Skip to main content

Advertisement

Log in

Marrow Adiposity and Hematopoiesis in Aging and Obesity: Exercise as an Intervention

  • Bone Marrow and Adipose Tissue (G Duque and B Lecka-Czernik, Section Editors)
  • Published:
Current Osteoporosis Reports Aims and scope Submit manuscript

Abstract

Purpose of Review

Changes in the bone marrow microenvironment, which accompany aging and obesity, including increased marrow adiposity, can compromise hematopoiesis. Here, we review deleterious shifts in molecular, cellular, and tissue activity and consider the potential of exercise to slow degenerative changes associated with aging and obesity.

Recent Findings

While bone marrow hematopoietic stem cells (HSC) are increased in frequency and myeloid-biased with age, the effect of obesity on HSC proliferation and differentiation remains controversial. HSC from both aged and obese environment have reduced hematopoietic reconstitution capacity following bone marrow transplant. Increased marrow adiposity affects HSC function, causing upregulation of myelopoiesis and downregulation of lymphopoiesis. Exercise, in contrast, can reduce marrow adiposity and restore hematopoiesis.

Summary

The impact of marrow adiposity on hematopoiesis is determined mainly through correlations. Mechanistic studies are needed to determine a causative relationship between marrow adiposity and declines in hematopoiesis, which could aid in developing treatments for conditions that arise from disruptions in the marrow microenvironment.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance ••Of major importance

  1. Travlos GS. Normal structure, function, and histology of the bone marrow. Toxicol Pathol. 2006;34(5):548–65. https://doi.org/10.1080/01926230600939856.

    Article  PubMed  Google Scholar 

  2. Augello A, De Bari C. The regulation of differentiation in mesenchymal stem cells. Hum Gene Ther. 2010;21(10):1226–38. https://doi.org/10.1089/hum.2010.173.

    Article  CAS  PubMed  Google Scholar 

  3. Luu YK, Capilla E, Rosen CJ, Gilsanz V, Pessin JE, Judex S, et al. Mechanical stimulation of mesenchymal stem cell proliferation and differentiation promotes osteogenesis while preventing dietary-induced obesity. J Bone Miner Res. 2009;24(1):50–61. https://doi.org/10.1359/jbmr.080817.

    Article  CAS  PubMed  Google Scholar 

  4. Alvarez-Viejo M, Menendez-Menendez Y, Blanco-Gelaz MA, Ferrero-Gutierrez A, Fernandez-Rodriguez MA, Gala J, et al. Quantifying mesenchymal stem cells in the mononuclear cell fraction of bone marrow samples obtained for cell therapy. Transplant Proc. 2013;45(1):434–9. https://doi.org/10.1016/j.transproceed.2012.05.091.

    Article  CAS  PubMed  Google Scholar 

  5. •• Patel VS, Chan ME, Pagnotti GM, Frechette DM, Rubin J, Rubin CT. Incorporating refractory period in mechanical stimulation mitigates obesity-induced adipose tissue dysfunction in adult mice. Obesity (Silver Spring). 2017;25(10):1745–53. https://doi.org/10.1002/oby.21958. This study demonstrates that obesity leads to systemic chronic inflammatory state by inducing increased infiltration of immune cells in the adipose tissue, which is restored via whole-body vibration. In addition, this study emphasizes that in adults, whole-body vibration treatment is only effective when it is separated with a rest period.

    Article  CAS  Google Scholar 

  6. Wang Y, Yu X, Chen E, Li L. Liver-derived human mesenchymal stem cells: a novel therapeutic source for liver diseases. Stem Cell Res Ther. 2016;7(1):71. https://doi.org/10.1186/s13287-016-0330-3.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Seeberger KL, Dufour JM, Shapiro AM, Lakey JR, Rajotte RV, Korbutt GS. Expansion of mesenchymal stem cells from human pancreatic ductal epithelium. Lab Investig. 2006;86(2):141–53. https://doi.org/10.1038/labinvest.3700377.

    Article  CAS  PubMed  Google Scholar 

  8. Yin L, Zhu Y, Yang J, Ni Y, Zhou Z, Chen Y, et al. Adipose tissue-derived mesenchymal stem cells differentiated into hepatocyte-like cells in vivo and in vitro. Mol Med Rep. 2015;11(3):1722–32. https://doi.org/10.3892/mmr.2014.2935.

