Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Combating osteoporosis and obesity with exercise: leveraging cell mechanosensitivity

Abstract

Osteoporosis, a condition of skeletal decline that undermines quality of life, is treated with pharmacological interventions that are associated with poor adherence and adverse effects. Complicating efforts to improve clinical outcomes, the incidence of obesity is increasing, predisposing the population to a range of musculoskeletal complications and metabolic disorders. Pharmacological management of obesity has yet to deliver notable reductions in weight and debilitating complications are rarely avoided. By contrast, exercise shows promise as a non-invasive and non-pharmacological method of regulating both osteoporosis and obesity. The principal components of exercise — mechanical signals — promote bone and muscle anabolism while limiting formation and expansion of fat mass. Mechanical regulation of bone and marrow fat might be achieved by regulating functions of differentiated cells in the skeletal tissue while biasing lineage selection of their common progenitors — mesenchymal stem cells. An inverse relationship between adipocyte versus osteoblast fate selection from stem cells is implicated in clinical conditions such as childhood obesity and increased marrow adiposity in type 2 diabetes mellitus, as well as contributing to skeletal frailty. Understanding how exercise-induced mechanical signals can be used to improve bone quality while decreasing fat mass and metabolic dysfunction should lead to new strategies to treat chronic diseases such as osteoporosis and obesity.

Key points

  • Ageing and inactivity each contribute towards a local and systemic environment conducive to poor bone quality, increased systemic adiposity, marrow adipogenesis and inflammation.

  • Mesenchymal stem cells and their lineage-differentiated progeny (for example, osteoblasts) are mechanosensitive, such that increased mechanical signals (such as exercise) stimulate muscle and bone anabolism.

  • Mechanical signals suppress obesity end points, including fat gain, adipocyte lipid acquisition, chronic inflammation and indices associated with type 2 diabetes mellitus.

  • Transduction of mechanical signals across the plasma membrane of stem cells into the nucleus activates signalling cascades and cytoskeletal adaptations to initiate osteogenic, chondrogenic and myogenic differentiation and inhibit adipocyte differentiation.

  • Mechanical signals, such as those induced through low-intensity vibration, need not be large in magnitude, or long in duration, to influence bone or fat phenotypes.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Exercise and mechanical signals are anabolic to skeletal tissue and muscle and slow excessive bone resorption, counteracting the negative effects of a high-fat diet and sedentary lifestyle on bone and fat.
Fig. 2: Exercise suppresses expansion of marrow adipocytes and strengthens bone in obese mice.
Fig. 3: Mechanotransductive responses of mesenchymal stem cells to dynamic mechanical stimuli are achieved through the internal stiffening of the cell via cytoplasmic-bound actin proteins.

Similar content being viewed by others

References

  1. Ruff, C. B., Larsen, C. S. & Hayes, W. C. Structural changes in the femur with the transition to agriculture on the Georgia coast. Am. J. Phys. Anthropol. 64, 125–136 (1984).

    CAS  PubMed  Google Scholar 

  2. Larsen, C. S. Biological changes in human-populations with agriculture. Annu. Rev. Anthropol. 24, 185–213 (1995).

    Google Scholar 

  3. Ruff, C. B. Gracilization of the modern human skeleton — the latent strength in our slender bones teaches lessons about human lives, current and past. Am. Sci. 94, 508–514 (2006).

    Google Scholar 

  4. Nowlan, N. C., Jepsen, K. J. & Morgan, E. F. Smaller, weaker, and less stiff bones evolve from changes in subsistence strategy. Osteoporos. Int. 22, 1967–1980 (2011).

    CAS  PubMed  Google Scholar 

  5. Bilezikian, J. P. Osteoporosis in men. J. Clin. Endocrinol. Metab. 84, 3431–3434 (1999).

    CAS  PubMed  Google Scholar 

  6. Hu, F. B., Li, T. Y., Colditz, G. A., Willett, W. C. & Manson, J. E. Television watching and other sedentary behaviors in relation to risk of obesity and type 2 diabetes mellitus in women. JAMA 289, 1785–1791 (2003).

    PubMed  Google Scholar 

  7. Manson, J. E., Skerrett, P. J., Greenland, P. & VanItallie, T. B. The escalating pandemics of obesity and sedentary lifestyle. A call to action for clinicians. Arch. Intern. Med. 164, 249–258 (2004).

    PubMed  Google Scholar 

  8. Watson, S. L. et al. High-intensity resistance and impact training improves bone mineral density and physical function in postmenopausal women with osteopenia and osteoporosis: the LIFTMOR randomized controlled trial. J. Bone Miner. Res. 33, 211–220 (2018).

    PubMed  Google Scholar 

  9. Wright, N. C. et al. The recent prevalence of osteoporosis and low bone mass in the United States based on bone mineral density at the femoral neck or lumbar spine. J. Bone Miner. Res. 29, 2520–2526 (2014).

    PubMed  PubMed Central  Google Scholar 

  10. NIH Consensus Development Panel on Osteoporosis Prevention, Diagnosis, and Therapy. Osteoporosis prevention, diagnosis, and therapy. JAMA 285, 785–795 (2001).

    Google Scholar 

  11. Brown, M. Skeletal muscle and bone: effect of sex steroids and aging. Adv. Physiol. Educ. 32, 120–126 (2008).

    PubMed  Google Scholar 

  12. Compston, J. E. Sex steroids and bone. Physiol. Rev. 81, 419–447 (2001).

    CAS  PubMed  Google Scholar 

  13. Manolagas, S. C., O’Brien, C. A. & Almeida, M. The role of estrogen and androgen receptors in bone health and disease. Nat. Rev. Endocrinol. 9, 699–712 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Rosenberg, I. H. Sarcopenia: origins and clinical relevance. Clin. Geriatr. Med. 27, 337–339 (2011).

    PubMed  Google Scholar 

  15. Black, D. M. & Rosen, C. J. Clinical practice. Postmenopausal osteoporosis. N. Engl. J. Med. 374, 254–262 (2016).

    CAS  PubMed  Google Scholar 

  16. Rossouw, J. E. et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women’s Health Initiative randomized controlled trial. JAMA 288, 321–333 (2002).

    CAS  PubMed  Google Scholar 

  17. Shane, E. et al. Atypical subtrochanteric and diaphyseal femoral fractures: second report of a task force of the American Society for Bone and Mineral Research. J. Bone Miner. Res. 29, 1–23 (2014).

    PubMed  Google Scholar 

  18. Siris, E. S. et al. Adherence to bisphosphonate therapy and fracture rates in osteoporotic women: relationship to vertebral and nonvertebral fractures from 2 US claims databases. Mayo Clin. Proc. 81, 1013–1022 (2006).

    PubMed  Google Scholar 

  19. Cramer, J. A., Gold, D. T., Silverman, S. L. & Lewiecki, E. M. A systematic review of persistence and compliance with bisphosphonates for osteoporosis. Osteoporos. Int. 18, 1023–1031 (2007).

    CAS  PubMed  Google Scholar 

  20. Khosla, S. et al. Addressing the crisis in the treatment of osteoporosis: a path forward. J. Bone Miner. Res. 32, 424–430 (2016).

    PubMed  Google Scholar 

  21. Centers for Disease Control and Prevention. Childhood obesity facts. CDC http://www.cdc.gov/obesity/data/childhood.html (2018).

  22. Ogden, C. L., Carroll, M. D., Kit, B. K. & Flegal, K. M. Prevalence of childhood and adult obesity in the United States, 2011–2012. JAMA 311, 806–814 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Kahn, S. E., Hull, R. L. & Utzschneider, K. M. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature 444, 840–846 (2006).

    CAS  PubMed  Google Scholar 

  24. Van Gaal, L. F., Mertens, I. L. & De Block, C. E. Mechanisms linking obesity with cardiovascular disease. Nature 444, 875–880 (2006).

    PubMed  Google Scholar 

  25. Wearing, S. C., Hennig, E. M., Byrne, N. M., Steele, J. R. & Hills, A. P. The biomechanics of restricted movement in adult obesity. Obes. Rev. 7, 13–24 (2006).

    CAS  PubMed  Google Scholar 

  26. Ko, S., Stenholm, S. & Ferrucci, L. Characteristic gait patterns in older adults with obesity—results from the Baltimore Longitudinal Study of Aging. J. Biomech. 43, 1104–1110 (2010).

    PubMed  PubMed Central  Google Scholar 

  27. Messier, S. P. Osteoarthritis of the knee and associated factors of age and obesity: effects on gait. Med. Sci. Sports Exerc. 26, 1446–1452 (1994).

    CAS  PubMed  Google Scholar 

  28. Felson, D. T., Anderson, J. J., Naimark, A., Walker, A. M. & Meenan, R. F. Obesity and knee osteoarthritis. The Framingham Study. Ann. Intern. Med. 109, 18–24 (1988).

    CAS  PubMed  Google Scholar 

  29. Hart, D. J. & Spector, T. D. The relationship of obesity, fat distribution and osteoarthritis in women in the general population: the Chingford Study. J. Rheumatol 20, 331–335 (1993).

    CAS  PubMed  Google Scholar 

  30. Calle, E. E. & Kaaks, R. Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nat. Rev. Cancer 4, 579–591 (2004).

    CAS  PubMed  Google Scholar 

  31. Lashinger, L. M., Ford, N. A. & Hursting, S. D. Interacting inflammatory and growth factor signals underlie the obesity-cancer link. J. Nutr. 144, 109–113 (2014).

    CAS  PubMed  Google Scholar 

  32. International Agency for Research on Cancer, Stewart, B. W. & Wild, C. P. World cancer report 2014. WHO https://www.who.int/cancer/publications/WRC_2014/en/ (2014).

  33. Olson, O. C., Quail, D. F. & Joyce, J. A. Obesity and the tumor microenvironment. Science 358, 1130–1131 (2017).

    CAS  PubMed  Google Scholar 

  34. Tilg, H. & Moschen, A. R. Adipocytokines: mediators linking adipose tissue, inflammation and immunity. Nat. Rev. Immunol. 6, 772–783 (2006).

    CAS  PubMed  Google Scholar 

  35. Adler, B. J., Kaushansky, K. & Rubin, C. T. Obesity-driven disruption of haematopoiesis and the bone marrow niche. Nat. Rev. Endocrinol. 10, 737–748 (2014).

    CAS  PubMed  Google Scholar 

  36. Kennedy, D. E. & Knight, K. L. Bone marrow fat induces inflammation that inhibits B lymphopoiesis. J. Immunol. 196 (Suppl), 122.11 (2016).

