Acessibilidade / Reportar erro

Diabetes and bone

Abstract

Globally, one in 11 adults has diabetes mellitus of which 90% have type 2 diabetes. The numbers for osteoporosis are no less staggering: 1 in 3 women has a fracture after menopause, and the same is true for 1 in 5 men after the age of 50 years. Aging is associated with several physiological changes that cause insulin resistance and impaired insulin secretion, which in turn lead to hyperglycemia. The negative balance between bone resorption and formation is a natural process that appears after the fourth decade of life and lasts for the following decades, eroding the bone structure and increasing the risk of fractures. Not incidentally, it has been acknowledged that diabetes mellitus, regardless of whether type 1 or 2, is associated with an increased risk of fracture. The nuances that differentiate bone damage in the two main forms of diabetes are part of the intrinsic heterogeneity of diabetes, which is enhanced when associated with a condition as complex as osteoporosis. This narrative review addresses the main parameters related to the increased risk of fractures in individuals with diabetes, and the mutual factors affecting the treatment of diabetes mellitus and osteoporosis. Arch Endocrinol Metab. 2022;66(5):633-41

Keywords
Glucose; fracture; insulin; FRAX; diagnosis

INTRODUCTION

Diabetes mellitus (DM) and osteoporosis are two of the most important diseases that affect human beings, overburden countries’ healthcare systems, result in high costs, and reduce life expectancy. Moreover, both diseases affect the quality of life, with DM leading to cardiovascular, ocular, renal, and neural damage and osteoporosis robbing its patients of their autonomy and mobility.

Multiple pieces of evidence indicate that not only muscle and adipose tissue contribute to the regulation of energy metabolism but that peptides produced by bone cells are also part of the orchestration that involves the metabolic control of glucose, lipids, and proteins (11 Paula FJ, Rosen CJ. Obesity, diabetes mellitus and last but not least, osteoporosis. Arq Bras Endocrinol Metabol. 2010;54(2):150-7.33 de Paula FJ, Rosen CJ. Vitamin D safety and requirements. Arch Biochem Biophys. 2012;523(1):64-72.). Similarly, several hormones are important for the regulation of energy metabolism and influence bone remodeling (e.g., insulin and epinephrine). As expected, both type 1 DM (T1DM) and type 2 DM (T2DM) are associated with an increased risk of fracture. The mechanisms involved in the deterioration of bone strength are not yet clearly elucidated, but the loss of bone density is certainly not the main determinant of fracture susceptibility in DM. This aspect is more evident in T2DM, where bone density is preserved or even increased, but it is also observed in T1DM, where the increase in fracture risk is much greater than the loss of bone density would suggest (44 Van Hulten V, Rasmussen N, Driessen JHM, Burden AM, Kvist A, van den Bergh JP. Fracture Patterns in Type 1 and Type 2 Diabetes Mellitus: A Narrative Review of Recent Literature. Curr Osteoporos Rep. 2021;19(6):644-55.). Experimental and clinical research on the mechanisms and various diagnostic and treatment aspects of osteoporosis in diabetes has incited great interest. However, in clinical practice, there is a gap in the diagnosis and preventive treatment of osteoporosis in patients with DM. Therefore, this narrative review aims to provide an up-to-date overview of the primary aspects that link osteoporosis to DM, including the effects of DM treatment drugs that increase the risk of fracture and the possible diabetogenic effects of drugs used to treat osteoporosis.

Bone density in type 1 diabetes mellitus

The introduction of insulin therapy and improvement of insulin replacement in patients with T1DM have mitigated the emergence of microvascular complications and allowed an increase in this population’s life expectancy. However, with the increasing number of studies on the comorbidities of diabetes, it has been acknowledged that T1DM increases the risk of fractures (55 Palermo A, D’Onofrio L, Buzzetti R, Manfrini S, Napoli N. Pathophysiology of Bone Fragility in Patients with Diabetes. Calcif Tissue Int. 2017;100(2):122-32.,66 Napoli N, Chandran M, Pierroz DD, Abrahamsen B, Schwartz AV, Ferrari SL, et al. Mechanisms of diabetes mellitus-induced bone fragility. Nat Rev Endocrinol. 2017;13(4):208-19.). The relative risk for all fractures in T1DM is 3.16 (95% confidence interval [CI] 1.51-6.63; P = 0.002) according to a meta-analysis performed by Shah and cols. (77 Shah VN, Shah CS, Snell-Bergeon JK. Type 1 diabetes and risk of fracture: meta-analysis and review of the literature. Diabet Med. 2015;32(9):1134-42.). Women still are four times and men two times more likely to have fractures when diagnosed with T1DM (77 Shah VN, Shah CS, Snell-Bergeon JK. Type 1 diabetes and risk of fracture: meta-analysis and review of the literature. Diabet Med. 2015;32(9):1134-42.).

Studies remain contradictory regarding the bone density of individuals with T1DM, and there appear to be sex differences. A prospective study followed-up men and women with T1DM for 5 years and observed that bone density did not change in women during that period, but there was a reduction in bone density in the femoral neck of men (88 Hamilton EJ, Rakic V, Davis WA, Paul Chubb SA, Kamber N, Prince RL, et al. A five-year prospective study of bone mineral density in men and women with diabetes: the Fremantle Diabetes Study. Acta Diabetol. 2012;49(2):153-8.). In addition, a meta-analysis reported a slight reduction in bone density only in the femoral neck of adults with T1DM while it remained stable in the lumbar spine (99 Shah VN, Harrall KK, Shah CS, Gallo TL, Joshee P, Snell-Bergeon JK, et al. Bone mineral density at femoral neck and lumbar spine in adults with type 1 diabetes: a meta-analysis and review of the literature. Osteoporos Int. 2017;28(9):2601-10.).

The traditional lean phenotype of T1DM is being changed by the combination of excess calorie intake and compensation with high insulin doses. This combination leads to weight gain, with changes in body composition and fat accumulation, which may increase the risk of developing obesity-related diseases (1010 Van der Schueren B, Ellis D, Faradji RN, Al-Ozairi E, Rosen J, Mathieu C. Obesity in people living with type 1 diabetes. Lancet Diabetes Endocrinol. 2021;9(11):776-85.).

Body weight has a positive relationship with bone density (1111 Reid IR. Fat and bone. Arch Biochem Biophys. 2010;503(1):20-7.). Thus, recent studies have observed that individuals with T1DM with normal or increased weight have bone density similar to normoglycemic individuals (1212 Slade JM, Coe LM, Meyer RA, McCabe LR. Human bone marrow adiposity is linked with serum lipid levels not T1-diabetes. J Diabetes Complications. 2012;26(1):1-9.,1313 Roggen I, Gies I, Vanbesien J, Louis O, De Schepper J. Trabecular bone mineral density and bone geometry of the distal radius at completion of pubertal growth in childhood type 1 diabetes. Horm Res Paediatr. 2013;79(2):68-74.). Therefore, as in T2DM, the increased risk of fractures in T1DM may be related to a decrease in bone quality. This hypothesis is supported by several studies showing that the trabecular bone score (TBS), an indirect measure of trabecular bone microarchitecture, is lower in T1DM than in a control group (1414 Loxton P, Narayan K, Munns CF, Craig ME. Bone Mineral Density and Type 1 Diabetes in Children and Adolescents: A Meta-analysis. Diabetes Care. 2021;44(8):1898-905.,1515 Neumann T, Lodes S, Kastner B, Lehmann T, Hans D, Lamy O, et al. Trabecular bone score in type 1 diabetes--a cross-sectional study. Osteoporos Int. 2016;27(1):127-33.).

Bone density in type 2 diabetes mellitus

Regardless of sample size, studies show that T2DM does not negatively affect bone density, even when compared with a group of similar body weight (1616 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.,1717 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.). Later prospective studies clearly showed that although T2DM does not result in specific loss of bone density, it is associated with a higher risk of fracture (1818 Schwartz AV, Sellmeyer DE, Ensrud KE, Cauley JA, Tabor HK, Schreiner PJ, et al. Older women with diabetes have an increased risk of fracture: a prospective study. J Clin Endocrinol Metab. 2001;86(1):32-8.,1919 Vestergaard P, Rejnmark L, Mosekilde L. Relative fracture risk in patients with diabetes mellitus, and the impact of insulin and oral antidiabetic medication on relative fracture risk. Diabetologia. 2005;48(7):1292-9.). Individuals with diabetes are 32% more likely to have a fracture compared to those without diabetes (2020 Wang H, Ba Y, Xing Q, Du JL. Diabetes mellitus and the risk of fractures at specific sites: a meta-analysis. BMJ Open. 2019;9(1):e024067.). The Rotterdam study evaluated the bone density of 792 subjects, divided into control and T2DM groups. The results clearly showed a greater bone density in the T2DM group than in the control group. However, the same individuals were followed-up for 6.8 years, and it was observed that the T2DM group, regardless of sex, had a higher frequency of fractures than the control group (2121 de L, II, van der Klift M, de Laet CE, van Daele PL, Hofman A, Pols HA. Bone mineral density and fracture risk in type-2 diabetes mellitus: the Rotterdam Study. Osteoporos Int. 2005;16(12):1713-20.). Thus, it can be concluded that the densitometry test alone underestimates the risk of fractures in individuals with T2DM.

