Acessibilidade / Reportar erro

Bone, fat, and muscle interactions in health and disease

Abstract

Energy metabolism is a point of integration among the various organs and tissues of the human body, not only in terms of consumption of energy substrates but also because it concentrates a wide interconnected network controlled by endocrine factors. Thus, not only do tissues consume substrates, but they also participate in modulating energy metabolism. Soft mesenchymal tissues, in particular, play a key role in this process. The recognition that high energy consumption is involved in bone remodeling has been accompanied by evidence showing that osteoblasts and osteocytes produce factors that influence, for example, insulin sensitivity and appetite. Additionally, there are significant interactions between muscle, adipose, and bone tissues to control mutual tissue trophism. Not by chance, trophic and functional changes in these tissues go hand in hand from the beginning of an individual’s development until aging. Likewise, metabolic and nutritional diseases deeply affect the musculoskeletal system and adipose tissue. The present narrative review highlights the importance of the interaction of the mesenchymal tissues for bone development and maintenance and the impact on bone from diseases marked by functional and trophic disorders of adipose and muscle tissues. Arch Endocrinol Metab. 2022;66(5):611-20

Keywords
Osteoporosis; obesity; diabetes mellitus; energy metabolism; mineral metabolism

INTRODUCTION

In the evolution of species, the development of mesenchymal tissues has endowed living beings with special skills for survival in adverse environments such as those with food shortages or low temperatures and situations requiring physical confrontation against aggressors or movements with different degrees of sophistication and speed. No matter the type of mesenchymal tissue – soft (fat and muscle) or hard (bone) – it is possible to easily identify the main function of each of these important body components. In addition to mechanical functions, these tissues have clear storage roles, i.e., storage of proteins by muscle, lipids by fat, and minerals by bone. The metabolic importance of the muscle tissue in adjusting glucose or free fatty acid uptake depending on feeding status has long been recognized (11 Paula FJ, Gouveia LM, Paccola GM, Piccinato CE, Moreira AC, Foss MC. Androgen-related effects on peripheral glucose metabolism in women with congenital adrenal hyperplasia. Horm Metab Res. 1994;26(11):552-6.). However, more time was required for the understanding of the actual importance of the adipose tissue as a site of metabolic regulation beyond its function of energy storage. The list of additional functions of the adipose tissue includes thermal insulation, thermogenesis, mechanical impact absorption, lubrication (joints), and cosmetic (22 de Paula FJA, Rosen CJ. Structure and Function of Bone Marrow Adipocytes. Compr Physiol. 2017;8(1):315-49.). In this regard, it is worth noting that only in this century was due attention given to aspects other than mineral ones in the study of bone metabolism, allowing the recognition of the importance of energy consumption by bone tissue and participation of bone tissue as an active site in controlling energy modulation. A non-trivial aspect that naturally connects mesenchymal tissues is their same embryonic origin from somites derived from paraxial (muscle and bone) and lateral (adipose) mesoderm (33 Pourquie O. Vertebrate somitogenesis. Annu Rev Cell Dev Biol. 2001;17:311-50.). Not by chance, nutritional disorders and metabolic diseases like obesity and diabetes mellitus (DM) have a high impact on the structure, function, development, and maintenance of bone mass (44 Paula FJ, Rosen CJ. Obesity, diabetes mellitus and last but not least, osteoporosis. Arq Bras Endocrinol Metabol. 2010;54(2):150-7.,55 Hofbauer LC, Busse B, Eastell R, Ferrari S, Frost M, Muller R, et al. Bone fragility in diabetes: novel concepts and clinical implications. Lancet Diabetes Endocrinol. 2022;10(3):207-20.). In recent decades, research advances such as the development of imaging technologies, molecular biology, and genetic engineering have allowed unprecedented progress in the recognition of the functional plasticity of mesenchymal tissues. In this process, the complex mutual endocrine interaction between mesenchymal tissues has become increasingly clear, i.e., one tissue stimulating or inhibiting the trophism of the other (66 Kim H, Wrann CD, Jedrychowski M, Vidoni S, Kitase Y, Nagano K, et al. Irisin Mediates Effects on Bone and Fat via alpha V Integrin Receptors. Cell. 2018;175(7):1756-68-e17.). Thus, there is great interest in unveiling the participation of muscle and adipose tissue in the development of bone damage in several diseases.

For the preparation of this narrative review, we performed a PubMed search using the following keywords: bone AND muscle, bone AND adipose tissue, osteoporosis, obesity, weight loss, and sarcopenia. We then made a careful selection of the articles which were to be included in the manuscript. This narrative review has been specially designed with these considerations in mind to present the reader with an up-to-date view on this topic.

ADIPOSE TISSUE AND SKELETON INTERACTION

No other element in the human body surpasses the adipose tissue’s ability to expand. Thus, adipose tissue plays a crucial role in determining body weight. Body weight, in turn, has a strong positive association with bone mass (77 Reid IR. Fat and bone. Arch Biochem Biophys. 2010;503(1):20-7.). Despite conflicting data regarding which tissue – muscle or fat – has a greater influence on bone mass, there is no doubt that both play a strong role in the development and maintenance of bone mass (88 Reid IR. Relationships between fat and bone. Osteoporos Int. 2008;19(5):595-606.). During the growth phase in childhood and adolescence, the concomitant development of mesenchymal tissues suggests a possible mutual and positive interrelationship between them (99 Alessandri SB, Pereira Fde A, Villela RA, Antonini SR, Elias PC, Martinelli CE Jr., et al. Bone mineral density and body composition in girls with idiopathic central precocious puberty before and after treatment with a gonadotropin-releasing hormone agonist. Clinics (Sao Paulo). 2012;67(6):591-6.). Along the same lines, it is possible to assume a non-linear relationship between the adipose tissue and the other mesenchymal tissues. During aging and in diseases such as DM and obesity, the expansion of adipose tissue is not associated with muscle or bone strengthening or performance (22 de Paula FJA, Rosen CJ. Structure and Function of Bone Marrow Adipocytes. Compr Physiol. 2017;8(1):315-49.,1010 Vilaca KH, Carneiro JA, Ferriolli E, Lima NK, de Paula FJ, Moriguti JC. Body composition, physical performance and muscle quality of active elderly women. Arch Gerontol Geriatr. 2014;59(1):44-8.1212 Sotunde OF, Kruger HS, Wright HH, Havemann-Nel L, Kruger IM, Wentzel-Viljoen E, et al. Lean Mass Appears to Be More Strongly Associated with Bone Health than Fat Mass in Urban Black South African Women. J Nutr Health Aging. 2015;19(6):628-36.). From a metabolic point of view, it is well known that adipose tissue can exert either a beneficial effect by favoring insulin sensitivity or a detrimental effect by producing an environment that generates insulin resistance (1313 Moreno-Mendez E, Quintero-Fabian S, Fernandez-Mejia C, Lazo-de-la-Vega-Monroy ML. Early-life programming of adipose tissue. Nutr Res Rev. 2020;33(2):244-59.). Likewise, it is quite possible that, in some circumstances, adipose tissue can have a negative or positive effect on the skeleton. In the last two decades, several groups have been dedicated to deciphering the mechanisms of influence of adipose tissue on bone (1414 Karsenty G, Khosla S. The crosstalk between bone remodeling and energy metabolism: A translational perspective. Cell Metab. 2022;34(6):805-17.,1515 de Araujo IM, Parreiras ESLT, Carvalho AL, Elias J Jr, Salmon CEG, de Paula FJA. Insulin resistance negatively affects bone quality not quantity: the relationship between bone and adipose tissue. Osteoporos Int. 2020;31(6):1125-33.).

In recent years, there has been a marked advance in knowledge about possible mechanisms involved in the intricate interaction between bone and adipose tissue (1414 Karsenty G, Khosla S. The crosstalk between bone remodeling and energy metabolism: A translational perspective. Cell Metab. 2022;34(6):805-17.). The current scenario shows that this process comprehends a complex communication network involving the participation of endocrine factors originating in adipocytes that act directly on their receptors expressed in bone cells (e.g., leptin and adiponectin in osteoblasts and chemerin in osteoclasts) (Table 1), but also indirectly through hypothalamic activation of the sympathetic system. However, these hormones’ central and peripheral actions do not always converge. For example, pioneering studies by Karsenty and Khosla have shown by various means that leptin has a central action, stimulating the sympathetic nervous system (1414 Karsenty G, Khosla S. The crosstalk between bone remodeling and energy metabolism: A translational perspective. Cell Metab. 2022;34(6):805-17.,1616 Ducy P, Amling M, Takeda S, Priemel M, Schilling AF, Beil FT, et al. Leptin inhibits bone formation through a hypothalamic relay: a central control of bone mass. Cell. 2000;100(2):197-207.). Consequently, there is inhibition of osteoblastic activity and increase in osteoclastic action, resulting in a negative balance in bone remodeling, which – ideally – should be for renewal and maintenance of bone mass in adults. On the other hand, studies indicate that leptin acts directly on osteoblasts, increasing their activity (1717 Philbrick KA, Wong CP, Branscum AJ, Turner RT, Iwaniec UT. Leptin stimulates bone formation in ob/ob mice at doses having minimal impact on energy metabolism. J Endocrinol. 2017;232(3):461-74.). An additional aspect that complicates the understanding of the final effect of leptin is the occurrence of resistance to its action. However, there is no evidence of leptin resistance in terms of its effects on bone.

