Warning: fopen(/home/virtual/enm-kes/journal/upload/ip_log/ip_log_2024-03.txt): failed to open stream: Permission denied in /home/virtual/lib/view_data.php on line 88 Warning: fwrite() expects parameter 1 to be resource, boolean given in /home/virtual/lib/view_data.php on line 89 New Insights into Calorie Restriction Induced Bone Loss
Skip Navigation
Skip to contents

Endocrinol Metab : Endocrinology and Metabolism

clarivate
OPEN ACCESS
SEARCH
Search

Articles

Page Path
HOME > Endocrinol Metab > Volume 38(2); 2023 > Article
Review Article
Calcium & bone metabolism New Insights into Calorie Restriction Induced Bone Loss
Keypoint
Caloric restriction is known for its benefits on lifespan and weight reduction, but it can negatively affect the skeletal system. Studies have shown that it suppresses bone growth and increases fracture risk. The exact mechanisms are not fully understood, but reduced mechanical loading and increased bone marrow adipose tissue are believed to play a role. This review explores the impact of caloric restriction on the skeleton, the role of bone marrow adipose tissue in its bone loss effects, and strategies to minimize these effects on skeletal health.
Linyi Liuorcid, Clifford J. Rosenorcid
Endocrinology and Metabolism 2023;38(2):203-213.
DOI: https://doi.org/10.3803/EnM.2023.1673
Published online: April 27, 2023

MaineHealth Institute for Research, Scarborough, ME, USA

Corresponding author: Clifford J. Rosen. MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA Tel: +1-207-396-8157, Fax: +1-207-885-8174 E-mail: Clifford.Rosen@mainehealth.org
• Received: February 2, 2023   • Revised: March 28, 2023   • Accepted: March 30, 2023