Insulin resistance is clearly related to the emergence of cardiovascular diseases and T2DM. However, a recent study found that there is no relationship between bone density and the various parameters related to insulin resistance (2222 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.). On the other hand, insulin resistance in osteoblasts may decrease osteoblastic activity, as observed in a mice model (2323 Wei J, Ferron M, Clarke CJ, Hannun YA, Jiang H, Blaner WS, et al. Bone-specific insulin resistance disrupts whole-body glucose homeostasis via decreased osteocalcin activation. J Clin Invest. 2014;124(4):1-13.). A low rate of bone remodeling is common in several conditions associated with insulin resistance (2424 de Paula FJA, Rosen CJ. Marrow Adipocytes: Origin, Structure, and Function. Annu Rev Physiol. 2020;82:461-84.,2525 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.)

Structural and metabolic bone changes in type 2 diabetes mellitus

Bone remodeling

Osteocalcin is considered a biochemical marker of bone remodeling and is involved in several physiological processes, such as the maintenance of normal bone mineralization and deceleration of growth-cartilage mineralization. A hallmark of bone disease in T2DM is decreased bone formation, biochemically translated as a reduction in serum osteocalcin (1717 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.). In a recent study, not only was it found that the production of osteocalcin was impaired but also that serum levels of osteocalcin are negatively associated with bone density. These data suggest that low bone remodeling activity is a determining factor in maintaining bone density in T2DM (1717 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.). These data have been replicated in the literature. A previous study reported that individuals with diabetes had reduced osteocalcin when compared to a control group, and serum levels of osteocalcin were also negatively associated with those of glucose and insulin as well as with the Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) score (2626 Liang Y, Tan A, Liang D, Yang X, Liao M, Gao Y, et al. Low osteocalcin level is a risk factor for impaired glucose metabolism in a Chinese male population. J Diabetes Investig. 2016;7(4):522-8.). In hyperglycemic conditions, the Wnt/β-catenin pathway is suppressed, thereby modulating bone formation owing to the increase of its inhibitors Dickkopf-related protein 1 (DKK1) and sclerostin (2727 Gaudio A, Privitera F, Pulvirenti I, Canzonieri E, Rapisarda R, Fiore CE. The relationship between inhibitors of the Wnt signalling pathway (sclerostin and Dickkopf-1) and carotid intima-media thickness in postmenopausal women with type 2 diabetes mellitus. Diab Vasc Dis Res. 2014;11(1):48-52.). An increase in DKK1 and sclerostin was observed in children with T1DM, and this increase was correlated to hyperglycemia (2828 Faienza MF, Ventura A, Delvecchio M, Fusillo A, Piacente L, Aceto G, et al. High Sclerostin and Dickkopf-1 (DKK-1) Serum Levels in Children and Adolescents With Type 1 Diabetes Mellitus. J Clin Endocrinol Metab. 2017;102(4):1174-81.).

Advanced glycation endproducts and ferroptosis

The accumulation of advanced glycation endproducts (AGEs) in bone possibly decreases bone elasticity by structural modification of collagen, altering the biomechanical properties of bone (2929 Saito M, Kida Y, Kato S, Marumo K. Diabetes, collagen, and bone quality. Curr Osteoporos Rep. 2014;12(2):181-8.). In vitro studies indicate that pentosidine decreases osteoclast differentiation and activity (3030 Valcourt U, Merle B, Gineyts E, Viguet-Carrin S, Delmas PD, Garnero P. Non-enzymatic glycation of bone collagen modifies osteoclastic activity and differentiation. J Biol Chem. 2007;282(8):5691-703.). AGEs appear to suppress cell differentiation of both osteoblasts and osteoclasts in a dose-dependent manner (3131 Park SY, Choi KH, Jun JE, Chung HY. Effects of Advanced Glycation End Products on Differentiation and Function of Osteoblasts and Osteoclasts. J Korean Med Sci. 2021;36(37):e239.). One study found a relationship between increased pentosidine levels and an increased incidence of clinical fractures in individuals with diabetes (relative risk [RR] 1.42; 95% CI) (3232 Schwartz AV, Garnero P, Hillier TA, Sellmeyer DE, Strotmeyer ES, Feingold KR, et al. Pentosidine and increased fracture risk in older adults with type 2 diabetes. J Clin Endocrinol Metab. 2009;94(7):2380-6.).

Ferroptosis has recently been recognized as a cell death programming mechanism induced by an increased production of reactive oxygen species and iron-dependent lipid peroxidation. In a recent study, Yang and cols. found that the metabolic environment of DM is favorable to lipid peroxidation, iron overload, and activation of ferroptosis. Additionally, their study showed that inhibition of this process can rescue osteocytes. Therefore, these authors not only point to a new mechanism for the emergence of bone damage in diabetes but also to a potential treatment path (3333 Yang Y, Lin Y, Wang M, Yuan K, Wang Q, Mu P, et al. Targeting ferroptosis suppresses osteocyte glucolipotoxicity and alleviates diabetic osteoporosis. Bone Res. 2022;10(1):26.).

Bone marrow adipose tissue

Studies of bone marrow adiposity in T2DM remain controversial. A recent study evaluated 78 individuals, divided into a control group and groups for patients with obesity and diabetes. Bone marrow adiposity was similar between the groups, and a trend toward a negative association between lumbar-spine bone density and bone marrow adiposity was noted (1717 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.). On the other hand, another study reported that women with T2DM have higher levels of saturated lipids and lower levels of unsaturated lipids in their bone marrow than women without diabetes (3434 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(8):1721-8.). Moreover, a previous study found that saturated lipids are associated with an increased risk of fracture in T2DM (3434 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(8):1721-8.).

Osteoblasts and adipocytes originate from the same mesenchymal stem cells (2424 de Paula FJA, Rosen CJ. Marrow Adipocytes: Origin, Structure, and Function. Annu Rev Physiol. 2020;82:461-84.). It is possible that an increase in adipocyte differentiation may occur to the detriment of the osteoblast lineage. An expanded bone marrow adipose tissue may produce inflammatory cytokines, promoting osteoclastogenesis. Furthermore, in mice with streptozotocin-induced T1DM, an increase in osteoclastogenesis was observed, related to an increased expression of the receptor activator of nuclear factor kappa-Β ligand (RANKL) protein in bone marrow adipocytes (3535 Yang J, Chen S, Zong Z, Yang L, Liu D, Bao Q, et al. The increase in bone resorption in early-stage type I diabetic mice is induced by RANKL secreted by increased bone marrow adipocytes. Biochem Biophys Res Commun. 2020;525(2):433-9.).

Bone microarchitecture

High-resolution peripheral quantitative computed tomography (HR-pQCT) can quantitatively detail cortical and trabecular bone parameters. The results obtained by HR-pQCT performed in individuals with T2DM suggest the occurrence of intracortical porosity in parallel with an increase in trabecular volume (3636 Burghardt AJ, Issever AS, Schwartz AV, Davis KA, Masharani U, Majumdar S, et al. High-resolution peripheral quantitative computed tomographic imaging of cortical and trabecular bone microarchitecture in patients with type 2 diabetes mellitus. J Clin Endocrinol Metab. 2010;95(11):5045-55.). In individuals with T1DM and microvascular disease, significant deficits in cortical and trabecular bone microarchitecture parameters appear to occur in the distal radius and tibia compared to control subjects (3737 Shanbhogue VV, Hansen S, Frost M, Jorgensen NR, Hermann AP, Henriksen JE, et al. Bone Geometry, Volumetric Density, Microarchitecture, and Estimated Bone Strength Assessed by HR-pQCT in Adult Patients With Type 1 Diabetes Mellitus. J Bone Miner Res. 2015;30(12):2188-99.). One study using the microindentation technique evaluated individuals with and without diabetes (3838 Furst JR, Bandeira LC, Fan WW, Agarwal S, Nishiyama KK, McMahon DJ, et al. Advanced Glycation Endproducts and Bone Material Strength in Type 2 Diabetes. J Clin Endocrinol Metab. 2016;101(6):2502-10.). The bone material strength index (BMSi) was impaired in postmenopausal women with diabetes. Another study not only confirmed the BMSi data but also observed a negative correlation with blood glucose (3939 Farr JN, Khosla S. Determinants of bone strength and quality in diabetes mellitus in humans. Bone. 2016;82:28-34.). The texture pattern of bone can be evaluated by the TBS, which is an estimate based on the variations of gray levels in the pixels of images of the lumbar spine from bone densitometry examinations. A previous study found a reduction in the TBS in T2DM and reported that TBS values were higher in individuals with better glycemic control (4040 Dhaliwal R, Cibula D, Ghosh C, Weinstock RS, Moses AM. Bone quality assessment in type 2 diabetes mellitus. Osteoporos Int. 2014;25(7):1969-73.). Figure 1 shows the mechanisms involved in bone fragility.