Table 1
Endocrine interactions between adipose and bone tissues

Decreased circulatory levels of leptin are a common finding in young women with hypothalamic amenorrhea. A previous study evaluated metreleptin therapy in women with low body weight and documented hypoleptinemia. Those who completed 2 years of therapy exhibited significant increases in lumbar spine bone mineral density (BMD) (4%), as well as in lumbar spine bone mineral content (6%). Additionally, a reduction was observed in serum levels of carboxy-terminal cross-linked telopeptide of type I collagen (CTX) and N-terminal procollagen type I propeptide (P1NP) during the second year of treatment (1818 Sienkiewicz E, Magkos F, Aronis KN. Long-term metreleptin treatment increases bone mineral density and content at the lumbar spine of lean hypoleptinemic women. Metabolism. 2011;60(9):1211-21.).

The ability to withstand mechanical impact, one of the main properties of bone, depends on a set of factors. One of these factors is the amount of bone mass, which can be easily estimated using bone densitometry. However, other attributes (grouped as bone quality parameters) conferring strength to bone tissue still lack standardized estimation. Predisposition to fracture also occurs in some situations where bone mass is not particularly affected (1919 Seeman E, Delmas PD. Bone quality--the material and structural basis of bone strength and fragility. N Engl J Med. 2006;354(21):2250-61.). Curiously, the most frequent situations in which this occurs are associated with significant changes in adipose tissue, including obesity, type 2 DM, and hypercortisolism (2020 Schwartz AV. Epidemiology of fractures in type 2 diabetes. Bone. 2016;82:2-8.2222 Compston J. Glucocorticoid-induced osteoporosis: an update. Endocrine. 2018;61(1):7-16.). These situations are addressed below.

OBESITY AND INSULIN RESISTANCE

Obesity is the most frequent metabolic disorder affecting humans. The World Health Organization (WHO) defines obesity as a state of fat accumulation that poses a risk to health in individuals with a body mass index (BMI) ≥ 30 kg/m2. The prevalence of obesity varies among countries and is reported at 3.7% in Japan (2323 Bluher M. Obesity: global epidemiology and pathogenesis. Nat Rev Endocrinol. 2019;15(5):288-98.) and 25.9% in Brazil (2424 Ferreira APS, Szwarcwald CL, Damacena GN, Souza Junior PRB. Increasing trends in obesity prevalence from 2013 to 2019 and associated factors in Brazil. Rev Bras Epidemiol. 2021;24(Suppl 2):e210009.). Obesity reflects the body’s remarkable ability to store excess energy, with the subcutaneous adipose tissue being the ideal storage site. Under conditions of continuing positive balance between energy consumption and expenditure, lipids are deposited at visceral sites, and when the limit in storage capacity is reached, they are exported to other tissues (2525 Pellegrinelli V, Carobbio S, Vidal-Puig A. Adipose tissue plasticity: how fat depots respond differently to pathophysiological cues. Diabetologia. 2016;59(6):1075-88.). Adipocytes fully engorged with lipids and assimilation of lipids by parts of other tissues lead to structural and functional alteration and inflammation (2626 Kahn CR, Wang G, Lee KY. Altered adipose tissue and adipocyte function in the pathogenesis of metabolic syndrome. J Clin Invest. 2019;129(10):3990-4000.). The secretory profile of the adipose tissue changes from anti-inflammatory to proinflammatory, with the production of TNF-α, IL6, and PAI1 prevailing over, for example, that of adiponectin (2626 Kahn CR, Wang G, Lee KY. Altered adipose tissue and adipocyte function in the pathogenesis of metabolic syndrome. J Clin Invest. 2019;129(10):3990-4000.,2727 Goossens GH. The role of adipose tissue dysfunction in the pathogenesis of obesity-related insulin resistance. Physiol Behav. 2008;94(2):206-18.). Metabolic abnormalities, oxidative stress, and insulin resistance add to this chronic inflammatory condition leading to type 2 DM, steatohepatitis, and cardiovascular diseases (2828 Dietrich P, Hellerbrand C. Non-alcoholic fatty liver disease, obesity and the metabolic syndrome. Best Pract Res Clin Gastroenterol. 2014;28(4):637-53.,2929 Singh C, Jain S, Dhawan V, Kalra N, Kumari S. Uric acid as a predictor of endothelial dysfunction in patients with metabolic syndrome. Arch Endocrinol Metab. 2021;64(6):810-5.).

The impact of obesity on the skeleton may not be positive, but it is at least different from that occurring in other organs and tissues. First, bone harbors its own adipose tissue, and bone marrow adipose tissue (MAT) in humans curiously does not appear to be a site primarily geared toward fat storage under conditions of excessive energy supply (1515 de Araujo IM, Parreiras ESLT, Carvalho AL, Elias J Jr, Salmon CEG, de Paula FJA. Insulin resistance negatively affects bone quality not quantity: the relationship between bone and adipose tissue. Osteoporos Int. 2020;31(6):1125-33.,3030 de Paula FJA, Rosen CJ. Marrow Adipocytes: Origin, Structure, and Function. Annu Rev Physiol. 2020;82:461-84.). Usually, increased bone marrow adipogenesis is associated with impaired maintenance of bone mass, a situation that occurs with aging, hypercortisolism, and, paradoxically, anorexia nervosa (22 de Paula FJA, Rosen CJ. Structure and Function of Bone Marrow Adipocytes. Compr Physiol. 2017;8(1):315-49.,1515 de Araujo IM, Parreiras ESLT, Carvalho AL, Elias J Jr, Salmon CEG, de Paula FJA. Insulin resistance negatively affects bone quality not quantity: the relationship between bone and adipose tissue. Osteoporos Int. 2020;31(6):1125-33.,3131 Batista SL, de Araujo IM, Carvalho AL, Alencar M, Nahas AK, Elias J Jr, et al. Beyond the metabolic syndrome: Visceral and marrow adipose tissues impair bone quantity and quality in Cushing’s disease. PLoS One. 2019;14(10):e0223432.,3232 Devlin MJ, Rosen CJ. The bone-fat interface: basic and clinical implications of marrow adiposity. Lancet Diabetes Endocrinol. 2015;3(2):141-7.). However, several studies have shown that insulin resistance does not correlate negatively with bone mass. Likewise, parameters strongly associated with insulin resistance, such as visceral adipose tissue (VAT) and the amount of intrahepatic lipids (IHL), are not negatively associated with bone mass (3333 de Araujo IM, Salmon CE, Nahas AK, Nogueira-Barbosa MH, Elias J Jr, de Paula FJ. Marrow adipose tissue spectrum in obesity and type 2 diabetes mellitus. Eur J Endocrinol. 2017;176(1):21-30.3535 Bland VL, Klimentidis YC, Bea JW, Roe DJ, Funk JL, Going SB. Cross-sectional associations between adipose tissue depots and areal bone mineral density in the UK Biobank imaging study. Osteoporos Int. 2022;33(2):391-402.). Despite a lack of consensus regarding these results, they are perfectly aligned with data established in the literature showing normal or increased bone mass in situations of insulin resistance, such as those occurring in obesity and type 2 DM. Notably, in generalized congenital lipodystrophy, a situation of severe insulin resistance, BMD is increased (3636 Lima JG, Nobrega LHC, Lima NN, Dos Santos MCF, Baracho MFP, Winzenrieth R, et al. Normal bone density and trabecular bone score, but high serum sclerostin in congenital generalized lipodystrophy. Bone. 2017;101:21-5.).

The initial perception that obesity was protective against fractures has changed after prospective studies have shown that individuals with obesity present fractures as frequently as those with normal weight (3737 Premaor MO, Comim FV, Compston JE. Obesity and fractures. Arq Bras Endocrinol Metabol. 2014;58(5):470-7.). However, this finding was based on variable data that depended on the characteristics of the population, sex, age, and, in particular, fracture site. Some meta-analyses have shown a decreased risk of femoral neck fractures in obese individuals (3838 Tang X, Liu G, Kang J, Hou Y, Jiang F, Yuan W, et al. Obesity and risk of hip fracture in adults: a meta-analysis of prospective cohort studies. PLoS One. 2013;8(4):e55077.). A meta-analysis including cohorts from several countries has reported that obese compared with non-obese women had a lower risk of hip and distal forearm fractures but a higher risk of humeral/elbow fractures. In contrast, some studies have shown an increased risk of hip fractures in obese individuals. For example, a prospective Korean study has found a positive association between BMI and risk of hip fracture only in women with BMI above 25 kg/m2 (3939 Kim SH, Yi SW, Yi JJ, Kim YM, Won YJ. Association Between Body Mass Index and the Risk of Hip Fracture by Sex and Age: A Prospective Cohort Study. J Bone Miner Res. 2018;33(9):1603-11.). This finding was supported by the results of a meta-analysis indicating a positive relationship between waist circumference and hip fracture (4040 Li X, Gong X, Jiang W. Abdominal obesity and risk of hip fracture: a meta-analysis of prospective studies. Osteoporos Int. 2017;28(10):2747-57.). A lack of consensus is also observed regarding a higher risk of vertebral fractures in obese individuals. Despite this lack of consensus, most results suggest that obesity is associated with a higher risk of peripheral fractures affecting the humerus, tibia, and ankle (3737 Premaor MO, Comim FV, Compston JE. Obesity and fractures. Arq Bras Endocrinol Metabol. 2014;58(5):470-7.).