    Article  CAS  PubMed  Google Scholar 

  9. Rubin CT, Capilla E, Luu YK, Busa B, Crawford H, Nolan DJ, et al. Adipogenesis is inhibited by brief, daily exposure to high-frequency, extremely low-magnitude mechanical signals. Proc Natl Acad Sci U S A. 2007;104(45):17879–84. https://doi.org/10.1073/pnas.0708467104.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. da Silva SV, Renovato-Martins M, Ribeiro-Pereira C, Citelli M, Barja-Fidalgo C. Obesity modifies bone marrow microenvironment and directs bone marrow mesenchymal cells to adipogenesis. Obesity (Silver Spring). 2016;24(12):2522–32. https://doi.org/10.1002/oby.21660.

    Article  Google Scholar 

  11. Chan ME, Adler BJ, Green DE, Rubin CT. Bone structure and B-cell populations, crippled by obesity, are partially rescued by brief daily exposure to low-magnitude mechanical signals. FASEB J. 2012;26(12):4855–63. https://doi.org/10.1096/fj.12-209841.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. •• Adler BJ, Green DE, Pagnotti GM, Chan ME, Rubin CT. High fat diet rapidly suppresses B lymphopoiesis by disrupting the supportive capacity of the bone marrow niche. PLoS One. 2014;9(3):e90639. https://doi.org/10.1371/journal.pone.0090639. This paper demonstrates that high-fat diet leads to increase in marrow adiposity, paralleled by reduced B lymphopoiesis and increased myelopoiesis.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Orkin SH, Zon LI. Hematopoiesis: an evolving paradigm for stem cell biology. Cell. 2008;132(4):631–44. https://doi.org/10.1016/j.cell.2008.01.025.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Notta F, Doulatov S, Laurenti E, Poeppl A, Jurisica I, Dick JE. Isolation of single human hematopoietic stem cells capable of long-term multilineage engraftment. Science. 2011;333(6039):218–21. https://doi.org/10.1126/science.1201219.

    Article  CAS  PubMed  Google Scholar 

  15. Tesio M, Tang Y, Mudder K, Saini M, von Paleske L, Macintyre E, et al. Hematopoietic stem cell quiescence and function are controlled by the CYLD-TRAF2-p38MAPK pathway. J Exp Med. 2015;212(4):525–38. https://doi.org/10.1084/jem.20141438.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Challen GA, Boles N, Lin KK, Goodell MA. Mouse hematopoietic stem cell identification and analysis. Cytometry A. 2009;75(1):14–24. https://doi.org/10.1002/cyto.a.20674.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Pang WW, Price EA, Sahoo D, Beerman I, Maloney WJ, Rossi DJ, et al. Human bone marrow hematopoietic stem cells are increased in frequency and myeloid-biased with age. Proc Natl Acad Sci U S A. 2011;108(50):20012–7. https://doi.org/10.1073/pnas.1116110108.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Franco RS. Measurement of red cell lifespan and aging. Transfus Med Hemother. 2012;39(5):302–7. https://doi.org/10.1159/000342232.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Dzierzak E, Philipsen S. Erythropoiesis: development and differentiation. Cold Spring Harb Perspect Med. 2013;3(4):a011601. https://doi.org/10.1101/cshperspect.a011601.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Cohen JA, Leeksma CH. Determination of the life span of human blood platelets using labelled diisopropylfluorophosphonate. J Clin Investig. 1956;35(9):964–9. https://doi.org/10.1172/jci103356.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Thon JN, Italiano JE. Platelet formation. Semin Hematol. 2010;47(3):220–6. https://doi.org/10.1053/j.seminhematol.2010.03.005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Sprent J, Tough DF. Lymphocyte life-span and memory. Science. 1994;265(5177):1395–400.

    Article  CAS  PubMed  Google Scholar 

  23. Pillay J, den Braber I, Vrisekoop N, Kwast LM, de Boer RJ, Borghans JA, et al. In vivo labeling with 2H2O reveals a human neutrophil lifespan of 5.4 days. Blood. 2010;116(4):625–7. https://doi.org/10.1182/blood-2010-01-259028.

