ a b c Little-Letsinger SE, Pagnotti GM, McGrath C, Styner M (December 2020). "Exercise and Diet: Uncovering Prospective Mediators of Skeletal Fragility in Bone and Marrow Adipose Tissue". Current Osteoporosis Reports. 18 (6): 774–789. doi:10.1007/s11914-020-00634-y. PMC 7736569. PMID 33068251.
  21. ^ Southmayd EA, Williams NI, Mallinson RJ, De Souza MJ (August 2019). "Energy Deficiency Suppresses Bone Turnover in Exercising Women With Menstrual Disturbances". The Journal of Clinical Endocrinology and Metabolism. 104 (8): 3131–3145. doi:10.1210/jc.2019-00089. PMID 30896746.
  22. ^ McGrath C, Little-Letsinger SE, Sankaran JS, Sen B, Xie Z, Styner MA, et al. (2022-01-25). "Exercise Increases Bone in SEIPIN Deficient Lipodystrophy, Despite Low Marrow Adiposity". Frontiers in Endocrinology. 12: 782194. doi:10.3389/fendo.2021.782194. PMC 8822583. PMID 35145475.
  23. ^ Krings A, Rahman S, Huang S, Lu Y, Czernik PJ, Lecka-Czernik B (February 2012). "Bone marrow fat has brown adipose tissue characteristics, which are attenuated with aging and diabetes". Bone. 50 (2): 546–552. doi:10.1016/j.bone.2011.06.016. PMC 3214232. PMID 21723971.
  24. ^ a b Suchacki KJ, Tavares AA, Mattiucci D, Scheller EL, Papanastasiou G, Gray C, et al. (June 2020). "Bone marrow adipose tissue is a unique adipose subtype with distinct roles in glucose homeostasis". Nature Communications. 11 (1): 3097. Bibcode:2020NatCo..11.3097S. doi:10.1038/s41467-020-16878-2. PMC 7303125. PMID 32555194.
  25. ^ Craft CS, Robles H, Lorenz MR, Hilker ED, Magee KL, Andersen TL, et al. (November 2019). "Bone marrow adipose tissue does not express UCP1 during development or adrenergic-induced remodeling". Scientific Reports. 9 (1): 17427. Bibcode:2019NatSR...917427C. doi:10.1038/s41598-019-54036-x. PMC 6874537. PMID 31758074.
  26. ^ Scheller EL, Khandaker S, Learman BS, Cawthorn WP, Anderson LM, Pham HA, et al. (January 2019). "Bone marrow adipocytes resist lipolysis and remodeling in response to β-adrenergic stimulation". Bone. 118: 32–41. doi:10.1016/j.bone.2018.01.016. PMC 6062480. PMID 29360620.
  27. ^ Attané C, Estève D, Chaoui K, Iacovoni JS, Corre J, Moutahir M, et al. (January 2020). "Human Bone Marrow Is Comprised of Adipocytes with Specific Lipid Metabolism". Cell Reports. 30 (4): 949–958.e6. doi:10.1016/j.celrep.2019.12.089. PMID 31995765. S2CID 210949460.
  28. ^ Ye R, Scherer PE (April 2013). "Adiponectin, driver or passenger on the road to insulin sensitivity?". Molecular Metabolism. 2 (3): 133–141. doi:10.1016/j.molmet.2013.04.001. PMC 3773837. PMID 24049728.
  29. ^ Tilg H, Moschen AR (October 2006). "Adipocytokines: mediators linking adipose tissue, inflammation and immunity". Nature Reviews. Immunology. 6 (10): 772–783. doi:10.1038/nri1937. PMID 16998510. S2CID 29865593.
  30. ^ Wronska A, Kmiec Z (June 2012). "Structural and biochemical characteristics of various white adipose tissue depots". Acta Physiologica. 205 (2): 194–208. doi:10.1111/j.1748-1716.2012.02409.x. PMID 22226221. S2CID 22915022.
  31. ^ Masuzaki H, Paterson J, Shinyama H, Morton NM, Mullins JJ, Seckl JR, Flier JS (December 2001). "A transgenic model of visceral obesity and the metabolic syndrome". Science. 294 (5549): 2166–2170. Bibcode:2001Sci...294.2166M. doi:10.1126/science.1066285. PMID 11739957. S2CID 768303.
  32. ^ Bredella MA, Lin E, Gerweck AV, Landa MG, Thomas BJ, Torriani M, et al. (November 2012). "Determinants of bone microarchitecture and mechanical properties in obese men". The Journal of Clinical Endocrinology and Metabolism. 97 (11): 4115–4122. doi:10.1210/jc.2012-2246. PMC 3485587. PMID 22933540.
  33. ^ Cohen A, Dempster DW, Recker RR, Lappe JM, Zhou H, Zwahlen A, et al. (June 2013). "Abdominal fat is associated with lower bone formation and inferior bone quality in healthy premenopausal women: a transiliac bone biopsy study". The Journal of Clinical Endocrinology and Metabolism. 98 (6): 2562–2572. doi:10.1210/jc.2013-1047. PMC 3667251. PMID 23515452.
  34. ^ Wu J, Cohen P, Spiegelman BM (February 2013). "Adaptive thermogenesis in adipocytes: is beige the new brown?". Genes & Development. 27 (3): 234–250. doi:10.1101/gad.211649.112. PMC 3576510. PMID 23388824.
  35. ^ Al-Nbaheen M, Vishnubalaji R, Ali D, Bouslimi A, Al-Jassir F, Megges M, et al. (February 2013). "Human stromal (mesenchymal) stem cells from bone marrow, adipose tissue and skin exhibit differences in molecular phenotype and differentiation potential". Stem Cell Reviews and Reports. 9 (1): 32–43. doi:10.1007/s12015-012-9365-8. PMC 3563956. PMID 22529014.
  36. ^ Gimble JM, Zvonic S, Floyd ZE, Kassem M, Nuttall ME (May 2006). "Playing with bone and fat". Journal of Cellular Biochemistry. 98 (2): 251–266. doi:10.1002/jcb.20777. PMID 16479589. S2CID 19746472.
  37. ^ Cawthorn WP, Scheller EL, Learman BS, Parlee SD, Simon BR, Mori H, et al. (August 2014). "Bone marrow adipose tissue is an endocrine organ that contributes to increased circulating adiponectin during caloric restriction". Cell Metabolism. 20 (2): 368–375. doi:10.1016/j.cmet.2014.06.003. PMC 4126847. PMID 24998914.
  38. ^ Duque G, Li W, Adams M, Xu S, Phipps R (May 2011). "Effects of risedronate on bone marrow adipocytes in postmenopausal women". Osteoporosis International. 22 (5): 1547–1553. doi:10.1007/s00198-010-1353-8. PMID 20661545. S2CID 27850362.