Several factors can be listed as responsible for the loss of bone strength in obesity and insulin resistance, such as vitamin (D) deficiency, hypothalamic hypogonadism, sarcopenia, changes in microbiota, chronic inflammation, and sedentary lifestyle (1010 Vilaca KH, Carneiro JA, Ferriolli E, Lima NK, de Paula FJ, Moriguti JC. Body composition, physical performance and muscle quality of active elderly women. Arch Gerontol Geriatr. 2014;59(1):44-8.,4141 de Paula FJ, Rosen CJ. Vitamin D safety and requirements. Arch Biochem Biophys. 2012;523(1):64-72.4343 Ozaki D, Kubota R, Maeno T, Abdelhakim M, Hitosugi N. Association between gut microbiota, bone metabolism, and fracture risk in postmenopausal Japanese women. Osteoporos Int. 2021;32(1):145-56.). From a clinical perspective, it is evident that bone densitometry underestimates the risk of fracture in obesity (4444 Evans AL, Paggiosi MA, Eastell R, Walsh JS. Bone density, microstructure and strength in obese and normal weight men and women in younger and older adulthood. J Bone Miner Res. 2015;30(5):920-8.). Therefore, there is need for improvement in the clinical assessment of the susceptibility to fracture in this situation. Even a more sophisticated imaging method like high-resolution peripheral quantitative computed tomography (HRpQCT) may not capture bone changes in obesity, as observed in a 2015 study by Evans and cols. (4444 Evans AL, Paggiosi MA, Eastell R, Walsh JS. Bone density, microstructure and strength in obese and normal weight men and women in younger and older adulthood. J Bone Miner Res. 2015;30(5):920-8.). Trabecular bone score (TBS) is a bone texture index obtained from lumbar spine densitometry images and a predictor of fracture risk independent of BMD or bone size (4545 Silva BC, Leslie WD, Resch H, Lamy O, Lesnyak O, Binkley N, et al. Trabecular bone score: a noninvasive analytical method based upon the DXA image. J Bone Miner Res. 2014;29(3):518-30.4747 Guagnelli MA, Winzenrieth R, Deleze M, Cons-Molina F, Clark P. Description of Normative Spine TBS Data for Men and Women in Mexican Population. J Clin Densitom. 2021;24(1):129-34.). This index can capture bone changes in obese individuals and may help determine risk factors for assessing fracture susceptibility in this condition. Despite significant advances in knowledge regarding the mechanisms and assessment of bone fragility in obesity, many aspects must still be unraveled. Hopefully, more options for recognizing and preventing bone fragility in obesity should emerge soon.

DIABETES MELLITUS

According to the International Diabetes Federation (IDF), DM affected 463 million people in 2019, corresponding to a worldwide prevalence of 9% (4848 Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, et al. IDF Diabetes Atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract. 2022;183:109119.). The classic macroangiopathic and microangiopathic complications associated with this metabolic disease overload the countries’ health systems and are among the leading causes of chronic kidney disease, blindness, amputations, and cardiovascular diseases. Until recently, only type 1 DM was considered a secondary cause of osteoporosis. It is now recognized that type 2 DM, responsible for about 90% of DM cases, is also associated with fractures, significantly increasing the burden for health providers in terms of caring for patients with osteoporosis and DM due to the coexistence of both diseases (4949 Lin YC, Wu J, Kuo SF, Cheung YC, Sung CM, Fan CM, et al. Vertebral Fractures in Type 2 Diabetes Patients: Utility of Trabecular Bone Score and Relationship With Serum Bone Turnover Biomarkers. J Clin Densitom. 2020;23(1):37-43.,5050 Paschou SA, Dede AD, Anagnostis PG, Vryonidou A, Morganstein D, Goulis DG. Type 2 Diabetes and Osteoporosis: A Guide to Optimal Management. J Clin Endocrinol Metab. 2017;102(10):3621-34.).

As in obesity, bone mass is not negatively affected in patients with type 2 DM, with studies showing that bone mass in these patients is normal or increased (5151 Cutrim DM, Pereira FA, de Paula FJ, Foss MC. Lack of relationship between glycemic control and bone mineral density in type 2 diabetes mellitus. Braz J Med Biol Res. 2007;40(2):221-7.,5252 Walsh JS, Vilaca T. Obesity, Type 2 Diabetes and Bone in Adults. Calcif Tissue Int. 2017;100(5):528-35.). In type 1 DM, in turn, bone mass is lost in the initial phase of the disease and does not seem to be recovered later (5353 Kayath MJ, Dib SA, Vieira JG. Prevalence and magnitude of osteopenia associated with insulin-dependent diabetes mellitus. J Diabetes Complications. 1994;8(2):97-104.,5454 Kayath MJ, Tavares EF, Dib SA, Vieira JG. Prospective bone mineral density evaluation in patients with insulin-dependent diabetes mellitus. J Diabetes Complications. 1998;12(3):133-9.). This fact must contribute to the higher susceptibility to fractures observed in type 1 DM compared with type 2 DM. However, several other hormonal and metabolic factors (e.g., IGF1 deficiency, hypoinsulinemia, and low body weight) must also play a role, considering that recent studies have shown that bone mass loss in type 1 DM is not prominent (5555 Carvalho AL, Massaro B, Silva L, Salmon CEG, Fukada SY, Nogueira-Barbosa MH, et al. Emerging Aspects of the Body Composition, Bone Marrow Adipose Tissue and Skeletal Phenotypes in Type 1 Diabetes Mellitus. J Clin Densitom. 2019;22(3):420-8.). Therefore, bone fragility is an additional mark of the heterogeneity among different forms of DM that are united by the occurrence of hyperglycemia.

The image linked to type 1 DM of emaciation, weight loss, and low body fat is part of the past for those who have access to new technologies and receive insulin in multiple doses or through infusion pump (5656 Walsh J, Roberts R, Bailey TS, Heinemann L. Insulin Titration Guidelines for Patients With Type 1 Diabetes: It Is About Time! J Diabetes Sci Technol. 2022:19322968221087261.). Several studies have shown a high rate of overweight/obesity in type 1 DM (5757 Gomes MB, Conte D, Drummond KRG, Mallmann F, Pinheiro AA, Leal FSL, et al. Overweight/obesity in adolescents with type 1 diabetes belonging to an admixed population. A Brazilian multicenter study. Diabetol Metab Syndr. 2022;14(1):1.). This new body profile of patients with type 1 DM may impose both positive and negative impacts on the skeleton, as discussed in the obesity section of this article. Recent studies have shown that bone loss in type 1 DM is not very significant (5454 Kayath MJ, Tavares EF, Dib SA, Vieira JG. Prospective bone mineral density evaluation in patients with insulin-dependent diabetes mellitus. J Diabetes Complications. 1998;12(3):133-9.). Therefore, as in type 2 DM, changes in bone quality must also play an important role in bone fragility in type 1 DM. Body fat distribution has been less studied in type 1 DM. Using magnetic resonance imaging, Carvalho and cols. have shown that, compared with a control group, individuals with type 1 DM with good or poor metabolic control have quantitatively similar subcutaneous adipose tissue (SAT), VAT, and IHL (5555 Carvalho AL, Massaro B, Silva L, Salmon CEG, Fukada SY, Nogueira-Barbosa MH, et al. Emerging Aspects of the Body Composition, Bone Marrow Adipose Tissue and Skeletal Phenotypes in Type 1 Diabetes Mellitus. J Clin Densitom. 2019;22(3):420-8.). The study also showed no differences in the amount of MAT between patients with type 1 DM and controls (5454 Kayath MJ, Tavares EF, Dib SA, Vieira JG. Prospective bone mineral density evaluation in patients with insulin-dependent diabetes mellitus. J Diabetes Complications. 1998;12(3):133-9.).