    Article  CAS  PubMed  Google Scholar 

  24. Summers C, Rankin SM, Condliffe AM, Singh N, Peters AM, Chilvers ER. Neutrophil kinetics in health and disease. Trends Immunol. 2010;31(8):318–24. https://doi.org/10.1016/j.it.2010.05.006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Park YM, Bochner BS. Eosinophil survival and apoptosis in health and disease. Allergy Asthma Immunol Res. 2010;2(2):87–101. https://doi.org/10.4168/aair.2010.2.2.87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Min B, Brown MA, Legros G. Understanding the roles of basophils: breaking dawn. Immunology. 2012;135(3):192–7. https://doi.org/10.1111/j.1365-2567.2011.03530.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Janssen WJ, Bratton DL, Jakubzick CV, Henson PM. Myeloid cell turnover and clearance. Microbiol Spectr. 2016;4(6). https://doi.org/10.1128/microbiolspec.MCHD-0005-2015.

  28. Velten L, Haas SF, Raffel S, Blaszkiewicz S, Islam S, Hennig BP, et al. Human haematopoietic stem cell lineage commitment is a continuous process. Nat Cell Biol. 2017;19(4):271–81. https://doi.org/10.1038/ncb3493.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Majumdar MK, Thiede MA, Haynesworth SE, Bruder SP, Gerson SL. Human marrow-derived mesenchymal stem cells (MSCs) express hematopoietic cytokines and support long-term hematopoiesis when differentiated toward stromal and osteogenic lineages. J Hematother Stem Cell Res. 2000;9(6):841–8. https://doi.org/10.1089/152581600750062264.

    Article  CAS  PubMed  Google Scholar 

  30. Mishima S, Nagai A, Abdullah S, Matsuda C, Taketani T, Kumakura S, et al. Effective ex vivo expansion of hematopoietic stem cells using osteoblast-differentiated mesenchymal stem cells is CXCL12 dependent. Eur J Haematol. 2010;84(6):538–46. https://doi.org/10.1111/j.1600-0609.2010.01419.x.

    Article  CAS  PubMed  Google Scholar 

  31. Carrancio S, Blanco B, Romo C, Muntion S, Lopez-Holgado N, Blanco JF, et al. Bone marrow mesenchymal stem cells for improving hematopoietic function: an in vitro and in vivo model. Part 2: effect on bone marrow microenvironment. PLoS One. 2011;6(10):e26241. https://doi.org/10.1371/journal.pone.0026241.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Bartholomew A, Sturgeon C, Siatskas M, Ferrer K, McIntosh K, Patil S, et al. Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo. Exp Hematol. 2002;30(1):42–8.

    Article  PubMed  Google Scholar 

  33. Nemeth K, Leelahavanichkul A, Yuen PS, Mayer B, Parmelee A, Doi K, et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med. 2009;15(1):42–9. https://doi.org/10.1038/nm.1905.

    Article  CAS  PubMed  Google Scholar 

  34. Roddy GW, Oh JY, Lee RH, Bartosh TJ, Ylostalo J, Coble K, et al. Action at a distance: systemically administered adult stem/progenitor cells (MSCs) reduce inflammatory damage to the cornea without engraftment and primarily by secretion of TNF-alpha stimulated gene/protein 6. Stem Cells. 2011;29(10):1572–9. https://doi.org/10.1002/stem.708.

    Article  CAS  PubMed  Google Scholar 

  35. Taichman RS, Emerson SG. The role of osteoblasts in the hematopoietic microenvironment. Stem Cells. 1998;16(1):7–15. https://doi.org/10.1002/stem.160007.

    Article  CAS  PubMed  Google Scholar 

  36. Takahashi N, Udagawa N, Akatsu T, Tanaka H, Shionome M, Suda T. Role of colony-stimulating factors in osteoclast development. J Bone Miner Res. 1991;6(9):977–85. https://doi.org/10.1002/jbmr.5650060912.