  39. ^ Yeung DK, Griffith JF, Antonio GE, Lee FK, Woo J, Leung PC (August 2005). "Osteoporosis is associated with increased marrow fat content and decreased marrow fat unsaturation: a proton MR spectroscopy study". Journal of Magnetic Resonance Imaging. 22 (2): 279–285. doi:10.1002/jmri.20367. PMID 16028245. S2CID 2941430.
  40. ^ Li X, Kuo D, Schafer AL, Porzig A, Link TM, Black D, Schwartz AV (April 2011). "Quantification of vertebral bone marrow fat content using 3 Tesla MR spectroscopy: reproducibility, vertebral variation, and applications in osteoporosis". Journal of Magnetic Resonance Imaging. 33 (4): 974–979. doi:10.1002/jmri.22489. PMC 3072841. PMID 21448966.
  41. ^ Syed FA, Oursler MJ, Hefferanm TE, Peterson JM, Riggs BL, Khosla S (September 2008). "Effects of estrogen therapy on bone marrow adipocytes in postmenopausal osteoporotic women". Osteoporosis International. 19 (9): 1323–1330. doi:10.1007/s00198-008-0574-6. PMC 2652842. PMID 18274695.
  42. ^ Khosla S, Riggs BL, Atkinson EJ, Oberg AL, McDaniel LJ, Holets M, et al. (January 2006). "Effects of sex and age on bone microstructure at the ultradistal radius: a population-based noninvasive in vivo assessment". Journal of Bone and Mineral Research. 21 (1): 124–131. doi:10.1359/jbmr.050916. PMC 1352156. PMID 16355281.
  43. ^ Glatt V, Canalis E, Stadmeyer L, Bouxsein ML (August 2007). "Age-related changes in trabecular architecture differ in female and male C57BL/6J mice". Journal of Bone and Mineral Research. 22 (8): 1197–1207. doi:10.1359/jbmr.070507. PMID 17488199.
  44. ^ Tchkonia T, Morbeck DE, Von Zglinicki T, Van Deursen J, Lustgarten J, Scrable H, et al. (October 2010). "Fat tissue, aging, and cellular senescence". Aging Cell. 9 (5): 667–684. doi:10.1111/j.1474-9726.2010.00608.x. PMC 2941545. PMID 20701600.
  45. ^ Kassem M, Marie PJ (April 2011). "Senescence-associated intrinsic mechanisms of osteoblast dysfunctions". Aging Cell. 10 (2): 191–197. doi:10.1111/j.1474-9726.2011.00669.x. PMID 21210937.
  46. ^ Moerman EJ, Teng K, Lipschitz DA, Lecka-Czernik B (December 2004). "Aging activates adipogenic and suppresses osteogenic programs in mesenchymal marrow stroma/stem cells: the role of PPAR-gamma2 transcription factor and TGF-beta/BMP signaling pathways". Aging Cell. 3 (6): 379–389. doi:10.1111/j.1474-9728.2004.00127.x. PMC 1850101. PMID 15569355.
  47. ^ Stevens JR, Miranda-Carboni GA, Singer MA, Brugger SM, Lyons KM, Lane TF (October 2010). "Wnt10b deficiency results in age-dependent loss of bone mass and progressive reduction of mesenchymal progenitor cells". Journal of Bone and Mineral Research. 25 (10): 2138–2147. doi:10.1002/jbmr.118. PMC 3153316. PMID 20499361.
  48. ^ Bennett CN, Ouyang H, Ma YL, Zeng Q, Gerin I, Sousa KM, et al. (December 2007). "Wnt10b increases postnatal bone formation by enhancing osteoblast differentiation". Journal of Bone and Mineral Research. 22 (12): 1924–1932. doi:10.1359/jbmr.070810. PMID 17708715.
  49. ^ Chen Q, Shou P, Zheng C, Jiang M, Cao G, Yang Q, et al. (July 2016). "Fate decision of mesenchymal stem cells: adipocytes or osteoblasts?". Cell Death and Differentiation. 23 (7): 1128–1139. doi:10.1038/cdd.2015.168. PMC 4946886. PMID 26868907.
  50. ^ Zhou BO, Yu H, Yue R, Zhao Z, Rios JJ, Naveiras O, Morrison SJ (August 2017). "Bone marrow adipocytes promote the regeneration of stem cells and haematopoiesis by secreting SCF". Nature Cell Biology. 19 (8): 891–903. doi:10.1038/ncb3570. PMC 5536858. PMID 28714970.
  51. ^ Bielohuby M, Matsuura M, Herbach N, Kienzle E, Slawik M, Hoeflich A, Bidlingmaier M (February 2010). "Short-term exposure to low-carbohydrate, high-fat diets induces low bone mineral density and reduces bone formation in rats". Journal of Bone and Mineral Research. 25 (2): 275–284. doi:10.1359/jbmr.090813. PMID 19653818.
  52. ^ Spatz JM, Ellman R, Cloutier AM, Louis L, van Vliet M, Suva LJ, et al. (April 2013). "Sclerostin antibody inhibits skeletal deterioration due to reduced mechanical loading". Journal of Bone and Mineral Research. 28 (4): 865–874. doi:10.1002/jbmr.1807. PMC 4076162. PMID 23109229.
  53. ^ Rosen CJ, Ackert-Bicknell CL, Adamo ML, Shultz KL, Rubin J, Donahue LR, et al. (November 2004). "Congenic mice with low serum IGF-I have increased body fat, reduced bone mineral density, and an altered osteoblast differentiation program". Bone. 35 (5): 1046–1058. doi:10.1016/j.bone.2004.07.008. PMID 15542029.
  54. ^ Naveiras O, Nardi V, Wenzel PL, Hauschka PV, Fahey F, Daley GQ (July 2009). "Bone-marrow adipocytes as negative regulators of the haematopoietic microenvironment". Nature. 460 (7252): 259–263. Bibcode:2009Natur.460..259N. doi:10.1038/nature08099. PMC 2831539. PMID 19516257.
  55. ^ David V, Martin A, Lafage-Proust MH, Malaval L, Peyroche S, Jones DB, et al. (May 2007). "Mechanical loading down-regulates peroxisome proliferator-activated receptor gamma in bone marrow stromal cells and favors osteoblastogenesis at the expense of adipogenesis". Endocrinology. 148 (5): 2553–2562. doi:10.1210/en.2006-1704. PMID 17317771.
  56. ^ Majka SM, Miller HL, Sullivan T, Erickson PF, Kong R, Weiser-Evans M, et al. (October 2012). "Adipose lineage specification of bone marrow-derived myeloid cells". Adipocyte. 1 (4): 215–229. doi:10.4161/adip.21496. PMC 3609111. PMID 23700536.
  57. ^ Majka SM, Miller HL, Helm KM, Acosta AS, Childs CR, Kong R, Klemm DJ (2014). "Analysis and Isolation of Adipocytes by Flow Cytometry". Methods of Adipose Tissue Biology, Part A. Methods in Enzymology. Vol. 537. pp. 281–96. doi:10.1016/b978-0-12-411619-1.00015-x. ISBN 9780124116191. PMC 4143162. PMID 24480352.