    Google Scholar 

  37. Singh, L., Tyagi, S., Myers, D. & Duque, G. Good, bad, or ugly: the biological roles of bone marrow fat. Curr. Osteoporos. Rep. 16, 130–137 (2018).

    PubMed  Google Scholar 

  38. National Osteoporosis Foundation. Exercise for your bone health. NOF https://cdn.nof.org/wp-content/uploads/2016/02/Exercise-for-Your-Bone-Health.pdf (2013).

  39. Styner, M. et al. Bone marrow fat accumulation accelerated by high fat diet is suppressed by exercise. Bone 64, 39–46 (2014).

    PubMed  PubMed Central  Google Scholar 

  40. Bortz, W. M. 2nd. Disuse and aging. JAMA 248, 1203–1208 (1982).

    PubMed  Google Scholar 

  41. Wolff, J. The Law Of Bone Remodeling (Springer, 1986).

  42. Lang, T. et al. Cortical and trabecular bone mineral loss from the spine and hip in long-duration spaceflight. J. Bone Miner. Res. 19, 1006–1012 (2004).

    PubMed  Google Scholar 

  43. Jones, H. H., Priest, J. D., Hayes, W. C., Tichenor, C. C. & Nagel, D. A. Humeral hypertrophy in response to exercise. J. Bone Joint Surg. Am. 59, 204–208 (1977).

    CAS  PubMed  Google Scholar 

  44. Heinonen, A. et al. Bone mineral density in female athletes representing sports with different loading characteristics of the skeleton. Bone 17, 197–203 (1995).

    CAS  PubMed  Google Scholar 

  45. Vlachopoulos, D. et al. Longitudinal adaptations of bone mass, geometry, and metabolism in adolescent male athletes: the PRO-BONE study. J. Bone Miner. Res. 32, 2269–2277 (2017).

    CAS  PubMed  Google Scholar 

  46. Gabel, L., Macdonald, H. M., Nettlefold, L. & McKay, H. A. Physical activity, sedentary time, and bone strength from childhood to early adulthood: a mixed longitudinal HR-pQCT study. J. Bone Miner. Res. 32, 1525–1536 (2017).

    CAS  PubMed  Google Scholar 

  47. Leichter, I. et al. Gain in mass density of bone following strenuous physical activity. J. Orthop. Res. 7, 86–90 (1989).

    CAS  PubMed  Google Scholar 

  48. McKay, H. A. et al. “Bounce at the Bell”: a novel program of short bouts of exercise improves proximal femur bone mass in early pubertal children. Br. J. Sports Med. 39, 521–526 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Heinonen, A., Sievanen, H., Kannus, P., Oja, P. & Vuori, I. Effects of unilateral strength training and detraining on bone mineral mass and estimated mechanical characteristics of the upper limb bones in young women. J. Bone Miner. Res. 11, 490–501 (1996).

    CAS  PubMed  Google Scholar 

  50. Rubin, C. T., Seeherman, H., Qin, Y. X. & Gross, T. S. The mechanical consequences of load bearing in the equine third metacarpal across speed and gait: the nonuniform distributions of normal strain, shear strain, and strain energy density. FASEB J. 27, 1887–1894 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Rubin, C. et al. Differentiation of the bone-tissue remodeling response to axial and torsional loading in the turkey ulna. J. Bone Joint Surg. Am. 78, 1523–1533 (1996).

    CAS  PubMed  Google Scholar 

  52. Patel, V. S. et al. Incorporating refractory period in mechanical stimulation mitigates obesity-induced adipose tissue dysfunction in adult mice. Obesity 25, 1745–1753 (2017).

    CAS  PubMed  Google Scholar 

  53. Wallace, B. A. & Cumming, R. G. Systematic review of randomized trials of the effect of exercise on bone mass in pre- and postmenopausal women. Calcif. Tissue Int. 67, 10–18 (2000).

    CAS  PubMed  Google Scholar 

  54. Warden, S. J. et al. Bone adaptation to a mechanical loading program significantly increases skeletal fatigue resistance. J. Bone Miner. Res. 20, 809–816 (2005).

    PubMed  Google Scholar 

  55. Rubin, C. T. & Lanyon, L. E. Regulation of bone mass by mechanical strain magnitude. Calcif. Tissue Int. 37, 411–417 (1985).

    CAS  PubMed  Google Scholar 

  56. Tjandrawinata, R. R., Vincent, V. L. & Hughes-Fulford, M. Vibrational force alters mRNA expression in osteoblasts. FASEB J. 11, 493–497 (1997).

    CAS  PubMed  Google Scholar 

  57. Ko, K. S. & McCulloch, C. A. Intercellular mechanotransduction: cellular circuits that coordinate tissue responses to mechanical loading. Biochem. Biophys. Res. Commun. 285, 1077–1083 (2001).

    CAS  PubMed  Google Scholar 

  58. Rubin, J., Rubin, C. & Jacobs, C. R. Molecular pathways mediating mechanical signaling in bone. Gene 367, 1–16 (2006).

    CAS  PubMed  Google Scholar 

  59. Thompson, W. R. et al. Osteocyte specific responses to soluble and mechanical stimuli in a stem cell derived culture model. Sci. Rep. 5, 11049 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Uzer, G. et al. Cell mechanosensitivity to extremely low-magnitude signals is enabled by a LINCed nucleus. Stem Cells 33, 2063–2076 (2015).

    PubMed  PubMed Central  Google Scholar 

  61. Uzer, G., Fuchs, R. K., Rubin, J. & Thompson, W. R. Concise review: plasma and nuclear membranes convey mechanical information to regulate mesenchymal stem cell lineage. Stem Cells 34, 1455–1463 (2016).

    PubMed  PubMed Central  Google Scholar 

  62. Case, N. & Rubin, J. Beta-catenin—a supporting role in the skeleton. J. Cell. Biochem. 110, 545–553 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Sen, B. et al. Mechanically induced focal adhesion assembly amplifies anti-adipogenic pathways in mesenchymal stem cells. Stem Cells 29, 1829–1836 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Sen, B. et al. Mechanical strain inhibits adipogenesis in mesenchymal stem cells by stimulating a durable beta-catenin signal. Endocrinology 149, 6065–6075 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Samelson, E. J. et al. Diabetes and deficits in cortical bone density, microarchitecture, and bone size: Framingham HR-pQCT study. J. Bone Miner. Res. 33, 54–62 (2018).

    PubMed  Google Scholar 

  66. Murfee, W. L. et al. High-frequency, low-magnitude vibrations suppress the number of blood vessels per muscle fiber in mouse soleus muscle. J. Appl. Physiol. 98, 2376–2380 (2005).

    PubMed  Google Scholar 

  67. Case, N. et al. Mechanical input restrains PPARgamma2 expression and action to preserve mesenchymal stem cell multipotentiality. Bone 52, 454–464 (2013).

    CAS  PubMed  Google Scholar 

  68. Luu, Y. K. et al. Mechanical stimulation of mesenchymal stem cell proliferation and differentiation promotes osteogenesis while preventing dietary-induced obesity. J. Bone Miner. Res. 24, 50–61 (2009).

    CAS  PubMed  Google Scholar 

  69. Styner, M., Sen, B., Xie, Z., Case, N. & Rubin, J. Indomethacin promotes adipogenesis of mesenchymal stem cells through a cyclooxygenase independent mechanism. J. Cell. Biochem. 111, 1042–1050 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Sen, B. et al. Mechanical signal influence on mesenchymal stem cell fate is enhanced by incorporation of refractory periods into the loading regimen. J. Biomech. 44, 593–599 (2011).

    PubMed  Google Scholar 

  71. Globus, R. K., Bikle, D. D. & Morey-Holton, E. The temporal response of bone to unloading. Endocrinology 118, 733–742 (1986).

    CAS  PubMed  Google Scholar 

  72. Bikle, D. D., Sakata, T. & Halloran, B. P. The impact of skeletal unloading on bone formation. Gravit. Space Biol. Bull. 16, 45–54 (2003).

    PubMed  Google Scholar 

  73. Rubin, C., Xu, G. & Judex, S. The anabolic activity of bone tissue, suppressed by disuse, is normalized by brief exposure to extremely low-magnitude mechanical stimuli. FASEB J. 15, 2225–2229 (2001).

    CAS  PubMed  Google Scholar 

  74. Rubin, C. et al. Quantity and quality of trabecular bone in the femur are enhanced by a strongly anabolic, noninvasive mechanical intervention. J. Bone Miner. Res. 17, 349–357 (2002).

    PubMed  Google Scholar 

  75. Judex, S. et al. Genetically linked site-specificity of disuse osteoporosis. J. Bone Miner. Res. 19, 607–613 (2004).

    PubMed  Google Scholar 

  76. Squire, M., Brazin, A., Keng, Y. & Judex, S. Baseline bone morphometry and cellular activity modulate the degree of bone loss in the appendicular skeleton during disuse. Bone 42, 341–349 (2008).

    PubMed  Google Scholar 

  77. Trudel, G. et al. Bone marrow fat accumulation after 60 days of bed rest persisted 1 year after activities were resumed along with hemopoietic stimulation: the Women International Space Simulation for Exploration study. J. Appl. Physiol. 107, 540–548 (2009).

    PubMed  Google Scholar 

  78. Rubin, J. et al. Mechanical strain differentially regulates endothelial nitric-oxide synthase and receptor activator of nuclear kappa B ligand expression via ERK1/2 MAPK. J. Biol. Chem. 278, 34018–34025 (2003).

    CAS  PubMed  Google Scholar 

  79. Tchkonia, T., Zhu, Y., van Deursen, J., Campisi, J. & Kirkland, J. L. Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. J. Clin. Invest. 123, 966–972 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Childs, B. G., Durik, M., Baker, D. J. & van Deursen, J. M. Cellular senescence in aging and age-related disease: from mechanisms to therapy. Nat. Med. 21, 1424–1435 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Stattin, K., Michaelsson, K., Larsson, S. C., Wolk, A. & Byberg, L. Leisure-time physical activity and risk of fracture: a cohort study of 66,940 men and women. J. Bone Miner. Res. 32, 1599–1606 (2017).

    PubMed  Google Scholar 

  82. Lindstrom, J. et al. The Finnish Diabetes Prevention Study (DPS): lifestyle intervention and 3-year results on diet and physical activity. Diabetes Care 26, 3230–3236 (2003).

    PubMed  Google Scholar 

  83. Hu, F. B. et al. Adiposity as compared with physical activity in predicting mortality among women. N. Engl. J. Med. 351, 2694–2703 (2004).