ANOREXIA NERVOSA

Weight loss by self-imposition of fasting is the hallmark of anorexia nervosa, a psychiatric disease that affects mainly adolescent and young adult women. Among the psychiatric diseases, anorexia nervosa is distinguished by a high rate of death by suicide (5858 Arcelus J, Mitchell AJ, Wales J, Nielsen S. Mortality rates in patients with anorexia nervosa and other eating disorders. A meta-analysis of 36 studies. Arch Gen Psychiatry. 2011;68(7):724-31.) and is associated with several morbidities, including osteoporosis. The fracture risk is about three times greater in patients with this condition, and low bone mass is likely an important predisposing factor (5959 Lucas AR, Melton LJ 3rd, Crowson CS, O’Fallon WM. Long-term fracture risk among women with anorexia nervosa: a population-based cohort study. Mayo Clin Proc. 1999;74(10):972-7.). Therefore, the skeleton follows the wasting process that affects muscle and adipose tissues in a state of food restriction. Part of the function of muscle and adipose tissues, as storage sites for energy substrates, is to provide, respectively, amino acids and fatty acids to meet energy needs, orchestrated by the well-recognized adaptation of hypoinsulinemia and elevation of insulin counterregulatory hormones (6060 Santos Ferreira DL, Hübel C, Herle M, Abdulkadir M, Loos RJF, Bryant-Waugh R, et al. Associations between Blood Metabolic Profile at 7 Years Old and Eating Disorders in Adolescence: Findings from the Avon Longitudinal Study of Parents and Children. Metabolites. 2019;9(9).,6161 Estour B, Germain N, Diconne E, Frere D, Cottet-Emard JM, Carrot G, et al. Hormonal profile heterogeneity and short-term physical risk in restrictive anorexia nervosa. J Clin Endocrinol Metab. 2010;95(5):2203-10.). The bone repercussions during adaptations caused by malnutrition in anorexia nervosa are well recognized, i.e., growth arrest and loss of bone mass gain in the development phase and bone loss in adulthood. Hormonal abnormalities in patients with anorexia nervosa include hypothalamic hypogonadism, increased cortisol, and low levels of IGF1, insulin, and leptin (6161 Estour B, Germain N, Diconne E, Frere D, Cottet-Emard JM, Carrot G, et al. Hormonal profile heterogeneity and short-term physical risk in restrictive anorexia nervosa. J Clin Endocrinol Metab. 2010;95(5):2203-10.,6262 Fazeli PK, Faje AT, Meenaghan E, Russell ST, Resulaj M, Lee H, et al. IGF-1 is associated with estimated bone strength in anorexia nervosa. Osteoporos Int. 2020;31(2):259-65.).

A peculiar aspect of the wide heterogeneity of adipose tissue is the expansion of MAT under conditions of food restriction, as shown experimentally in mice fed with 30% fewer calories than controls (6363 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(9):2078-88.). Studies in women with anorexia nervosa have shown the same pattern and demonstrated that the increase in MAT correlates negatively with bone mass (6464 Fazeli PK, Faje AT, Bredella MA, Polineni S, Russell S, Resulaj M, et al. Changes in marrow adipose tissue with short-term changes in weight in premenopausal women with anorexia nervosa. Eur J Endocrinol. 2019;180(3):189-99.). Thus, MAT expansion in anorexia nervosa is aligned with observations from common conditions in which osteoporosis occurs, such as aging and glucocorticoid-induced osteoporosis (6565 Maciel JG, de Araujo IM, Carvalho AL, Simao MN, Bastos CM, Troncon LE, et al. Marrow Fat Quality Differences by Sex in Healthy Adults. J Clin Densitom. 2017;20(1):106-13.,6666 Rosen CJ, Bouxsein ML. Mechanisms of disease: is osteoporosis the obesity of bone? Nat Clin Pract Rheumatol. 2006;2(1):35-43.). Bone remodeling markers suggest a disconnect between bone formation and resorption, with studies showing a reduction in bone formation markers and an increase in markers that reflect resorption (6767 Maimoun L, Garnero P, Mura T, Nocca D, Lefebvre P, Philibert P, et al. Specific Effects of Anorexia Nervosa and Obesity on Bone Mineral Density and Bone Turnover in Young Women. J Clin Endocrinol Metab. 2020;105(4).). Since osteoblasts and adipocytes share a common origin from the same mesenchymal stem cell, it is reasonable to consider that the direction of the differentiation occurs toward one cell lineage at the expense of the other, with adipocytes prevailing in this case (6868 Horowitz MC, Berry R, Holtrup B, Sebo Z, Nelson T, Fretz JA, et al. Bone marrow adipocytes. Adipocyte. 2017;6(3):193-204.). The role of the paracrine activity of adipocytes in determining the fate of mesenchymal stem cells under these circumstances remains to be determined.

WEIGHT LOSS AFTER SURGICAL INTERVENTIONS IN THE GASTROINTESTINAL TRACT (BARIATRIC SURGERY AND SHORT BOWEL SYNDROME)

Given the current obesity pandemic and the frustrating limitation of clinical alternatives for obesity treatment (6969 Rolls BJ. What is the role of portion control in weight management? Int J Obes (Lond). 2014;38 Suppl 1:S1-8.,7070 Jaacks LM, Vandevijvere S, Pan A, McGowan CJ, Wallace C, Imamura F, et al. The obesity transition: stages of the global epidemic. Lancet Diabetes Endocrinol. 2019;7(3):231-40.), aggressive management through restrictive and malabsorptive surgery has emerged in recent decades as the most efficient option to mitigate morbidity and mortality of severe obesity (7171 Carlsson LMS, Sjoholm K, Jacobson P, Andersson-Assarsson JC, Svensson PA, Taube M, et al. Life Expectancy after Bariatric Surgery in the Swedish Obese Subjects Study. N Engl J Med. 2020;383(16):1535-43.). This approach has drawbacks; the loss of bone mass and increased susceptibility to fracture are among the most important.

There is a paucity of prospective data evaluating both the bone loss and metabolic benefits of bariatric surgery in severely obese individuals (7272 Alencar M, Araujo IM, Parreiras ESLT, Nogueira-Barbosa MH, Salgado W Jr, Elias J Jr, et al. Hashtag bone: detrimental effects on bone contrast with metabolic benefits one and five years after Roux-en-Y gastric bypass. Braz J Med Biol Res. 2021;54(12):e11499.). However, a recent cross-sectional study has shown metabolic benefits obtained with bariatric surgery, which persist for 5 years after the procedure, even in patients whose BMI remains in the range of obesity. After bariatric surgery, the VAT/SAT distribution ratio and the amount of IHL were similar in individuals with obesity or normal weight. In contrast, individuals submitted to bariatric surgery present early elevation of remodeling marker of bone resorption, a pattern that is maintained for at least 5 years after surgery (7272 Alencar M, Araujo IM, Parreiras ESLT, Nogueira-Barbosa MH, Salgado W Jr, Elias J Jr, et al. Hashtag bone: detrimental effects on bone contrast with metabolic benefits one and five years after Roux-en-Y gastric bypass. Braz J Med Biol Res. 2021;54(12):e11499.). Two previous studies, one cross-sectional (7272 Alencar M, Araujo IM, Parreiras ESLT, Nogueira-Barbosa MH, Salgado W Jr, Elias J Jr, et al. Hashtag bone: detrimental effects on bone contrast with metabolic benefits one and five years after Roux-en-Y gastric bypass. Braz J Med Biol Res. 2021;54(12):e11499.) and the other prospective (7373 Lindeman KG, Greenblatt LB, Rourke C, Bouxsein ML, Finkelstein FL, Yu WE. Longitudinal 5-year evaluation of bone density and microarchitecture after Roux-en-Y gastric bypass surgery. J Clin Endocrinol Metab 2018;103(11):4104-12.), have suggested precocious and sustained increased levels of the biochemical marker of bone resorption CTX. In the former, Alencar and cols. described that the groups evaluated 1 and 5 years after Roux-en-Y bypass (RYGB) showed levels of serum CTX that were, respectively, 97% and 51% higher than those in the control group (7272 Alencar M, Araujo IM, Parreiras ESLT, Nogueira-Barbosa MH, Salgado W Jr, Elias J Jr, et al. Hashtag bone: detrimental effects on bone contrast with metabolic benefits one and five years after Roux-en-Y gastric bypass. Braz J Med Biol Res. 2021;54(12):e11499.). In a prospective study, serum CTX levels have been found to be 150% above the basal levels 5 years after RYGB (7373 Lindeman KG, Greenblatt LB, Rourke C, Bouxsein ML, Finkelstein FL, Yu WE. Longitudinal 5-year evaluation of bone density and microarchitecture after Roux-en-Y gastric bypass surgery. J Clin Endocrinol Metab 2018;103(11):4104-12.). On the other hand, the literature shows more modest or no difference in the biochemical markers of bone formation after bariatric surgery (7272 Alencar M, Araujo IM, Parreiras ESLT, Nogueira-Barbosa MH, Salgado W Jr, Elias J Jr, et al. Hashtag bone: detrimental effects on bone contrast with metabolic benefits one and five years after Roux-en-Y gastric bypass. Braz J Med Biol Res. 2021;54(12):e11499.,7373 Lindeman KG, Greenblatt LB, Rourke C, Bouxsein ML, Finkelstein FL, Yu WE. Longitudinal 5-year evaluation of bone density and microarchitecture after Roux-en-Y gastric bypass surgery. J Clin Endocrinol Metab 2018;103(11):4104-12.). For instance, Alencar and cols. have also observed that, compared with a control group, individuals submitted to RYGB show similar serum osteocalcin levels both 1 and 5 years after the procedure. However, circulatory levels of P1NP were 34% higher than baseline 5 years after RYGB. These results are in line with previous data showing that RYGB has detrimental effects on bone microstructure as well as bone strength. The impact on bone after RYGB appears early in the first year and persists for at least a few years after weight loss plateaus.