Figure 1
Mechanisms involved in bone fragility in T2DM. AGEs: advanced glycosylation end products.

Medications

Insulin

Insulin resistance does not seem to be a detrimental factor for bone density from a quantitative point of view, and the literature shows evidence of an increase in bone mineral density at all sites in patients with T2DM (2222 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.,4141 Leslie WD, Aubry-Rozier B, Lamy O, Hans D, Manitoba Bone Density P. TBS (trabecular bone score) and diabetes-related fracture risk. J Clin Endocrinol Metab. 2013;98(2):602-9.). Lipodystrophic syndrome (LDS) can be considered a natural model of hyperinsulinemia, and studies have shown that bone mineral density is preserved in both partial familial forms (4242 Fernandez-Pombo A, Ossandon-Otero JA, Guillin-Amarelle C, Sanchez-Iglesias S, Castro AI, Gonzalez-Mendez B, et al. Bone mineral density in familial partial lipodystrophy. Clin Endocrinol (Oxf). 2018;88(1):44-50.) and generalized congenital forms (4343 Lima JG, Lima NN, Nobrega LH, Jeronimo SM. Conversations between insulin and bone: Potential mechanism of high bone density in patients with Berardinelli-Seip Congenital Lipodystrophy. Med Hypotheses. 2016;97:94-7.) of LDS. However, several studies have reported that the use of insulin is associated with an increase in fractures, with an approximately 38% higher risk among patients using insulin than those using oral antidiabetics (4444 Losada-Grande E, Hawley S, Soldevila B, Martinez-Laguna D, Nogues X, Diez-Perez A, et al. Insulin use and Excess Fracture Risk in Patients with Type 2 Diabetes: A Propensity-Matched cohort analysis. Sci Rep. 2017;7(1):3781.). Furthermore, in a meta-analysis with 138,690 individuals (4545 Zhang Y, Chen Q, Liang Y, Dong Y, Mo X, Zhang L, et al. Insulin use and fracture risk in patients with type 2 diabetes: A meta-analysis of 138,690 patients. Exp Ther Med. 2019;17(5):3957-64.), insulin use was positively associated with fracture risk (RR = 1.24, 95% CI 1.07-1.44; P = 0.004). This higher risk can be explained by episodes of hypoglycemia, and the consequent occurrence of falls (4646 Kennedy RL, Henry J, Chapman AJ, Nayar R, Grant P, Morris AD. Accidents in patients with insulin-treated diabetes: increased risk of low-impact falls but not motor vehicle crashes--a prospective register-based study. J Trauma. 2002;52(4):660-6.). It must be noted that the type of insulin can influence this scenario. Insulin analogs such as glargine have a lower risk of hypoglycemia and a lower association with risk of fractures than neutral protamine Hagedorn insulin (4747 Pscherer S, Kostev K, Dippel FW, Rathmann W. Fracture risk in patients with type 2 diabetes under different antidiabetic treatment regimens: a retrospective database analysis in primary care. Diabetes Metab Syndr Obes. 2016;9:17-23.). Notably, other factors can interfere in the relationship between insulin use and fractures, such as the duration of the disease and coexistence of complications, especially diabetic microvascular disease, retinopathy, and neuropathy (4848 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.).

Metformin

Biguanides have been used for over seven decades, and metformin is considered a first-line treatment for T2DM. However, its role in bone metabolism remains controversial. Some preclinical studies have demonstrated a positive in vitro action of this medication on the differentiation and mineralization of osteoblastic cells through the activation of the adenosine monophosphate (AMP)-activated protein kinase (AMPK) pathway (4949 Kanazawa I, Yamaguchi T, Yano S, Yamauchi M, Sugimoto T. Metformin enhances the differentiation and mineralization of osteoblastic MC3T3-E1 cells via AMP kinase activation as well as eNOS and BMP-2 expression. Biochem Biophys Res Commun. 2008;375(3):414-9.). Metformin also has an osteogenic effect on the recruitment of bone marrow progenitor cells for differentiation into osteoblasts, mediated by the activation of AMPK and the runt-related transcription factor (Runx2), both in vitro and in vivo (5050 Molinuevo MS, Schurman L, McCarthy AD, Cortizo AM, Tolosa MJ, Gangoiti MV, et al. Effect of metformin on bone marrow progenitor cell differentiation: in vivo and in vitro studies. J Bone Miner Res. 2010;25(2):211-21.). Conversely, in a study with in vivo murine models, no effect of metformin use on bone density or fracture healing was observed (5151 Jeyabalan J, Viollet B, Smitham P, Ellis SA, Zaman G, Bardin C, et al. The anti-diabetic drug metformin does not affect bone mass in vivo or fracture healing. Osteoporos Int. 2013;24(10):2659-70.). Some clinical studies reported that metformin use was associated with reduced risk of fractures (hazard ratio [HR] 0.81; 95% CI 0.70-0.93) (1919 Vestergaard P, Rejnmark L, Mosekilde L. Relative fracture risk in patients with diabetes mellitus, and the impact of insulin and oral antidiabetic medication on relative fracture risk. Diabetologia. 2005;48(7):1292-9.). These results were not confirmed in other studies, which did not observe a reduction in the risk of fractures in postmenopausal women or in men (5252 Colhoun HM, Livingstone SJ, Looker HC, Morris AD, Wild SH, Lindsay RS, et al. Hospitalised hip fracture risk with rosiglitazone and pioglitazone use compared with other glucose-lowering drugs. Diabetologia. 2012;55(11):2929-37.,5353 Kanazawa I, Yamaguchi T, Yamamoto M, Sugimoto T. Relationship between treatments with insulin and oral hypoglycemic agents versus the presence of vertebral fractures in type 2 diabetes mellitus. J Bone Miner Metab. 2010;28(5):554-60.). Therefore, the effect of metformin appears to be either positive or neutral in relation to bone density and fracture risk, making it safe for use in patients with osteoporosis.

Sulfonylureas

Sulfonylureas are a class of insulin secretagogue medications. Regarding bone remodeling markers, they seem to have a neutral or reducing effect on C-terminal (CTX) and N-terminal telopeptides of type I collagen (5353 Kanazawa I, Yamaguchi T, Yamamoto M, Sugimoto T. Relationship between treatments with insulin and oral hypoglycemic agents versus the presence of vertebral fractures in type 2 diabetes mellitus. J Bone Miner Metab. 2010;28(5):554-60.,5454 Zinman B, Haffner SM, Herman WH, Holman RR, Lachin JM, Kravitz BG, et al. Effect of rosiglitazone, metformin, and glyburide on bone biomarkers in patients with type 2 diabetes. J Clin Endocrinol Metab. 2010;95(1):134-42.). As for the risk of fractures, a meta-analysis with 11 studies involving more than 200,000 individuals showed that the risk of having a fracture in individuals using sulfonylureas was 1.14 (95% CI 1.08-1.19), similar to the use of thiazolidinediones (TDZs), higher than for metformin, but lower than for insulin (5555 Zhang Z, Cao Y, Tao Y, E M, Tang J, Liu Y, et al. Sulfonylurea and fracture risk in patients with type 2 diabetes mellitus: A meta-analysis. Diabetes Res Clin Pract. 2020;159:107990.). Therefore, in view of the increased risk of hypoglycemia associated with this class of drugs, their use should be avoided in individuals at high risk of fracture.