    CAS  PubMed  Google Scholar 

  84. Hinton, P. S., Nigh, P. & Thyfault, J. Effectiveness of resistance training or jumping-exercise to increase bone mineral density in men with low bone mass: a 12-month randomized, clinical trial. Bone 79, 203–212 (2015).

    PubMed  PubMed Central  Google Scholar 

  85. Dalsky, G. P. et al. Weight-bearing exercise training and lumbar bone-mineral content in postmenopausal women. Ann. Intern. Med. 108, 824–828 (1988).

    CAS  PubMed  Google Scholar 

  86. Nilsson, M., Sundh, D., Mellstrom, D. & Lorentzon, M. Current physical activity is independently associated with cortical bone size and bone strength in elderly Swedish women. J. Bone Miner. Res. 32, 473–485 (2017).

    CAS  PubMed  Google Scholar 

  87. Ensrud, K. E. & Crandall, C. J. Osteoporosis. Ann. Intern. Med. 167, ITC17–ITC32 (2017).

    PubMed  Google Scholar 

  88. Ness, K. K. et al. Skeletal, neuromuscular and fitness impairments among children with newly diagnosed acute lymphoblastic leukemia. Leuk. Lymphoma 56, 1004–1011 (2015).

    CAS  PubMed  Google Scholar 

  89. Maratova, K. et al. Muscle functions and bone strength are impaired in adolescents with type 1 diabetes. Bone 106, 22–27 (2018).

    PubMed  Google Scholar 

  90. Joyce, E. D. et al. Association of muscle strength and bone mineral density in adult survivors of childhood acute lymphoblastic leukemia. Arch. Phys. Med. Rehabil. 92, 873–879 (2011).

    PubMed  PubMed Central  Google Scholar 

  91. Ness, K. K. et al. Physiologic frailty as a sign of accelerated aging among adult survivors of childhood cancer: a report from the St Jude Lifetime cohort study. J. Clin. Oncol. 31, 4496–4503 (2013).

    PubMed  PubMed Central  Google Scholar 

  92. Huang, H. P. et al. Adherence to prescribed exercise time and intensity declines as the exercise program proceeds: findings from women under treatment for breast cancer. Support. Care Cancer 23, 2061–2071 (2015).

    PubMed  Google Scholar 

  93. Hemmatian, H., Bakker, A. D., Klein-Nulend, J. & van Lenthe, G. H. Aging, osteocytes, and mechanotransduction. Curr. Osteoporos. Rep. 15, 401–411 (2017).

    PubMed  PubMed Central  Google Scholar 

  94. Bonewald, L. F. The amazing osteocyte. J. Bone Miner. Res. 26, 229–238 (2011).

    CAS  PubMed  Google Scholar 

  95. Joldersma, M., Klein-Nulend, J., Oleksik, A. M., Heyligers, I. C. & Burger, E. H. Estrogen enhances mechanical stress-induced prostaglandin production by bone cells from elderly women. Am. J. Physiol. Endocrinol. Metab. 280, E436–E442 (2001).

    CAS  PubMed  Google Scholar 

  96. Joldersma, M., Burger, E. H., Semeins, C. M. & Klein-Nulend, J. Mechanical stress induces COX-2 mRNA expression in bone cells from elderly women. J. Biomech. 33, 53–61 (2000).

    CAS  PubMed  Google Scholar 

  97. Sterck, J. G., Klein-Nulend, J., Lips, P. & Burger, E. H. Response of normal and osteoporotic human bone cells to mechanical stress in vitro. Am. J. Physiol. 274, E1113–E1120 (1998).

    CAS  PubMed  Google Scholar 

  98. Rubin, C. T., Bain, S. D. & McLeod, K. J. Suppression of the osteogenic response in the aging skeleton. Calcif. Tissue Int. 50, 306–313 (1992).

    CAS  PubMed  Google Scholar 

  99. Willie, B. M. et al. Diminished response to in vivo mechanical loading in trabecular and not cortical bone in adulthood of female C57Bl/6 mice coincides with a reduction in deformation to load. Bone 55, 335–346 (2013).

    PubMed  Google Scholar 

  100. Strube, P. et al. Sex-specific compromised bone healing in female rats might be associated with a decrease in mesenchymal stem cell quantity. Bone 45, 1065–1072 (2009).

    CAS  PubMed  Google Scholar 

  101. Wiley, C. D. & Campisi, J. From ancient pathways to aging cells-connecting metabolism and cellular senescence. Cell Metab. 23, 1013–1021 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Farr, J. N. et al. Targeting cellular senescence prevents age-related bone loss in mice. Nat. Med. 23, 1072–1079 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Zhu, Y. et al. Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors. Aging Cell 15, 428–435 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Chiche, A. et al. Injury-induced senescence enables in vivo reprogramming in skeletal muscle. Cell Stem Cell 20, 407–414 (2017).

    CAS  PubMed  Google Scholar 

  105. Akunuru, S. & Geiger, H. Aging, clonality, and rejuvenation of hematopoietic stem cells. Trends Mol. Med. 22, 701–712 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Qin, Y. et al. Myostatin inhibits osteoblastic differentiation by suppressing osteocyte-derived exosomal microRNA-218: a novel mechanism in muscle-bone communication. J. Biol. Chem. 292, 11021–11033 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Evans, W. J. & Campbell, W. W. Sarcopenia and age-related-changes in body-composition and functional-capacity. J. Nutr. 123, 465–468 (1993).

    CAS  PubMed  Google Scholar 

  108. Lamberts, S. W., van den Beld, A. W. & van der Lely, A. J. The endocrinology of aging. Science 278, 419–424 (1997).

    CAS  PubMed  Google Scholar 

  109. Hu, Z. Y. et al. MicroRNA-29 induces cellular senescence in aging muscle through multiple signaling pathways. Aging 6, 160–175 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Evans, W. J. Skeletal muscle loss: cachexia, sarcopenia, and inactivity. Am. J. Clin. Nutr. 91, 1123S–1127S (2010).

    CAS  PubMed  Google Scholar 

  111. Morse, C. I., Thom, J. M., Reeves, N. D., Birch, K. M. & Narici, M. V. In vivo physiological cross-sectional area and specific force are reduced in the gastrocnemius of elderly men. J. Appl. Physiol. 99, 1050–1055 (2005).

    PubMed  Google Scholar 

  112. [No authors listed.] Sarcopenia: European consensus on definition and diagnosis: report of the European Working Group on Sarcopenia in Older People [Japanese]. Nihon Ronen Igakkai Zasshi 49, 788–805 (2012).

  113. Cruz-Jentoft, A. J. et al. Sarcopenia: European consensus on definition and diagnosis: report of the European Working Group on Sarcopenia in Older People. Age Ageing 39, 412–423 (2010).

    PubMed  PubMed Central  Google Scholar 

  114. Han, A., Bokshan, S. L., Marcaccio, S. E., DePasse, J. M. & Daniels, A. H. Diagnostic criteria and clinical outcomes in sarcopenia research: a literature review. J. Clin. Med. 7, 70 (2018).

    PubMed Central  Google Scholar 

  115. Janssen, I., Shepard, D. S., Katzmarzyk, P. T. & Roubenoff, R. The healthcare costs of sarcopenia in the United States. J. Am. Geriatr. Soc. 52, 80–85 (2004).

    PubMed  Google Scholar 

  116. Cruz-Jentoft, A. J. et al. Prevalence of and interventions for sarcopenia in ageing adults: a systematic review. Report of the International Sarcopenia Initiative (EWGSOP and IWGS). Age Ageing 43, 748–759 (2014).

    PubMed  PubMed Central  Google Scholar 

  117. Phillips, S. K., Rook, K. M., Siddle, N. C., Bruce, S. A. & Woledge, R. C. Muscle weakness in women occurs at an earlier age than in men, but strength is preserved by hormone replacement therapy. Clin. Sci. 84, 95–98 (1993).

    CAS  PubMed  Google Scholar 

  118. Huang, J. et al. Crosstalk between MLO-Y4 osteocytes and C2C12 muscle cells is mediated by the Wnt/beta-catenin pathway. JBMR Plus 1, 86–100 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Marks, A. R. Intracellular calcium-release channels: regulators of cell life and death. Am. J. Physiol. 272, H597–H605 (1997).

    CAS  PubMed  Google Scholar 

  120. Wehrens, X. H. et al. FKBP12.6 deficiency and defective calcium release channel (ryanodine receptor) function linked to exercise-induced sudden cardiac death. Cell 113, 829–840 (2003).

    CAS  PubMed  Google Scholar 

  121. Bellinger, A. M. et al. Hypernitrosylated ryanodine receptor calcium release channels are leaky in dystrophic muscle. Nat. Med. 15, 325–330 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Andersson, D. C. et al. Ryanodine receptor oxidation causes intracellular calcium leak and muscle weakness in aging. Cell Metab. 14, 196–207 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Guise, T. A. et al. Basic mechanisms responsible for osteolytic and osteoblastic bone metastases. Clin. Cancer Res. 12, 6213S–6216S (2006).

    CAS  PubMed  Google Scholar 

  124. Mundy, G. R. Metastasis to bone: causes, consequences and therapeutic opportunities. Nat. Rev. Cancer 2, 584–593 (2002).

    CAS  PubMed  Google Scholar 

  125. Janssens, K. et al. Mutations in the gene encoding the latency-associated peptide of TGF-beta 1 cause Camurati-Engelmann disease. Nat. Genet. 26, 273–275 (2000).

    CAS  PubMed  Google Scholar 

  126. Hauschka, P. V., Mavrakos, A. E., Iafrati, M. D., Doleman, S. E. & Klagsbrun, M. Growth factors in bone matrix. Isolation of multiple types by affinity chromatography on heparin-Sepharose. J. Biol. Chem. 261, 12665–12674 (1986).

    CAS  PubMed  Google Scholar 

  127. Waning, D. L. et al. Excess TGF-beta mediates muscle weakness associated with bone metastases in mice. Nat. Med. 21, 1262–1271 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Mundy, G. R., Yoneda, T. & Hiraga, T. Preclinical studies with zoledronic acid and other bisphosphonates: impact on the bone microenvironment. Semin. Oncol. 28, 35–44 (2001).

    CAS  PubMed  Google Scholar 

  129. Waning, D. L. & Guise, T. A. Molecular mechanisms of bone metastasis and associated muscle weakness. Clin. Cancer Res. 20, 3071–3077 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Tuttle, L. J., Sinacore, D. R., Cade, W. T. & Mueller, M. J. Lower physical activity is associated with higher intermuscular adipose tissue in people with type 2 diabetes and peripheral neuropathy. Phys. Ther. 91, 923–930 (2011).