Notably, bone mass measured by bone densitometry may not be an efficient way to detect bone catabolic states in individuals with severe obesity, as these individuals generally start from a high bone mass value (7272 Alencar M, Araujo IM, Parreiras ESLT, Nogueira-Barbosa MH, Salgado W Jr, Elias J Jr, et al. Hashtag bone: detrimental effects on bone contrast with metabolic benefits one and five years after Roux-en-Y gastric bypass. Braz J Med Biol Res. 2021;54(12):e11499.). However, after bariatric surgery, the BMD/body weight ratio has been shown to be significantly lower at 5 years compared with 1 year (7272 Alencar M, Araujo IM, Parreiras ESLT, Nogueira-Barbosa MH, Salgado W Jr, Elias J Jr, et al. Hashtag bone: detrimental effects on bone contrast with metabolic benefits one and five years after Roux-en-Y gastric bypass. Braz J Med Biol Res. 2021;54(12):e11499.). Another study has shown that MAT in adolescents undergoing bariatric surgery is increased in the lumbar spine and reduced in long bones (7474 Misra M, Bredella MA. Bone Metabolism in Adolescents Undergoing Bariatric Surgery. J Clin Endocrinol Metab. 2021;106(2):326-36.). Thus, it is possible that, in addition to decreased food intake and absorption of nutrients (e.g., calcium and vitamin D) and reduced serum levels of IGF1 and insulin, the increased MAT may contribute to a negative balance in the process of bone remodeling (7575 Liu C, Wu D, Zhang JF, Xu D, Xu WF, Chen Y, et al. Changes in Bone Metabolism in Morbidly Obese Patients After Bariatric Surgery: A Meta-Analysis. Obes Surg. 2016;26(1):91-7.).

SHORT BOWEL SYNDROME

Individuals with anorexia nervosa and those submitted to bariatric surgery experience caloric restriction through different mechanisms. In contrast, patients with short bowel syndrome (SBS) have an alternative supply of nutrients through parenteral means, allowing for part of these patients to regain their body weight. This aspect may determine significant differences between SBS and the two other conditions (anorexia nervosa and bariatric surgery) from a metabolic point of view, but from the perspective of bone, the three conditions share negative consequences on the skeleton (7676 Braga CB, Bizari L, Suen VM, Marchini JS, Paula FJ, Cunha SF. Bone mineral density in short bowel syndrome: correlation with BMI and serum vitamins C, E and K. Arch Endocrinol Metab. 2015;59(3):252-8.). Indeed, individuals with SBS have a high prevalence of osteoporosis and hepatic steatosis (7777 Parreiras ESLT, de Araujo IM, Elias J Jr, Nogueira-Barbosa MH, Suen VMM, Marchini JS, et al. Short bowel syndrome: influence of nutritional therapy and incretin GLP1 on bone marrow adipose tissue. Ann N Y Acad Sci. 2018;1415(1):47-56.,7878 Parreiras ESLT, de Araujo IM, Elias J Jr, Nogueira-Barbosa MH, Suen VMM, Marchini JS, et al. Osteoporosis and Hepatic Steatosis: 2 Closely Related Complications in Short-Bowel Syndrome. JPEN J Parenter Enteral Nutr. 2020;44(7):1271-9.). Another aspect is that the parenteral supply of nutrients in SBS not only seems to limit MAT expansion but also changes the pattern of correlation between MAT and bone mass, which in this case is positive, suggesting that – depending on the circumstances – MAT may have a positive or negative effect on the maintenance of bone mass. Finally, it is worth noting that other variables such as incretins (GLP1 and GIP) and the microbiota are potential protagonists in bone changes in SBS, although their roles have yet to be better determined (7777 Parreiras ESLT, de Araujo IM, Elias J Jr, Nogueira-Barbosa MH, Suen VMM, Marchini JS, et al. Short bowel syndrome: influence of nutritional therapy and incretin GLP1 on bone marrow adipose tissue. Ann N Y Acad Sci. 2018;1415(1):47-56.).

MUSCULOSKELETAL INTERACTION

Muscle and skeleton are interconnected, forming an interdependent unit in a lever system that allows movement. The mechanical interaction between both serves as a positive mutual stimulus, depending on the appropriate nutritional environment (7979 Li G, Zhang L, Wang D, AIQudsy L, Jiang JX, Xu H, et al. Muscle-bone crosstalk and potential therapies for sarco-osteoporosis. J Cell Biochem. 2019;120(9):14262-73.,8080 Bonewald L. Use it or lose it to age: A review of bone and muscle communication. Bone. 2019;120:212-8.). The mechanical interaction is contemplated in the mechanostat model proposed by Frost (8181 Frost HM. Bone’s mechanostat: a 2003 update. Anat Rec A Discov Mol Cell Evol Biol. 2003;275(2):1081-101.). The musculoskeletal mechanical coupling is the most visible portion of the model, and its importance can be highlighted in several studies in models ranging from neural injury (8282 Jiang SD, Dai LY, Jiang LS. Osteoporosis after spinal cord injury. Osteoporos Int. 2006;17(2):180-92.,8383 Jiang SD, Jiang LS, Dai LY. Mechanisms of osteoporosis in spinal cord injury. Clin Endocrinol (Oxf). 2006;65(5):555-65.), simulation of weight subtraction using tail suspension (8484 Falcai MJ, Louzada MJ, de Paula FJ, Okubo R, Volpon JB. A modified technique of rat tail suspension for longer periods of observation. Aviat Space Environ Med. 2012;83(12):1176-80.,8585 Komori T. Animal models for osteoporosis. Eur J Pharmacol. 2015;759:287-94.), and microgravity experiments in space (8686 Vico L, van Rietbergen B, Vilayphiou N, Linossier MT, Locrelle H, Normand M, et al. Cortical and Trabecular Bone Microstructure Did Not Recover at Weight-Bearing Skeletal Sites and Progressively Deteriorated at Non-Weight-Bearing Sites During the Year Following International Space Station Missions. J Bone Miner Res. 2017;32(10):2010-21.,8787 Dadwal UC, Maupin KA, Zamarioli A, Tucker A, Harris JS, Fischer JP, et al. The effects of spaceflight and fracture healing on distant skeletal sites. Sci Rep. 2019;9(1):11419.). In fact, osteoporosis and sarcopenia go hand in hand in different physiological situations and are particularly present in the context of aging and in various diseases (8787 Dadwal UC, Maupin KA, Zamarioli A, Tucker A, Harris JS, Fischer JP, et al. The effects of spaceflight and fracture healing on distant skeletal sites. Sci Rep. 2019;9(1):11419.). New evidence shows that this interaction also occurs at molecular and biochemical levels (Table 2) and opens up the possibility of therapeutic interventions that may preserve the quality of life in older age and in various diseases (8888 Brotto M, Bonewald L. Bone and muscle: Interactions beyond mechanical. Bone. 2015;80:109-14.).

Table 2
Endocrine interactions between muscle and bone tissue

Myokine is a term used generically to refer to factors secreted by muscle tissue. The first myokine to be identified was myostatin (a potent inhibitor of myocyte proliferation), with others being later identified, including irisin (which induces browning of white adipose tissue), IL8 (which induces angiogenesis), and IL15 (which reduces adipose tissue) (8989 Seale P, Bjork B, Yang W, Kajimura S, Chin S, Kuang S, et al. PRDM16 controls a brown fat/skeletal muscle switch. Nature. 2008;454(7207):961-7.9191 Quinn LS, Anderson BG, Strait-Bodey L, Stroud AM, Argiles JM. Oversecretion of interleukin-15 from skeletal muscle reduces adiposity. Am J Physiol Endocrinol Metab. 2009;296(1):E191-202.). The musculoskeletal interaction is definitely not a one-way path, as factors derived from bone also influence muscle metabolism. Osteocalcin, a pleiotropic peptide, may have a role in muscle trophism (9292 Karsenty G, Mera P. Molecular bases of the crosstalk between bone and muscle. Bone. 2018;115:43-9.). Excess FGF23, as observed in osteomalacia-inducing tumors, is distinguished by the presentation of muscle weakness and pain that is probably not explained by hypophosphatemia alone (9393 Minisola S, Peacock M, Fukumoto S, Cipriani C, Pepe J, Tella SH, et al. Tumour-induced osteomalacia. Nat Rev Dis Primers. 2017;3:17044.). Furthermore, in chronic kidney disease, FGF23 may have a deleterious effect on the myocardium (9494 Prie D. FGF23 and Cardiovascular Risk. Ann Endocrinol (Paris). 2021;82(3-4):141-3.).