  58. ^ Bernstein RL, Hyun WC, Davis JH, Fulwyler MJ, Pershadsingh HA (July 1989). "Flow cytometric analysis of mature adipocytes". Cytometry. 10 (4): 469–474. doi:10.1002/cyto.990100416. PMID 2766892.
  59. ^ Bredella MA, Torriani M, Ghomi RH, Thomas BJ, Brick DJ, Gerweck AV, et al. (January 2011). "Vertebral bone marrow fat is positively associated with visceral fat and inversely associated with IGF-1 in obese women". Obesity. 19 (1): 49–53. doi:10.1038/oby.2010.106. PMC 3593350. PMID 20467419.
  60. ^ de Paula FJ, Dick-de-Paula I, Bornstein S, Rostama B, Le P, Lotinun S, et al. (September 2011). "VDR haploinsufficiency impacts body composition and skeletal acquisition in a gender-specific manner". Calcified Tissue International. 89 (3): 179–191. doi:10.1007/s00223-011-9505-1. PMC 3157554. PMID 21637996.
  61. ^ Fazeli PK, Bredella MA, Freedman L, Thomas BJ, Breggia A, Meenaghan E, et al. (September 2012). "Marrow fat and preadipocyte factor-1 levels decrease with recovery in women with anorexia nervosa". Journal of Bone and Mineral Research. 27 (9): 1864–1871. doi:10.1002/jbmr.1640. PMC 3415584. PMID 22508185.
  62. ^ Rantalainen T, Nikander R, Heinonen A, Cervinka T, Sievänen H, Daly RM (May 2013). "Differential effects of exercise on tibial shaft marrow density in young female athletes". The Journal of Clinical Endocrinology and Metabolism. 98 (5): 2037–2044. doi:10.1210/jc.2012-3748. hdl:10536/DRO/DU:30060423. PMID 23616150.
  63. ^ Scheller EL, Troiano N, Vanhoutan JN, Bouxsein MA, Fretz JA, Xi Y, et al. (2014). "Use of Osmium Tetroxide Staining with Microcomputerized Tomography to Visualize and Quantify Bone Marrow Adipose Tissue in Vivo". Methods of Adipose Tissue Biology, Part A. Methods in Enzymology. Vol. 537. pp. 123–39. doi:10.1016/b978-0-12-411619-1.00007-0. ISBN 9780124116191. PMC 4097010. PMID 24480344.
  64. ^ Pham TT, Ivaska KK, Hannukainen JC, Virtanen KA, Lidell ME, Enerbäck S, et al. (July 2020). "Human Bone Marrow Adipose Tissue is a Metabolically Active and Insulin-Sensitive Distinct Fat Depot". The Journal of Clinical Endocrinology and Metabolism. 105 (7): 2300–2310. doi:10.1210/clinem/dgaa216. PMC 7247553. PMID 32311037.
  65. ^ "The International Bone Marrow Adiposity Society". BMAS Website.
  66. ^ Tratwal J, Labella R, Bravenboer N, Kerckhofs G, Douni E, Scheller EL, et al. (2020). "Reporting Guidelines, Review of Methodological Standards, and Challenges Toward Harmonization in Bone Marrow Adiposity Research. Report of the Methodologies Working Group of the International Bone Marrow Adiposity Society". Frontiers in Endocrinology. 11: 65. doi:10.3389/fendo.2020.00065. PMC 7059536. PMID 32180758.
  67. ^ Lucas S, Tencerova M, von der Weid B, Andersen TL, Attané C, Behler-Janbeck F, et al. (2021). "Guidelines for Biobanking of Bone Marrow Adipose Tissue and Related Cell Types: Report of the Biobanking Working Group of the International Bone Marrow Adiposity Society". Frontiers in Endocrinology. 12: 744527. doi:10.3389/fendo.2021.744527. PMC 8503265. PMID 34646237.
  68. ^ Hardouin P, Marie PJ, Rosen CJ (December 2016). "New insights into bone marrow adipocytes: Report from the First European Meeting on Bone Marrow Adiposity (BMA 2015)". Bone. 93: 212–215. doi:10.1016/j.bone.2015.11.013. PMID 26608519.
  69. ^ van der Eerden B, van Wijnen A (October 2017). "Meeting report of the 2016 bone marrow adiposity meeting". Adipocyte. 6 (4): 304–313. doi:10.1080/21623945.2017.1313374. PMC 5736244. PMID 28410005.
  70. ^ Corsi A, Palmisano B, Tratwal J, Riminucci M, Naveiras O (2019). "Brief Report From the 3rd International Meeting on Bone Marrow Adiposity (BMA 2017)". Frontiers in Endocrinology. 10: 336. doi:10.3389/fendo.2019.00336. PMC 6546805. PMID 31191458.
  71. ^ Penel G, Kerckhofs G, Chauveau C (2019). "Brief Report From the 4th International Meeting on Bone Marrow Adiposity (BMA2018)". Frontiers in Endocrinology. 10: 691. doi:10.3389/fendo.2019.00691. PMC 6813723. PMID 31681168.
  72. ^ Scheller EL, McGee-Lawrence ME, Lecka-Czernik B (2021). "Report From the 6th International Meeting on Bone Marrow Adiposity (BMA2020)". Frontiers in Endocrinology. 12: 712088. doi:10.3389/fendo.2021.712088. PMC 8323480. PMID 34335478.
  73. ^ Labella R, Little-Letsinger S, Avilkina V, Sarkis R, Tencerova M, Vlug A, Palmisano B (2022). "Next Generation Bone Marrow Adiposity Researchers: Report From the 1st BMAS Summer School 2021". Frontiers in Endocrinology. 13: 879588. doi:10.3389/fendo.2022.879588. PMC 9043644. PMID 35498418.

Further reading edit

  • . University of Michigan. 3 July 2014. Archived from the original on 15 March 2015.
  • "Another reason to exercise: Burning bone fat a key to better bone health". Science Daily. 18 May 2017.
  • "Why are our bones full of fat? The secrets of bone marrow adipose tissue". Society for Endocrinology. Winter 2017.