    Article  CAS  PubMed  Google Scholar 

  37. Marks SC Jr, Mackay CA, Jackson ME, Larson EK, Cielinski MJ, Stanley ER, et al. The skeletal effects of colony-stimulating factor-1 in toothless (osteopetrotic) rats: persistent metaphyseal sclerosis and the failure to restore subepiphyseal osteoclasts. Bone. 1993;14(4):675–80.

    Article  PubMed  Google Scholar 

  38. Sanchez-Fernandez MA, Gallois A, Riedl T, Jurdic P, Hoflack B. Osteoclasts control osteoblast chemotaxis via PDGF-BB/PDGF receptor beta signaling. PLoS One. 2008;3(10):e3537. https://doi.org/10.1371/journal.pone.0003537.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Bonewald LF. The amazing osteocyte. J Bone Miner Res. 2011;26(2):229–38. https://doi.org/10.1002/jbmr.320.

    Article  CAS  PubMed  Google Scholar 

  40. Fulzele K, Krause DS, Panaroni C, Saini V, Barry KJ, Liu X, et al. Myelopoiesis is regulated by osteocytes through Gsα-dependent signaling. Blood. 2013;121(6):930–9. https://doi.org/10.1182/blood-2012-06-437160.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Yoneshiro T, Aita S, Matsushita M, Okamatsu-Ogura Y, Kameya T, Kawai Y, et al. Age-related decrease in cold-activated brown adipose tissue and accumulation of body fat in healthy humans. Obesity (Silver Spring). 2011;19(9):1755–60. https://doi.org/10.1038/oby.2011.125.

    Article  Google Scholar 

  42. Trayhurn P, Beattie JH. Physiological role of adipose tissue: white adipose tissue as an endocrine and secretory organ. Proc Nutr Soc. 2001;60(3):329–39.

    Article  CAS  PubMed  Google Scholar 

  43. Spalding KL, Arner E, Westermark PO, Bernard S, Buchholz BA, Bergmann O, et al. Dynamics of fat cell turnover in humans. Nature. 2008;453(7196):783–7. https://doi.org/10.1038/nature06902.

    Article  CAS  PubMed  Google Scholar 

  44. 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.

    Article  CAS  PubMed  Google Scholar 

  45. Moore SG, Dawson KL. Red and yellow marrow in the femur: age-related changes in appearance at MR imaging. Radiology. 1990;175(1):219–23. https://doi.org/10.1148/radiology.175.1.2315484.

    Article  CAS  PubMed  Google Scholar 

  46. Scheller EL, Rosen CJ. What’s the matter with MAT? Marrow adipose tissue, metabolism, and skeletal health. Ann N Y Acad Sci. 2014;1311:14–30. https://doi.org/10.1111/nyas.12327.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Huggins C, Blocksom BH. Changes in outlying bone marrow accompanying a local increase of temperature within physiological limits. J Exp Med. 1936;64(2):253–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Naveiras O, Nardi V, Wenzel PL, Hauschka PV, Fahey F, Daley GQ. Bone-marrow adipocytes as negative regulators of the haematopoietic microenvironment. Nature. 2009;460(7252):259–63. https://doi.org/10.1038/nature08099.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Scheller EL, Doucette CR, Learman BS, Cawthorn WP, Khandaker S, Schell B, et al. Corrigendum: region-specific variation in the properties of skeletal adipocytes reveals regulated and constitutive marrow adipose tissues. Nat Commun. 2016;7:13775. https://doi.org/10.1038/ncomms13775.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Lihn AS, Pedersen SB, Richelsen B. Adiponectin: action, regulation and association to insulin sensitivity. Obes Rev. 2005;6(1):13–21. https://doi.org/10.1111/j.1467-789X.2005.00159.x.

    Article  CAS  PubMed  Google Scholar 

  51. 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. https://doi.org/10.1016/j.cmet.2014.06.003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Suchacki KJ, Cawthorn WP, Rosen CJ. Bone marrow adipose tissue: formation, function and regulation. Curr Opin Pharmacol. 2016;28:50–6. https://doi.org/10.1016/j.coph.2016.03.001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Keune JA, Wong CP, Branscum AJ, Iwaniec UT, Turner RT. Bone marrow adipose tissue deficiency increases disuse-induced bone loss in male mice. Sci Rep. 2017;7:46325. https://doi.org/10.1038/srep46325.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Ogawa T, Kitagawa M, Hirokawa K. Age-related changes of human bone marrow: a histometric estimation of proliferative cells, apoptotic cells, T cells, B cells and macrophages. Mech Ageing Dev. 2000;117(1–3):57–68.