Incretin mimetics

Incretin mimetics include dipeptidyl-peptidase 4 inhibitors (iDPP4) and glucagon-like peptide (GLP1) analogs. Their role in bone metabolism appears to be related to the differentiation of mesenchymal cells into osteoblasts, which express receptors for GLP1 and glucose-dependent insulinotropic peptide (GIP) (5656 de Paula FJA, Rosen CJ. Structure and Function of Bone Marrow Adipocytes. Compr Physiol. 2017;8(1):315-49.). However, available data from human studies are not conclusive. A meta-analysis of more than 9,000 patients using iDPP4 compared to placebo (5757 Monami M, Dicembrini I, Antenore A, Mannucci E. Dipeptidyl peptidase-4 inhibitors and bone fractures: a meta-analysis of randomized clinical trials. Diabetes Care. 2011;34(11):2474-6.) showed a reduced risk of fractures (odds ratio [OR] = 0.90, 95% CI 0.37-0.99), a finding that was not confirmed in another meta-analysis of observational real-life studies (5858 Hidayat K, Du X, Shi BM. Risk of fracture with dipeptidyl peptidase-4 inhibitors, glucagon-like peptide-1 receptor agonists, or sodium-glucose cotransporter-2 inhibitors in real-world use: systematic review and meta-analysis of observational studies. Osteoporos Int. 2019;30(10):1923-40.). Regarding GLP1, one systematic review (5959 Cheng L, Hu Y, Li YY, Cao X, Bai N, Lu TT, et al. Glucagon-like peptide-1 receptor agonists and risk of bone fracture in patients with type 2 diabetes: A meta-analysis of randomized controlled trials. Diabetes Metab Res Rev. 2019;35(7):e3168.) including 38 studies found protection against all fractures (OR = 0.71, 95% CI 0.56-0.91). Another meta-analysis only found reduced risk of hip fractures in the GLP1 subgroup (5858 Hidayat K, Du X, Shi BM. Risk of fracture with dipeptidyl peptidase-4 inhibitors, glucagon-like peptide-1 receptor agonists, or sodium-glucose cotransporter-2 inhibitors in real-world use: systematic review and meta-analysis of observational studies. Osteoporos Int. 2019;30(10):1923-40.). Therefore, these medications do not appear to have detrimental effects on bone density.

Sodium-glucose cotransporter 2 (SGLT2) inhibitors

Sodium-glucose cotransporter 2 (SGLT2i) inhibitors are a group of medications that show a wide range of effects beyond glycemic control, with benefits in cardiovascular outcomes and diabetic kidney disease. Initially, there were concerns regarding bone health, as the CANVAS study associated the use of canagliflozin with reduced bone mineral density in the hip (6060 Bilezikian JP, Watts NB, Usiskin K, Polidori D, Fung A, Sullivan D, et al. Evaluation of Bone Mineral Density and Bone Biomarkers in Patients With Type 2 Diabetes Treated With Canagliflozin. J Clin Endocrinol Metab. 2016;101(1):44-51.) and an increased risk of extremity fractures (6161 Watts NB, Bilezikian JP, Usiskin K, Edwards R, Desai M, Law G, et al. Effects of Canagliflozin on Fracture Risk in Patients With Type 2 Diabetes Mellitus. J Clin Endocrinol Metab. 2016;101(1):157-66.). The CREDENCE trial evaluated, as a secondary analysis, the incidence of bone fracture in T2DM submitted to canagliflozin therapy, but showing impairment in kidney function. In this study, no difference was found in occurrence of fractures between the canagliflozin and placebo groups. As such, further studies will be necessary to clearly define the skeletal effects of canagliflozin (6262 Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, et al. Canagliflozin and Renal Outcomes in Type 2 Diabetes and Nephropathy. N Engl J Med. 2019;380(24):2295-306.). In subsequent meta-analyses and systematic reviews involving canagliflozin, dapagliflozin and empagliflozin, as well as real-world evidence in the literature, no increased risk of fractures was observed (5858 Hidayat K, Du X, Shi BM. Risk of fracture with dipeptidyl peptidase-4 inhibitors, glucagon-like peptide-1 receptor agonists, or sodium-glucose cotransporter-2 inhibitors in real-world use: systematic review and meta-analysis of observational studies. Osteoporos Int. 2019;30(10):1923-40.,6363 Ruanpeng D, Ungprasert P, Sangtian J, Harindhanavudhi T. Sodium-glucose cotransporter 2 (SGLT2) inhibitors and fracture risk in patients with type 2 diabetes mellitus: A meta-analysis. Diabetes Metab Res Rev. 2017;33(6).,6464 Azharuddin M, Adil M, Ghosh P, Sharma M. Sodium-glucose cotransporter 2 inhibitors and fracture risk in patients with type 2 diabetes mellitus: A systematic literature review and Bayesian network meta-analysis of randomized controlled trials. Diabetes Res Clin Pract. 2018;146:180-90.).

Thiazolidinediones

Thiazolidinediones are peroxisome proliferator-activated receptor gamma (PPARγ) agonists. They improve insulin sensitivity, act on the redistribution of visceral adipose tissue to the subcutaneous tissue, and reduce lipotoxicity (6565 Barroso I, Gurnell M, Crowley VE, Agostini M, Schwabe JW, Soos MA, et al. Dominant negative mutations in human PPARgamma associated with severe insulin resistance, diabetes mellitus and hypertension. Nature. 1999;402(6764):880-3.,6666 Kawai M, Rosen CJ. PPARgamma: a circadian transcription factor in adipogenesis and osteogenesis. Nat Rev Endocrinol. 2010;6(11):629-36.). In bone marrow adipose tissue, PPARγ stimulates the differentiation of mesenchymal stem cells into adipocytes impeding the osteoblast formation pathway. Medications (such as TZDs) or stimuli that favor the activation of PPARγ impair bone formation, leading to reduced bone mineral density and predisposition to fractures (5656 de Paula FJA, Rosen CJ. Structure and Function of Bone Marrow Adipocytes. Compr Physiol. 2017;8(1):315-49.). In the literature review, an increased risk of fractures was observed in women (OR = 1.94; 95% CI 1.60-2.35; P < 0.001) but not in men (OR = 1.02; 95% CI 0.83-1.27; P = 0.83), and this risk was similar with both rosiglitazone and pioglitazone. In addition, the use of TZDs was also associated with a reduction in bone mineral density in the lumbar spine, total hip, and femoral neck, and a correlation, albeit not significant, was found between the cumulative use of TZDs and risk of fractures (6767 Zhu ZN, Jiang YF, Ding T. Risk of fracture with thiazolidinediones: an updated meta-analysis of randomized clinical trials. Bone. 2014;68:115-23.). These data were reinforced by a meta-analysis study, confirming the association of this class with an increased risk of fractures (6868 Hidayat K, Du X, Wu MJ, Shi BM. The use of metformin, insulin, sulphonylureas, and thiazolidinediones and the risk of fracture: Systematic review and meta-analysis of observational studies. Obes Rev. 2019;20(10):1494-503.). In view of this evidence, the use of TZDs should be avoided in postmenopausal women owing to the increased risk of fractures.

Clinical management of osteoporotic patients with diabetes

In 2018, the International Osteoporosis Foundation (IOF) published recommendations for the diagnosis and management of bone fragility in diabetes (6969 Ferrari SL, Abrahamsen B, Napoli N, Akesson K, Chandran M, Eastell R, et al. Diagnosis and management of bone fragility in diabetes: an emerging challenge. Osteoporos Int. 2018;29(12):2585-96.). The criteria for the diagnosis and treatment of osteoporosis in patients with diabetes are the same as for the general population: the presence of fragility, osteoporotic fractures, or low bone mineral density in postmenopausal women. However, as mentioned above, a significant proportion of individuals with T2DM have normal or increased bone density, and the risk of fracture is underestimated by densitometry. To improve fracture risk estimation, the IOF proposed several adaptations to the fracture risk analysis in patients with T2DM.