Copyright © 2023 Korean Endocrine Society

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • 3,079 Views
  • 170 Download
  • 2 Web of Science
  • 2 Crossref
  • 1 Scopus
  • Caloric restriction (CR) is now a popular lifestyle choice due to its ability in experimental animals to improve lifespan, reduce body weight, and lessen oxidative stress. However, more and more emerging evidence suggests this treatment requires careful consideration because of its detrimental effects on the skeletal system. Experimental and clinical studies show that CR can suppress bone growth and raise the risk of fracture, but the specific mechanisms are poorly understood. Reduced mechanical loading has long been thought to be the primary cause of weight loss-induced bone loss from calorie restriction. Despite fat loss in peripheral depots with calorie restriction, bone marrow adipose tissue (BMAT) increases, and this may play a significant role in this pathological process. Here, we update recent advances in our understanding of the effects of CR on the skeleton, the possible pathogenic role of BMAT in CR-induced bone loss, and some strategies to mitigate any potential side effects on the skeletal system.
Many studies have indicated that caloric restriction (CR) is a good strategy to prolong life expectancy and improve life quality in most species [1,2]. CR is also thought to be beneficial for weight loss, blood glucose control, and reducing oxygen stress [3,4]. However, more and more studies have pointed out that CR should be adopted very cautiously due to its well-documented side effects on the skeletal system [5-9]. The mechanisms by which CR leads to bone loss are still not clear. Recently, many scholars have speculated that bone marrow adipose tissue (BMAT) may exert a pivotal effect on this [10]. Therefore, figuring out the effects of CR on bone metabolism and the role of BMAT in this can lead to a better understanding of CR-related bone diseases and the treatment of bone loss associated with it.
In human research, there is evidence suggesting that weight loss can result in the reduced bone quantity and lower bone mineral density (BMD) in older adults [8,11,12]. In one study, 47 postmenopausal women underwent a 1-year CR trial and found weight loss caused bone loss at the ultradistal radius, 1/3 radius, and total hip [13]. Another study involving 1705 healthy and aging men followed up for 2 years showed that, compared to the people who gained weight during these 2 years, weight-loss participants lost 0.8% more of their hip BMD annually [14]. The effects of CR on younger individuals are controversial. A reduction in bone quantity has not been demonstrated in many studies in younger adults [15,16]. Nevertheless, a 2-year intervention in 218 healthy men and women whose ages were between 20 and 50 (i.e., Comprehensive Assessment of Long-term Effects of Reducing Intake of Energy [CALERIE]) concluded that the calorie restriction group had 2% lower BMD at the femoral neck, lumber spine, and total hip compared to those consuming a regular diet [9].
There is controversy regarding which skeletal areas are most affected by weight loss. Previous studies reported that the bone loss occurred mainly in the trabecular sites [13,17], while other research suggested that cortical bone was also strongly influenced in this process, especially in the weight-bearing sites [18,19]. Liu et al. [20] recently conducted a trial to examine how long-term (over 40 years) and recent (over 6 years) weight loss affected bone metabolism and discovered that both types of weight loss were related to decreased thickness and density of trabecular and cortical bone. Likewise, in rodents, food restriction can also result in reduced bone quantity and bone microstructure [21-23]. Devlin et al. [24] used young male mice (3 weeks of age) to test the effects of 30% calorie restriction on bone mass. The results demonstrated that CR mice were closely associated with lower skeletal parameters in both trabecular and cortical bone [24]. Conversely, another study using young female mice concluded that CR only suppressed cortical bone area, but increased the connectivity of trabeculae [25]. Our previous study showed that feeding female mice (8 weeks of age) a 30% CR diet for 4 and 8 weeks, respectively, showed CR only decreased cortical bone and didn’t influence trabecular bone mass [26]. These opposing results possibly reflect the different background strains of mice, age at onset of CR, sex, severity of the CR, and treatment duration used in various studies.
Except for bone quantity, bone quality is also an important factor in bone health, and several studies have pointed to a distinction between bone mass and bone quality [27,28]. Obese people with a higher weight, for example, have been shown to have compromised bone quality, yet BMD is normal or high [29-31]. In terms of CR, one study showed men who had accelerated bone loss were associated with a lower trabecular bone volume fraction (BV/TV), lower cortical thickness, and higher trabecular separation [32]. This finding was also confirmed in anorexia nervosa patients. One study on anorexia nervosa suggested that, compared with normal weight individuals, anorexia nervosa patients had decreased several related skeletal parameters both in trabecular bone and cortical bone, and this change was distinct from BMD [33,34]. However, another team’s trial concluded that moderate weight loss did not impair the bone quality of trabecular and cortical bone in obese men [35]. One possible explanation is that the subjects chosen for the studies were of varying ages or anatomical sites and responded differently to the CR. Bone strength is an important factor in the overall health of the skeleton, but we currently lack accurate methods for measuring it [4]. In animal experiments, biomechanical testing is a common method used to measure bone strength [24]. The results of three-point bending tests from one study concluded that 40% calorie restriction had little effect on bone stability and strength [36]. Another study in which male mice had caloric-restricted food for 10 weeks showed that diet restriction could reduce the ultimate force, stiffness, and second area moment of inertia of the femur [37]. Weight loss was also associated with a lower maximal force and yield point force in female rats, according to research [38]. Based on the evidence, we can conclude that different diet restriction strategies had different effects on the biomechanical properties of the bone. Moreover, even in the models that concluded food restriction caused lower bone strength, the skeleton still grew rather than stopping. In the future, more exploration and research will be needed on how to apply the bone strength-related measurements to our clinical practice.
Previously, the reduction in bone quantity and quality caused by CR was thought to be associated with weight loss. The skeleton system is a dynamic organ that can respond to various mechanical loads. Mechanical stress induced by body weight is a pivotal factor in maintaining bone homeostasis and increasing bone volume and strength [39,40]. Lower body weight has been demonstrated to have lower BMD and higher fracture incidence [41,42]. However, recent research indicates that bone mechanical stress is not the only factor affecting bone mass. Hyldstrup et al. [43] studied the effect of obesity on bone metabolism and concluded that BMD could also increase in non-weight-bearing regions. Similarly, Fogelholm et al. [44] found that BMD in non-weight-bearing regions was more closely related to weight loss than BMD in weight-bearing regions. Therefore, it is likely that other factors play an important role in the process of bone loss due to weight loss.
Unlike white adipose tissue (WAT) and brown adipose tissue, BMAT, another type of fat tissue in the bone marrow that accounts for more than 10% of total body fat, has not been well defined until recently [45,46]. Adipose tissue in the bone marrow is present from birth and was previously thought to be a passive filler in response to bone loss and the aging process. Recent research suggests that BMAT is important in responding to a variety of challenges, such as aging, diabetes, nutritional changes, and temperature differences [47-51]. In addition, BMAT is also a metabolic organ that can secrete endocrine cytokines to influence energy metabolism [45,52,53]. Furthermore, studies in animal models and humans have shown that there are two different types of BMAT, suggesting BMAT is a heterogeneous tissue. Regulated BMAT (rBMAT), which exists in the proximal regions surrounded by hematopoietic elements, is comprised of single, smaller adipocytes, while constitutive BMAT (cBMAT), which exists in the distal regions, features larger adipocytes but less marrow elements [54]. As noted, rBMAT has more active hematopoiesis and behaves more strongly under nutritional stress than cBMAT [54,55], but one study by Li et al. [56] suggested that 12 weeks of CR could also expand the cBMAT in the tibia, indicating that cBMAT may also have an important impact on skeleton metabolism.
It is well established that CR will decrease the mass and size of adipocytes in WAT, especially in visceral adipose tissue [57-61]. BMAT, on the other hand, accumulates in the presence of CR in both animal models and clinical diseases [24-26,55,62,63]. Anorexia nervosa is a psychiatric disease characterized by self-imposed starvation, which will cause extreme weight loss [19]. Paradoxically, clinical evidence suggests that anorexia nervosa reduces peripheral fat but increases fat tissues in the bone marrow, and when weight was regained, there was a corresponding decrease in BMAT [64,65]. Fazeli et al. [66] studied the response of BMAT to high fat and energy restriction in 23 volunteers and discovered that CR significantly increased vertebral BMAT.