    Article  CAS  PubMed  Google Scholar 

  55. Pearce DJ, Anjos-Afonso F, Ridler CM, Eddaoudi A, Bonnet D. Age-dependent increase in side population distribution within hematopoiesis: implications for our understanding of the mechanism of aging. Stem Cells. 2007;25(4):828–35. https://doi.org/10.1634/stemcells.2006-0405.

    Article  CAS  PubMed  Google Scholar 

  56. Yahata T, Takanashi T, Muguruma Y, Ibrahim AA, Matsuzawa H, Uno T, et al. Accumulation of oxidative DNA damage restricts the self-renewal capacity of human hematopoietic stem cells. Blood. 2011;118(11):2941–50. https://doi.org/10.1182/blood-2011-01-330050.

    Article  CAS  PubMed  Google Scholar 

  57. Kuranda K, Vargaftig J, de la Rochere P, Dosquet C, Charron D, Bardin F, et al. Age-related changes in human hematopoietic stem/progenitor cells. Aging Cell. 2011;10(3):542–6. https://doi.org/10.1111/j.1474-9726.2011.00675.x.

    Article  CAS  PubMed  Google Scholar 

  58. Ambrosi TH, Scialdone A, Graja A, Gohlke S, Jank AM, Bocian C, et al. Adipocyte accumulation in the bone marrow during obesity and aging impairs stem cell-based hematopoietic and bone regeneration. Cell Stem Cell. 2017;20(6):771–84.e6. https://doi.org/10.1016/j.stem.2017.02.009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Griffith JF, Yeung DK, Ma HT, Leung JC, Kwok TC, Leung PC. Bone marrow fat content in the elderly: a reversal of sex difference seen in younger subjects. J Magn Reson Imaging. 2012;36(1):225–30. https://doi.org/10.1002/jmri.23619.

    Article  PubMed  Google Scholar 

  60. Tuljapurkar SR, McGuire TR, Brusnahan SK, Jackson JD, Garvin KL, Kessinger MA, et al. Changes in human bone marrow fat content associated with changes in hematopoietic stem cell numbers and cytokine levels with aging. J Anat. 2011;219(5):574–81. https://doi.org/10.1111/j.1469-7580.2011.01423.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Reinisch A, Etchart N, Thomas D, Hofmann NA, Fruehwirth M, Sinha S, et al. Epigenetic and in vivo comparison of diverse MSC sources reveals an endochondral signature for human hematopoietic niche formation. Blood. 2015;125(2):249–60. https://doi.org/10.1182/blood-2014-04-572255.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Trottier MD, Naaz A, Li Y, Fraker PJ. Enhancement of hematopoiesis and lymphopoiesis in diet-induced obese mice. Proc Natl Acad Sci U S A. 2012;109(20):7622–9. https://doi.org/10.1073/pnas.1205129109.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. do Carmo LS, Rogero MM, Paredes-Gamero EJ, Nogueira-Pedro A, Xavier JG, Cortez M, et al. A high-fat diet increases interleukin-3 and granulocyte colony-stimulating factor production by bone marrow cells and triggers bone marrow hyperplasia and neutrophilia in Wistar rats. Exp Biol Med (Maywood). 2013;238(4):375–84. https://doi.org/10.1177/1535370213477976.

    Article  Google Scholar 

  64. van den Berg SM, Seijkens TT, Kusters PJ, Beckers L, den Toom M, Smeets E, et al. Diet-induced obesity in mice diminishes hematopoietic stem and progenitor cells in the bone marrow. FASEB J. 2016;30(5):1779–88. https://doi.org/10.1096/fj.201500175.