    PubMed  PubMed Central  Google Scholar 

  131. Bittel, D. C. et al. Adipose tissue content, muscle performance and physical function in obese adults with type 2 diabetes mellitus and peripheral neuropathy. J. Diabetes Compl. 29, 250–257 (2015).

    Google Scholar 

  132. Papadakis, M. A. et al. Growth hormone replacement in healthy older men improves body composition but not functional ability. Ann. Intern. Med. 124, 708–716 (1996).

    CAS  PubMed  Google Scholar 

  133. Brioche, T. et al. Growth hormone replacement therapy prevents sarcopenia by a dual mechanism: improvement of protein balance and of antioxidant defenses. J. Gerontol. A Biol. Sci. Med. Sci. 69, 1186–1198 (2014).

    CAS  PubMed  Google Scholar 

  134. Jorgensen, J. O. et al. Beneficial effects of growth hormone treatment in GH-deficient adults. Lancet 1, 1221–1225 (1989).

    CAS  PubMed  Google Scholar 

  135. Mikkelsen, U. R. et al. Skeletal muscle morphology and regulatory signalling in endurance-trained and sedentary individuals: the influence of ageing. Exp. Gerontol. 93, 54–67 (2017).

    CAS  PubMed  Google Scholar 

  136. Rogers, M. A. & Evans, W. J. Changes in skeletal muscle with aging: effects of exercise training. Exerc. Sport Sci. Rev. 21, 65–102 (1993).

    CAS  PubMed  Google Scholar 

  137. Ryan, A. S. Insulin resistance with aging: effects of diet and exercise. Sports Med. 30, 327–346 (2000).

    CAS  PubMed  Google Scholar 

  138. Ivy, J. L. Role of exercise training in the prevention and treatment of insulin resistance and non-insulin-dependent diabetes mellitus. Sports Med. 24, 321–336 (1997).

    CAS  PubMed  Google Scholar 

  139. Baar, K. & Esser, K. Phosphorylation of p70(S6k) correlates with increased skeletal muscle mass following resistance exercise. Am. J. Physiol. 276, C120–C127 (1999).

    CAS  PubMed  Google Scholar 

  140. Kubica, N., Bolster, D. R., Farrell, P. A., Kimball, S. R. & Jefferson, L. S. Resistance exercise increases muscle protein synthesis and translation of eukaryotic initiation factor 2Bepsilon mRNA in a mammalian target of rapamycin-dependent manner. J. Biol. Chem. 280, 7570–7580 (2005).

    CAS  PubMed  Google Scholar 

  141. Ji, L. L., Gomez-Cabrera, M. C., Steinhafel, N. & Vina, J. Acute exercise activates nuclear factor (NF)-kappaB signaling pathway in rat skeletal muscle. FASEB J. 18, 1499–1506 (2004).

    CAS  PubMed  Google Scholar 

  142. Senf, S. M., Dodd, S. L. & Judge, A. R. FOXO signaling is required for disuse muscle atrophy and is directly regulated by Hsp70. Am. J. Physiol. Cell Physiol. 298, C38–C45 (2010).

    CAS  PubMed  Google Scholar 

  143. Kavazis, A. N., Smuder, A. J. & Powers, S. K. Effects of short-term endurance exercise training on acute doxorubicin-induced FoxO transcription in cardiac and skeletal muscle. J. Appl. Physiol. 117, 223–230 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Iyer, S. et al. FOXOs attenuate bone formation by suppressing Wnt signaling. J. Clin. Invest. 123, 3409–3419 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Stitt, T. N. et al. The IGF-1/PI3K/Akt pathway prevents expression of muscle atrophy-induced ubiquitin ligases by inhibiting FOXO transcription factors. Mol. Cell 14, 395–403 (2004).

    CAS  PubMed  Google Scholar 

  146. Bodine, S. C. et al. Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nat. Cell Biol. 3, 1014–1019 (2001).

    CAS  PubMed  Google Scholar 

  147. Salanova, M. et al. Nitrosative stress in human skeletal muscle attenuated by exercise countermeasure after chronic disuse. Redox Biol. 1, 514–526 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Salanova, M., Schiffl, G., Rittweger, J., Felsenberg, D. & Blottner, D. Ryanodine receptor type-1 (RyR1) expression and protein S-nitrosylation pattern in human soleus myofibres following bed rest and exercise countermeasure. Histochem. Cell Biol. 130, 105–118 (2008).

    CAS  PubMed  Google Scholar 

  149. Frechette, D. M., Krishnamoorthy, D., Adler, B. J., Chan, M. E. & Rubin, C. T. Diminished satellite cells and elevated adipogenic gene expression in muscle as caused by ovariectomy are averted by low-magnitude mechanical signals. J. Appl. Physiol. 119, 27–36 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Frechette, D. M. et al. Mechanical signals protect stem cell lineage selection, preserving the bone and muscle phenotypes in obesity. Ann. NY Acad. Sci. 1409, 33–50 (2017).

    CAS  PubMed  Google Scholar 

  151. Mera, P., Laue, K., Wei, J., Berger, J. M. & Karsenty, G. Osteocalcin is necessary and sufficient to maintain muscle mass in older mice. Mol. Metab. 5, 1042–1047 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Mera, P. et al. Osteocalcin signaling in myofibers is necessary and sufficient for optimum adaptation to exercise. Cell Metab. 23, 1078–1092 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Febbraio, M. A., Hiscock, N., Sacchetti, M., Fischer, C. P. & Pedersen, B. K. Interleukin-6 is a novel factor mediating glucose homeostasis during skeletal muscle contraction. Diabetes 53, 1643–1648 (2004).

    CAS  PubMed  Google Scholar 

  154. Karsenty, G. & Mera, P. Molecular bases of the crosstalk between bone and muscle. Bone 11, 43–49 (2017).

    Google Scholar 

  155. Messi, M. L. et al. Resistance training enhances skeletal muscle innervation without modifying the number of satellite cells or their myofiber association in obese older adults. J. Gerontol. A Biol. Sci. Med. Sci. 71, 1273–1280 (2016).

    PubMed  Google Scholar 

  156. Stewart, V. H., Saunders, D. H. & Greig, C. A. Responsiveness of muscle size and strength to physical training in very elderly people: a systematic review. Scand. J. Med. Sci. Sports 24, e1–e10 (2014).

    CAS  PubMed  Google Scholar 

  157. Thompson, L. V. Effects of age and training on skeletal muscle physiology and performance. Phys. Ther. 74, 71–81 (1994).

    CAS  PubMed  Google Scholar 

  158. Pedersen, B. K. & Saltin, B. Evidence for prescribing exercise as therapy in chronic disease. Scand. J. Med. Sci. Sports 16 (Suppl. 1), 3–63 (2006).

    PubMed  Google Scholar 

  159. Hedlund, P. B., Yanaihara, N. & Fuxe, K. Evidence for specific N-terminal galanin fragment binding sites in the rat brain. Eur. J. Pharmacol. 224, 203–205 (1992).

    CAS  PubMed  Google Scholar 

  160. Kelly, O. J. & Gilman, J. C. Can unconventional exercise be helpful in the treatment, management and prevention of osteosarcopenic obesity? Curr. Aging Sci. 10, 106–121 (2017).

    Google Scholar 

  161. Tiedemann, A., O’Rourke, S., Sesto, R. & Sherrington, C. A. 12-week Iyengar yoga program improved balance and mobility in older community-dwelling people: a pilot randomized controlled trial. J. Gerontol. A Biol. Sci. Med. Sci. 68, 1068–1075 (2013).

    PubMed  Google Scholar 

  162. Courneya, K. S. et al. Predictors of adherence to different types and doses of supervised exercise during breast cancer chemotherapy. Int. J. Behav. Nutr. Phys. Act 11, 85 (2014).

    PubMed  PubMed Central  Google Scholar 

  163. Cox, C. L. et al. Modifying bone mineral density, physical function, and quality of life in children with acute lymphoblastic leukemia. Pediatr. Blood Cancer 65, e26929 (2018).

    Google Scholar 

  164. Schoenau, E., Neu, C. M., Beck, B., Manz, F. & Rauch, F. Bone mineral content per muscle cross-sectional area as an index of the functional muscle-bone unit. J. Bone Miner. Res. 17, 1095–1101 (2002).

    PubMed  Google Scholar 

  165. Hall, B. K. & Herring, S. W. Paralysis and growth of the musculoskeletal system in the embryonic chick. J. Morphol. 206, 45–56 (1990).

    CAS  PubMed  Google Scholar 

  166. Sharir, A., Stern, T., Rot, C., Shahar, R. & Zelzer, E. Muscle force regulates bone shaping for optimal load-bearing capacity during embryogenesis. Development 138, 3247–3259 (2011).

    CAS  PubMed  Google Scholar 

  167. Hamrick, M. W., McGee-Lawrence, M. E. & Frechette, D. M. Fatty infiltration of skeletal muscle: mechanisms and comparisons with bone marrow adiposity. Front. Endocrinol. 7, 69 (2016).

    Google Scholar 

  168. Long, P., Liu, F., Piesco, N. P., Kapur, R. & Agarwal, S. Signaling by mechanical strain involves transcriptional regulation of proinflammatory genes in human periodontal ligament cells in vitro. Bone 30, 547–552 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Pagnotti, G. M. & Styner, M. Exercise regulation of marrow adipose tissue. Front. Endocrinol. 7, 94 (2016).

    Google Scholar 

  170. Cawthorn, W. P. & Scheller, E. L. Editorial: bone marrow adipose tissue: formation, function, and impact on health and disease. Front. Endocrinol. 8, 112 (2017).

    Google Scholar 

  171. Fantuzzi, G. Adipose tissue, adipokines, and inflammation. J. Allergy Clin. Immunol. 115, 911–919; quiz 920 (2005).

    CAS  PubMed  Google Scholar 

  172. Xue, Y. et al. Adipokines in psoriatic arthritis patients: the correlations with osteoclast precursors and bone erosions. PLOS ONE 7, e46740 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Patel, V. S., Ete Chan, M., Rubin, J. & Rubin, C. T. Marrow adiposity and hematopoiesis in aging and obesity: exercise as an intervention. Curr. Osteoporos. Rep. 16, 105–115 (2018).

    PubMed  PubMed Central  Google Scholar 

  174. Neumann, E., Junker, S., Schett, G., Frommer, K. & Muller-Ladner, U. Adipokines in bone disease. Nat. Rev. Rheumatol. 12, 296–302 (2016).