Conclusions about the weight loss caused by surgery on BMAT are still controversial. Patients undergoing vertical sleeve gastrectomy showed that BMAT increases, while in people who received the Roux-en-Y gastric bypass, BMAT decreased after surgery [51,67,68]. In murine models, CR significantly increases the content of BMAT in adult and growing mice, as well as in mice of different sexes, accompanied by a decrease in serum leptin and insulin like growth factor 1 levels [24-26,55]. Our recent research also showed that 30% CR lasting for 4 or 8 weeks could induce bone marrow adiposity in both male and female mice.
To date, the mechanisms of how CR reduces bone integrity have not been fully elucidated. More research indicates BMAT may play an important role in CR-induced bone loss due to the inverse association between bone mass and marrow adipose tissue [24-26]. There are some theoretical foundations and experimental results to support this conclusion. Mesenchymal stem cells in bone marrow can give rise to various cell types like osteoblasts, adipocytes, and chondrocytes [9,69]. The accrual of adipose tissue under the condition of CR would blunt the ability of bone marrow stem cells (BMSCs) to differentiate into osteoblasts. Our research also showed that BMSCs from the CR group would form more lipid droplets but fewer calcium deposits after 9 days of adipogenic induction and 14 days of osteogenic induction. Moreover, the increased adipocytes in bone marrow could secrete some cytokines or adipokines to negatively influence the skeletal system. Peroxisome proliferator-receptor activated gamma (Pparγ), a major adipocyte transcriptional factor, increases during the CR process [55]. Wan et al. [70] found that heterozygous Pparγ +/– mice had more bone and less BMAT. In an in vitro study, Pparγ activation had an inhibitory effect on osteogenic differentiation [71-73]. Recently, adipsin, another adipokine secreted by fat tissue, was associated with bone loss [55]. Adipsin, also known as complement factor D, was secreted almost exclusively from marrow adipocytes and its expression significantly increases with CR, where the induction reached ninefold compared to the ad libitum group [55,74,75]. More importantly, when comparing the responses of wild type mice and adipsin knockout (KO) mice to CR, the researchers found that adipsin KO mice could successfully reverse the inhibitory impacts of CR on bone and decrease the adiposity of BMAT [55]. In addition to the above-mentioned reasons, many researchers have pointed out there are other related mechanisms in this process. Receptor activator of nuclear factors κB ligand (RANKL), an important cytokine for osteoclast differentiation, was previously shown to be produced primarily by osteoblasts and osteocytes [76,77], but bone marrow adipocytes (BMAds) have now been shown to be a critical source of RANKL production, affecting the balance between bone formation and bone resorption [78-80]. In humans, Attane et al. [81] showed that adipocytes in the bone marrow were devoid of lipolytic activity, which was different from classic white adipocytes. Similarly, research from Li et al. [82] concluded BMAds-induced lipolysis was essential to maintain bone mass under CR in a mouse model. Therefore, prolonged energy restriction may also contribute to the lack of bone mass by impairing the lipolysis of BMAds.
Parathyroid hormone
Parathyroid hormone (PTH), a hormone from the parathyroid glands, has been convinced to have some beneficial effects on bone and is a potent therapy for osteoporosis [83-85]. Although the specific mechanism involved in PTH is not very clear, many studies have pointed out that PTH can target different cell types. For bone formation, PTH could increase osteoblast numbers and promote osteoblast activity [86-88]. PTH has also been shown to stimulate osteoclasts and bone resorption by increasing the expression of RANKL [89,90]. Besides osteoblasts and osteoclasts, PTH could affect the osteocytes to change the expressions of fibroblast growth factor-23 and sclerostin [91,92]. Furthermore, recent research has shown that PTH can recruit marrow stromal cells (MSCs) and change the balance between bone and fat by shifting the marrow lineage [93,94]. PTH treatment shifted MSCs’ commitment to the osteogenic lineage at the expense of the adipogenic lineage. Similar research suggested PTH related protein (PTHrP) could downregulate the differentiation of adipocytes, and overexpression of PTHrP in mice resulted in the accrual of osteoblasts [95,96]. Because CR caused MSCs to prefer differentiating toward adipocytes over osteoblasts, PTH treatment might mitigate the negative effects of CR on the skeletal system to some extent. Our previous study also showed that PTH administration to CR suppressed bone marrow adiposity and increased bone mass. Meanwhile, PTH promoted the lipolysis of adipocytes, and this fuel could transfer to osteoblasts during bone formation [26].
Vitamin D
Vitamin D, including vitamin D2 and vitamin D3, is hydroxylated in the liver to 25-hydoxyvitamin (25-OHD/calcidiol) and then converted in the kidney and bone cells to 1, 25-dihydroxyvitamin (1, 25(OH)D/calcitriol) [97]. Calcidiol, a major circulating form, is usually used for estimating vitamin D status, whereas calcitriol is the active metabolite for biological functions [97]. In order to function, vitamin D first needs to be combined with the vitamin D receptor (VDR), which can be expressed in various cells like BMSCs, osteoblasts, and osteoclast precursor lineages [98,99]. Vitamin D has been well recognized as having a close relationship with bone health. Vitamin D deficiency increased the risk of falling in older people, and supplementing with 700 to 800 IU of vitamin D daily can reduce this risk [100,101]. Animal evidence also suggested that VDR deficient mice have significantly impaired development and growth [102]. Although the specific mechanisms are still not very clear, it has been confirmed that calcitriol can promote the differentiation of BMSCs into osteoblasts [97,103,104]. Vitamin D also plays a role in osteoclastogenesis. Physiological doses of vitamin D could reduce bone resorption, while supraphysiological levels could increase the osteoclasts’ activity by enhancing RANKL expression [105]. Recently, studies have demonstrated calcitriol could retard the adipogenesis of bone marrow, which is consistent with the notion that bone and adipose tissue have an inverse relationship in the bone marrow [106-108]. One randomized double-blind trial studied the effect of vitamin D supplementation on the calcium level under the CR process and concluded that supplementing vitamin D during the CR could normalize serum calcium [109]. Taken together evidence suggests that vitamin D might be a potential treatment modality to decrease the inhibitory impacts of CR on the skeletal system.
Exercise
The effects of physical exercise on weight-stable individuals are controversial. Some evidence suggested that exercise-induced weight loss was harmful to bone health, while other evidence indicated that exercise had a positive impact on the skeletal system [6,110-112]. The different results may be due to the varying exercise prescriptions, the duration of the trial, and the different ages and genders chosen for the test. However, when it came to CR-induced bone loss, most studies showed that exercise, particularly resistance training, could mitigate this effect [113-115]. Emerging cellular evidence suggested exercise played a role in bone remodeling through different cell types. Exercise has been proven to enhance the proliferation and osteoblastic differentiation potential of BMSCs [116]. In this process, several signaling pathways are involved. The PTH signaling pathway could be activated by exercise, which changes the commitment of MSCs to osteoblasts at the expense of adipocytes [117]. Another pivotal mediator for bone remodeling, the Wnt signaling pathway, has also been reported to play a positive role. Exercise treatment caused β-catenin, a key component of the canonical Wnt pathway, to move to the nucleus while decreasing PPARg expression [118]. In addition to BMSCs, osteocytes are also important targets for exercise. Exercise was demonstrated to inhibit the apoptosis of osteocytes and promote the release of some cytokines like irisin that can influence bone turnover [119,120]. Irisin, a cleaved product from fibronectin type III domain-containing protein 5 (FNDC5), could increase the survival ability of osteocytes and sclerostin production through binding to its receptor, αV integrin, which suggested irisin generated by exercise might be a potential treatment target for CR-induced bone loss. Finally, there are several studies suggesting exercise can suppress bone marrow adiposity [121,122].
Although CR is beneficial for human beings in regards to some tissues, its inhibitory effects on the skeletal system should be addressed. It may not only cause bone loss but also a significant decrease in bone quality, increasing the risk of fracture. As osteoporosis is a major public health problem worldwide and the cost of treating osteoporosis-related fractures has increased over the years, which makes it a heavy burden for both government and individuals, the possibility of osteoporosis due to CR is an issue that must be considered before considering treatment options.
Recently, BMAT has garnered more interest due to its increase in various pathological conditions, including CR, and has been suggested as a possible cause of bone loss. Evidence such as the fact that osteoblasts and adipocytes are derived from the same mesenchymal cells and that some cytokines secreted by adipocytes suppress osteoblast differentiation is suggesting that BMAT may become an important target for the future treatment of diet-induced bone loss (Fig. 1).