    Article  PubMed  Google Scholar 

  65. Singer K, DelProposto J, Morris DL, Zamarron B, Mergian T, Maley N, et al. Diet-induced obesity promotes myelopoiesis in hematopoietic stem cells. Mol Metab. 2014;3(6):664–75. https://doi.org/10.1016/j.molmet.2014.06.005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Takahashi K, Mizuarai S, Araki H, Mashiko S, Ishihara A, Kanatani A, et al. Adiposity elevates plasma MCP-1 levels leading to the increased CD11b-positive monocytes in mice. J Biol Chem. 2003;278(47):46654–60. https://doi.org/10.1074/jbc.M309895200.

    Article  CAS  PubMed  Google Scholar 

  67. Nanji AA, Freeman JB. Relationship between body weight and total leukocyte count in morbid obesity. Am J Clin Pathol. 1985;84(3):346–7.

    Article  CAS  PubMed  Google Scholar 

  68. Womack J, Tien PC, Feldman J, Shin JH, Fennie K, Anastos K, et al. Obesity and immune cell counts in women. Metabolism. 2007;56(7):998–1004. https://doi.org/10.1016/j.metabol.2007.03.008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Nteeba J, Ortinau LC, Perfield JW 2nd, Keating AF. Diet-induced obesity alters immune cell infiltration and expression of inflammatory cytokine genes in mouse ovarian and peri-ovarian adipose depot tissues. Mol Reprod Dev. 2013;80(11):948–58. https://doi.org/10.1002/mrd.22231.

    Article  CAS  PubMed  Google Scholar 

  70. Caer C, Rouault C, Le Roy T, Poitou C, Aron-Wisnewsky J, Torcivia A, et al. Immune cell-derived cytokines contribute to obesity-related inflammation, fibrogenesis and metabolic deregulation in human adipose tissue. Sci Rep. 2017;7(1):3000. https://doi.org/10.1038/s41598-017-02660-w.

    Article  PubMed  PubMed Central  Google Scholar 

  71. 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. https://doi.org/10.1002/jcp.24954.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. 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 (Lausanne). 2016;7:102. https://doi.org/10.3389/fendo.2016.00102.

    Google Scholar 

  73. •• 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. https://doi.org/10.1016/j.bone.2014.03.044. This paper demonstrates that high-fat diet leads to increased marrow adiposity, which gets suppressed by exercise.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Bredella MA, Torriani M, Ghomi RH, Thomas BJ, Brick DJ, Gerweck AV, et al. Vertebral bone marrow fat is positively associated with visceral fat and inversely associated with IGF-1 in obese women. Obesity (Silver Spring). 2011;19(1):49–53. https://doi.org/10.1038/oby.2010.106.

    Article  CAS  Google Scholar 

  75. Caselli A, Olson TS, Otsuru S, Chen X, Hofmann TJ, Nah HD, et al. IGF-1-mediated osteoblastic niche expansion enhances long-term hematopoietic stem cell engraftment after murine bone marrow transplantation. Stem Cells. 2013;31(10):2193–204. https://doi.org/10.1002/stem.1463.

    Article  CAS  PubMed  Google Scholar 

  76. Zhou D, Deoliveira D, Kang Y, Choi SS, Li Z, Chao NJ, et al. Insulin-like growth factor 1 mitigates hematopoietic toxicity after lethal total body irradiation. Int J Radiat Oncol Biol Phys. 2013;85(4):1141–8. https://doi.org/10.1016/j.ijrobp.2012.08.014.

    Article  CAS  PubMed  Google Scholar 

  77. Liang X, Su YP, Kong PY, Zeng DF, Chen XH, Peng XG, et al. Human bone marrow mesenchymal stem cells expressing SDF-1 promote hematopoietic stem cell function of human mobilised peripheral blood CD34+ cells in vivo and in vitro. Int J Radiat Biol. 2010;86(3):230–7. https://doi.org/10.3109/09553000903422555.

    Article  CAS  PubMed  Google Scholar 

  78. Lapidot T, Kollet O. The essential roles of the chemokine SDF-1 and its receptor CXCR4 in human stem cell homing and repopulation of transplanted immune-deficient NOD/SCID and NOD/SCID/B2m(null) mice. Leukemia. 2002;16(10):1992–2003. https://doi.org/10.1038/sj.leu.2402684.