    CAS  PubMed  Google Scholar 

  175. Kudo, O. et al. Interleukin-6 and interleukin-11 support human osteoclast formation by a RANKL-independent mechanism. Bone 32, 1–7 (2003).

    CAS  PubMed  Google Scholar 

  176. Cawthorn, W. P. et al. Bone marrow adipose tissue is an endocrine organ that contributes to increased circulating adiponectin during caloric restriction. Cell Metab. 20, 368–375 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  177. Basurto, L. et al. Adiponectin is associated with low bone mineral density in elderly men. Eur. J. Endocrinol. 160, 289–293 (2009).

    CAS  PubMed  Google Scholar 

  178. Barbour, K. E. et al. The effects of adiponectin and leptin on changes in bone mineral density. Osteoporosis Int. 23, 1699–1710 (2012).

    CAS  Google Scholar 

  179. Liu, L. et al. Rosiglitazone inhibits bone regeneration and causes significant accumulation of fat at sites of new bone formation. Calcif. Tissue Int. 91, 139–148 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  180. Styner, M. et al. Exercise regulation of marrow fat in the setting of PPARgamma agonist treatment in female C57BL/6 mice. Endocrinology 156, 2753–2761 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  181. Grey, A. et al. Pioglitazone increases bone marrow fat in type 2 diabetes: results from a randomized controlled trial. Eur. J. Endocrinol. 166, 1087–1091 (2012).

    CAS  PubMed  Google Scholar 

  182. Kannel, W. B., Gordon, T. & Castelli, W. P. Obesity, lipids, and glucose intolerance. The Framingham Study. Am. J. Clin. Nutr. 32, 1238–1245 (1979).

    CAS  PubMed  Google Scholar 

  183. Adler, B. J., Green, D. E., Pagnotti, G. M., Chan, M. E. & Rubin, C. T. High fat diet rapidly suppresses B lymphopoiesis by disrupting the supportive capacity of the bone marrow niche. PLOS ONE 9, e90639 (2014).

    PubMed  PubMed Central  Google Scholar 

  184. Ambrosi, T. H. et al. Adipocyte accumulation in the bone marrow during obesity and aging impairs stem cell-based hematopoietic and bone regeneration. Cell Stem Cell 20, 771–784 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Doucette, C. R. 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. 230, 2032–2037 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  186. Scheller, E. L. et al. Changes in skeletal integrity and marrow adiposity during high-fat diet and after weight loss. Front. Endocrinol. 7, 102 (2016).

    Google Scholar 

  187. Styner, M. et al. Exercise decreases marrow adipose tissue through β-oxidation in obese running mice. J. Bone Miner. Res. 32, 1692–1702 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  188. Trottier, M. D., Naaz, A., Li, Y. & Fraker, P. J. Enhancement of hematopoiesis and lymphopoiesis in diet-induced obese mice. Proc. Natl Acad. Sci. USA 109, 7622–7629 (2012).

    CAS  PubMed  Google Scholar 

  189. do Carmo, L. S. 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. 238, 375–384 (2013).

    Google Scholar 

  190. van den Berg, S. M. et al. Diet-induced obesity in mice diminishes hematopoietic stem and progenitor cells in the bone marrow. FASEB J. 30, 1779–1788 (2016).

    PubMed  Google Scholar 

  191. Singer, K. et al. Diet-induced obesity promotes myelopoiesis in hematopoietic stem cells. Mol. Metab. 3, 664–675 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  192. Chan, M. E., Adler, B. J., Green, D. E. & Rubin, C. T. Bone structure and B cell populations, crippled by obesity, are partially rescued by brief daily exposure to low-magnitude mechanical signals. FASEB J. 26, 4855–4863 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  193. Nanji, A. A. & Freeman, J. B. Relationship between body weight and total leukocyte count in morbid obesity. Am. J. Clin. Pathol. 84, 346–347 (1985).

    CAS  PubMed  Google Scholar 

  194. Womack, J. et al. Obesity and immune cell counts in women. Metabolism 56, 998–1004 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  195. Ryder, E. et al. Association of obesity with leukocyte count in obese individuals without metabolic syndrome. Diabetes Metab. Syndr. 8, 197–204 (2014).

    Google Scholar 

  196. Pecht, T., Gutman-Tirosh, A., Bashan, N. & Rudich, A. Peripheral blood leucocyte subclasses as potential biomarkers of adipose tissue inflammation and obesity subphenotypes in humans. Obes. Rev. 15, 322–337 (2014).

    CAS  PubMed  Google Scholar 

  197. Xu, X. et al. Obesity is associated with more activated neutrophils in African American male youth. Int. J. Obes. 39, 26–32 (2015).

    CAS  Google Scholar 

  198. Ghigliotti, G. et al. Adipose tissue immune response: novel triggers and consequences for chronic inflammatory conditions. Inflammation 37, 1337–1353 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  199. Nteeba, J., Ortinau, L. C., Perfield, J. W. 2nd & Keating, A. F. 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. 80, 948–958 (2013).

    CAS  PubMed  Google Scholar 

  200. Caer, C. et al. Immune cell-derived cytokines contribute to obesity-related inflammation, fibrogenesis and metabolic deregulation in human adipose tissue. Sci. Rep. 7, 3000 (2017).

    PubMed  PubMed Central  Google Scholar 

  201. Rubin, C. T. et al. Adipogenesis is inhibited by brief, daily exposure to high-frequency, extremely low-magnitude mechanical signals. Proc. Natl Acad. Sci. USA 104, 17879–17884 (2007).

    CAS  PubMed  Google Scholar 

  202. Kahn, B. B. & Flier, J. S. Obesity and insulin resistance. J. Clin. Invest. 106, 473–481 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  203. Qatanani, M. & Lazar, M. A. Mechanisms of obesity-associated insulin resistance: many choices on the menu. Gene Dev. 21, 1443–1455 (2007).

    CAS  PubMed  Google Scholar 

  204. Souza, P. P. & Lerner, U. H. The role of cytokines in inflammatory bone loss. Immunol. Invest. 42, 555–622 (2013).

    CAS  PubMed  Google Scholar 

  205. Schett, G. Effects of inflammatory and anti-inflammatory cytokines on the bone. Eur. J. Clin. Invest. 41, 1361–1366 (2011).

    CAS  PubMed  Google Scholar 

  206. McLean, R. R. Proinflammatory cytokines and osteoporosis. Curr. Osteoporos. Rep. 7, 134–139 (2009).

    PubMed  Google Scholar 

  207. Kimble, R. B. et al. Simultaneous block of interleukin-1 and tumor necrosis factor is required to completely prevent bone loss in the early postovariectomy period. Endocrinology 136, 3054–3061 (1995).

    CAS  PubMed  Google Scholar 

  208. Rubin, C. T. & Lanyon, L. E. Regulation of bone formation by applied dynamic loads. J. Bone Joint Surg. Am. 66, 397–402 (1984).

    CAS  PubMed  Google Scholar 

  209. O’Connor, J. A., Lanyon, L. E. & MacFie, H. The influence of strain rate on adaptive bone remodelling. J. Biomech. 15, 767–781 (1982).

    PubMed  Google Scholar 

  210. Rath, B., Nam, J., Knobloch, T. J., Lannutti, J. J. & Agarwal, S. Compressive forces induce osteogenic gene expression in calvarial osteoblasts. J. Biomech. 41, 1095–1103 (2008).

    PubMed  PubMed Central  Google Scholar 

  211. Poliachik, S. L., Threet, D., Srinivasan, S. & Gross, T. S. 32-wk-old C3H/HeJ mice actively respond to mechanical loading. Bone 42, 653–659 (2008).

    PubMed  PubMed Central  Google Scholar 

  212. Rubin, C., Turner, A. S., Bain, S., Mallinckrodt, C. & McLeod, K. Anabolism. Low mechanical signals strengthen long bones. Nature 412, 603–604 (2001).

    CAS  PubMed  Google Scholar 

  213. Oxlund, B. S., Ortoft, G., Andreassen, T. T. & Oxlund, H. Low-intensity, high-frequency vibration appears to prevent the decrease in strength of the femur and tibia associated with ovariectomy of adult rats. Bone 32, 69–77 (2003).

    CAS  PubMed  Google Scholar 

  214. Tanaka, S. M. et al. Effects of broad frequency vibration on cultured osteoblasts. J. Biomech. 36, 73–80 (2003).

    PubMed  Google Scholar 

  215. Garman, R., Gaudette, G., Donahue, L. R., Rubin, C. & Judex, S. Low-level accelerations applied in the absence of weight bearing can enhance trabecular bone formation. J. Orthop. Res. 25, 732–740 (2007).

    PubMed  Google Scholar 

  216. Wren, T. A. et al. Effect of high-frequency, low-magnitude vibration on bone and muscle in children with cerebral palsy. J. Pediatr. Orthop. 30, 732–738 (2010).

    PubMed  PubMed Central  Google Scholar 

  217. Pre, D. et al. The differentiation of human adipose-derived stem cells (hASCs) into osteoblasts is promoted by low amplitude, high frequency vibration treatment. Bone 49, 295–303 (2011).

    CAS  PubMed  Google Scholar 

  218. Robinson, T. L. et al. Gymnasts exhibit higher bone mass than runners despite similar prevalence of amenorrhea and oligomenorrhea. J. Bone Miner. Res. 10, 26–35 (1995).

    CAS  PubMed  Google Scholar 

  219. Verschueren, S. M. et al. Effect of 6-month whole body vibration training on hip density, muscle strength, and postural control in postmenopausal women: a randomized controlled pilot study. J. Bone Miner. Res. 19, 352–359 (2004).

    PubMed  Google Scholar 

  220. Judex, S. & Rubin, C. T. Is bone formation induced by high-frequency mechanical signals modulated by muscle activity? J. Musculoskelet. Neuronal Interact. 10, 3–11 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  221. McGarry, J. G., Klein-Nulend, J., Mullender, M. G. & Prendergast, P. J. A comparison of strain and fluid shear stress in stimulating bone cell responses — a computational and experimental study. FASEB J. 19, 482–484 (2005).

    CAS  PubMed  Google Scholar 

  222. Sikavitsas, V. I., Bancroft, G. N., Holtorf, H. L., Jansen, J. A. & Mikos, A. G. Mineralized matrix deposition by marrow stromal osteoblasts in 3D perfusion culture increases with increasing fluid shear forces. Proc. Natl Acad. Sci. USA 100, 14683–14688 (2003).

    CAS  PubMed  Google Scholar 

  223. Bancroft, G. N. et al. Fluid flow increases mineralized matrix deposition in 3D perfusion culture of marrow stromal osteoblasts in a dose-dependent manner. Proc. Natl Acad. Sci. USA 99, 12600–12605 (2002).