CONFLICTS OF INTEREST

No potential conflict of interest relevant to this article was reported.

Fig. 1.
Caloric restriction induces bone loss and bone marrow adiposity. Caloric restriction induces bone marrow stem cells (BMSCs) to differentiate into adipocytes at the expense of osteoblasts, in which some adipokines such as peroxisome proliferator-receptor activated gamma (Pparγ) and adipsin play a role.
enm-2023-1673f1.jpg
  • 1. Madeo F, Carmona-Gutierrez D, Hofer SJ, Kroemer G. Caloric restriction mimetics against age-associated disease: targets, mechanisms, and therapeutic potential. Cell Metab 2019;29:592–610.ArticlePubMed
  • 2. Mattison JA, Colman RJ, Beasley TM, Allison DB, Kemnitz JW, Roth GS, et al. Caloric restriction improves health and survival of rhesus monkeys. Nat Commun 2017;8:14063.ArticlePubMedPMCPDF
  • 3. Redman LM, Smith SR, Burton JH, Martin CK, Il’yasova D, Ravussin E. Metabolic slowing and reduced oxidative damage with sustained caloric restriction support the rate of living and oxidative damage theories of aging. Cell Metab 2018;27:805–15.e4.ArticlePubMedPMC
  • 4. Huang TH, Ables GP. Dietary restrictions, bone density, and bone quality. Ann N Y Acad Sci 2016;1363:26–39.ArticlePubMed
  • 5. Villareal DT, Fontana L, Weiss EP, Racette SB, Steger-May K, Schechtman KB, et al. Bone mineral density response to caloric restriction-induced weight loss or exercise-induced weight loss: a randomized controlled trial. Arch Intern Med 2006;166:2502–10.ArticlePubMed
  • 6. Villareal DT, Shah K, Banks MR, Sinacore DR, Klein S. Effect of weight loss and exercise therapy on bone metabolism and mass in obese older adults: a one-year randomized controlled trial. J Clin Endocrinol Metab 2008;93:2181–7.ArticlePubMedPMCPDF
  • 7. Morley JE, Chahla E, Alkaade S. Antiaging, longevity and calorie restriction. Curr Opin Clin Nutr Metab Care 2010;13:40–5.ArticlePubMed
  • 8. Schwartz AV, Johnson KC, Kahn SE, Shepherd JA, Nevitt MC, Peters AL, et al. Effect of 1 year of an intentional weight loss intervention on bone mineral density in type 2 diabetes: results from the Look AHEAD randomized trial. J Bone Miner Res 2012;27:619–27.ArticlePubMed
  • 9. Villareal DT, Fontana L, Das SK, Redman L, Smith SR, Saltzman E, et al. Effect of two-year caloric restriction on bone metabolism and bone mineral density in non-obese younger adults: a randomized clinical trial. J Bone Miner Res 2016;31:40–51.ArticlePubMedPMCPDF
  • 10. Pachon-Pena G, Bredella MA. Bone marrow adipose tissue in metabolic health. Trends Endocrinol. Metab 2022;33:401–8.
  • 11. Svendsen OL, Hassager C, Christiansen C. Effect of an energy-restrictive diet, with or without exercise, on lean tissue mass, resting metabolic rate, cardiovascular risk factors, and bone in overweight postmenopausal women. Am J Med 1993;95:131–40.ArticlePubMed
  • 12. Avenell A, Richmond PR, Lean ME, Reid DM. Bone loss associated with a high fibre weight reduction diet in postmenopausal women. Eur J Clin Nutr 1994;48:561–6.PubMed
  • 13. Sukumar D, Ambia-Sobhan H, Zurfluh R, Schlussel Y, Stahl TJ, Gordon CL, et al. Areal and volumetric bone mineral density and geometry at two levels of protein intake during caloric restriction: a randomized, controlled trial. J Bone Miner Res 2011;26:1339–48.ArticlePubMedPMC
  • 14. Bleicher K, Cumming RG, Naganathan V, Travison TG, Sambrook PN, Blyth FM, et al. The role of fat and lean mass in bone loss in older men: findings from the CHAMP study. Bone 2011;49:1299–305.ArticlePubMed
  • 15. Riedt CS, Schlussel Y, von Thun N, Ambia-Sobhan H, Stahl T, Field MP, et al. Premenopausal overweight women do not lose bone during moderate weight loss with adequate or higher calcium intake. Am J Clin Nutr 2007;85:972–80.ArticlePubMedPMC
  • 16. Redman LM, Rood J, Anton SD, Champagne C, Smith SR, Ravussin E, et al. Calorie restriction and bone health in young, overweight individuals. Arch Intern Med 2008;168:1859–66.ArticlePubMedPMC
  • 17. Villalon KL, Gozansky WS, Van Pelt RE, Wolfe P, Jankowski CM, Schwartz RS, et al. A losing battle: weight regain does not restore weight loss-induced bone loss in postmenopausal women. Obesity (Silver Spring) 2011;19:2345–50.ArticlePubMedPMCPDF
  • 18. Frolich J, Hansen S, Winkler LA, Andresen AK, Hermann AP, Stoving RK. The role of body weight on bone in anorexia nervosa: a HR-pQCT study. Calcif Tissue Int 2017;101:24–33.ArticlePubMedPMCPDF
  • 19. Fazeli PK, Klibanski A. Effects of anorexia nervosa on bone metabolism. Endocr Rev 2018;39:895–910.ArticlePubMedPMC
  • 20. Liu CT, Sahni S, Xu H, McLean RR, Broe KE, Hannan MT, et al. Long-term and recent weight change are associated with reduced peripheral bone density, deficits in bone microarchitecture, and decreased bone strength: the Framingham Osteoporosis Study. J Bone Miner Res 2018;33:1851–8.ArticlePubMedPMCPDF
  • 21. LaMothe JM, Hepple RT, Zernicke RF. Selected contribution: bone adaptation with aging and long-term caloric restriction in Fischer 344 x Brown-Norway F1-hybrid rats. J Appl Physiol (1985) 2003;95:1739–45.PubMed
  • 22. Ables GP, Perrone CE, Orentreich D, Orentreich N. Methionine-restricted C57BL/6J mice are resistant to diet-induced obesity and insulin resistance but have low bone density. PLoS One 2012;7:e51357.ArticlePubMedPMC
  • 23. Cao JJ. Caloric restriction combined with exercise is effective in reducing adiposity and mitigating bone structural deterioration in obese rats. Ann N Y Acad Sci 2018;1433:41–52.ArticlePubMedPDF