bone, marrow, adipose, tissue, bmat, sometimes, referred, marrow, adipose, tissue, type, deposit, bone, marrow, increases, states, bone, density, osteoporosis, anorexia, nervosa, caloric, restriction, skeletal, unweighting, such, that, which, occurs, space, tr. Bone marrow adipose tissue BMAT sometimes referred to as marrow adipose tissue MAT is a type of fat deposit in bone marrow It increases in states of low bone density osteoporosis 1 2 anorexia nervosa caloric restriction 3 4 skeletal unweighting such as that which occurs in space travel 5 6 and anti diabetes therapies 7 BMAT decreases in anaemia leukaemia and hypertensive heart failure in response to hormones such as oestrogen leptin and growth hormone with exercise induced weight loss or bariatric surgery in response to chronic cold exposure and in response to pharmacological agents such as bisphosphonates teriparatide and metformin 8 Bone marrow adipose tissueDetailsSystemMusculoskeletal or locomotor IdentifiersLatinadipose ossium medullaAnatomical terminology edit on Wikidata Bone marrow adipocytes are derived from mesenchymal stem cell MSC differentiation Contents 1 Anatomy 2 Physiology 2 1 Exercise regulation 2 2 Relationships to other types of fat 2 3 Impact on bone health 2 4 Maintenance of hematopoietic stem cells 3 Measurement 4 Scientific societies 4 1 The International Bone Marrow Adiposity Society BMAS 4 1 1 BMAS meetings 4 2 American Society for Bone and Mineral Research 4 3 Endocrine Society 5 References 6 Further readingAnatomy editBone marrow adipocytes BMAds 9 originate from mesenchymal stem cell MSC progenitors that also give rise to osteoblasts among other cell types 10 Thus it is thought that BMAT results from preferential MSC differentiation into the adipocyte rather than osteoblast lineage in the setting of osteoporosis 11 Since BMAT is increased in the setting of obesity 12 13 14 and is suppressed by endurance exercise 15 12 16 17 or vibration 18 it is likely that BMAT physiology in the setting of mechanical input exercise approximates that of white adipose tissue WAT Physiology editExercise regulation edit The first study to demonstrate exercise regulation of BMAT in rodents was published in 2014 12 Now exercise regulation of BMAT has been confirmed in a human 19 adding clinical importance Several studies demonstrated exercise reduction of BMAT which occurs along with an increase in bone quantity 17 15 16 20 Since exercise increases bone quantity reduces BMAT and increases expression of markers of fatty acid oxidation in bone BMAT is thought to be providing needed fuel for exercise induced bone formation or anabolism 16 A notable exception occurs in the setting of caloric restriction exercise suppression of BMAT does not yield an increase in bone formation and even appears to cause bone loss 4 21 20 Indeed energy availability appears to be a factor in the ability of exercise to regulate BMAT 4 Another exception occurs in lipodystrophy a condition with reduced overall adipose stores exercise induced anabolism is possible even with minimal BMAT stores 22 Relationships to other types of fat edit BMAT has been reported to have qualities of both white and brown fat 23 However more recent functional and omics studies have shown that BMAT is a unique adipose depot that is molecularly and functionally distinct to WAT or BAT 24 25 26 27 Subcutaneous white fat contain excess energy indicating a clear evolutionary advantage during times of scarcity WAT is also the source of adipokines and inflammatory markers which have both positive e g adiponectin 28 and negative 29 effects on metabolic and cardiovascular endpoints Visceral abdominal fat VAT is a distinct type of WAT that is proportionally associated with negative metabolic and cardiovascular morbidity 30 regenerates cortisol 31 and recently has been tied to decreased bone formation 32 33 Both types of WAT substantially differ from brown adipose tissue BAT as by a group of proteins that help BAT s thermogenic role 34 BMAT by its specific marrow location and its adipocyte origin from at least LepR marrow MSC is separated from non bone fat storage by larger expression of bone transcription factors 35 and likely indicates a different fat phenotype 36 Recently BMAT was noted to produce a greater proportion of adiponectin an adipokine associated with improved metabolism than WAT 37 suggesting an endocrine function for this depot akin but different from that of WAT Impact on bone health edit BMAT increases in states of bone fragility BMAT is thought to result from preferential MSC differentiation into an adipocyte rather than osteoblast lineage in osteoporosis 11 20 based on the inverse relationship between bone and BMAT in bone fragile osteoporotic states An increase in BMAT is noted in osteoporosis clinical studies measured by MR spectroscopy 38 39 40 Estrogen therapy in postmenopausal osteoporosis reduces BMAT 41 Antiresorptive therapies like risedronate or zoledronate also decrease BMAT while increasing bone density supporting an inverse relationship between bone quantity and BMAT During aging bone quantity declines 42 43 and fat redistributes from subcutaneous to ectopic sites such as bone marrow muscle and liver 44 Aging is associated with lower osteogenic and greater adipogenic biasing of MSC 45 This aging related biasing of MSC away from osteoblast lineage may represent higher basal PPARg expression 46 or decreased Wnt10b 47 48 49 Thus bone fragility osteoporosis and osteoporotic fractures are thought to be linked to mechanisms which promote BMAT accumulation citation needed Maintenance of hematopoietic stem cells edit BMAds secrete factors that promote HSC renewal in most bones 50 Hematopoietic cells also known as blood cells reside in the bone marrow along with BMAds These hematopoietic cells are derived from hematopoietic stem cells HSC which give rise to diverse cells cells of the blood immune system as well as cells that break down bone osteoclasts HSC renewal occurs in the marrow stem cell niche a microenvironment that contains cells and secreted factors that promote appropriate renewal and differentiation of HSC The study of the stem cell niche is relevant to the field of oncology in order to improve therapy for multiple hematologic cancers As such cancers are often treated with bone marrow transplantation there is interest in improving the renewal of HSC citation needed Measurement editIn order to understand the physiology of BMAT various analytic methods have been applied BMAds are difficult to isolate and quantify because