Dual-energy X-ray absorptiometry (DXA)

Specifically for patients with diabetes, the IOF suggests to reduce the threshold of therapeutic intervention to a T-score of ≤−2.0 standard deviations (SD), instead of −2.5 SD as recommended for the general population. This suggestion is based on data showing that low bone density is a risk factor for fractures in T2DM and occurs at a higher T-score level than in the non-diabetic population. Densitometric evaluation should start at age 50 or 5 years after the diagnosis of T2DM in the absence of other risk factors, and it should be repeated every 2 or 3 years, as needed by each patient (6969 Ferrari SL, Abrahamsen B, Napoli N, Akesson K, Chandran M, Eastell R, et al. Diagnosis and management of bone fragility in diabetes: an emerging challenge. Osteoporos Int. 2018;29(12):2585-96.). Owing to the higher risk of vertebral fractures in this population (7070 Koromani F, Oei L, Shevroja E, Trajanoska K, Schoufour J, Muka T, et al. Vertebral Fractures in Individuals With Type 2 Diabetes: More Than Skeletal Complications Alone. Diabetes Care. 2020;43(1):137-44.), evaluation of subclinical fractures by spine radiographs or by a vertebral fracture assessment is recommended. Recently, a group of experts suggested the incorporation of TBS into the FRAX algorithm to refine the analysis of fracture risk in T2DM (7171 Shevroja E, Lamy O, Kohlmeier L, Koromani F, Rivadeneira F, Hans D. Use of Trabecular Bone Score (TBS) as a Complementary Approach to Dual-energy X-ray Absorptiometry (DXA) for Fracture Risk Assessment in Clinical Practice. J Clin Densitom. 2017;20(3):334-45.). Figure 2 shows different options for the evaluation of bone structure, strength and metabolism in T2DM.

Figure 2
Different options to evaluate bone microstructure (HRpQCT, bone histomorphometry and TBS), strength (BMSi) and metabolism (biochemical markers).

World Health Organization fracture-risk algorithm (FRAX®)

The FRAX risk stratification calculator includes T1DM as a risk factor for secondary osteoporosis in patients aged > 40 years. However, there are no parameters that include the increased risk inherent to T2DM, and again, the risk of fracture is underestimated in these patients. In this case, there are several suggestions in the literature for adjusting FRAX. Leslie and cols. suggested four alternatives: 1. select rheumatoid arthritis (RA) in the risk factors; 2. reduce the informed femoral neck T-score by 0.5 SD; 3. increase the patient’s age by 10 years; and 4. correct FRAX with the TBS if available (7272 Leslie WD, Rubin MR, Schwartz AV, Kanis JA. Type 2 diabetes and bone. J Bone Miner Res. 2012;27(11):2231-7.). The IOF recommends choosing one of these adjustments for FRAX and suggests using the RA option in the risk factors as equivalent to T2DM.

Non-pharmacological treatment

There are no specific nutritional recommendations for patients with T2DM and osteoporosis. The Brazilian guidelines for the treatment of osteoporosis recommend men and women aged > 50 years to consume 1,200 mg of calcium a day, preferably through milk and dairy products or supplements, and the use of maintenance doses of vitamin D, 1,000-2,000 IU daily, targeting 30 ng/mL (7373 Radominski SC, Bernardo W, Paula AP, Albergaria BH, Moreira C, Fernandes CE, et al. Diretrizes brasileiras para o diagnóstico e tratamento da osteoporose em mulheres na pós-menopausa. Rev Bras Reumatol. 2017;57:s452-66.). In the context of diabetes, hypoglycemia is an adverse event that must be avoided due to the risk of falls. In frail or elderly patients, strict control of diabetes is not recommended. Regular aerobic physical activity (such as walking) and muscle strengthening are indicated both to improve glycemic control and to prevent sarcopenia and falls (7474 Peres-Ueno MJ, Capato LL, Porto JM, Adao IF, Gomes JM, Herrero C, et al. Association between vertebral fragility fractures, muscle strength and physical performance: a cross-sectional study. Ann Phys Rehabil Med. 2022:101680.,7575 Cangussu-Oliveira LM, Porto JM, Freire Junior RC, Capato LL, Gomes JM, Herrero C, et al. Association between the trunk muscle function performance and the presence of vertebral fracture in older women with low bone mass. Aging Clin Exp Res. 2020;32(6):1067-76.). In addition, medications that reduce bone density, such as glitazones, should be replaced or discontinued, and alcohol consumption and smoking should be discouraged.

Pharmacological treatment

Several lines of evidence suggest that bone cells have endocrine activity, modulating energy metabolism (e.g., through osteocalcin and lipocalin) (7676 Anastasilakis AD, Tsourdi E, Tabacco G, Naciu AM, Napoli N, Vescini F, et al. The Impact of Antiosteoporotic Drugs on Glucose Metabolism and Fracture Risk in Diabetes: Good or Bad News? J Clin Med. 2021;10(5).). Therefore, interference in bone remodeling could potentially affect glucose metabolism (7777 Mendonca ML, Batista SL, Nogueira-Barbosa MH, Salmon CE, Paula FJ. Primary Hyperparathyroidism: The Influence of Bone Marrow Adipose Tissue on Bone Loss and of Osteocalcin on Insulin Resistance. Clinics (Sao Paulo). 2016;71(8):464-9.). However, few studies have addressed whether anti-osteoporosis medications have any impact on glucose metabolism.

There is no consensus in the literature regarding the effects of bisphosphonates (BPPs) on blood glucose. A large retrospective study conducted in the United Kingdom showed a reduced risk of diabetes with the use of BPPs (7878 Toulis KA, Nirantharakumar K, Ryan R, Marshall T, Hemming K. Bisphosphonates and glucose homeostasis: a population-based, retrospective cohort study. J Clin Endocrinol Metab. 2015;100(5):1933-40.), and some prospective studies showed improvement in fasting glucose and hemoglobin A1c (HbA1c) as well as a reduced risk of T2DM (7979 Vestergaard P. Risk of newly diagnosed type 2 diabetes is reduced in users of alendronate. Calcif Tissue Int. 2011;89(4):265-70.,8080 Chan DC, Yang RS, Ho CH, Tsai YS, Wang JJ, Tsai KT. The use of alendronate is associated with a decreased incidence of type 2 diabetes mellitus--a population-based cohort study in Taiwan. PLoS One. 2015;10(4):e0123279.). However, these findings are not described in the pivotal studies of alendronate and zoledronic acid (the FIT and HORIZON studies, respectively), which did not find any differences in fasting glucose values or in the incidence of diabetes (8181 Schwartz AV, Schafer AL, Grey A, Vittinghoff E, Palermo L, Lui LY, et al. Effects of antiresorptive therapies on glucose metabolism: results from the FIT, HORIZON-PFT, and FREEDOM trials. J Bone Miner Res. 2013;28(6):1348-54.).

RANKL may reduce glucose tolerance. Therefore, denosumab, an anti-RANKL antibody, could have some beneficial effect on insulin sensitivity (8282 Bonnet N, Bourgoin L, Biver E, Douni E, Ferrari S. RANKL inhibition improves muscle strength and insulin sensitivity and restores bone mass. J Clin Invest. 2019;129(8):3214-23.). However, no significant beneficial effect of denosumab on fasting blood glucose or on the incidence of diabetes was found in the FREEDOM trial (8181 Schwartz AV, Schafer AL, Grey A, Vittinghoff E, Palermo L, Lui LY, et al. Effects of antiresorptive therapies on glucose metabolism: results from the FIT, HORIZON-PFT, and FREEDOM trials. J Bone Miner Res. 2013;28(6):1348-54.). Additionally, in another study, the administration of denosumab to 48 postmenopausal women with osteoporosis did not change HOMA-IR or fasting glucose after 24 weeks, although there was a decrease in CTX and osteocalcin (8383 Lasco A, Morabito N, Basile G, Atteritano M, Gaudio A, Giorgianni GM, et al. Denosumab Inhibition of RANKL and Insulin Resistance in Postmenopausal Women with Osteoporosis. Calcif Tissue Int. 2016;98(2):123-8.).