  • 24. Devlin MJ, Cloutier AM, Thomas NA, Panus DA, Lotinun S, Pinz I, et al. Caloric restriction leads to high marrow adiposity and low bone mass in growing mice. J Bone Miner Res 2010;25:2078–88.ArticlePubMedPMC
  • 25. Devlin MJ, Brooks DJ, Conlon C, van Vliet M, Louis L, Rosen CJ, et al. Daily leptin blunts marrow fat but does not impact bone mass in calorie-restricted mice. J Endocrinol 2016;229:295–306.ArticlePubMedPMC
  • 26. Maridas DE, Rendina-Ruedy E, Helderman RC, DeMambro VE, Brooks D, Guntur AR, et al. Progenitor recruitment and adipogenic lipolysis contribute to the anabolic actions of parathyroid hormone on the skeleton. FASEB J 2019;33:2885–98.ArticlePubMedPMCPDF
  • 27. Seeman E, Delmas PD. Bone quality: the material and structural basis of bone strength and fragility. N Engl J Med 2006;354:2250–61.ArticlePubMed
  • 28. Villareal DT, Kotyk JJ, Armamento-Villareal RC, Kenguva V, Seaman P, Shahar A, et al. Reduced bone mineral density is not associated with significantly reduced bone quality in men and women practicing long-term calorie restriction with adequate nutrition. Aging Cell 2011;10:96–102.ArticlePubMedPMC
  • 29. Dimitri P, Wales JK, Bishop N. Fat and bone in children: differential effects of obesity on bone size and mass according to fracture history. J Bone Miner Res 2010;25:527–36.ArticlePubMed
  • 30. Premaor MO, Compston JE, Fina Aviles F, Pages-Castella A, Nogues X, Diez-Perez A, et al. The association between fracture site and obesity in men: a population-based cohort study. J Bone Miner Res 2013;28:1771–7.ArticlePubMed
  • 31. Choi YJ, Kim DJ, Lee Y, Chung YS. Insulin is inversely associated with bone mass, especially in the insulin-resistant population: the Korea and US National Health and Nutrition Examination Surveys. J Clin Endocrinol Metab 2014;99:1433–41.ArticlePubMed
  • 32. Cauley JA, Burghardt AJ, Harrison SL, Cawthon PM, Schwartz AV, Connor EB, et al. Accelerated bone loss in older men: effects on bone microarchitecture and strength. J Bone Miner Res 2018;33:1859–69.ArticlePubMedPMCPDF
  • 33. Faje AT, Karim L, Taylor A, Lee H, Miller KK, Mendes N, et al. Adolescent girls with anorexia nervosa have impaired cortical and trabecular microarchitecture and lower estimated bone strength at the distal radius. J Clin Endocrinol Metab 2013;98:1923–9.ArticlePubMedPMC
  • 34. Bredella MA, Misra M, Miller KK, Madisch I, Sarwar A, Cheung A, et al. Distal radius in adolescent girls with anorexia nervosa: trabecular structure analysis with high-resolution flat-panel volume CT. Radiology 2008;249:938–46.ArticlePubMedPMC
  • 35. Pop LC, Sukumar D, Tomaino K, Schlussel Y, Schneider SH, Gordon CL, et al. Moderate weight loss in obese and overweight men preserves bone quality. Am J Clin Nutr 2015;101:659–67.ArticlePubMedPMC
  • 36. Behrendt AK, Kuhla A, Osterberg A, Polley C, Herlyn P, Fischer DC, et al. Dietary restriction-induced alterations in bone phenotype: effects of lifelong versus short-term caloric restriction on femoral and vertebral bone in C57BL/6 mice. J Bone Miner Res 2016;31:852–63.ArticlePubMed
  • 37. Hamrick MW, Ding KH, Ponnala S, Ferrari SL, Isales CM. Caloric restriction decreases cortical bone mass but spares trabecular bone in the mouse skeleton: implications for the regulation of bone mass by body weight. J Bone Miner Res 2008;23:870–8.ArticlePubMed
  • 38. Shen CL, Zhu W, Gao W, Wang S, Chen L, Chyu MC. Energy-restricted diet benefits body composition but degrades bone integrity in middle-aged obese female rats. Nutr Res 2013;33:668–76.ArticlePubMed
  • 39. Frost HM. Bone “mass” and the “mechanostat”: a proposal. Anat Rec 1987;219:1–9.ArticlePubMed
  • 40. Frost HM. Obesity, and bone strength and “mass”: a tutorial based on insights from a new paradigm. Bone 1997;21:211–4.ArticlePubMed
  • 41. Ensrud KE, Lipschutz RC, Cauley JA, Seeley D, Nevitt MC, Scott J, et al. Body size and hip fracture risk in older women: a prospective study. Am J Med 1997;103:274–80.PubMed
  • 42. Ravn P, Cizza G, Bjarnason NH, Thompson D, Daley M, Wasnich RD, et al. Low body mass index is an important risk factor for low bone mass and increased bone loss in early postmenopausal women. J Bone Miner Res 1999;14:1622–7.ArticlePubMed
  • 43. Hyldstrup L, Andersen T, McNair P, Breum L, Transbol I. Bone metabolism in obesity: changes related to severe overweight and dietary weight reduction. Acta Endocrinol (Copenh) 1993;129:393–8.ArticlePubMed
  • 44. Fogelholm GM, Sievanen HT, Kukkonen-Harjula TK, Pasanen ME. Bone mineral density during reduction, maintenance and regain of body weight in premenopausal, obese women. Osteoporos Int 2001;12:199–206.ArticlePubMedPDF
  • 45. Nandy A, Rendina-Ruedy E. Bone marrow adipocytes: good, bad, or just different? Best Pract Res Clin Endocrinol Metab 2021;35:101550.ArticlePubMedPMC
  • 46. Spencer JA, Ferraro F, Roussakis E, Klein A, Wu J, Runnels JM, et al. Direct measurement of local oxygen concentration in the bone marrow of live animals. Nature 2014;508:269–73.ArticlePubMedPMCPDF
  • 47. Deng P, Yuan Q, Cheng Y, Li J, Liu Z, Liu Y, et al. Loss of KDM4B exacerbates bone-fat imbalance and mesenchymal stromal cell exhaustion in skeletal aging. Cell Stem Cell 2021;28:1057–73.e7.ArticlePubMedPMC
  • 48. Yu EW, Greenblatt L, Eajazi A, Torriani M, Bredella MA. Marrow adipose tissue composition in adults with morbid obesity. Bone 2017;97:38–42.ArticlePubMedPMC
  • 49. Bredella MA, Gill CM, Gerweck AV, Landa MG, Kumar V, Daley SM, et al. Ectopic and serum lipid levels are positively associated with bone marrow fat in obesity. Radiology 2013;269:534–41.ArticlePubMedPMC