    CAS  PubMed  Google Scholar 

  224. Weinbaum, S., Cowin, S. C. & Zeng, Y. A model for the excitation of osteocytes by mechanical loading-induced bone fluid shear stresses. J. Biomech. 27, 339–360 (1994).

    CAS  PubMed  Google Scholar 

  225. Reich, K. M., Gay, C. V. & Frangos, J. A. Fluid shear stress as a mediator of osteoblast cyclic adenosine monophosphate production. J. Cell. Physiol. 143, 100–104 (1990).

    CAS  PubMed  Google Scholar 

  226. Qin, Y. X. & Hu, M. Intramedullary pressure induced by dynamic hydraulic pressure stimulation and its potential in treatment of osteopenia. Bone 48, S186 (2011).

    Google Scholar 

  227. Qin, Y. X. & Lam, H. Y. Intramedullary pressure and matrix strain induced by oscillatory skeletal muscle stimulation and its potential in adaptation. J. Biomech. 42, 140–145 (2009).

    PubMed  Google Scholar 

  228. Zhang, P., Su, M., Liu, Y. L., Hsu, A. & Yokota, H. Knee loading dynamically alters intramedullary pressure in mouse femora. Bone 40, 538–543 (2007).

    PubMed  Google Scholar 

  229. Chan, M. E., Uzer, G. & Rubin, C. T. The potential benefits and inherent risks of vibration as a non-drug therapy for the prevention and treatment of osteoporosis. Curr. Osteoporos. Rep. 11, 36–44 (2013).

    PubMed  PubMed Central  Google Scholar 

  230. Price, C., Zhou, X., Li, W. & Wang, L. Real-time measurement of solute transport within the lacunar-canalicular system of mechanically loaded bone: direct evidence for load-induced fluid flow. J. Bone Miner. Res. 26, 277–285 (2011).

    CAS  PubMed  Google Scholar 

  231. Coughlin, T. R. & Niebur, G. L. Fluid shear stress in trabecular bone marrow due to low-magnitude high-frequency vibration. J. Biomech. 45, 2222–2229 (2012).

    PubMed  Google Scholar 

  232. Lim, K. T. et al. Physical stimulation-based osteogenesis: effect of secretion in vitro on fluid dynamic shear stress of human alveolar bone-derived mesenchymal stem cells. IEEE Trans. Nanobioscience 15, 881–890 (2016).

    PubMed  Google Scholar 

  233. Williams, J. L., Iannotti, J. P., Ham, A., Bleuit, J. & Chen, J. H. Effects of fluid, shear-stress on bone-cells. Biorheology 31, 163–170 (1994).

    CAS  PubMed  Google Scholar 

  234. Vainionpaa, A. et al. Intensity of exercise is associated with bone density change in premenopausal women. Osteoporosis Int. 17, 455–463 (2006).

    CAS  Google Scholar 

  235. Cabrita, G. J. M. et al. Hematopoietic stem cells: from the bone to the bioreactor. Trends Biotechnol. 21, 233–240 (2003).

    CAS  PubMed  Google Scholar 

  236. Gordon, M. Stem cells handbook. Bone Marrow Transplant. 33, 1165 (2004).

    Google Scholar 

  237. Dickerson, D. A., Sander, E. A. & Nauman, E. A. Modeling the mechanical consequences of vibratory loading in the vertebral body: microscale effects. Biomech. Model. Mechanobiol. 7, 191–202 (2008).

    CAS  PubMed  Google Scholar 

  238. Bryant, J. D., David, T., Gaskell, P. H., King, S. & Lond, G. Rheology of bovine bone marrow. Proc. Inst. Mech. Eng. H 203, 71–75 (1989).

    CAS  PubMed  Google Scholar 

  239. Blecha, L. D., Rakotomanana, L., Razafimahery, F., Terrier, A. & Pioletti, D. P. Mechanical interaction between cells and fluid for bone tissue engineering scaffold: modulation of the interfacial shear stress. J. Biomech. 43, 933–937 (2010).

    CAS  PubMed  Google Scholar 

  240. Gimble, J. M., Robinson, C. E., Wu, X. & Kelly, K. A. The function of adipocytes in the bone marrow stroma: an update. Bone 19, 421–428 (1996).

    CAS  PubMed  Google Scholar 

  241. Justesen, J. et al. Adipocyte tissue volume in bone marrow is increased with aging and in patients with osteoporosis. Biogerontology 2, 165–171 (2001).

    CAS  PubMed  Google Scholar 

  242. Swift, J. et al. Nuclear lamin-A scales with tissue stiffness and enhances matrix-directed differentiation. Science 341, 1240104 (2013).

    PubMed  PubMed Central  Google Scholar 

  243. Engler, A. J., Sen, S., Sweeney, H. L. & Discher, D. E. Matrix elasticity directs stem cell lineage specification. Cell 126, 677–689 (2006).

    CAS  PubMed  Google Scholar 

  244. Thompson, W. R. et al. LARG GEF and ARHGAP18 orchestrate RhoA activity to control mesenchymal stem cell lineage. Bone 107, 172–180 (2018).

    CAS  PubMed  Google Scholar 

  245. Sen, B. et al. mTORC2 regulates mechanically induced cytoskeletal reorganization and lineage selection in marrow-derived mesenchymal stem cells. J. Bone Miner. Res. 29, 78–89 (2014).

    CAS  PubMed  Google Scholar 

  246. Sen, B. et al. Intranuclear actin regulates osteogenesis. Stem Cells 33, 3065–3076 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  247. Sen, B. et al. Mechanical loading regulates NFATc1 and beta-catenin signaling through a GSK3beta control node. J. Biol. Chem. 284, 34607–34617 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  248. Lombardi, M. L. et al. The interaction between nesprins and sun proteins at the nuclear envelope is critical for force transmission between the nucleus and cytoskeleton. J. Biol. Chem. 286, 26743–26753 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  249. Parsons, J. T., Horwitz, A. R. & Schwartz, M. A. Cell adhesion: integrating cytoskeletal dynamics and cellular tension. Nat. Rev. Mol. Cell Biol. 11, 633–643 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  250. Burridge, K., Fath, K., Kelly, T., Nuckolls, G. & Turner, C. Focal adhesions: transmembrane junctions between the extracellular matrix and the cytoskeleton. Annu. Rev. Cell Biol. 4, 487–525 (1988).

    CAS  PubMed  Google Scholar 

  251. Grashoff, C. et al. Measuring mechanical tension across vinculin reveals regulation of focal adhesion dynamics. Nature 466, 263–266 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  252. Turner, C. E., Glenney, J. R. Jr & Burridge, K. Paxillin: a new vinculin-binding protein present in focal adhesions. J. Cell Biol. 111, 1059–1068 (1990).

    CAS  PubMed  Google Scholar 

  253. Pasapera, A. M., Schneider, I. C., Rericha, E., Schlaepfer, D. D. & Waterman, C. M. Myosin II activity regulates vinculin recruitment to focal adhesions through FAK-mediated paxillin phosphorylation. J. Cell Biol. 188, 877–890 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  254. Machesky, L. M. & Insall, R. H. Scar1 and the related Wiskott-Aldrich syndrome protein, WASP, regulate the actin cytoskeleton through the Arp2/3 complex. Curr. Biol. 8, 1347–1356 (1998).

    CAS  PubMed  Google Scholar 

  255. Marchand, J. B., Kaiser, D. A., Pollard, T. D. & Higgs, H. N. Interaction of WASP/Scar proteins with actin and vertebrate Arp2/3 complex. Nat. Cell Biol. 3, 76–82 (2001).

    CAS  PubMed  Google Scholar 

  256. Mullins, R. D., Heuser, J. A. & Pollard, T. D. The interaction of Arp2/3 complex with actin: nucleation, high affinity pointed end capping, and formation of branching networks of filaments. Proc. Natl Acad. Sci. USA 95, 6181–6186 (1998).

    CAS  PubMed  Google Scholar 

  257. Jaffe, A. B. & Hall, A. Rho GTPases: biochemistry and biology. Annu. Rev. Cell Dev. Biol. 21, 247–269 (2005).

    CAS  PubMed  Google Scholar 

  258. Riddick, N., Ohtani, K. & Surks, H. K. Targeting by myosin phosphatase-RhoA interacting protein mediates RhoA/ROCK regulation of myosin phosphatase. J. Cell. Biochem. 103, 1158–1170 (2008).

    CAS  PubMed  Google Scholar 

  259. Bhadriraju, K. et al. Activation of ROCK by RhoA is regulated by cell adhesion, shape, and cytoskeletal tension. Exp. Cell Res. 313, 3616–3623 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  260. Thompson, W. R. et al. Mechanically activated Fyn utilizes mTORC2 to regulate RhoA and adipogenesis in mesenchymal stem cells. Stem Cells 31, 2528–2537 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  261. Case, N. et al. Mechanical regulation of glycogen synthase kinase 3beta (GSK3beta) in mesenchymal stem cells is dependent on Akt protein serine 473 phosphorylation via mTORC2 protein. J. Biol. Chem. 286, 39450–39456 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  262. Stewart-Hutchinson, P. J., Hale, C. M., Wirtz, D. & Hodzic, D. Structural requirements for the assembly of LINC complexes and their function in cellular mechanical stiffness. Exp. Cell Res. 314, 1892–1905 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  263. Neelam, S. et al. Direct force probe reveals the mechanics of nuclear homeostasis in the mammalian cell. Proc. Natl Acad. Sci. USA 112, 5720–5725 (2015).

    CAS  PubMed  Google Scholar 

  264. Holaska, J. M., Kowalski, A. K. & Wilson, K. L. Emerin caps the pointed end of actin filaments: evidence for an actin cortical network at the nuclear inner membrane. PLOS Biol. 2, E231 (2004).

    PubMed  PubMed Central  Google Scholar 

  265. Le, H. Q. et al. Mechanical regulation of transcription controls Polycomb-mediated gene silencing during lineage commitment. Nat. Cell Biol. 18, 864–875 (2016).