Primary hyperparathyroidism is associated with hyperglycemia. Therefore, there was concern about the effects of parathyroid hormone analogs on glucose metabolism (7777 Mendonca ML, Batista SL, Nogueira-Barbosa MH, Salmon CE, Paula FJ. Primary Hyperparathyroidism: The Influence of Bone Marrow Adipose Tissue on Bone Loss and of Osteocalcin on Insulin Resistance. Clinics (Sao Paulo). 2016;71(8):464-9.,8484 Paula FJ, Lanna CM, Shuhama T, Foss MC. Effect of metabolic control on parathyroid hormone secretion in diabetic patients. Braz J Med Biol Res. 2001;34(9):1139-45.). In the case of teriparatide (TPD), these findings remain undefined. Two studies conducted by Anastasilakis’ group showed different findings. The first analyzed 25 women using TPD and 19 women with primary hyperparathyroidism before and after surgical treatment. While the former group did not show any significant changes in glucose homeostasis, the latter showed greater insulin resistance, which improved after surgical resolution (8585 Anastasilakis A, Goulis DG, Koukoulis G, Kita M, Slavakis A, Avramidis A. Acute and chronic effect of teriparatide on glucose metabolism in women with established osteoporosis. Exp Clin Endocrinol Diabetes. 2007;115(2):108-11.). In the other study, 23 postmenopausal women with osteoporosis were evaluated with a 75-g oral glucose tolerance test after 6 months of TPD use. The results showed a slight, subclinical, but significant increase in glycemia in the glycemic curves (8686 Anastasilakis AD, Efstathiadou Z, Plevraki E, Koukoulis GN, Slavakis A, Kita M, et al. Effect of exogenous intermittent recombinant human PTH 1-34 administration and chronic endogenous parathyroid hormone excess on glucose homeostasis and insulin sensitivity. Horm Metab Res. 2008;40(10):702-7.). On the other hand, treatment with TPD was positively associated with an improvement in HbA1c in patients with glucocorticoid-induced osteoporosis (8787 Mazziotti G, Maffezzoni F, Doga M, Hofbauer LC, Adler RA, Giustina A. Outcome of glucose homeostasis in patients with glucocorticoid-induced osteoporosis undergoing treatment with bone active-drugs. Bone. 2014;67:175-80.).

Another anabolic medication available in clinical practice is the anti-sclerostin antibody romosozumab. Owing to its more recent use, there are few data in the literature on its association with glycemic metabolism. A 2022 meta-analysis (8888 Frysz M, Gergei I, Scharnagl H, Smith GD, Zheng J, Lawlor DA, et al. Circulating Sclerostin Levels Are Positively Related to Coronary Artery Disease Severity and Related Risk Factors. J Bone Miner Res. 2022;37(2):273-84.) evaluated 5,069 patients and observed that high sclerostin levels were associated with a higher risk of diabetes (OR = 1.25, 95% CI 1.12-1.37) and high fasting glucose levels (OR = 1.15, 95% CI 1.04-1.26). Indirectly, these data are inconsistent with the findings of the ARCH study, which cast doubt on the cardiovascular safety of romosozumab. These findings were not observed in other safety studies with the same medication (8989 Saag KG, Petersen J, Brandi ML, Karaplis AC, Lorentzon M, Thomas T, et al. Romosozumab or Alendronate for Fracture Prevention in Women with Osteoporosis. N Engl J Med. 2017;377(15):1417-27.). Therefore, further studies are needed to better characterize the cardiovascular effects associated with this medication.

Thus, as there is no strong evidence of any detrimental effects of osteoporosis treatment on diabetes, the approach to osteoporotic patients with diabetes still follows the general recommendations, and BPPs remain the first-line treatment. In older patients with impaired kidney function or those using multiple oral medications, denosumab may be preferred. Finally, the use of anabolic medications remains indicated in patients with severe osteoporosis, previous fractures, or in those at very high risk of fractures (6969 Ferrari SL, Abrahamsen B, Napoli N, Akesson K, Chandran M, Eastell R, et al. Diagnosis and management of bone fragility in diabetes: an emerging challenge. Osteoporos Int. 2018;29(12):2585-96.).

In conclusion, this article emphasizes that bone fragility should be part of the investigation protocols for the chronic complications of T1DM and T2DM, particularly in individuals who have had diabetes for more than five years. While anti-osteoporosis agents do not appear to negatively impact glucose metabolism, some oral antidiabetic drugs may impair bone density maintenance. This should be considered in the therapeutic choice, particularly for postmenopausal women.

Acknowledgment:

the support received from the following sources enabled the creation of this review and the studies that were conducted.: F.J.A.P.: São Paulo State Foundation for Research Support (Fapesp) grant nº 2018/18071-5 and Brazilian National Council for the Scientific and Technological Development (CNPq) grant nº 309316/2021-9 and Faepa. I.M.A.: Fapesp grant nº 2021/03152-2.