  • 50. Patsch JM, Li X, Baum T, Yap SP, Karampinos DC, Schwartz AV, et al. Bone marrow fat composition as a novel imaging biomarker in postmenopausal women with prevalent fragility fractures. J Bone Miner Res 2013;28:1721–8.ArticlePubMedPMC
  • 51. Bredella MA, Greenblatt LB, Eajazi A, Torriani M, Yu EW. Effects of Roux-en-Y gastric bypass and sleeve gastrectomy on bone mineral density and marrow adipose tissue. Bone 2017;95:85–90.ArticlePubMedPMC
  • 52. 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;20:368–75.ArticlePubMedPMC
  • 53. Caers J, Deleu S, Belaid Z, De Raeve H, Van Valckenborgh E, De Bruyne E, et al. Neighboring adipocytes participate in the bone marrow microenvironment of multiple myeloma cells. Leukemia 2007;21:1580–4.ArticlePubMedPDF
  • 54. Scheller EL, Doucette CR, Learman BS, Cawthorn WP, Khandaker S, Schell B, et al. Region-specific variation in the properties of skeletal adipocytes reveals regulated and constitutive marrow adipose tissues. Nat Commun 2015;6:7808.ArticlePubMedPMCPDF
  • 55. Aaron N, Kraakman MJ, Zhou Q, Liu Q, Costa S, Yang J, et al. Adipsin promotes bone marrow adiposity by priming mesenchymal stem cells. Elife 2021;10:e69209.ArticlePubMedPMCPDF
  • 56. Li Z, Bagchi DP, Zhu J, Bowers E, Yu H, Hardij J, et al. Constitutive bone marrow adipocytes suppress local bone formation. JCI Insight 2022;7:e160915.ArticlePubMedPMC
  • 57. Larson-Meyer DE, Heilbronn LK, Redman LM, Newcomer BR, Frisard MI, Anton S, et al. Effect of calorie restriction with or without exercise on insulin sensitivity, beta-cell function, fat cell size, and ectopic lipid in overweight subjects. Diabetes Care 2006;29:1337–44.PubMed
  • 58. Duarte FO, Sene-Fiorese M, Manzoni MS, de Freitas LF, Cheik NC, Garcia de Oliveira Duarte AC, et al. Caloric restriction and refeeding promoted different metabolic effects in fat depots and impaired dyslipidemic profile in rats. Nutrition 2008;24:177–86.ArticlePubMed
  • 59. Maio MT, Hannan JL, Komolova M, Adams MA. Caloric restriction prevents visceral adipose tissue accumulation and maintains erectile function in aging rats. J Sex Med 2012;9:2273–83.ArticlePubMed
  • 60. Racette SB, Weiss EP, Villareal DT, Arif H, Steger-May K, Schechtman KB, et al. One year of caloric restriction in humans: feasibility and effects on body composition and abdominal adipose tissue. J Gerontol A Biol Sci Med Sci 2006;61:943–50.ArticlePubMedPMCPDF
  • 61. Murphy JC, McDaniel JL, Mora K, Villareal DT, Fontana L, Weiss EP. Preferential reductions in intermuscular and visceral adipose tissue with exercise-induced weight loss compared with calorie restriction. J Appl Physiol (1985) 2012;112:79–85.ArticlePubMedPMC
  • 62. Bredella MA, Fazeli PK, Miller KK, Misra M, Torriani M, Thomas BJ, et al. Increased bone marrow fat in anorexia nervosa. J Clin Endocrinol Metab 2009;94:2129–36.ArticlePubMedPMC
  • 63. Bredella MA, Karzar NH, Singhal V, Bose A, Animashaun A, Mitchell DM, et al. Impact of sleeve gastrectomy on bone outcomes in adolescents vs. adults with obesity. Bone 2021;149:115975.ArticlePubMedPMC
  • 64. Ecklund K, Vajapeyam S, Feldman HA, Buzney CD, Mulkern RV, Kleinman PK, et al. Bone marrow changes in adolescent girls with anorexia nervosa. J Bone Miner Res 2010;25:298–304.ArticlePubMedPMC
  • 65. Fazeli PK, Bredella MA, Freedman L, Thomas BJ, Breggia A, Meenaghan E, et al. Marrow fat and preadipocyte factor-1 levels decrease with recovery in women with anorexia nervosa. J Bone Miner Res 2012;27:1864–71.ArticlePubMedPMC
  • 66. Fazeli PK, Bredella MA, Pachon-Pena G, Zhao W, Zhang X, Faje AT, et al. The dynamics of human bone marrow adipose tissue in response to feeding and fasting. JCI Insight 2021;6:e138636.ArticlePubMedPMC
  • 67. Bredella MA, Singhal V, Hazhir Karzar N, Animashaun A, Bose A, Stanford FC, et al. Effects of sleeve gastrectomy on bone marrow adipose tissue in adolescents and young adults with obesity. J Clin Endocrinol Metab 2020;105:e3961–70.ArticlePubMedPMCPDF
  • 68. Blom-Hogestol IK, Mala T, Kristinsson JA, Hauge EM, Brunborg C, Gulseth HL, et al. Changes in bone marrow adipose tissue one year after Roux-en-Y Gastric bypass: a prospective cohort study. J Bone Miner Res 2019;34:1815–23.ArticlePubMedPDF
  • 69. Tencerova M, Kassem M. The bone marrow-derived stromal cells: commitment and regulation of adipogenesis. Front Endocrinol (Lausanne) 2016;7:127.ArticlePubMedPMC
  • 70. Wan Y, Chong LW, Evans RM. PPAR-gamma regulates osteoclastogenesis in mice. Nat Med 2007;13:1496–503.ArticlePubMedPDF
  • 71. Khan E, Abu-Amer Y. Activation of peroxisome proliferator-activated receptor-gamma inhibits differentiation of preosteoblasts. J Lab Clin Med 2003;142:29–34.PubMed
  • 72. Lecka-Czernik B, Gubrij I, Moerman EJ, Kajkenova O, Lipschitz DA, Manolagas SC, et al. Inhibition of Osf2/Cbfa1 expression and terminal osteoblast differentiation by PPARgamma2. J Cell Biochem 1999;74:357–71.ArticlePubMed
  • 73. Chen Q, Shou P, Zheng C, Jiang M, Cao G, Yang Q, et al. Fate decision of mesenchymal stem cells: adipocytes or osteoblasts? Cell Death Differ 2016;23:1128–39.ArticlePubMedPMCPDF
  • 74. Rosen BS, Cook KS, Yaglom J, Groves DL, Volanakis JE, Damm D, et al. Adipsin and complement factor D activity: an immune-related defect in obesity. Science 1989;244:1483–7.ArticlePubMed
  • 75. Tontonoz P, Hu E, Spiegelman BM. Stimulation of adipogenesis in fibroblasts by PPAR gamma 2, a lipid-activated transcription factor. Cell 1994;79:1147–56.ArticlePubMed