    CAS  PubMed  Google Scholar 

  266. Sen, B. et al. Intranuclear actin structure modulates mesenchymal stem cell differentiation. Stem Cells 35, 1624–1635 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  267. Kutscheidt, S. et al. FHOD1 interaction with nesprin-2G mediates TAN line formation and nuclear movement. Nat. Cell Biol. 16, 708–715 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  268. Chancellor, T. J., Lee, J., Thodeti, C. K. & Lele, T. Actomyosin tension exerted on the nucleus through nesprin-1 connections influences endothelial cell adhesion, migration, and cyclic strain-induced reorientation. Biophys. J. 99, 115–123 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  269. Chen, C. Y. et al. Accumulation of the inner nuclear envelope protein Sun1 is pathogenic in progeric and dystrophic laminopathies. Cell 149, 565–577 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  270. Thakar, K., May, C. K., Rogers, A. & Carroll, C. W. Opposing roles for distinct LINC complexes in regulation of the small GTPase RhoA. Mol. Biol. Cell 28, 182–191 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  271. Uzer, G. et al. Sun-mediated mechanical LINC between nucleus and cytoskeleton regulates betacatenin nuclear access. J. Biomech. 74, 32–40 (2018).

    PubMed  PubMed Central  Google Scholar 

  272. Shiu, J. Y., Aires, L., Lin, Z. & Vogel, V. Nanopillar force measurements reveal actin-cap-mediated YAP mechanotransduction. Nat. Cell Biol. 20, 262–271 (2018).

    CAS  PubMed  Google Scholar 

  273. Elosegui-Artola, A. et al. Force triggers YAP nuclear entry by regulating transport across nuclear pores. Cell 171, 1397–1410 (2017).

    CAS  PubMed  Google Scholar 

  274. Koike, M. et al. beta-Catenin shows an overlapping sequence requirement but distinct molecular interactions for its bidirectional passage through nuclear pores. J. Biol. Chem. 279, 34038–34047 (2004).

    CAS  PubMed  Google Scholar 

  275. Tolwinski, N. S. & Wieschaus, E. A nuclear function for armadillo/beta-catenin. PLOS Biol. 2, E95 (2004).

    PubMed  PubMed Central  Google Scholar 

  276. Neumann, S. et al. Nesprin-2 interacts with {alpha}-catenin and regulates Wnt signaling at the nuclear envelope. J. Biol. Chem. 285, 34932–34938 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  277. Broers, J. L. et al. Decreased mechanical stiffness in LMNA−/− cells is caused by defective nucleo-cytoskeletal integrity: implications for the development of laminopathies. Hum. Mol. Genet. 13, 2567–2580 (2004).

    CAS  PubMed  Google Scholar 

  278. Stephens, A. D., Banigan, E. J., Adam, S. A., Goldman, R. D. & Marko, J. F. Chromatin and lamin A determine two different mechanical response regimes of the cell nucleus. Mol. Biol. Cell 28, 1984–1996 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  279. Khatau, S. B. et al. The differential formation of the LINC-mediated perinuclear actin cap in pluripotent and somatic cells. PLOS ONE 7, e36689 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  280. Vidal, C., Bermeo, S., Fatkin, D. & Duque, G. Role of the nuclear envelope in the pathogenesis of age-related bone loss and osteoporosis. Bonekey Rep. 1, 62 (2012).

    PubMed  PubMed Central  Google Scholar 

  281. McBeath, R., Pirone, D. M., Nelson, C. M., Bhadriraju, K. & Chen, C. S. Cell shape, cytoskeletal tension, and RhoA regulate stem cell lineage commitment. Dev. Cell 6, 483–495 (2004).

    CAS  PubMed  Google Scholar 

  282. Bermeo, S., Vidal, C., Zhou, H. & Duque, G. Lamin A/C acts as an essential factor in mesenchymal stem cell differentiation through the regulation of the dynamics of the Wnt/beta-catenin pathway. J. Cell. Biochem. 116, 2344–2353 (2015).

    CAS  PubMed  Google Scholar 

  283. Constantinescu, D., Gray, H. L., Sammak, P. J., Schatten, G. P. & Csoka, A. B. Lamin A/C expression is a marker of mouse and human embryonic stem cell differentiation. Stem Cells 24, 177–185 (2006).

    CAS  PubMed  Google Scholar 

  284. Akter, R., Rivas, D., Geneau, G., Drissi, H. & Duque, G. Effect of lamin A/C knockdown on osteoblast differentiation and function. J. Bone Miner. Res. 24, 283–293 (2009).

    CAS  PubMed  Google Scholar 

  285. Tong, J. et al. Lamin A/C deficiency is associated with fat infiltration of muscle and bone. Mech. Ageing Dev. 132, 552–559 (2011).

    CAS  PubMed  Google Scholar 

  286. Li, W. et al. Decreased bone formation and osteopenia in lamin a/c-deficient mice. PLOS ONE 6, e19313 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  287. Armstrong, V. J. et al. Estrogen receptor a is required for strain-related beta-catenin signaling in osteoblasts. J. Bone Miner. Res. 22, S95 (2007).

    Google Scholar 

  288. Bonewald, L. F. Mechanosensation and transduction in osteocytes. Bonekey Osteovision 3, 7–15 (2006).

    PubMed  PubMed Central  Google Scholar 

  289. Bradford, P. G., Gerace, K. V., Roland, R. L. & Chrzan, B. G. Estrogen regulation of apoptosis in osteoblasts. Physiol. Behav. 99, 181–185 (2010).

    CAS  PubMed  Google Scholar 

  290. Jessop, H. L. et al. Mechanical strain and estrogen activate estrogen receptor alpha in bone cells. J. Bone Miner. Res. 16, 1045–1055 (2001).

    CAS  PubMed  Google Scholar 

  291. Wehrle, E. et al. The impact of low-magnitude high-frequency vibration on fracture healing is profoundly influenced by the oestrogen status in mice. Dis. Model. Mech. 8, 93–104 (2015).

    CAS  PubMed  Google Scholar 

  292. Guo, Y. et al. Mechanical strain promotes osteoblast ECM formation and improves its osteoinductive potential. Biomed. Eng. Online 11, 80 (2012).

    PubMed  PubMed Central  Google Scholar 

  293. Kamel, M. A. et al. Fluid flow shear stress and prostaglandin E2 activates beta-catenin signaling in MLO-Y4 osteocytic and 2T3 osteoblastic cells. J. Bone Miner. Res. 22, S375 (2007).

    Google Scholar 

  294. Srinivasan, S., Weimer, D. A., Agans, S. C., Bain, S. D. & Gross, T. S. Low-magnitude mechanical loading becomes osteogenic when rest is inserted between each load cycle. J. Bone Miner. Res. 17, 1613–1620 (2002).

    PubMed  PubMed Central  Google Scholar 

  295. Wright, L. E. et al. Single-limb irradiation induces local and systemic bone loss in a murine model. J. Bone Miner. Res. 30, 1268–1279 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  296. Krishnamoorthy, D. et al. Marrow adipogenesis and bone loss that parallels estrogen deficiency is slowed by low-intensity mechanical signals. Osteoporos. Int. 27, 747–756 (2016).

    CAS  PubMed  Google Scholar 

  297. Limonard, E. J. et al. Short-term effect of estrogen on human bone marrow fat. J. Bone Miner. Res. 30, 2058–2066 (2015).

    CAS  PubMed  Google Scholar 

  298. Tuomilehto, J. et al. Prevention of type 2 diabetes mellitus by changes in lifestyle among subjects with impaired glucose tolerance. N. Engl. J. Med. 344, 1343–1350 (2001).

    CAS  PubMed  Google Scholar 

  299. Wallace, I. J. et al. Focal enhancement of the skeleton to exercise correlates with responsivity of bone marrow mesenchymal stem cells rather than peak external forces. J. Exp. Biol. 218, 3002–3009 (2015).

    PubMed  PubMed Central  Google Scholar 

  300. Bonewald, L. F. & Johnson, M. L. Osteocytes, mechanosensing and Wnt signaling. Bone 42, 606–615 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  301. Klein-Nulend, J., Semeins, C. M., Ajubi, N. E., Nijweide, P. J. & Burger, E. H. Pulsating fluid flow increases nitric oxide (NO) synthesis by osteocytes but not periosteal fibroblasts—correlation with prostaglandin upregulation. Biochem. Biophys. Res. Commun. 217, 640–648 (1995).

    CAS  PubMed  Google Scholar 

  302. Qin, Y. X., Lin, W. & Rubin, C. The pathway of bone fluid flow as defined by in vivo intramedullary pressure and streaming potential measurements. Ann. Biomed. Eng. 30, 693–702 (2002).

    PubMed  Google Scholar 

  303. Qin, Y. X., Kaplan, T., Saldanha, A. & Rubin, C. Fluid pressure gradients, arising from oscillations in intramedullary pressure, is correlated with the formation of bone and inhibition of intracortical porosity. J. Biomech. 36, 1427–1437 (2003).

    PubMed  Google Scholar 

  304. Rubin, C. T. & Lanyon, L. E. Kappa Delta Award paper. Osteoregulatory nature of mechanical stimuli: function as a determinant for adaptive remodeling in bone. J. Orthop. Res. 5, 300–310 (1987).

    CAS  PubMed  Google Scholar 

  305. Jarvinen, T. L. et al. Randomized controlled study of effects of sudden impact loading on rat femur. J. Bone Miner. Res. 13, 1475–1482 (1998).

    CAS  PubMed  Google Scholar 

  306. Gross, T. S., Edwards, J. L., McLeod, K. J. & Rubin, C. T. Strain gradients correlate with sites of periosteal bone formation. J. Bone Miner. Res. 12, 982–988 (1997).

    CAS  PubMed  Google Scholar 

  307. Lanyon, L. E. & Rubin, C. T. Static versus dynamic loads as an influence on bone remodelling. J. Biomech. 17, 897–905 (1984).

    CAS  PubMed  Google Scholar 

  308. Reyes, M. L., Hernandez, M., Holmgren, L. J., Sanhueza, E. & Escobar, R. G. High-frequency, low-intensity vibrations increase bone mass and muscle strength in upper limbs, improving autonomy in disabled children. J. Bone Miner. Res. 26, 1759–1766 (2011).

    PubMed  Google Scholar 

  309. Dumas, V. et al. Extracellular matrix produced by osteoblasts cultured under low-magnitude, high-frequency stimulation is favourable to osteogenic differentiation of mesenchymal stem cells. Calcif. Tissue Int. 87, 351–364 (2010).

    CAS  PubMed  Google Scholar 

  310. Raso, V., Natale, V. M., Duarte, A. J., Greve, J. M. & Shephard, R. J. Immunological parameters in elderly women: correlations with aerobic power, muscle strength and mood state. Brain Behav. Immun. 26, 597–606 (2012).

    CAS  PubMed  Google Scholar 

  311. Wenger, K. H. et al. Effect of whole-body vibration on bone properties in aging mice. Bone 47, 746–755 (2010).

    PubMed  Google Scholar 

  312. Bacabac, R. G. et al. Bone cell responses to high-frequency vibration stress: does the nucleus oscillate within the cytoplasm? FASEB J. 20, 858–864 (2006).