they are interspersed with bony and hematopoietic elements Until recently qualitative measurements of BMAT have relied on bone histology 51 52 which is subject to site selection bias and cannot adequately quantify the volume of fat in the marrow Nevertheless histological techniques and fixation make possible visualization of BMAT quantification of BMAd size and BMAT s association with the surrounding endosteum milieu of cells and secreted factors 53 54 55 Recent advances in cell surface and intracellular marker identification and single cell analyses led to greater resolution and high throughput ex vivo quantification Flow cytometric quantification can be used to purify adipocytes from the stromal vascular fraction of most fat depots 56 Early research with such machinery cited adipocytes as too large and fragile for cytometer based purification rendering them susceptible to lysis however recent advances have been made to mitigate this 57 nevertheless this methodology continues to pose technical challenges 58 and is inaccessible to much of the research community To improve quantification of BMAT novel imaging techniques have been developed as a means to visualize and quantify BMAT Although proton magnetic resonance spectroscopy 1H MRS has been used with success to quantify vertebral BMAT in humans 59 it is difficult to employ in laboratory animals 60 Magnetic resonance imaging MRI provides BMAT assessment in the vertebral skeleton 61 in conjunction with mCT based marrow density measurements 62 A volumetric method to identify quantify and localize BMAT in rodent bone has been recently developed requiring osmium staining of bones and mCT imaging 63 followed by advanced image analysis of osmium bound lipid volume in mm3 relative to bone volume 12 16 15 This technique provides reproducible quantification and visualization of BMAT enabling the ability to consistently quantify changes in BMAT with diet exercise and agents that constrain precursor lineage allocation Although the osmium method is quantitatively precise osmium is toxic and cannot be compared across batched experiments Recently researchers developed and validated 16 a 9 4T MRI scanner technique that allows localization and volumetric 3D quantification that can be compared across experiments as in 4 Several studies have also analysed BMAT function in vivo using positron emission tomography computed tomography PET CT combined with the tracer 18F Fluorodeoxyglucose FDG This allows glucose uptake a measure of metabolic activity to be quantified in living organisms including humans Two recent studies found that unlike brown adipose tissue BMAT does not increase glucose uptake in response to cold exposure demonstrating that BMAT is functionally distinct from BAT 24 64 The full extent of BMAT s impact on systemic metabolic homeostasis remains to be determined Methods for Quantification of Bone Marrow Adipose Tissue BMAT nbsp This figure demonstrates the use of the osmium mCT method with advanced image processing to quantify BMAT In this figure running exercise is shown to suppress BMAT despite PPARg agonist Fat binder osmium is imaged via mCT A in n 5 per group overlaid images Quantification of osmium as BMAT bone volume in the whole femur is shown a significant due to Rosi b significant due to exercise Rosi rosiglizaone CTL control E exercise nbsp This figure demonstrates the use of MRI imaging 9 4T scanner along with advanced image processing to quantify BMAT The images and graph demonstrate that BMAT is higher in obese compared with lean mice B6 mice were fed HFD from age 4 wk until age 16 wk BMAT was quantified by MRI A n 10 superimposed group average images are shown B BMAT normalized to bone volume in each group nbsp Representative distal femur histologic section of a 16 week old healthy C57BL 6 mouse demonstrating a typical quantity of marrow adipocytes nbsp Representative distal femur histologic section of a 16 week old C57BL 6 mouse after 6 weeks of calorie restriction demonstrating an increased quantity of marrow adipocytes Scientific societies editThe International Bone Marrow Adiposity Society BMAS editBecause of the increasing interest in BMAT from both researchers and clinicians in 2018 The International Bone Marrow Adiposity Society BMAS was founded 65 Work to build the society began in Lille France in 2015 when the first International Meeting on Bone Marrow Adiposity BMA2015 was held The meeting was a great success and led to a second international meeting BMA2016 in August 2016 held in Rotterdam The Netherlands Both meetings were a success in that they for the first time brought together scientists and physicians from different backgrounds bone metabolism cancer obesity and diabetes to share ideas and advance research into and our understanding of the patho physiological role of BMAds nbsp Logo for The International Bone Marrow Adiposity Society This success led to a network of researchers discussing the formation of a new society focusing on bone marrow adiposity BMA This network worked together in 2016 2017 to lay the foundations for this society which was then discussed further during the third international meeting held in Lausanne Switzerland in 2017 BMA2017 The statues were then signed at the fourth international meeting held in 2018 again in Lille BMA2018 As discussed in the following section there have since been three further international meetings held in Odense Denmark in 2019 BMA2019 virtually in 2020 BMA2020 and in Athens Greece in 2022 BMA2022 The first BMAS Summer School was held virtually in the summer of 2021 Since its foundation BMAS working groups have published three position papers relating to nomenclature 9 methodologies 66 and biobanking for BMA research 67 These working groups remain active with other working groups also focussing on clinical and translational issues public engagement and young researchers Next Generation BMAS BMAS meetings edit BMA2015 Lille France 68 BMA2016 Rotterdam Netherlands 69 BMA2017 Lausanne Switzerland 70 BMA2018 Lille France 71 BMA2019 Odense Denmark BMA2020 virtual BMA meeting 72 BMA Summer School 2021 virtual 73 BMA2022 Athens Greece American Society for Bone and Mineral Research edit ASBMR has published hundreds of presentations and articles on BMAT featured in the ASBMR annual meetings The Journal of Bone and Mineral Research JBMR JBMRPlus and the Primer on the Metabolic Bone Diseases and Disorders of Mineral Metabolism Endocrine Society edit Endocrine society features many presentations and articles on BMAT References edit nbsp This