    Article  CAS  PubMed  Google Scholar 

  79. Takeshita S, Fumoto T, Naoe Y, Ikeda K. Age-related marrow adipogenesis is linked to increased expression of RANKL. J Biol Chem. 2014;289(24):16699–710. https://doi.org/10.1074/jbc.M114.547919.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Holt V, Caplan AI, Haynesworth SE. Identification of a subpopulation of marrow MSC-derived medullary adipocytes that express osteoclast-regulating molecules: marrow adipocytes express osteoclast mediators. PLoS One. 2014;9(10):e108920. https://doi.org/10.1371/journal.pone.0108920.

    Article  PubMed  PubMed Central  Google Scholar 

  81. • Kennedy DE, Knight KL. Inhibition of B lymphopoiesis by adipocytes and IL-1-producing myeloid-derived suppressor cells. J Immunol. 2015;195(6):2666–74. https://doi.org/10.4049/jimmunol.1500957. This study demonstrates that MAT-induced depletion in B lymphopoiesis is mediated by secretion of IL-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. • Huang JY, Zhou QL, Huang CH, Song Y, Sharma AG, Liao Z, et al. Neutrophil elastase regulates emergency myelopoiesis preceding systemic inflammation in diet-induced obesity. J Biol Chem. 2017;292(12):4770–6. https://doi.org/10.1074/jbc.C116.758748. This study demonstrates that obesity-induced myeloid bias is mediated via secretion of neutrophil elastase.

    Article  CAS  PubMed  Google Scholar 

  83. Walji TA, Turecamo SE, Sanchez AC, Anthony BA, Abou-Ezzi G, Scheller EL, et al. Marrow adipose tissue expansion coincides with insulin resistance in MAGP1-deficient mice. Front Endocrinol (Lausanne). 2016;7:87. https://doi.org/10.3389/fendo.2016.00087.

    PubMed Central  Google Scholar 

  84. 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. https://doi.org/10.1002/jcp.25976.

    Article  CAS  PubMed  Google Scholar 

  85. Ma YH, Schwartz AV, Sigurdsson S, Hue TF, Lang TF, Harris TB, et al. Circulating sclerostin associated with vertebral bone marrow fat in older men but not women. J Clin Endocrinol Metab. 2014;99(12):E2584–90. https://doi.org/10.1210/jc.2013-4493.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Kohli SS, Kohli VS. Role of RANKL-RANK/osteoprotegerin molecular complex in bone remodeling and its immunopathologic implications. Indian J Endocrinol Metab. 2011;15(3):175–81. https://doi.org/10.4103/2230-8210.83401.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. • 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; https://doi.org/10.1002/jbmr.3159. This study demonstrates that reduction in high-fat diet-induced accumulation in marrow adiposity during exercise is mediated via basal lyposis and β-oxidation, which is partially mediated by perilipin 3.

  88. Shen W, Chen J, Gantz M, Punyanitya M, Heymsfield SB, Gallagher D, et al. MRI-measured pelvic bone marrow adipose tissue is inversely related to DXA-measured bone mineral in younger and older adults. Eur J Clin Nutr. 2012;66(9):983–8. https://doi.org/10.1038/ejcn.2012.35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. 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;32:1692–702. https://doi.org/10.1002/jbmr.3159.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Rantalainen T, Nikander R, Heinonen A, Cervinka T, Sievanen H, Daly RM. Differential effects of exercise on tibial shaft marrow density in young female athletes. J Clin Endocrinol Metab. 2013;98(5):2037–44. https://doi.org/10.1210/jc.2012-3748.

    Article  CAS  PubMed  Google Scholar 

  91. Covington JD, Noland RC, Hebert RC, Masinter BS, Smith SR, Rustan AC, et al. Perilipin 3 differentially regulates skeletal muscle lipid oxidation in active, sedentary, and type 2 diabetic males. J Clin Endocrinol Metab. 2015;100(10):3683–92. https://doi.org/10.1210/jc.2014-4125.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Patel S, Yang W, Kozusko K, Saudek V, Savage DB. Perilipins 2 and 3 lack a carboxy-terminal domain present in perilipin 1 involved in sequestering ABHD5 and suppressing basal lipolysis. Proc Natl Acad Sci U S A. 2014;111(25):9163–8. https://doi.org/10.1073/pnas.1318791111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Trudel G, Coletta E, Cameron I, Belavy DL, Lecompte M, Armbrecht G, et al. Resistive exercises, with or without whole body vibration, prevent vertebral marrow fat accumulation during 60 days of head-down tilt bed rest in men. J Appl Physiol. 2012;112(11):1824–31. https://doi.org/10.1152/japplphysiol.00029.2012.