  • 76. Suda T, Takahashi N, Udagawa N, Jimi E, Gillespie MT, Martin TJ. Modulation of osteoclast differentiation and function by the new members of the tumor necrosis factor receptor and ligand families. Endocr Rev 1999;20:345–57.ArticlePubMed
  • 77. Xiong J, Onal M, Jilka RL, Weinstein RS, Manolagas SC, O’Brien CA. Matrix-embedded cells control osteoclast formation. Nat Med 2011;17:1235–41.ArticlePubMedPMCPDF
  • 78. Fan Y, Hanai JI, Le PT, Bi R, Maridas D, DeMambro V, et al. Parathyroid hormone directs bone marrow mesenchymal cell fate. Cell Metab 2017;25:661–72.ArticlePubMedPMC
  • 79. Yu W, Zhong L, Yao L, Wei Y, Gui T, Li Z, et al. Bone marrow adipogenic lineage precursors promote osteoclastogenesis in bone remodeling and pathologic bone loss. J Clin Invest 2021;131:e140214.ArticlePubMedPMC
  • 80. Hu Y, Li X, Zhi X, Cong W, Huang B, Chen H, et al. RANKL from bone marrow adipose lineage cells promotes osteoclast formation and bone loss. EMBO Rep 2021;22:e52481.ArticlePubMedPMCPDF
  • 81. Attane C, Esteve D, Chaoui K, Iacovoni JS, Corre J, Moutahir M, et al. Human bone marrow is comprised of adipocytes with specific lipid metabolism. Cell Rep 2020;30:949–58.e6.ArticlePubMed
  • 82. Li Z, Bowers E, Zhu J, Yu H, Hardij J, Bagchi DP, et al. Lipolysis of bone marrow adipocytes is required to fuel bone and the marrow niche during energy deficits. Elife 2022;11:e78496.ArticlePubMedPMCPDF
  • 83. Shimizu E, Selvamurugan N, Westendorf JJ, Partridge NC. Parathyroid hormone regulates histone deacetylases in osteoblasts. Ann N Y Acad Sci 2007;1116:349–53.ArticlePubMed
  • 84. Neer RM, Arnaud CD, Zanchetta JR, Prince R, Gaich GA, Reginster JY, et al. Effect of parathyroid hormone (1-34) on fractures and bone mineral density in postmenopausal women with osteoporosis. N Engl J Med 2001;344:1434–41.ArticlePubMed
  • 85. Marcus R, Wang O, Satterwhite J, Mitlak B. The skeletal response to teriparatide is largely independent of age, initial bone mineral density, and prevalent vertebral fractures in postmenopausal women with osteoporosis. J Bone Miner Res 2003;18:18–23.ArticlePubMed
  • 86. Dobnig H, Turner RT. Evidence that intermittent treatment with parathyroid hormone increases bone formation in adult rats by activation of bone lining cells. Endocrinology 1995;136:3632–8.ArticlePubMed
  • 87. Lotinun S, Sibonga JD, Turner RT. Differential effects of intermittent and continuous administration of parathyroid hormone on bone histomorphometry and gene expression. Endocrine 2002;17:29–36.ArticlePubMed
  • 88. Esen E, Lee SY, Wice BM, Long F. PTH promotes bone anabolism by stimulating aerobic glycolysis via IGF signaling. J Bone Miner Res 2015;30:1959–68.ArticlePubMedPMC
  • 89. Ben-awadh AN, Delgado-Calle J, Tu X, Kuhlenschmidt K, Allen MR, Plotkin LI, et al. Parathyroid hormone receptor signaling induces bone resorption in the adult skeleton by directly regulating the RANKL gene in osteocytes. Endocrinology 2014;155:2797–809.ArticlePubMedPMCPDF
  • 90. Delgado-Calle J, Hancock B, Likine EF, Sato AY, McAndrews K, Sanudo C, et al. MMP14 is a novel target of PTH signaling in osteocytes that controls resorption by regulating soluble RANKL production. FASEB J 2018;32:2878–90.ArticlePubMedPMCPDF
  • 91. Rhee Y, Bivi N, Farrow E, Lezcano V, Plotkin LI, White KE, et al. Parathyroid hormone receptor signaling in osteocytes increases the expression of fibroblast growth factor-23 in vitro and in vivo. Bone 2011;49:636–43.ArticlePubMedPMC
  • 92. Kramer I, Keller H, Leupin O, Kneissel M. Does osteocytic SOST suppression mediate PTH bone anabolism? Trends Endocrinol Metab 2010;21:237–44.ArticlePubMed
  • 93. Wang J, Wang X, Holz JD, Rutkowski T, Wang Y, Zhu Z, et al. Runx1 is critical for PTH-induced onset of mesenchymal progenitor cell chondrogenic differentiation. PLoS One 2013;8:e74255.ArticlePubMedPMC
  • 94. Yu B, Zhao X, Yang C, Crane J, Xian L, Lu W, et al. Parathyroid hormone induces differentiation of mesenchymal stromal/stem cells by enhancing bone morphogenetic protein signaling. J Bone Miner Res 2012;27:2001–14.ArticlePubMedPMC
  • 95. Chan GK, Deckelbaum RA, Bolivar I, Goltzman D, Karaplis AC. PTHrP inhibits adipocyte differentiation by downregulating PPAR gamma activity via a MAPK-dependent pathway. Endocrinology 2001;142:4900–9.ArticlePubMed
  • 96. Calvi LM, Sims NA, Hunzelman JL, Knight MC, Giovannetti A, Saxton JM, et al. Activated parathyroid hormone/parathyroid hormone-related protein receptor in osteoblastic cells differentially affects cortical and trabecular bone. J Clin Invest 2001;107:277–86.ArticlePubMedPMC
  • 97. Christakos S, Dhawan P, Liu Y, Peng X, Porta A. New insights into the mechanisms of vitamin D action. J Cell Biochem 2003;88:695–705.ArticlePubMed
  • 98. Feldman D, Wesley Pike J, Adams JS. Vitamin D; 3rd ed. London: Elsevier; 2011. Chapter 61, Vitamin D and osteoporosis. p. 1129–44.
  • 99. Geng S, Zhou S, Glowacki J. Effects of 25-hydroxyvitamin D (3) on proliferation and osteoblast differentiation of human marrow stromal cells require CYP27B1/1α-hydroxylase. J Bone Miner Res 2011;26:1145–53.ArticlePubMedPMC
  • 100. Cranney A, Horsley T, O’Donnell S, Weiler H, Puil L, Ooi D, et al. Effectiveness and safety of vitamin D in relation to bone health. Evid Rep Technol Assess (Full Rep) 2007;158:1–235.
  • 101. Bischoff-Ferrari HA, Dawson-Hughes B, Staehelin HB, Orav JE, Stuck AE, Theiler R, et al. Fall prevention with supplemental and active forms of vitamin D: a meta-analysis of randomized controlled trials. BMJ 2009;339:b3692.ArticlePubMedPMC
