Next Article in Journal
HUS1 as a Potential Therapeutic Target in Urothelial Cancer
Next Article in Special Issue
Simple Predictors for Cardiac Fibrosis in Patients with Type 2 Diabetes Mellitus: The Role of Circulating Biomarkers and Pulse Wave Velocity
Previous Article in Journal
The Need to Control Thoughts in Eating Disorder Outpatients: A Longitudinal Study on Its Modification and Association with Eating Disorder Symptom Improvement
Previous Article in Special Issue
Sex Differences in Cardiovascular Prevention in Type 2: Diabetes in a Real-World Practice Database
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Evaluation of Quality and Bone Microstructure Alterations in Patients with Type 2 Diabetes: A Narrative Review

by
José Ignacio Martínez-Montoro
1,
Beatriz García-Fontana
2,3,4,*,
Cristina García-Fontana
2,3,4,* and
Manuel Muñoz-Torres
2,3,4,5,*
1
Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Faculty of Medicine, University of Malaga, 29010 Malaga, Spain
2
Bone Metabolic Unit, Endocrinology and Nutrition Division, University Hospital Clínico San Cecilio, 18016 Granada, Spain
3
Instituto de Investigación Biosanitaria de Granada (Ibs. GRANADA), 18012 Granada, Spain
4
Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
5
Department of Medicine, University of Granada, 18016 Granada, Spain
*
Authors to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(8), 2206; https://doi.org/10.3390/jcm11082206
Submission received: 2 March 2022 / Revised: 30 March 2022 / Accepted: 13 April 2022 / Published: 14 April 2022
(This article belongs to the Special Issue Clinical Research on Type 2 Diabetes and Its Complications)

Abstract

:
Bone fragility is a common complication in subjects with type 2 diabetes mellitus (T2DM). However, traditional techniques for the evaluation of bone fragility, such as dual-energy X-ray absorptiometry (DXA), do not perform well in this population. Moreover, the Fracture Risk Assessment Tool (FRAX) usually underestimates fracture risk in T2DM. Importantly, novel technologies for the assessment of one microarchitecture in patients with T2DM, such as the trabecular bone score (TBS), high-resolution peripheral quantitative computed tomography (HR-pQCT), and microindentation, are emerging. Furthermore, different serum and urine bone biomarkers may also be useful for the evaluation of bone quality in T2DM. Hence, in this article, we summarize the limitations of conventional tools for the evaluation of bone fragility and review the current evidence on novel approaches for the assessment of quality and bone microstructure alterations in patients with T2DM.

1. Introduction

In the last few decades, type 2 diabetes mellitus (T2DM) has dramatically increased in prevalence worldwide, resulting in significant burdens on patients suffering from this condition and healthcare systems [1]. Of note, the rising prevalence of this disease is associated with the development of a wide range of complications, including retinopathy, nephropathy, neuropathy, and cardiovascular disease [1,2]. These complications often affect the quality of life of patients with T2DM, including their physical and psychological functioning [3]. Although some of these comorbidities have a well-known impact on the quality of life [4,5], others have received less attention [6].
Mounting evidence reveals that bone fragility is common in T2DM [7]. Several studies have shown that T2DM constitutes an independent risk factor for osteoporotic fractures, presenting a particularly strong association with hip fractures [8,9,10,11]. Indeed, a number of meta-analyses have confirmed that T2DM is associated with an increased risk of incident hip, vertebral, and non-vertebral fractures [12,13,14]. Since T2DM has a strong relationship with hip fractures that need replacement surgery using total hip arthroplasty, new techniques have been developed in this field [15,16]. Importantly, increases in the incidence of fractures lead to greater costs and healthcare resource utilization in this population [17]. Moreover, fractures are associated with functional impairment and reduction of health-related quality of life [18,19]. Given the important health and socioeconomic impact of skeletal fragility and fractures, individuals with T2DM, especially those with major diabetes-related determinants and other conventional risk factors for osteoporosis, should be assessed for the presence of bone fragility and their fracture risk [20]. However, traditional imaging techniques and fracture risk assessment tools may not be accurate for this purpose in patients with T2DM [21].
In this review, we summarize the main limitations of commonly used methods to evaluate bone fragility and estimate fracture risk in patients with T2DM, and we also discuss the potential role of novel strategies in the evaluation of quality and bone microstructure alterations in this population. Although some of these issues have been addressed in previous works [22], the current knowledge on novel techniques and biomarkers for the evaluation of bone fragility in T2DM is still limited. We have updated all the information available on the pathogenic mechanisms that explain bone fragility in patients with T2DM. In addition, we have reviewed the role of new technologies and biomarkers in the assessment of bone fragility in T2DM, considering the main clinical studies currently available.