article incorporates text by Gabriel M Pagnotti and Maya Styner available under the CC BY 4 0 license Cohen A Dempster DW Stein EM Nickolas TL Zhou H McMahon DJ et al August 2012 Increased marrow adiposity in premenopausal women with idiopathic osteoporosis The Journal of Clinical Endocrinology and Metabolism 97 8 2782 2791 doi 10 1210 jc 2012 1477 PMC 3410269 PMID 22701013 Meunier P Aaron J Edouard C Vignon G October 1971 Osteoporosis and the replacement of cell populations of the marrow by adipose tissue A quantitative study of 84 iliac bone biopsies Clinical Orthopaedics and Related Research 80 147 154 doi 10 1097 00003086 197110000 00021 PMID 5133320 Fazeli PK Horowitz MC MacDougald OA Scheller EL Rodeheffer MS Rosen CJ Klibanski A March 2013 Marrow fat and bone new perspectives The Journal of Clinical Endocrinology and Metabolism 98 3 935 945 doi 10 1210 jc 2012 3634 PMC 3590487 PMID 23393168 a b c d McGrath C Sankaran JS Misaghian Xanthos N Sen B Xie Z Styner MA et al January 2020 Exercise Degrades Bone in Caloric Restriction Despite Suppression of Marrow Adipose Tissue MAT Journal of Bone and Mineral Research 35 1 106 115 doi 10 1002 jbmr 3872 PMC 6980282 PMID 31509274 Ahdjoudj S Lasmoles F Holy X Zerath E Marie PJ April 2002 Transforming growth factor beta2 inhibits adipocyte differentiation induced by skeletal unloading in rat bone marrow stroma Journal of Bone and Mineral Research 17 4 668 677 doi 10 1359 jbmr 2002 17 4 668 PMID 11918224 S2CID 23060094 Wronski TJ Morey ER 1982 01 01 Skeletal abnormalities in rats induced by simulated weightlessness Metabolic Bone Disease amp Related Research 4 1 69 75 doi 10 1016 0221 8747 82 90011 X PMID 7121257 Rubin MR Manavalan JS Agarwal S McMahon DJ Nino A Fitzpatrick LA Bilezikian JP October 2014 Effects of rosiglitazone vs metformin on circulating osteoclast and osteogenic precursor cells in postmenopausal women with type 2 diabetes mellitus The Journal of Clinical Endocrinology and Metabolism 99 10 E1933 E1942 doi 10 1210 jc 2013 3666 PMID 24905061 Cawthorn W 2020 Bone Marrow Adipose Tissue Encyclopedia of Bone Biology Oxford UK Academic Press pp 156 177 doi 10 1016 B978 0 12 801238 3 11207 3 ISBN 978 0 12 814082 6 S2CID 213762507 a b Bravenboer N Bredella MA Chauveau C Corsi A Douni E Ferris WF et al 2020 Standardised Nomenclature Abbreviations and Units for the Study of Bone Marrow Adiposity Report of the Nomenclature Working Group of the International Bone Marrow Adiposity Society Frontiers in Endocrinology 10 923 doi 10 3389 fendo 2019 00923 PMC 6993042 PMID 32038486 Muruganandan S Roman AA Sinal CJ January 2009 Adipocyte differentiation of bone marrow derived mesenchymal stem cells cross talk with the osteoblastogenic program Cellular and Molecular Life Sciences 66 2 236 253 doi 10 1007 s00018 008 8429 z PMID 18854943 S2CID 5558912 a b Paccou J Hardouin P Cotten A Penel G Cortet B October 2015 The Role of Bone Marrow Fat in Skeletal Health Usefulness and Perspectives for Clinicians The Journal of Clinical Endocrinology and Metabolism 100 10 3613 3621 doi 10 1210 jc 2015 2338 PMID 26244490 a b c d Styner M Thompson WR Galior K Uzer G Wu X Kadari S et al July 2014 Bone marrow fat accumulation accelerated by high fat diet is suppressed by exercise Bone 64 39 46 doi 10 1016 j bone 2014 03 044 PMC 4041820 PMID 24709686 Scheller EL Khoury B Moller KL Wee NK Khandaker S Kozloff KM et al 2016 Changes in Skeletal Integrity and Marrow Adiposity during High Fat Diet and after Weight Loss Frontiers in Endocrinology 7 102 doi 10 3389 fendo 2016 00102 PMC 4961699 PMID 27512386 Doucette CR Horowitz MC Berry R MacDougald OA Anunciado Koza R Koza RA Rosen CJ September 2015 A High Fat Diet Increases Bone Marrow Adipose Tissue MAT But Does Not Alter Trabecular or Cortical Bone Mass in C57BL 6J Mice Journal of Cellular Physiology 230 9 2032 2037 doi 10 1002 jcp 24954 PMC 4580244 PMID 25663195 a b c Styner M Pagnotti GM Galior K Wu X Thompson WR Uzer G et al August 2015 Exercise Regulation of Marrow Fat in the Setting of PPARg Agonist Treatment in Female C57BL 6 Mice Endocrinology 156 8 2753 2761 doi 10 1210 en 2015 1213 PMC 4511140 PMID 26052898 a b c d e Styner M Pagnotti GM McGrath C Wu X Sen B Uzer G et al August 2017 Exercise Decreases Marrow Adipose Tissue Through ss Oxidation in Obese Running Mice Journal of Bone and Mineral Research 32 8 1692 1702 doi 10 1002 jbmr 3159 PMC 5550355 PMID 28436105 a b Pagnotti GM Styner M Uzer G Patel VS Wright LE Ness KK et al June 2019 Combating osteoporosis and obesity with exercise leveraging cell mechanosensitivity Nature Reviews Endocrinology 15 6 339 355 doi 10 1038 s41574 019 0170 1 PMC 6520125 PMID 30814687 Luu YK Pessin JE Judex S Rubin J Rubin CT April 2009 Mechanical Signals As a Non Invasive Means to Influence Mesenchymal Stem Cell Fate Promoting Bone and Suppressing the Fat Phenotype BoneKEy Osteovision 6 4 132 149 doi 10 1138 20090371 PMC 3255555 PMID 22241295 Belavy DL Quittner MJ Ridgers ND Shiekh A Rantalainen T Trudel G April 2018 Specific Modulation of Vertebral Marrow Adipose Tissue by Physical Activity Journal of Bone and Mineral Research 33 4 651 657 doi 10 1002 jbmr 3357 hdl 10536 DRO DU 30106029 PMID 29336053 a b c Little Letsinger SE Pagnotti GM McGrath C Styner M December 2020 Exercise and Diet Uncovering Prospective Mediators of Skeletal Fragility in Bone and Marrow Adipose Tissue Current Osteoporosis Reports 18 6 774 789 doi 10 1007 s11914 020 00634 y PMC 7736569 PMID 33068251 Southmayd EA Williams NI Mallinson RJ De Souza MJ August 2019 Energy Deficiency Suppresses Bone Turnover in Exercising Women With Menstrual Disturbances The Journal of Clinical Endocrinology and Metabolism 104 8 3131 3145 doi 10 1210 jc 2019 00089 PMID 30896746 McGrath C Little Letsinger SE Sankaran JS Sen B Xie Z Styner MA et al 2022 01 25 Exercise Increases Bone in SEIPIN Deficient Lipodystrophy Despite Low Marrow Adiposity Frontiers in Endocrinology 12 782194 doi 10 3389 fendo 2021 782194 PMC 8822583 PMID 35145475 Krings A Rahman S Huang S Lu Y Czernik PJ Lecka Czernik B February 2012 Bone marrow fat has brown adipose tissue characteristics which are attenuated with aging and diabetes Bone 50 2 546 552 doi 10 1016 j bone 2011 06 016 PMC 3214232 PMID 21723971 a b Suchacki KJ Tavares AA Mattiucci D Scheller EL Papanastasiou G Gray C et al June 2020 Bone marrow adipose tissue is a unique adipose subtype with distinct roles in glucose homeostasis Nature Communications 11 1 3097 Bibcode 2020NatCo 11 3097S doi 10 1038 s41467 020 16878 2 PMC 7303125 PMID 32555194 Craft CS Robles H Lorenz