  • 102. Li YC, Pirro AE, Amling M, Delling G, Baron R, Bronson R, et al. Targeted ablation of the vitamin D receptor: an animal model of vitamin D-dependent rickets type II with alopecia. Proc Natl Acad Sci U S A 1997;94:9831–5.ArticlePubMedPMC
  • 103. Piek E, Sleumer LS, van Someren EP, Heuver L, de Haan JR, de Grijs I, et al. Osteo-transcriptomics of human mesenchymal stem cells: accelerated gene expression and osteoblast differentiation induced by vitamin D reveals c-MYC as an enhancer of BMP2-induced osteogenesis. Bone 2010;46:613–27.ArticlePubMed
  • 104. Liu P, Oyajobi BO, Russell RG, Scutt A. Regulation of osteogenic differentiation of human bone marrow stromal cells: interaction between transforming growth factor-beta and 1,25(OH)(2) vitamin D(3) In vitro. Calcif Tissue Int 1999;65:173–80.PubMed
  • 105. Suda T, Ueno Y, Fujii K, Shinki T. Vitamin D and bone. J Cell Biochem 2003;88:259–66.ArticlePubMedPDF
  • 106. Kelly KA, Gimble JM. 1,25-Dihydroxy vitamin D3 inhibits adipocyte differentiation and gene expression in murine bone marrow stromal cell clones and primary cultures. Endocrinology 1998;139:2622–8.ArticlePubMed
  • 107. Duque G, Macoritto M, Kremer R. 1,25(OH)2D3 inhibits bone marrow adipogenesis in senescence accelerated mice (SAM-P/6) by decreasing the expression of peroxisome proliferator-activated receptor gamma 2 (PPARgamma2). Exp Gerontol 2004;39:333–8.PubMed
  • 108. Ding J, Nagai K, Woo JT. Insulin-dependent adipogenesis in stromal ST2 cells derived from murine bone marrow. Biosci Biotechnol Biochem 2003;67:314–21.ArticlePubMed
  • 109. Shapses SA, Sukumar D, Schneider SH, Schlussel Y, Sherrell RM, Field MP, et al. Vitamin D supplementation and calcium absorption during caloric restriction: a randomized double-blind trial. Am J Clin Nutr 2013;97:637–45.ArticlePubMedPMC
  • 110. Shah K, Armamento-Villareal R, Parimi N, Chode S, Sinacore DR, Hilton TN, et al. Exercise training in obese older adults prevents increase in bone turnover and attenuates decrease in hip bone mineral density induced by weight loss despite decline in bone-active hormones. J Bone Miner Res 2011;26:2851–9.ArticlePubMed
  • 111. Bolam KA, van Uffelen JG, Taaffe DR. The effect of physical exercise on bone density in middle-aged and older men: a systematic review. Osteoporos Int 2013;24:2749–62.ArticlePubMedPDF
  • 112. Zhao R, Zhao M, Xu Z. The effects of differing resistance training modes on the preservation of bone mineral density in postmenopausal women: a meta-analysis. Osteoporos Int 2015;26:1605–18.ArticlePubMedPDF
  • 113. Hosny IA, Elghawabi HS, Younan WB, Sabbour AA, Gobrial MA. Beneficial impact of aerobic exercises on bone mineral density in obese premenopausal women under caloric restriction. Skeletal Radiol 2012;41:423–7.ArticlePubMedPDF
  • 114. Beavers KM, Beavers DP, Martin SB, Marsh AP, Lyles MF, Lenchik L, et al. Change in bone mineral density during weight loss with resistance versus aerobic exercise training in older adults. J Gerontol A Biol Sci Med Sci 2017;72:1582–5.ArticlePubMedPMC
  • 115. Villareal DT, Aguirre L, Gurney AB, Waters DL, Sinacore DR, Colombo E, et al. Aerobic or resistance exercise, or both, in dieting obese older adults. N Engl J Med 2017;376:1943–55.ArticlePubMedPMC
  • 116. Hell RC, Ocarino NM, Boeloni JN, Silva JF, Goes AM, Santos RL, et al. Physical activity improves age-related decline in the osteogenic potential of rats’ bone marrow-derived mesenchymal stem cells. Acta Physiol (Oxf) 2012;205:292–301.ArticlePubMed
  • 117. Menuki K, Mori T, Sakai A, Sakuma M, Okimoto N, Shimizu Y, et al. Climbing exercise enhances osteoblast differentiation and inhibits adipogenic differentiation with high expression of PTH/PTHrP receptor in bone marrow cells. Bone 2008;43:613–20.ArticlePubMed
  • 118. Bu S, Chen Y, Wang S, Zhang F, Ji G. Treadmill training regulates β-catenin signaling through phosphorylation of GSK-3β in lumbar vertebrae of ovariectomized rats. Eur J Appl Physiol 2012;112:3295–304.ArticlePubMedPDF
  • 119. Swift JM, Swift SN, Nilsson MI, Hogan HA, Bouse SD, Bloomfield SA. Cancellous bone formation response to simulated resistance training during disuse is blunted by concurrent alendronate treatment. J Bone Miner Res 2011;26:2140–50.ArticlePubMed
  • 120. Kim H, Wrann CD, Jedrychowski M, Vidoni S, Kitase Y, Nagano K, et al. Irisin mediates effects on bone and fat via αV integrin receptors. Cell 2018 175:1756–68. e17.ArticlePubMedPMC
  • 121. Styner M, Pagnotti GM, Galior K, Wu X, Thompson WR, Uzer G, et al. Exercise regulation of marrow fat in the setting of PPARγ agonist treatment in female C57BL/6 mice. Endocrinology 2015;156:2753–61.ArticlePubMedPMC
  • 122. 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.ArticlePubMedPMCPDF

Figure & Data

References

    Citations

    Citations to this article as recorded by  
    • Bone Marrow Adipose Tissue Is Not Required for Reconstitution of the Immune System Following Irradiation in Male Mice
      Jessica A. Keune, Carmen P. Wong, Adam J. Branscum, Scott A. Menn, Urszula T. Iwaniec, Russell T. Turner
      International Journal of Molecular Sciences.2024; 25(4): 1980.     CrossRef
    • Obesity, diabetes and risk of bone fragility: How BMAT behavior is affected by metabolic disturbances and its influence on bone health
      Gregório Corrêa Guimarães, João Bosco Costa Coelho, João Gabriel Oliveira Silva, Ana Carolina Chalfun de Sant’Ana, Cássia Alves Carrilho de Sá, Júlia Marques Moreno, Lívia Marçal Reis, Camila Souza de Oliveira Guimarães
      Osteoporosis International.2023;[Epub]     CrossRef

    Figure

    Endocrinol Metab : Endocrinology and Metabolism