REFERENCES

  • 1
    Paula FJ, Rosen CJ. Obesity, diabetes mellitus and last but not least, osteoporosis. Arq Bras Endocrinol Metabol. 2010;54(2):150-7.
  • 2
    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.
  • 3
    de Paula FJ, Rosen CJ. Vitamin D safety and requirements. Arch Biochem Biophys. 2012;523(1):64-72.
  • 4
    Van Hulten V, Rasmussen N, Driessen JHM, Burden AM, Kvist A, van den Bergh JP. Fracture Patterns in Type 1 and Type 2 Diabetes Mellitus: A Narrative Review of Recent Literature. Curr Osteoporos Rep. 2021;19(6):644-55.
  • 5
    Palermo A, D’Onofrio L, Buzzetti R, Manfrini S, Napoli N. Pathophysiology of Bone Fragility in Patients with Diabetes. Calcif Tissue Int. 2017;100(2):122-32.
  • 6
    Napoli N, Chandran M, Pierroz DD, Abrahamsen B, Schwartz AV, Ferrari SL, et al. Mechanisms of diabetes mellitus-induced bone fragility. Nat Rev Endocrinol. 2017;13(4):208-19.
  • 7
    Shah VN, Shah CS, Snell-Bergeon JK. Type 1 diabetes and risk of fracture: meta-analysis and review of the literature. Diabet Med. 2015;32(9):1134-42.
  • 8
    Hamilton EJ, Rakic V, Davis WA, Paul Chubb SA, Kamber N, Prince RL, et al. A five-year prospective study of bone mineral density in men and women with diabetes: the Fremantle Diabetes Study. Acta Diabetol. 2012;49(2):153-8.
  • 9
    Shah VN, Harrall KK, Shah CS, Gallo TL, Joshee P, Snell-Bergeon JK, et al. Bone mineral density at femoral neck and lumbar spine in adults with type 1 diabetes: a meta-analysis and review of the literature. Osteoporos Int. 2017;28(9):2601-10.
  • 10
    Van der Schueren B, Ellis D, Faradji RN, Al-Ozairi E, Rosen J, Mathieu C. Obesity in people living with type 1 diabetes. Lancet Diabetes Endocrinol. 2021;9(11):776-85.
  • 11
    Reid IR. Fat and bone. Arch Biochem Biophys. 2010;503(1):20-7.
  • 12
    Slade JM, Coe LM, Meyer RA, McCabe LR. Human bone marrow adiposity is linked with serum lipid levels not T1-diabetes. J Diabetes Complications. 2012;26(1):1-9.
  • 13
    Roggen I, Gies I, Vanbesien J, Louis O, De Schepper J. Trabecular bone mineral density and bone geometry of the distal radius at completion of pubertal growth in childhood type 1 diabetes. Horm Res Paediatr. 2013;79(2):68-74.
  • 14
    Loxton P, Narayan K, Munns CF, Craig ME. Bone Mineral Density and Type 1 Diabetes in Children and Adolescents: A Meta-analysis. Diabetes Care. 2021;44(8):1898-905.
  • 15
    Neumann T, Lodes S, Kastner B, Lehmann T, Hans D, Lamy O, et al. Trabecular bone score in type 1 diabetes--a cross-sectional study. Osteoporos Int. 2016;27(1):127-33.
  • 16
    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.
  • 17
    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.
  • 18
    Schwartz AV, Sellmeyer DE, Ensrud KE, Cauley JA, Tabor HK, Schreiner PJ, et al. Older women with diabetes have an increased risk of fracture: a prospective study. J Clin Endocrinol Metab. 2001;86(1):32-8.
  • 19
    Vestergaard P, Rejnmark L, Mosekilde L. Relative fracture risk in patients with diabetes mellitus, and the impact of insulin and oral antidiabetic medication on relative fracture risk. Diabetologia. 2005;48(7):1292-9.
  • 20
    Wang H, Ba Y, Xing Q, Du JL. Diabetes mellitus and the risk of fractures at specific sites: a meta-analysis. BMJ Open. 2019;9(1):e024067.
  • 21
    de L, II, van der Klift M, de Laet CE, van Daele PL, Hofman A, Pols HA. Bone mineral density and fracture risk in type-2 diabetes mellitus: the Rotterdam Study. Osteoporos Int. 2005;16(12):1713-20.
  • 22
    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.
  • 23
    Wei J, Ferron M, Clarke CJ, Hannun YA, Jiang H, Blaner WS, et al. Bone-specific insulin resistance disrupts whole-body glucose homeostasis via decreased osteocalcin activation. J Clin Invest. 2014;124(4):1-13.
  • 24
    de Paula FJA, Rosen CJ. Marrow Adipocytes: Origin, Structure, and Function. Annu Rev Physiol. 2020;82:461-84.
  • 25
    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.
  • 26
    Liang Y, Tan A, Liang D, Yang X, Liao M, Gao Y, et al. Low osteocalcin level is a risk factor for impaired glucose metabolism in a Chinese male population. J Diabetes Investig. 2016;7(4):522-8.
  • 27
    Gaudio A, Privitera F, Pulvirenti I, Canzonieri E, Rapisarda R, Fiore CE. The relationship between inhibitors of the Wnt signalling pathway (sclerostin and Dickkopf-1) and carotid intima-media thickness in postmenopausal women with type 2 diabetes mellitus. Diab Vasc Dis Res. 2014;11(1):48-52.
  • 28
    Faienza MF, Ventura A, Delvecchio M, Fusillo A, Piacente L, Aceto G, et al. High Sclerostin and Dickkopf-1 (DKK-1) Serum Levels in Children and Adolescents With Type 1 Diabetes Mellitus. J Clin Endocrinol Metab. 2017;102(4):1174-81.
  • 29
    Saito M, Kida Y, Kato S, Marumo K. Diabetes, collagen, and bone quality. Curr Osteoporos Rep. 2014;12(2):181-8.
  • 30
    Valcourt U, Merle B, Gineyts E, Viguet-Carrin S, Delmas PD, Garnero P. Non-enzymatic glycation of bone collagen modifies osteoclastic activity and differentiation. J Biol Chem. 2007;282(8):5691-703.
  • 31
    Park SY, Choi KH, Jun JE, Chung HY. Effects of Advanced Glycation End Products on Differentiation and Function of Osteoblasts and Osteoclasts. J Korean Med Sci. 2021;36(37):e239.
  • 32
    Schwartz AV, Garnero P, Hillier TA, Sellmeyer DE, Strotmeyer ES, Feingold KR, et al. Pentosidine and increased fracture risk in older adults with type 2 diabetes. J Clin Endocrinol Metab. 2009;94(7):2380-6.
  • 33
    Yang Y, Lin Y, Wang M, Yuan K, Wang Q, Mu P, et al. Targeting ferroptosis suppresses osteocyte glucolipotoxicity and alleviates diabetic osteoporosis. Bone Res. 2022;10(1):26.
  • 34
    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(8):1721-8.
  • 35
    Yang J, Chen S, Zong Z, Yang L, Liu D, Bao Q, et al. The increase in bone resorption in early-stage type I diabetic mice is induced by RANKL secreted by increased bone marrow adipocytes. Biochem Biophys Res Commun. 2020;525(2):433-9.
  • 36
    Burghardt AJ, Issever AS, Schwartz AV, Davis KA, Masharani U, Majumdar S, et al. High-resolution peripheral quantitative computed tomographic imaging of cortical and trabecular bone microarchitecture in patients with type 2 diabetes mellitus. J Clin Endocrinol Metab. 2010;95(11):5045-55.
  • 37
    Shanbhogue VV, Hansen S, Frost M, Jorgensen NR, Hermann AP, Henriksen JE, et al. Bone Geometry, Volumetric Density, Microarchitecture, and Estimated Bone Strength Assessed by HR-pQCT in Adult Patients With Type 1 Diabetes Mellitus. J Bone Miner Res. 2015;30(12):2188-99.
  • 38
    Furst JR, Bandeira LC, Fan WW, Agarwal S, Nishiyama KK, McMahon DJ, et al. Advanced Glycation Endproducts and Bone Material Strength in Type 2 Diabetes. J Clin Endocrinol Metab. 2016;101(6):2502-10.
  • 39
    Farr JN, Khosla S. Determinants of bone strength and quality in diabetes mellitus in humans. Bone. 2016;82:28-34.
  • 40
    Dhaliwal R, Cibula D, Ghosh C, Weinstock RS, Moses AM. Bone quality assessment in type 2 diabetes mellitus. Osteoporos Int. 2014;25(7):1969-73.
  • 41
    Leslie WD, Aubry-Rozier B, Lamy O, Hans D, Manitoba Bone Density P. TBS (trabecular bone score) and diabetes-related fracture risk. J Clin Endocrinol Metab. 2013;98(2):602-9.
  • 42
    Fernandez-Pombo A, Ossandon-Otero JA, Guillin-Amarelle C, Sanchez-Iglesias S, Castro AI, Gonzalez-Mendez B, et al. Bone mineral density in familial partial lipodystrophy. Clin Endocrinol (Oxf). 2018;88(1):44-50.
  • 43
    Lima JG, Lima NN, Nobrega LH, Jeronimo SM. Conversations between insulin and bone: Potential mechanism of high bone density in patients with Berardinelli-Seip Congenital Lipodystrophy. Med Hypotheses. 2016;97:94-7.
  • 44
    Losada-Grande E, Hawley S, Soldevila B, Martinez-Laguna D, Nogues X, Diez-Perez A, et al. Insulin use and Excess Fracture Risk in Patients with Type 2 Diabetes: A Propensity-Matched cohort analysis. Sci Rep. 2017;7(1):3781.
  • 45
    Zhang Y, Chen Q, Liang Y, Dong Y, Mo X, Zhang L, et al. Insulin use and fracture risk in patients with type 2 diabetes: A meta-analysis of 138,690 patients. Exp Ther Med. 2019;17(5):3957-64.
  • 46
    Kennedy RL, Henry J, Chapman AJ, Nayar R, Grant P, Morris AD. Accidents in patients with insulin-treated diabetes: increased risk of low-impact falls but not motor vehicle crashes--a prospective register-based study. J Trauma. 2002;52(4):660-6.
  • 47
    Pscherer S, Kostev K, Dippel FW, Rathmann W. Fracture risk in patients with type 2 diabetes under different antidiabetic treatment regimens: a retrospective database analysis in primary care. Diabetes Metab Syndr Obes. 2016;9:17-23.
  • 48
    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.
  • 49
    Kanazawa I, Yamaguchi T, Yano S, Yamauchi M, Sugimoto T. Metformin enhances the differentiation and mineralization of osteoblastic MC3T3-E1 cells via AMP kinase activation as well as eNOS and BMP-2 expression. Biochem Biophys Res Commun. 2008;375(3):414-9.
  • 50