    Article  PubMed  Google Scholar 

  94. Krishnamoorthy D, Frechette DM, Adler BJ, Green DE, Chan ME, Rubin CT. Marrow adipogenesis and bone loss that parallels estrogen deficiency is slowed by low-intensity mechanical signals. Osteoporos Int. 2016;27(2):747–56. https://doi.org/10.1007/s00198-015-3289-5.

    Article  CAS  PubMed  Google Scholar 

  95. Baker JM, De Lisio M, Parise G. Endurance exercise training promotes medullary hematopoiesis. FASEB J. 2011;25(12):4348–57. https://doi.org/10.1096/fj.11-189043.

    Article  CAS  PubMed  Google Scholar 

  96. De Lisio M, Parise G. Characterization of the effects of exercise training on hematopoietic stem cell quantity and function. J Appl Physiol. 2012;113(10):1576–84. https://doi.org/10.1152/japplphysiol.00717.2012.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Bonsignore MR, Morici G, Santoro A, Pagano M, Cascio L, Bonanno A, et al. Circulating hematopoietic progenitor cells in runners. J Appl Physiol. 2002;93(5):1691–7. https://doi.org/10.1152/japplphysiol.00376.2002.

    Article  PubMed  Google Scholar 

  98. Kim HL, Cho HY, Park IY, Choi JM, Kim M, Jang HJ, et al. The positive association between peripheral blood cell counts and bone mineral density in postmenopausal women. Yonsei Med J. 2011;52(5):739–45. https://doi.org/10.3349/ymj.2011.52.5.739.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Klein-Nulend J, Sterck JG, Semeins CM, Lips P, Joldersma M, Baart JA, et al. Donor age and mechanosensitivity of human bone cells. Osteoporos Int. 2002;13(2):137–46. https://doi.org/10.1007/s001980200005.

    Article  CAS  PubMed  Google Scholar 

  100. Robling AG, Burr DB, Turner CH. Recovery periods restore mechanosensitivity to dynamically loaded bone. J Exp Biol. 2001;204(Pt 19):3389–99.

    CAS  PubMed  Google Scholar 

  101. Sen B, Xie Z, Case N, Styner M, Rubin CT, Rubin J. Mechanical signal influence on mesenchymal stem cell fate is enhanced by incorporation of refractory periods into the loading regimen. J Biomech. 2011;44(4):593–9. https://doi.org/10.1016/j.jbiomech.2010.11.022.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by National Institute of Health through National Institute of Arthritis and Musculoskeletal and Skin Diseases grant AR-43498 and National Institute of Biomedical Imaging and Bioengineering grant EB-14351.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Clinton T. Rubin.

Ethics declarations

Conflict of Interest

Janet Rubin, Vihitaben Patel, and M. Ete Chan declare no conflict of interest. Clinton Rubin has authored patents related to the use of mechanical signals to bias stem cell fate and mechanical regulation of metabolic diseases. He is also a Founder of Marodyne Medical. Other authors have nothing to disclose.

Human and Animal Rights and Informed Consent

All animal and human studies referred to herein, that were performed by any/all of the authors, were reviewed and approved by the university (SBU and/or UNC) human/animal user committees. In addition, any of the human studies referred to, performed by Dr. C. Rubin, were done with informed consent of the subjects.

Additional information

This article is part of the Topical Collection on Bone Marrow and Adipose Tissue

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Patel, V.S., Ete Chan, M., Rubin, J. et al. Marrow Adiposity and Hematopoiesis in Aging and Obesity: Exercise as an Intervention. Curr Osteoporos Rep 16, 105–115 (2018). https://doi.org/10.1007/s11914-018-0424-1

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11914-018-0424-1

Keywords

Navigation