MR Hilker ED Magee KL Andersen TL et al November 2019 Bone marrow adipose tissue does not express UCP1 during development or adrenergic induced remodeling Scientific Reports 9 1 17427 Bibcode 2019NatSR 917427C doi 10 1038 s41598 019 54036 x PMC 6874537 PMID 31758074 Scheller EL Khandaker S Learman BS Cawthorn WP Anderson LM Pham HA et al January 2019 Bone marrow adipocytes resist lipolysis and remodeling in response to b adrenergic stimulation Bone 118 32 41 doi 10 1016 j bone 2018 01 016 PMC 6062480 PMID 29360620 Attane C Esteve D Chaoui K Iacovoni JS Corre J Moutahir M et al January 2020 Human Bone Marrow Is Comprised of Adipocytes with Specific Lipid Metabolism Cell Reports 30 4 949 958 e6 doi 10 1016 j celrep 2019 12 089 PMID 31995765 S2CID 210949460 Ye R Scherer PE April 2013 Adiponectin driver or passenger on the road to insulin sensitivity Molecular Metabolism 2 3 133 141 doi 10 1016 j molmet 2013 04 001 PMC 3773837 PMID 24049728 Tilg H Moschen AR October 2006 Adipocytokines mediators linking adipose tissue inflammation and immunity Nature Reviews Immunology 6 10 772 783 doi 10 1038 nri1937 PMID 16998510 S2CID 29865593 Wronska A Kmiec Z June 2012 Structural and biochemical characteristics of various white adipose tissue depots Acta Physiologica 205 2 194 208 doi 10 1111 j 1748 1716 2012 02409 x PMID 22226221 S2CID 22915022 Masuzaki H Paterson J Shinyama H Morton NM Mullins JJ Seckl JR Flier JS December 2001 A transgenic model of visceral obesity and the metabolic syndrome Science 294 5549 2166 2170 Bibcode 2001Sci 294 2166M doi 10 1126 science 1066285 PMID 11739957 S2CID 768303 Bredella MA Lin E Gerweck AV Landa MG Thomas BJ Torriani M et al November 2012 Determinants of bone microarchitecture and mechanical properties in obese men The Journal of Clinical Endocrinology and Metabolism 97 11 4115 4122 doi 10 1210 jc 2012 2246 PMC 3485587 PMID 22933540 Cohen A Dempster DW Recker RR Lappe JM Zhou H Zwahlen A et al June 2013 Abdominal fat is associated with lower bone formation and inferior bone quality in healthy premenopausal women a transiliac bone biopsy study The Journal of Clinical Endocrinology and Metabolism 98 6 2562 2572 doi 10 1210 jc 2013 1047 PMC 3667251 PMID 23515452 Wu J Cohen P Spiegelman BM February 2013 Adaptive thermogenesis in adipocytes is beige the new brown Genes amp Development 27 3 234 250 doi 10 1101 gad 211649 112 PMC 3576510 PMID 23388824 Al Nbaheen M Vishnubalaji R Ali D Bouslimi A Al Jassir F Megges M et al February 2013 Human stromal mesenchymal stem cells from bone marrow adipose tissue and skin exhibit differences in molecular phenotype and differentiation potential Stem Cell Reviews and Reports 9 1 32 43 doi 10 1007 s12015 012 9365 8 PMC 3563956 PMID 22529014 Gimble JM Zvonic S Floyd ZE Kassem M Nuttall ME May 2006 Playing with bone and fat Journal of Cellular Biochemistry 98 2 251 266 doi 10 1002 jcb 20777 PMID 16479589 S2CID 19746472 Cawthorn WP Scheller EL Learman BS Parlee SD Simon BR Mori H et al August 2014 Bone marrow adipose tissue is an endocrine organ that contributes to increased circulating adiponectin during caloric restriction Cell Metabolism 20 2 368 375 doi 10 1016 j cmet 2014 06 003 PMC 4126847 PMID 24998914 Duque G Li W Adams M Xu S Phipps R May 2011 Effects of risedronate on bone marrow adipocytes in postmenopausal women Osteoporosis International 22 5 1547 1553 doi 10 1007 s00198 010 1353 8 PMID 20661545 S2CID 27850362 Yeung DK Griffith JF Antonio GE Lee FK Woo J Leung PC August 2005 Osteoporosis is associated with increased marrow fat content and decreased marrow fat unsaturation a proton MR spectroscopy study Journal of Magnetic Resonance Imaging 22 2 279 285 doi 10 1002 jmri 20367 PMID 16028245 S2CID 2941430 Li X Kuo D Schafer AL Porzig A Link TM Black D Schwartz AV April 2011 Quantification of vertebral bone marrow fat content using 3 Tesla MR spectroscopy reproducibility vertebral variation and applications in osteoporosis Journal of Magnetic Resonance Imaging 33 4 974 979 doi 10 1002 jmri 22489 PMC 3072841 PMID 21448966 Syed FA Oursler MJ Hefferanm TE Peterson JM Riggs BL Khosla S September 2008 Effects of estrogen therapy on bone marrow adipocytes in postmenopausal osteoporotic women Osteoporosis International 19 9 1323 1330 doi 10 1007 s00198 008 0574 6 PMC 2652842 PMID 18274695 Khosla S Riggs BL Atkinson EJ Oberg AL McDaniel LJ Holets M et al January 2006 Effects of sex and age on bone microstructure at the ultradistal radius a population based noninvasive in vivo assessment Journal of Bone and Mineral Research 21 1 124 131 doi 10 1359 jbmr 050916 PMC 1352156 PMID 16355281 Glatt V Canalis E Stadmeyer L Bouxsein ML August 2007 Age related changes in trabecular architecture differ in female and male C57BL 6J mice Journal of Bone and Mineral Research 22 8 1197 1207 doi 10 1359 jbmr 070507 PMID 17488199 Tchkonia T Morbeck DE Von Zglinicki T Van Deursen J Lustgarten J Scrable H et al October 2010 Fat tissue aging and cellular senescence Aging Cell 9 5 667 684 doi 10 1111 j 1474 9726 2010 00608 x PMC 2941545 PMID 20701600 Kassem M Marie PJ April 2011 Senescence associated intrinsic mechanisms of osteoblast dysfunctions Aging Cell 10 2 191 197 doi 10 1111 j 1474 9726 2011 00669 x PMID 21210937 Moerman EJ Teng K Lipschitz DA Lecka Czernik B December 2004 Aging activates adipogenic and suppresses osteogenic programs in mesenchymal marrow stroma stem cells the role of PPAR gamma2 transcription factor and TGF beta BMP signaling pathways Aging Cell 3 6 379 389 doi 10 1111 j 1474 9728 2004 00127 x PMC 1850101 PMID 15569355 Stevens JR Miranda Carboni GA Singer MA Brugger SM Lyons KM Lane TF October 2010 Wnt10b deficiency results in age dependent loss of bone mass and progressive reduction of mesenchymal progenitor cells Journal of Bone and Mineral Research 25 10 2138 2147 doi 10 1002 jbmr 118 PMC 3153316 PMID 20499361 Bennett CN Ouyang H Ma YL Zeng Q Gerin I Sousa KM et al December 2007 Wnt10b increases postnatal bone formation by enhancing osteoblast differentiation Journal of Bone and Mineral Research 22 12 1924 1932 doi 10 1359 jbmr 070810 PMID 17708715 Chen Q Shou P Zheng C Jiang M Cao G Yang Q et al July 2016 Fate decision of mesenchymal stem cells adipocytes or osteoblasts Cell Death and Differentiation 23 7 1128 1139 doi 10 1038 cdd 2015 168 PMC 4946886 PMID 26868907 Zhou BO Yu H Yue R Zhao Z Rios JJ Naveiras O Morrison SJ August 2017 Bone marrow adipocytes promote the regeneration of stem cells and haematopoiesis by secreting SCF Nature Cell Biology 19 8 891 903 doi 10 1038 ncb3570 PMC 5536858 PMID 