SARCOPENIA AND OSTEOPOROSIS

As mentioned above, osteoporosis and sarcopenia often coexist in elderly individuals. At first glance, it is possible to assume that a reduction in muscle activity alone results in a loss of bone maintenance due to overestimating the dependence of bone biomechanics on the muscular system. However, the picture may be more complex, and other factors can contribute to the bone fragility that settles in elderly individuals with sarcopenia. In reality, the aging process is a complex and incompletely understood occurrence. For example, osteocytes comprise 90%-95% of bone tissue cells; these are terminally differentiated cells established to remain encased in the calcified matrix for decades. Osteocytes become senescent and have their functional capacity modified by altered gene expression and change in secretome to a proinflammatory pattern. Currently, there is great interest in the development of agents that can target senescent cells by either eliminating them or reversing the profile of their secretome (9595 Farr JN, Khosla S. Cellular senescence in bone. Bone. 2019;121:121-33.). The relationship that the emergence of these cells has with sarcopenia and osteoporosis has yet to be determined.

IMPACT OF MUSCLE DISEASES ON BONE

Several rare genetic diseases may present with muscle atrophy, either due to a direct muscle defect or indirect atrophy secondary to damage to the innervation system. Spinal muscular atrophy (degeneration of alpha motor neurons of the anterior horn of the spinal cord) is a group of diseases that, in their most severe forms, produce intense muscle weakness before birth and multiple contractures due to defects in joint development secondary to lack of movement, resulting in arthrogryposis multiplex congenita. Also, frequent fractures can occur both before and after birth. Duchenne muscular dystrophy, an X-linked recessive genetic disease caused by dystrophin deficiency, is also associated with bone weakness. Boys affected with this condition show progressive loss of muscle function and become unable to walk after the first decade of life. Long bone fractures are frequent and can occur even before the dystrophy is diagnosed (9696 Perera N, Sampaio H, Woodhead H, Farrar M. Fracture in Duchenne Muscular Dystrophy: Natural History and Vitamin D Deficiency. J Child Neurol. 2016;31(9):1181-7.). The bone condition can be worsened by glucocorticoids, which may have an important role in the emergence of vertebral fractures. Individuals with spina bifida present impaired muscle function in the lower limbs but preserved upper limb muscles (9797 Veilleux LN, Rauch F. Muscle-Bone Interactions in Pediatric Bone Diseases. Curr Osteoporos Rep. 2017;15(5):425-32). A previous study has found that lower limb fractures are 10 times more frequent in children who are unable to walk than in those who have this ability preserved (9797 Veilleux LN, Rauch F. Muscle-Bone Interactions in Pediatric Bone Diseases. Curr Osteoporos Rep. 2017;15(5):425-32).

The bone effects from muscular dystrophies that start early in life usually have a common pattern. Long bones have low bone density in the trabecular-rich regions around metaphyses and thin cortical regions in diaphyses, reflecting, respectively, trabecular catabolism and low periosteal apposition. From a clinical point of view, there is a striking difference in patterns of fractures between children who are normal versus those who have muscular dystrophy, with fractures occurring more commonly in the upper limbs in normal children and in the lower limbs in children with muscular dystrophy.

In summary, mesenchymal tissues share important structural and metabolic functions and are distinguished by broad functional plasticity. Refinements in cellular and molecular investigations have shown that these tissues play a broad role in mutual regulation. Not only their normal functions are interdependent, but diseases that primarily affect one have repercussions on the others. Muscle, adipose, and bone tissues are deeply affected by deteriorations that occur during aging and in common diseases such as DM and obesity. Although a wide window of possibilities for innovative therapies in this area has opened up, the understanding of these interactions between muscle, adipose, and bone tissues serves as a warning that potential risks of mutual effects must be carefully considered.

  • Financial support: FJAP: National Council for Scientific and Technological Development (CNPq) 309316/2021-9; Fapesp: 2018/18071-5 and Fundação de Apoio ao Ensino Pesquisa e Assistência HCFMRP-USP (FAEPA). MMRS Fapesp: 2021/01475-9; IMA Fapesp: 2021/03152-2.