    CAS  PubMed  Google Scholar 

  313. Sherk, V. D. et al. Acute bone marker responses to whole-body vibration and resistance exercise in young women. J. Clin. Densitom. 16, 104–109 (2013).

    PubMed  Google Scholar 

  314. Pagnotti, G. M. et al. Low magnitude mechanical signals mitigate osteopenia without compromising longevity in an aged murine model of spontaneous granulosa cell ovarian cancer. Bone 51, 570–577 (2012).

    PubMed  PubMed Central  Google Scholar 

  315. Pagnotti, G. M. et al. Low intensity vibration mitigates tumor progression and protects bone quantity and quality in a murine model of myeloma. Bone 90, 69–79 (2016).

    PubMed  PubMed Central  Google Scholar 

  316. David, V. et al. Mechanical loading down-regulates peroxisome proliferator-activated receptor gamma in bone marrow stromal cells and favors osteoblastogenesis at the expense of adipogenesis. Endocrinology 148, 2553–2562 (2007).

    CAS  PubMed  Google Scholar 

  317. Maddalozzo, G. F., Iwaniec, U. T., Turner, R. T., Rosen, C. J. & Widrick, J. J. Whole-body vibration slows the acquisition of fat in mature female rats. Int. J. Obes. 32, 1348–1354 (2008).

    CAS  Google Scholar 

  318. Huang, R. P., Rubin, C. T. & McLeod, K. J. Changes in postural muscle dynamics as a function of age. J. Gerontol. A Biol. Sci. Med. Sci. 54, B352–B357 (1999).

    CAS  PubMed  Google Scholar 

  319. Adams, D. J. et al. Testing the daily stress stimulus theory of bone adaptation with natural and experimentally controlled strain histories. J. Biomech. 30, 671–678 (1997).

    CAS  PubMed  Google Scholar 

  320. Fritton, S. P., McLeod, K. J. & Rubin, C. T. Quantifying the strain history of bone: spatial uniformity and self- similarity of low-magnitude strains. J. Biomech. 33, 317–325 (2000).

    CAS  PubMed  Google Scholar 

  321. Rubin, C. et al. Mechanical strain, induced noninvasively in the high-frequency domain, is anabolic to cancellous bone, but not cortical bone. Bone 30, 445–452 (2002).

    CAS  PubMed  Google Scholar 

  322. Luu, Y. K. et al. Development of diet-induced fatty liver disease in the aging mouse is suppressed by brief daily exposure to low-magnitude mechanical signals. Int. J. Obes. 34, 401–405 (2010).

    CAS  Google Scholar 

  323. Rosen, C. J. et al. Congenic mice with low serum IGF-I have increased body fat, reduced bone mineral density, and an altered osteoblast differentiation program. Bone 35, 1046–1058 (2004).

    CAS  PubMed  Google Scholar 

  324. Mukherjee, S. et al. Pharmacologic targeting of a stem/progenitor population in vivo is associated with enhanced bone regeneration in mice. J. Clin. Invest. 118, 491–504 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  325. Luu, Y. K., Capilla, E., Pessin, J. E., Judex, S. & Rubin, C. T. in 2007 IEEE 33rd Annual Northeast Bioengineering Conference 203–204 (IEEE, Long Island, NY, 2007).

  326. Klein-Nulend, J. et al. Donor age and mechanosensitivity of human bone cells. Osteoporosis Int. 13, 137–146 (2002).

    CAS  Google Scholar 

  327. Gilsanz, V. et al. Low-level, high-frequency mechanical signals enhance musculoskeletal development of young women with low BMD. J. Bone Miner. Res. 21, 1464–1474 (2006).

    PubMed  Google Scholar 

  328. McGee-Lawrence, M. E. et al. Whole-body vibration mimics the metabolic effects of exercise in male leptin receptor-deficient mice. Endocrinology 158, 1160–1171 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  329. Robling, A. G., Burr, D. B. & Turner, C. H. Recovery periods restore mechanosensitivity to dynamically loaded bone. J. Exp. Biol. 204, 3389–3399 (2001).

    CAS  PubMed  Google Scholar 

  330. Guo, S. Insulin signaling, resistance, and the metabolic syndrome: insights from mouse models into disease mechanisms. J. Endocrinol. 220, T1–T23 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  331. Ward, K. et al. Low magnitude mechanical loading is osteogenic in children with disabling conditions. J. Bone Miner. Res. 19, 360–369 (2004).

    PubMed  Google Scholar 

  332. Lam, T. P. et al. Effect of whole body vibration (WBV) therapy on bone density and bone quality in osteopenic girls with adolescent idiopathic scoliosis: a randomized, controlled trial. Osteoporos. Int. 24, 1623–1636 (2013).

    CAS  PubMed  Google Scholar 

  333. Bianchi, M. et al. Effects of low-magnitude high-frequency vibration on bone density, bone resorption and muscular strength in ambulant children affected by Duchenne muscular dystrophy. J. Bone Miner. Res. 28 (Suppl. 1), LB–SU03 (2013).

    Google Scholar 

  334. Mogil, R. J. et al. Effect of low-magnitude, high-frequency mechanical stimulation on BMD among young childhood cancer survivors: a randomized clinical trial. JAMA Oncol. 2, 908–914 (2016).

    PubMed  PubMed Central  Google Scholar 

  335. Leonard, M. B. et al. Effect of low magnitude mechanical stimuli on bone density and structure in pediatric Crohn’s disease: a randomized placebo controlled trial. J. Bone Miner. Res. 31, 1177–1188 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  336. DiVasta, A. D. et al. The ability of low-magnitude mechanical signals to normalize bone turnover in adolescents hospitalized for anorexia nervosa. Osteoporosis Int. 28, 1255–1263 (2017).

    CAS  Google Scholar 

  337. Fritton, J. C., Rubin, C. T., Qin, Y. X. & McLeod, K. J. Whole-body vibration in the skeleton: development of a resonance-based testing device. Ann. Biomed. Eng. 25, 831–839 (1997).

    CAS  PubMed  Google Scholar 

  338. Muir, J., Kiel, D. P. & Rubin, C. T. Safety and severity of accelerations delivered from whole body vibration exercise devices to standing adults. J. Sci. Med. Sport 16, 526–531 (2013).

    PubMed  PubMed Central  Google Scholar 

  339. Kiel, D. P. et al. Insights from the conduct of a device trial in older persons: low magnitude mechanical stimulation for musculoskeletal health. Clin. Trials 7, 354–367 (2010).

    PubMed  PubMed Central  Google Scholar 

  340. Kiel, D. P. et al. Low-magnitude mechanical stimulation to improve bone density in persons of advanced age: a randomized, placebo-controlled trial. J. Bone Miner. Res. 30, 1319–1328 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  341. Asselin, P., Spungen, A. M., Muir, J. W., Rubin, C. T. & Bauman, W. A. Transmission of low-intensity vibration through the axial skeleton of persons with spinal cord injury as a potential intervention for preservation of bone quantity and quality. J. Spinal Cord Med. 34, 52–59 (2011).

    PubMed  PubMed Central  Google Scholar 

  342. International Organization for Standardization. Evaluation of human exposure to whole-body vibration [ISO 2631-1:1985] (ISO, 1985).

  343. Kiiski, J., Heinonen, A., Jarvinen, T. L., Kannus, P. & Sievanen, H. Transmission of vertical whole body vibration to the human body. J. Bone Miner. Res. 23, 1318–1325 (2008).

    PubMed  Google Scholar 

  344. Ozcivici, E. et al. Mechanical signals as anabolic agents in bone. Nat. Rev. Rheumatol. 6, 50–59 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  345. Martinac, B. Mechanosensitive ion channels: molecules of mechanotransduction. J. Cell Sci. 117, 2449–2460 (2004).

    CAS  PubMed  Google Scholar 

  346. Kung, C., Martinac, B. & Sukharev, S. Mechanosensitive channels in microbes. Annu. Rev. Microbiol. 64, 313–329 (2010).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Reviewer information

Nature Reviews Endocrinology thanks G. Duque, and other anonymous reviewers, for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

All authors provided a substantial contribution to the discussion of the material. C.T.R., J.R., G.M.P., T.A.G., M.S. and G.U. contributed to all aspects of this Review. V.S.P., L.E.W. and K.K.N. researched data for the article, contributed to discussion of the content and wrote the article.

Corresponding author

Correspondence to Clinton T. Rubin.

Ethics declarations

Competing interests

C.T.R. is a founder of Marodyne Medical, Inc. and BTT Health and has several patents issued and pending related to the ability of mechanical signals to control musculoskeletal and metabolic disorders. The other authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Glossary

Loading

In terms of mechanical loading, a singular or compound series of static or dynamic (time-varying) forces applied to a system via gravity or direct application from an external body, causing tension, shear or compression.

Unloading

A cell or body is considered mechanically unloaded if no static or dynamic strain is present, such as what might occur with bed rest or spaceflight (that is, microgravity).

Ground-reaction forces

As applicable to biomechanics, ground-reaction forces consist of the normal forces exerted by the ground on the body making contact with it, particularly resulting from a heel strike during walking or running.

Spectral content

Muscle contractive forces, specifically on bone, resonate within a discrete frequency range.

Load sensation

Mechanical loads are ‘sensed’ by cells through transduction of external or internal forces across cytoskeletal proteins into the nucleus.

Tissue senility

The ageing process is associated with the quiescence of regenerative cell populations residing in tissues throughout the body.

Muscle-specific force

Quantification of the contractile forces generated by muscles can be normalized to muscle size ex vivo.

Fluid shear

Fluidic forces applied tangentially across cell membranes or tissues.

Dynamic shear forces

Physiological fluids exert a gradient of pulsatile flow across vessel walls, mineralized bone and cells housed in the bone marrow microenvironment.

Tissue stiffness

In terms of bone, the stiffness of the tissue is correlated to its ability to resist deformation.

Nuclear stiffness

Nuclear stiffness refers to its rigidity and is directly related to polymeric structural proteins (that is, microtubules, intermediate filaments and microfilaments) found across the cytoskeleton, of which actin proteins provide substantial reinforcement.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pagnotti, G.M., Styner, M., Uzer, G. et al. Combating osteoporosis and obesity with exercise: leveraging cell mechanosensitivity. Nat Rev Endocrinol 15, 339–355 (2019). https://doi.org/10.1038/s41574-019-0170-1

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41574-019-0170-1

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research