28714970 Bielohuby M Matsuura M Herbach N Kienzle E Slawik M Hoeflich A Bidlingmaier M February 2010 Short term exposure to low carbohydrate high fat diets induces low bone mineral density and reduces bone formation in rats Journal of Bone and Mineral Research 25 2 275 284 doi 10 1359 jbmr 090813 PMID 19653818 Spatz JM Ellman R Cloutier AM Louis L van Vliet M Suva LJ et al April 2013 Sclerostin antibody inhibits skeletal deterioration due to reduced mechanical loading Journal of Bone and Mineral Research 28 4 865 874 doi 10 1002 jbmr 1807 PMC 4076162 PMID 23109229 Rosen CJ Ackert Bicknell CL Adamo ML Shultz KL Rubin J Donahue LR et al November 2004 Congenic mice with low serum IGF I have increased body fat reduced bone mineral density and an altered osteoblast differentiation program Bone 35 5 1046 1058 doi 10 1016 j bone 2004 07 008 PMID 15542029 Naveiras O Nardi V Wenzel PL Hauschka PV Fahey F Daley GQ July 2009 Bone marrow adipocytes as negative regulators of the haematopoietic microenvironment Nature 460 7252 259 263 Bibcode 2009Natur 460 259N doi 10 1038 nature08099 PMC 2831539 PMID 19516257 David V Martin A Lafage Proust MH Malaval L Peyroche S Jones DB et al May 2007 Mechanical loading down regulates peroxisome proliferator activated receptor gamma in bone marrow stromal cells and favors osteoblastogenesis at the expense of adipogenesis Endocrinology 148 5 2553 2562 doi 10 1210 en 2006 1704 PMID 17317771 Majka SM Miller HL Sullivan T Erickson PF Kong R Weiser Evans M et al October 2012 Adipose lineage specification of bone marrow derived myeloid cells Adipocyte 1 4 215 229 doi 10 4161 adip 21496 PMC 3609111 PMID 23700536 Majka SM Miller HL Helm KM Acosta AS Childs CR Kong R Klemm DJ 2014 Analysis and Isolation of Adipocytes by Flow Cytometry Methods of Adipose Tissue Biology Part A Methods in Enzymology Vol 537 pp 281 96 doi 10 1016 b978 0 12 411619 1 00015 x ISBN 9780124116191 PMC 4143162 PMID 24480352 Bernstein RL Hyun WC Davis JH Fulwyler MJ Pershadsingh HA July 1989 Flow cytometric analysis of mature adipocytes Cytometry 10 4 469 474 doi 10 1002 cyto 990100416 PMID 2766892 Bredella MA Torriani M Ghomi RH Thomas BJ Brick DJ Gerweck AV et al January 2011 Vertebral bone marrow fat is positively associated with visceral fat and inversely associated with IGF 1 in obese women Obesity 19 1 49 53 doi 10 1038 oby 2010 106 PMC 3593350 PMID 20467419 de Paula FJ Dick de Paula I Bornstein S Rostama B Le P Lotinun S et al September 2011 VDR haploinsufficiency impacts body composition and skeletal acquisition in a gender specific manner Calcified Tissue International 89 3 179 191 doi 10 1007 s00223 011 9505 1 PMC 3157554 PMID 21637996 Fazeli PK Bredella MA Freedman L Thomas BJ Breggia A Meenaghan E et al September 2012 Marrow fat and preadipocyte factor 1 levels decrease with recovery in women with anorexia nervosa Journal of Bone and Mineral Research 27 9 1864 1871 doi 10 1002 jbmr 1640 PMC 3415584 PMID 22508185 Rantalainen T Nikander R Heinonen A Cervinka T Sievanen H Daly RM May 2013 Differential effects of exercise on tibial shaft marrow density in young female athletes The Journal of Clinical Endocrinology and Metabolism 98 5 2037 2044 doi 10 1210 jc 2012 3748 hdl 10536 DRO DU 30060423 PMID 23616150 Scheller EL Troiano N Vanhoutan JN Bouxsein MA Fretz JA Xi Y et al 2014 Use of Osmium Tetroxide Staining with Microcomputerized Tomography to Visualize and Quantify Bone Marrow Adipose Tissue in Vivo Methods of Adipose Tissue Biology Part A Methods in Enzymology Vol 537 pp 123 39 doi 10 1016 b978 0 12 411619 1 00007 0 ISBN 9780124116191 PMC 4097010 PMID 24480344 Pham TT Ivaska KK Hannukainen JC Virtanen KA Lidell ME Enerback S et al July 2020 Human Bone Marrow Adipose Tissue is a Metabolically Active and Insulin Sensitive Distinct Fat Depot The Journal of Clinical Endocrinology and Metabolism 105 7 2300 2310 doi 10 1210 clinem dgaa216 PMC 7247553 PMID 32311037 The International Bone Marrow Adiposity Society BMAS Website Tratwal J Labella R Bravenboer N Kerckhofs G Douni E Scheller EL et al 2020 Reporting Guidelines Review of Methodological Standards and Challenges Toward Harmonization in Bone Marrow Adiposity Research Report of the Methodologies Working Group of the International Bone Marrow Adiposity Society Frontiers in Endocrinology 11 65 doi 10 3389 fendo 2020 00065 PMC 7059536 PMID 32180758 Lucas S Tencerova M von der Weid B Andersen TL Attane C Behler Janbeck F et al 2021 Guidelines for Biobanking of Bone Marrow Adipose Tissue and Related Cell Types Report of the Biobanking Working Group of the International Bone Marrow Adiposity Society Frontiers in Endocrinology 12 744527 doi 10 3389 fendo 2021 744527 PMC 8503265 PMID 34646237 Hardouin P Marie PJ Rosen CJ December 2016 New insights into bone marrow adipocytes Report from the First European Meeting on Bone Marrow Adiposity BMA 2015 Bone 93 212 215 doi 10 1016 j bone 2015 11 013 PMID 26608519 van der Eerden B van Wijnen A October 2017 Meeting report of the 2016 bone marrow adiposity meeting Adipocyte 6 4 304 313 doi 10 1080 21623945 2017 1313374 PMC 5736244 PMID 28410005 Corsi A Palmisano B Tratwal J Riminucci M Naveiras O 2019 Brief Report From the 3rd International Meeting on Bone Marrow Adiposity BMA 2017 Frontiers in Endocrinology 10 336 doi 10 3389 fendo 2019 00336 PMC 6546805 PMID 31191458 Penel G Kerckhofs G Chauveau C 2019 Brief Report From the 4th International Meeting on Bone Marrow Adiposity BMA2018 Frontiers in Endocrinology 10 691 doi 10 3389 fendo 2019 00691 PMC 6813723 PMID 31681168 Scheller EL McGee Lawrence ME Lecka Czernik B 2021 Report From the 6th International Meeting on Bone Marrow Adiposity BMA2020 Frontiers in Endocrinology 12 712088 doi 10 3389 fendo 2021 712088 PMC 8323480 PMID 34335478 Labella R Little Letsinger S Avilkina V Sarkis R Tencerova M Vlug A Palmisano B 2022 Next Generation Bone Marrow Adiposity Researchers Report From the 1st BMAS Summer School 2021 Frontiers in Endocrinology 13 879588 doi 10 3389 fendo 2022 879588 PMC 9043644 PMID 35498418 Further reading edit Bone marrow fat tissue secretes hormone that helps body stay healthy University of Michigan 3 July 2014 Archived from the original on 15 March 2015 Another reason to exercise Burning bone fat a key to better bone health Science Daily 18 May 2017 Why are our bones full of fat The secrets of bone marrow adipose tissue Society for Endocrinology Winter 2017 Retrieved from https en wikipedia org w index php title Bone marrow adipose tissue amp oldid 1203775472, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.