REFERENCES

  • 1
    Paula FJ, Gouveia LM, Paccola GM, Piccinato CE, Moreira AC, Foss MC. Androgen-related effects on peripheral glucose metabolism in women with congenital adrenal hyperplasia. Horm Metab Res. 1994;26(11):552-6.
  • 2
    de Paula FJA, Rosen CJ. Structure and Function of Bone Marrow Adipocytes. Compr Physiol. 2017;8(1):315-49.
  • 3
    Pourquie O. Vertebrate somitogenesis. Annu Rev Cell Dev Biol. 2001;17:311-50.
  • 4
    Paula FJ, Rosen CJ. Obesity, diabetes mellitus and last but not least, osteoporosis. Arq Bras Endocrinol Metabol. 2010;54(2):150-7.
  • 5
    Hofbauer LC, Busse B, Eastell R, Ferrari S, Frost M, Muller R, et al. Bone fragility in diabetes: novel concepts and clinical implications. Lancet Diabetes Endocrinol. 2022;10(3):207-20.
  • 6
    Kim H, Wrann CD, Jedrychowski M, Vidoni S, Kitase Y, Nagano K, et al. Irisin Mediates Effects on Bone and Fat via alpha V Integrin Receptors. Cell. 2018;175(7):1756-68-e17.
  • 7
    Reid IR. Fat and bone. Arch Biochem Biophys. 2010;503(1):20-7.
  • 8
    Reid IR. Relationships between fat and bone. Osteoporos Int. 2008;19(5):595-606.
  • 9
    Alessandri SB, Pereira Fde A, Villela RA, Antonini SR, Elias PC, Martinelli CE Jr., et al. Bone mineral density and body composition in girls with idiopathic central precocious puberty before and after treatment with a gonadotropin-releasing hormone agonist. Clinics (Sao Paulo). 2012;67(6):591-6.
  • 10
    Vilaca KH, Carneiro JA, Ferriolli E, Lima NK, de Paula FJ, Moriguti JC. Body composition, physical performance and muscle quality of active elderly women. Arch Gerontol Geriatr. 2014;59(1):44-8.
  • 11
    Zaidi M, Buettner C, Sun L, Iqbal J. Minireview: The link between fat and bone: does mass beget mass? Endocrinology. 2012;153(5):2070-5.
  • 12
    Sotunde OF, Kruger HS, Wright HH, Havemann-Nel L, Kruger IM, Wentzel-Viljoen E, et al. Lean Mass Appears to Be More Strongly Associated with Bone Health than Fat Mass in Urban Black South African Women. J Nutr Health Aging. 2015;19(6):628-36.
  • 13
    Moreno-Mendez E, Quintero-Fabian S, Fernandez-Mejia C, Lazo-de-la-Vega-Monroy ML. Early-life programming of adipose tissue. Nutr Res Rev. 2020;33(2):244-59.
  • 14
    Karsenty G, Khosla S. The crosstalk between bone remodeling and energy metabolism: A translational perspective. Cell Metab. 2022;34(6):805-17.
  • 15
    de Araujo IM, Parreiras ESLT, Carvalho AL, Elias J Jr, Salmon CEG, de Paula FJA. Insulin resistance negatively affects bone quality not quantity: the relationship between bone and adipose tissue. Osteoporos Int. 2020;31(6):1125-33.
  • 16
    Ducy P, Amling M, Takeda S, Priemel M, Schilling AF, Beil FT, et al. Leptin inhibits bone formation through a hypothalamic relay: a central control of bone mass. Cell. 2000;100(2):197-207.
  • 17
    Philbrick KA, Wong CP, Branscum AJ, Turner RT, Iwaniec UT. Leptin stimulates bone formation in ob/ob mice at doses having minimal impact on energy metabolism. J Endocrinol. 2017;232(3):461-74.
  • 18
    Sienkiewicz E, Magkos F, Aronis KN. Long-term metreleptin treatment increases bone mineral density and content at the lumbar spine of lean hypoleptinemic women. Metabolism. 2011;60(9):1211-21.
  • 19
    Seeman E, Delmas PD. Bone quality--the material and structural basis of bone strength and fragility. N Engl J Med. 2006;354(21):2250-61.
  • 20
    Schwartz AV. Epidemiology of fractures in type 2 diabetes. Bone. 2016;82:2-8.
  • 21
    Nielson CM, Srikanth P, Orwoll ES. Obesity and fracture in men and women: an epidemiologic perspective. J Bone Miner Res. 2012;27(1):1-10.
  • 22
    Compston J. Glucocorticoid-induced osteoporosis: an update. Endocrine. 2018;61(1):7-16.
  • 23
    Bluher M. Obesity: global epidemiology and pathogenesis. Nat Rev Endocrinol. 2019;15(5):288-98.
  • 24
    Ferreira APS, Szwarcwald CL, Damacena GN, Souza Junior PRB. Increasing trends in obesity prevalence from 2013 to 2019 and associated factors in Brazil. Rev Bras Epidemiol. 2021;24(Suppl 2):e210009.
  • 25
    Pellegrinelli V, Carobbio S, Vidal-Puig A. Adipose tissue plasticity: how fat depots respond differently to pathophysiological cues. Diabetologia. 2016;59(6):1075-88.
  • 26
    Kahn CR, Wang G, Lee KY. Altered adipose tissue and adipocyte function in the pathogenesis of metabolic syndrome. J Clin Invest. 2019;129(10):3990-4000.
  • 27
    Goossens GH. The role of adipose tissue dysfunction in the pathogenesis of obesity-related insulin resistance. Physiol Behav. 2008;94(2):206-18.
  • 28
    Dietrich P, Hellerbrand C. Non-alcoholic fatty liver disease, obesity and the metabolic syndrome. Best Pract Res Clin Gastroenterol. 2014;28(4):637-53.
  • 29
    Singh C, Jain S, Dhawan V, Kalra N, Kumari S. Uric acid as a predictor of endothelial dysfunction in patients with metabolic syndrome. Arch Endocrinol Metab. 2021;64(6):810-5.
  • 30
    de Paula FJA, Rosen CJ. Marrow Adipocytes: Origin, Structure, and Function. Annu Rev Physiol. 2020;82:461-84.
  • 31
    Batista SL, de Araujo IM, Carvalho AL, Alencar M, Nahas AK, Elias J Jr, et al. Beyond the metabolic syndrome: Visceral and marrow adipose tissues impair bone quantity and quality in Cushing’s disease. PLoS One. 2019;14(10):e0223432.
  • 32
    Devlin MJ, Rosen CJ. The bone-fat interface: basic and clinical implications of marrow adiposity. Lancet Diabetes Endocrinol. 2015;3(2):141-7.
  • 33
    de Araujo IM, Salmon CE, Nahas AK, Nogueira-Barbosa MH, Elias J Jr, de Paula FJ. Marrow adipose tissue spectrum in obesity and type 2 diabetes mellitus. Eur J Endocrinol. 2017;176(1):21-30.
  • 34
    de Paula FJ, de Araujo IM, Carvalho AL, Elias J Jr, Salmon CE, Nogueira-Barbosa MH. The Relationship of Fat Distribution and Insulin Resistance with Lumbar Spine Bone Mass in Women. PLoS One. 2015;10(6):e0129764.
  • 35
    Bland VL, Klimentidis YC, Bea JW, Roe DJ, Funk JL, Going SB. Cross-sectional associations between adipose tissue depots and areal bone mineral density in the UK Biobank imaging study. Osteoporos Int. 2022;33(2):391-402.
  • 36
    Lima JG, Nobrega LHC, Lima NN, Dos Santos MCF, Baracho MFP, Winzenrieth R, et al. Normal bone density and trabecular bone score, but high serum sclerostin in congenital generalized lipodystrophy. Bone. 2017;101:21-5.
  • 37
    Premaor MO, Comim FV, Compston JE. Obesity and fractures. Arq Bras Endocrinol Metabol. 2014;58(5):470-7.
  • 38
    Tang X, Liu G, Kang J, Hou Y, Jiang F, Yuan W, et al. Obesity and risk of hip fracture in adults: a meta-analysis of prospective cohort studies. PLoS One. 2013;8(4):e55077.
  • 39
    Kim SH, Yi SW, Yi JJ, Kim YM, Won YJ. Association Between Body Mass Index and the Risk of Hip Fracture by Sex and Age: A Prospective Cohort Study. J Bone Miner Res. 2018;33(9):1603-11.
  • 40
    Li X, Gong X, Jiang W. Abdominal obesity and risk of hip fracture: a meta-analysis of prospective studies. Osteoporos Int. 2017;28(10):2747-57.
  • 41
    de Paula FJ, Rosen CJ. Vitamin D safety and requirements. Arch Biochem Biophys. 2012;523(1):64-72.
  • 42
    Corona G, Vignozzi L, Sforza A, Mannucci E, Maggi M. Obesity and late-onset hypogonadism. Mol Cell Endocrinol. 2015;418 Pt 2:120-33.
  • 43
    Ozaki D, Kubota R, Maeno T, Abdelhakim M, Hitosugi N. Association between gut microbiota, bone metabolism, and fracture risk in postmenopausal Japanese women. Osteoporos Int. 2021;32(1):145-56.
  • 44
    Evans AL, Paggiosi MA, Eastell R, Walsh JS. Bone density, microstructure and strength in obese and normal weight men and women in younger and older adulthood. J Bone Miner Res. 2015;30(5):920-8.
  • 45
    Silva BC, Leslie WD, Resch H, Lamy O, Lesnyak O, Binkley N, et al. Trabecular bone score: a noninvasive analytical method based upon the DXA image. J Bone Miner Res. 2014;29(3):518-30.
  • 46
    Bonaccorsi G, Cafarelli FP, Cervellati C, De Guio F, Greco P, Giganti M, et al. A new corrective model to evaluate TBS in obese post-menopausal women: a cross-sectional study. Aging Clin Exp Res. 2020;32(7):1303-8.
  • 47
    Guagnelli MA, Winzenrieth R, Deleze M, Cons-Molina F, Clark P. Description of Normative Spine TBS Data for Men and Women in Mexican Population. J Clin Densitom. 2021;24(1):129-34.
  • 48
    Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, et al. IDF Diabetes Atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract. 2022;183:109119.
  • 49
    Lin YC, Wu J, Kuo SF, Cheung YC, Sung CM, Fan CM, et al. Vertebral Fractures in Type 2 Diabetes Patients: Utility of Trabecular Bone Score and Relationship With Serum Bone Turnover Biomarkers. J Clin Densitom. 2020;23(1):37-43.
  • 50
    Paschou SA, Dede AD, Anagnostis PG, Vryonidou A, Morganstein D, Goulis DG. Type 2 Diabetes and Osteoporosis: A Guide to Optimal Management. J Clin Endocrinol Metab. 2017;102(10):3621-34.
  • 51
    Cutrim DM, Pereira FA, de Paula FJ, Foss MC. Lack of relationship between glycemic control and bone mineral density in type 2 diabetes mellitus. Braz J Med Biol Res. 2007;40(2):221-7.