    Molinuevo MS, Schurman L, McCarthy AD, Cortizo AM, Tolosa MJ, Gangoiti MV, et al. Effect of metformin on bone marrow progenitor cell differentiation: in vivo and in vitro studies. J Bone Miner Res. 2010;25(2):211-21.
  • 51
    Jeyabalan J, Viollet B, Smitham P, Ellis SA, Zaman G, Bardin C, et al. The anti-diabetic drug metformin does not affect bone mass in vivo or fracture healing. Osteoporos Int. 2013;24(10):2659-70.
  • 52
    Colhoun HM, Livingstone SJ, Looker HC, Morris AD, Wild SH, Lindsay RS, et al. Hospitalised hip fracture risk with rosiglitazone and pioglitazone use compared with other glucose-lowering drugs. Diabetologia. 2012;55(11):2929-37.
  • 53
    Kanazawa I, Yamaguchi T, Yamamoto M, Sugimoto T. Relationship between treatments with insulin and oral hypoglycemic agents versus the presence of vertebral fractures in type 2 diabetes mellitus. J Bone Miner Metab. 2010;28(5):554-60.
  • 54
    Zinman B, Haffner SM, Herman WH, Holman RR, Lachin JM, Kravitz BG, et al. Effect of rosiglitazone, metformin, and glyburide on bone biomarkers in patients with type 2 diabetes. J Clin Endocrinol Metab. 2010;95(1):134-42.
  • 55
    Zhang Z, Cao Y, Tao Y, E M, Tang J, Liu Y, et al. Sulfonylurea and fracture risk in patients with type 2 diabetes mellitus: A meta-analysis. Diabetes Res Clin Pract. 2020;159:107990.
  • 56
    de Paula FJA, Rosen CJ. Structure and Function of Bone Marrow Adipocytes. Compr Physiol. 2017;8(1):315-49.
  • 57
    Monami M, Dicembrini I, Antenore A, Mannucci E. Dipeptidyl peptidase-4 inhibitors and bone fractures: a meta-analysis of randomized clinical trials. Diabetes Care. 2011;34(11):2474-6.
  • 58
    Hidayat K, Du X, Shi BM. Risk of fracture with dipeptidyl peptidase-4 inhibitors, glucagon-like peptide-1 receptor agonists, or sodium-glucose cotransporter-2 inhibitors in real-world use: systematic review and meta-analysis of observational studies. Osteoporos Int. 2019;30(10):1923-40.
  • 59
    Cheng L, Hu Y, Li YY, Cao X, Bai N, Lu TT, et al. Glucagon-like peptide-1 receptor agonists and risk of bone fracture in patients with type 2 diabetes: A meta-analysis of randomized controlled trials. Diabetes Metab Res Rev. 2019;35(7):e3168.
  • 60
    Bilezikian JP, Watts NB, Usiskin K, Polidori D, Fung A, Sullivan D, et al. Evaluation of Bone Mineral Density and Bone Biomarkers in Patients With Type 2 Diabetes Treated With Canagliflozin. J Clin Endocrinol Metab. 2016;101(1):44-51.
  • 61
    Watts NB, Bilezikian JP, Usiskin K, Edwards R, Desai M, Law G, et al. Effects of Canagliflozin on Fracture Risk in Patients With Type 2 Diabetes Mellitus. J Clin Endocrinol Metab. 2016;101(1):157-66.
  • 62
    Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, et al. Canagliflozin and Renal Outcomes in Type 2 Diabetes and Nephropathy. N Engl J Med. 2019;380(24):2295-306.
  • 63
    Ruanpeng D, Ungprasert P, Sangtian J, Harindhanavudhi T. Sodium-glucose cotransporter 2 (SGLT2) inhibitors and fracture risk in patients with type 2 diabetes mellitus: A meta-analysis. Diabetes Metab Res Rev. 2017;33(6).
  • 64
    Azharuddin M, Adil M, Ghosh P, Sharma M. Sodium-glucose cotransporter 2 inhibitors and fracture risk in patients with type 2 diabetes mellitus: A systematic literature review and Bayesian network meta-analysis of randomized controlled trials. Diabetes Res Clin Pract. 2018;146:180-90.
  • 65
    Barroso I, Gurnell M, Crowley VE, Agostini M, Schwabe JW, Soos MA, et al. Dominant negative mutations in human PPARgamma associated with severe insulin resistance, diabetes mellitus and hypertension. Nature. 1999;402(6764):880-3.
  • 66
    Kawai M, Rosen CJ. PPARgamma: a circadian transcription factor in adipogenesis and osteogenesis. Nat Rev Endocrinol. 2010;6(11):629-36.
  • 67
    Zhu ZN, Jiang YF, Ding T. Risk of fracture with thiazolidinediones: an updated meta-analysis of randomized clinical trials. Bone. 2014;68:115-23.
  • 68
    Hidayat K, Du X, Wu MJ, Shi BM. The use of metformin, insulin, sulphonylureas, and thiazolidinediones and the risk of fracture: Systematic review and meta-analysis of observational studies. Obes Rev. 2019;20(10):1494-503.
  • 69
    Ferrari SL, Abrahamsen B, Napoli N, Akesson K, Chandran M, Eastell R, et al. Diagnosis and management of bone fragility in diabetes: an emerging challenge. Osteoporos Int. 2018;29(12):2585-96.
  • 70
    Koromani F, Oei L, Shevroja E, Trajanoska K, Schoufour J, Muka T, et al. Vertebral Fractures in Individuals With Type 2 Diabetes: More Than Skeletal Complications Alone. Diabetes Care. 2020;43(1):137-44.
  • 71
    Shevroja E, Lamy O, Kohlmeier L, Koromani F, Rivadeneira F, Hans D. Use of Trabecular Bone Score (TBS) as a Complementary Approach to Dual-energy X-ray Absorptiometry (DXA) for Fracture Risk Assessment in Clinical Practice. J Clin Densitom. 2017;20(3):334-45.
  • 72
    Leslie WD, Rubin MR, Schwartz AV, Kanis JA. Type 2 diabetes and bone. J Bone Miner Res. 2012;27(11):2231-7.
  • 73
    Radominski SC, Bernardo W, Paula AP, Albergaria BH, Moreira C, Fernandes CE, et al. Diretrizes brasileiras para o diagnóstico e tratamento da osteoporose em mulheres na pós-menopausa. Rev Bras Reumatol. 2017;57:s452-66.
  • 74
    Peres-Ueno MJ, Capato LL, Porto JM, Adao IF, Gomes JM, Herrero C, et al. Association between vertebral fragility fractures, muscle strength and physical performance: a cross-sectional study. Ann Phys Rehabil Med. 2022:101680.
  • 75
    Cangussu-Oliveira LM, Porto JM, Freire Junior RC, Capato LL, Gomes JM, Herrero C, et al. Association between the trunk muscle function performance and the presence of vertebral fracture in older women with low bone mass. Aging Clin Exp Res. 2020;32(6):1067-76.
  • 76
    Anastasilakis AD, Tsourdi E, Tabacco G, Naciu AM, Napoli N, Vescini F, et al. The Impact of Antiosteoporotic Drugs on Glucose Metabolism and Fracture Risk in Diabetes: Good or Bad News? J Clin Med. 2021;10(5).
  • 77
    Mendonca ML, Batista SL, Nogueira-Barbosa MH, Salmon CE, Paula FJ. Primary Hyperparathyroidism: The Influence of Bone Marrow Adipose Tissue on Bone Loss and of Osteocalcin on Insulin Resistance. Clinics (Sao Paulo). 2016;71(8):464-9.
  • 78
    Toulis KA, Nirantharakumar K, Ryan R, Marshall T, Hemming K. Bisphosphonates and glucose homeostasis: a population-based, retrospective cohort study. J Clin Endocrinol Metab. 2015;100(5):1933-40.
  • 79
    Vestergaard P. Risk of newly diagnosed type 2 diabetes is reduced in users of alendronate. Calcif Tissue Int. 2011;89(4):265-70.
  • 80
    Chan DC, Yang RS, Ho CH, Tsai YS, Wang JJ, Tsai KT. The use of alendronate is associated with a decreased incidence of type 2 diabetes mellitus--a population-based cohort study in Taiwan. PLoS One. 2015;10(4):e0123279.
  • 81
    Schwartz AV, Schafer AL, Grey A, Vittinghoff E, Palermo L, Lui LY, et al. Effects of antiresorptive therapies on glucose metabolism: results from the FIT, HORIZON-PFT, and FREEDOM trials. J Bone Miner Res. 2013;28(6):1348-54.
  • 82
    Bonnet N, Bourgoin L, Biver E, Douni E, Ferrari S. RANKL inhibition improves muscle strength and insulin sensitivity and restores bone mass. J Clin Invest. 2019;129(8):3214-23.
  • 83
    Lasco A, Morabito N, Basile G, Atteritano M, Gaudio A, Giorgianni GM, et al. Denosumab Inhibition of RANKL and Insulin Resistance in Postmenopausal Women with Osteoporosis. Calcif Tissue Int. 2016;98(2):123-8.
  • 84
    Paula FJ, Lanna CM, Shuhama T, Foss MC. Effect of metabolic control on parathyroid hormone secretion in diabetic patients. Braz J Med Biol Res. 2001;34(9):1139-45.
  • 85
    Anastasilakis A, Goulis DG, Koukoulis G, Kita M, Slavakis A, Avramidis A. Acute and chronic effect of teriparatide on glucose metabolism in women with established osteoporosis. Exp Clin Endocrinol Diabetes. 2007;115(2):108-11.
  • 86
    Anastasilakis AD, Efstathiadou Z, Plevraki E, Koukoulis GN, Slavakis A, Kita M, et al. Effect of exogenous intermittent recombinant human PTH 1-34 administration and chronic endogenous parathyroid hormone excess on glucose homeostasis and insulin sensitivity. Horm Metab Res. 2008;40(10):702-7.
  • 87
    Mazziotti G, Maffezzoni F, Doga M, Hofbauer LC, Adler RA, Giustina A. Outcome of glucose homeostasis in patients with glucocorticoid-induced osteoporosis undergoing treatment with bone active-drugs. Bone. 2014;67:175-80.
  • 88
    Frysz M, Gergei I, Scharnagl H, Smith GD, Zheng J, Lawlor DA, et al. Circulating Sclerostin Levels Are Positively Related to Coronary Artery Disease Severity and Related Risk Factors. J Bone Miner Res. 2022;37(2):273-84.
  • 89
    Saag KG, Petersen J, Brandi ML, Karaplis AC, Lorentzon M, Thomas T, et al. Romosozumab or Alendronate for Fracture Prevention in Women with Osteoporosis. N Engl J Med. 2017;377(15):1417-27.

Publication Dates

  • Publication in this collection
    05 Dec 2022
  • Date of issue
    2022

History

  • Received
    06 July 2022
  • Accepted
    11 Aug 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