  • 52
    Walsh JS, Vilaca T. Obesity, Type 2 Diabetes and Bone in Adults. Calcif Tissue Int. 2017;100(5):528-35.
  • 53
    Kayath MJ, Dib SA, Vieira JG. Prevalence and magnitude of osteopenia associated with insulin-dependent diabetes mellitus. J Diabetes Complications. 1994;8(2):97-104.
  • 54
    Kayath MJ, Tavares EF, Dib SA, Vieira JG. Prospective bone mineral density evaluation in patients with insulin-dependent diabetes mellitus. J Diabetes Complications. 1998;12(3):133-9.
  • 55
    Carvalho AL, Massaro B, Silva L, Salmon CEG, Fukada SY, Nogueira-Barbosa MH, et al. Emerging Aspects of the Body Composition, Bone Marrow Adipose Tissue and Skeletal Phenotypes in Type 1 Diabetes Mellitus. J Clin Densitom. 2019;22(3):420-8.
  • 56
    Walsh J, Roberts R, Bailey TS, Heinemann L. Insulin Titration Guidelines for Patients With Type 1 Diabetes: It Is About Time! J Diabetes Sci Technol. 2022:19322968221087261.
  • 57
    Gomes MB, Conte D, Drummond KRG, Mallmann F, Pinheiro AA, Leal FSL, et al. Overweight/obesity in adolescents with type 1 diabetes belonging to an admixed population. A Brazilian multicenter study. Diabetol Metab Syndr. 2022;14(1):1.
  • 58
    Arcelus J, Mitchell AJ, Wales J, Nielsen S. Mortality rates in patients with anorexia nervosa and other eating disorders. A meta-analysis of 36 studies. Arch Gen Psychiatry. 2011;68(7):724-31.
  • 59
    Lucas AR, Melton LJ 3rd, Crowson CS, O’Fallon WM. Long-term fracture risk among women with anorexia nervosa: a population-based cohort study. Mayo Clin Proc. 1999;74(10):972-7.
  • 60
    Santos Ferreira DL, Hübel C, Herle M, Abdulkadir M, Loos RJF, Bryant-Waugh R, et al. Associations between Blood Metabolic Profile at 7 Years Old and Eating Disorders in Adolescence: Findings from the Avon Longitudinal Study of Parents and Children. Metabolites. 2019;9(9).
  • 61
    Estour B, Germain N, Diconne E, Frere D, Cottet-Emard JM, Carrot G, et al. Hormonal profile heterogeneity and short-term physical risk in restrictive anorexia nervosa. J Clin Endocrinol Metab. 2010;95(5):2203-10.
  • 62
    Fazeli PK, Faje AT, Meenaghan E, Russell ST, Resulaj M, Lee H, et al. IGF-1 is associated with estimated bone strength in anorexia nervosa. Osteoporos Int. 2020;31(2):259-65.
  • 63
    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(9):2078-88.
  • 64
    Fazeli PK, Faje AT, Bredella MA, Polineni S, Russell S, Resulaj M, et al. Changes in marrow adipose tissue with short-term changes in weight in premenopausal women with anorexia nervosa. Eur J Endocrinol. 2019;180(3):189-99.
  • 65
    Maciel JG, de Araujo IM, Carvalho AL, Simao MN, Bastos CM, Troncon LE, et al. Marrow Fat Quality Differences by Sex in Healthy Adults. J Clin Densitom. 2017;20(1):106-13.
  • 66
    Rosen CJ, Bouxsein ML. Mechanisms of disease: is osteoporosis the obesity of bone? Nat Clin Pract Rheumatol. 2006;2(1):35-43.
  • 67
    Maimoun L, Garnero P, Mura T, Nocca D, Lefebvre P, Philibert P, et al. Specific Effects of Anorexia Nervosa and Obesity on Bone Mineral Density and Bone Turnover in Young Women. J Clin Endocrinol Metab. 2020;105(4).
  • 68
    Horowitz MC, Berry R, Holtrup B, Sebo Z, Nelson T, Fretz JA, et al. Bone marrow adipocytes. Adipocyte. 2017;6(3):193-204.
  • 69
    Rolls BJ. What is the role of portion control in weight management? Int J Obes (Lond). 2014;38 Suppl 1:S1-8.
  • 70
    Jaacks LM, Vandevijvere S, Pan A, McGowan CJ, Wallace C, Imamura F, et al. The obesity transition: stages of the global epidemic. Lancet Diabetes Endocrinol. 2019;7(3):231-40.
  • 71
    Carlsson LMS, Sjoholm K, Jacobson P, Andersson-Assarsson JC, Svensson PA, Taube M, et al. Life Expectancy after Bariatric Surgery in the Swedish Obese Subjects Study. N Engl J Med. 2020;383(16):1535-43.
  • 72
    Alencar M, Araujo IM, Parreiras ESLT, Nogueira-Barbosa MH, Salgado W Jr, Elias J Jr, et al. Hashtag bone: detrimental effects on bone contrast with metabolic benefits one and five years after Roux-en-Y gastric bypass. Braz J Med Biol Res. 2021;54(12):e11499.
  • 73
    Lindeman KG, Greenblatt LB, Rourke C, Bouxsein ML, Finkelstein FL, Yu WE. Longitudinal 5-year evaluation of bone density and microarchitecture after Roux-en-Y gastric bypass surgery. J Clin Endocrinol Metab 2018;103(11):4104-12.
  • 74
    Misra M, Bredella MA. Bone Metabolism in Adolescents Undergoing Bariatric Surgery. J Clin Endocrinol Metab. 2021;106(2):326-36.
  • 75
    Liu C, Wu D, Zhang JF, Xu D, Xu WF, Chen Y, et al. Changes in Bone Metabolism in Morbidly Obese Patients After Bariatric Surgery: A Meta-Analysis. Obes Surg. 2016;26(1):91-7.
  • 76
    Braga CB, Bizari L, Suen VM, Marchini JS, Paula FJ, Cunha SF. Bone mineral density in short bowel syndrome: correlation with BMI and serum vitamins C, E and K. Arch Endocrinol Metab. 2015;59(3):252-8.
  • 77
    Parreiras ESLT, de Araujo IM, Elias J Jr, Nogueira-Barbosa MH, Suen VMM, Marchini JS, et al. Short bowel syndrome: influence of nutritional therapy and incretin GLP1 on bone marrow adipose tissue. Ann N Y Acad Sci. 2018;1415(1):47-56.
  • 78
    Parreiras ESLT, de Araujo IM, Elias J Jr, Nogueira-Barbosa MH, Suen VMM, Marchini JS, et al. Osteoporosis and Hepatic Steatosis: 2 Closely Related Complications in Short-Bowel Syndrome. JPEN J Parenter Enteral Nutr. 2020;44(7):1271-9.
  • 79
    Li G, Zhang L, Wang D, AIQudsy L, Jiang JX, Xu H, et al. Muscle-bone crosstalk and potential therapies for sarco-osteoporosis. J Cell Biochem. 2019;120(9):14262-73.
  • 80
    Bonewald L. Use it or lose it to age: A review of bone and muscle communication. Bone. 2019;120:212-8.
  • 81
    Frost HM. Bone’s mechanostat: a 2003 update. Anat Rec A Discov Mol Cell Evol Biol. 2003;275(2):1081-101.
  • 82
    Jiang SD, Dai LY, Jiang LS. Osteoporosis after spinal cord injury. Osteoporos Int. 2006;17(2):180-92.
  • 83
    Jiang SD, Jiang LS, Dai LY. Mechanisms of osteoporosis in spinal cord injury. Clin Endocrinol (Oxf). 2006;65(5):555-65.
  • 84
    Falcai MJ, Louzada MJ, de Paula FJ, Okubo R, Volpon JB. A modified technique of rat tail suspension for longer periods of observation. Aviat Space Environ Med. 2012;83(12):1176-80.
  • 85
    Komori T. Animal models for osteoporosis. Eur J Pharmacol. 2015;759:287-94.
  • 86
    Vico L, van Rietbergen B, Vilayphiou N, Linossier MT, Locrelle H, Normand M, et al. Cortical and Trabecular Bone Microstructure Did Not Recover at Weight-Bearing Skeletal Sites and Progressively Deteriorated at Non-Weight-Bearing Sites During the Year Following International Space Station Missions. J Bone Miner Res. 2017;32(10):2010-21.
  • 87
    Dadwal UC, Maupin KA, Zamarioli A, Tucker A, Harris JS, Fischer JP, et al. The effects of spaceflight and fracture healing on distant skeletal sites. Sci Rep. 2019;9(1):11419.
  • 88
    Brotto M, Bonewald L. Bone and muscle: Interactions beyond mechanical. Bone. 2015;80:109-14.
  • 89
    Seale P, Bjork B, Yang W, Kajimura S, Chin S, Kuang S, et al. PRDM16 controls a brown fat/skeletal muscle switch. Nature. 2008;454(7207):961-7.
  • 90
    Pedersen BK, Akerstrom TC, Nielsen AR, Fischer CP. Role of myokines in exercise and metabolism. J Appl Physiol (1985). 2007;103(3):1093-8.
  • 91
    Quinn LS, Anderson BG, Strait-Bodey L, Stroud AM, Argiles JM. Oversecretion of interleukin-15 from skeletal muscle reduces adiposity. Am J Physiol Endocrinol Metab. 2009;296(1):E191-202.
  • 92
    Karsenty G, Mera P. Molecular bases of the crosstalk between bone and muscle. Bone. 2018;115:43-9.
  • 93
    Minisola S, Peacock M, Fukumoto S, Cipriani C, Pepe J, Tella SH, et al. Tumour-induced osteomalacia. Nat Rev Dis Primers. 2017;3:17044.
  • 94
    Prie D. FGF23 and Cardiovascular Risk. Ann Endocrinol (Paris). 2021;82(3-4):141-3.
  • 95
    Farr JN, Khosla S. Cellular senescence in bone. Bone. 2019;121:121-33.
  • 96
    Perera N, Sampaio H, Woodhead H, Farrar M. Fracture in Duchenne Muscular Dystrophy: Natural History and Vitamin D Deficiency. J Child Neurol. 2016;31(9):1181-7.
  • 97
    Veilleux LN, Rauch F. Muscle-Bone Interactions in Pediatric Bone Diseases. Curr Osteoporos Rep. 2017;15(5):425-32
  • 98
    Kajimura D, Lee HW, Riley KJ, Arteaga-Solis E, Ferron M, Zhou B, et al. Adiponectin regulates bone mass via opposite central and peripheral mechanisms through FoxO1. Cell metabolism. 2013;17(6):901-15.
  • 99
    Li J, Zhang T, Huang C, Xu M, Xie W, Pei Q, et al. Chemerin located in bone marrow promotes osteogenic differentiation and bone formation via Akt/Gsk3beta/beta-catenin axis in mice. J cell Physiol. 2021;236(8):6042-54.

Publication Dates

  • Publication in this collection
    05 Dec 2022
  • Date of issue
    Sep-Oct 2022

History

  • Received
    05 June 2022
  • Accepted
    14 Sept 2022
Sociedade Brasileira de Endocrinologia e Metabologia Rua Botucatu, 572 - Conjuntos 81/83, 04023-061 São Paulo SP Brasil, Tel: (55 11) 5575-0311 - São Paulo - SP - Brazil
E-mail: aem.editorial.office@endocrino.org.br