2. Search Strategy and Limitations of the Review

We conducted a comprehensive literature search of articles published in PubMed until March 2022. Peer-reviewed articles related to T2DM and bone fragility published in English were selected, with special attention to clinical studies evaluating bone mineral density (BMD) by dual energy X-ray absorptiometry (DXA) in patients with T2DM, as well as clinical studies assessing bone microstructure through the trabecular bone score (TBS), high-resolution peripheral quantitative computed tomography (HR-pQCT), and microindentation in this population. Finally, we included clinical studies related to the evaluation of novel non-invasive biomarkers of bone quality and fracture risk prediction in T2DM. Original human research articles, including randomized controlled trials, prospective and retrospective observational studies, and cross-sectional studies were considered. The largest studies, as well as the most recent and solid available evidence, were prioritized. Remarkably, a considerable number of the available studies were conducted in postmenopausal women with T2DM; therefore, these results have to be considered cautiously in subjects with T2DM and different characteristics. Moreover, several studies included in this review had a cross-sectional design; thus, further large-scale long-term prospective studies are needed in this field.

3. Determinants of Skeletal Fragility and Increased Risk of Fracture in T2DM

Several determinants have been identified in the pathogenesis of bone fragility and increased fracture risk in subjects with T2DM [23] (Figure 1). Notably, a longer duration of T2DM was reported to be an independent risk factor for major osteoporotic fractures in women aged ≥40 and with ≥10 years of diabetes duration [24], and a recent meta-analysis showed a greater increase in the risk of both hip and non-vertebral fractures in subjects with longer diabetes duration [13]. Besides this, poor glycemic control is closely linked to fracture risk, as several large-scale population-based cohort studies have demonstrated [25,26,27]. In this regard, the generation of advanced glycation end-products (AGEs) resulting from chronic exposure to hyperglycemia is one of the key mechanisms in the pathophysiology of bone fragility in T2DM [23]. As such, non-enzymatic glycosylation of collagen leads to the formation of collagen-AGEs, which are involved in the development of impaired bone mineralization and quality through different alterations of the extracellular matrix, a reduction of alkaline phosphatase activity in osteoblasts, and an overactivation of the receptor for AGEs (the latter associated with the release of pro-inflammatory cytokines and reactive oxygen species—ROS—by osteoclasts) [23,28]. On the other hand, it is also postulated that the main event related to bone fragility in T2DM is an overall inhibition of bone cells function and decreased bone turnover [23,29]. This effect may be driven in part by insulin resistance [30].
In addition to chronic hyperglycemia and AGE formation, other mechanisms play a role in bone fragility in T2DM, as previously reviewed [7,23,31]. Among them, a pro-inflammatory state and oxidative stress, along with adipokine dysregulation and marrow adiposity, have a strong influence on bone metabolism [7,31]. Loss of incretin effect has also been implicated in the pathogenesis of skeletal fragility in T2DM [31,32]. Microvascular disease and impaired vascular bone intercommunication determine alterations of bone quality and microarchitecture [7,31]. Ischemic heart disease has also been reported to be associated with an increased risk of vertebral fractures in T2DM [33]. Vitamin D deficiency, commonly found in patients with T2DM, could play a role in both T2DM development and bone fragility [34]. Pathological changes in gut microbiota composition in T2DM may also trigger bone alterations in this population [35].
Further to this, glucose-lowering agents may also be crucial contributors to the reported associations between T2DM and bone fragility [36,37]. The potential benefits of some drugs for bone density and fracture risk (i.e., metformin, glucagon-like peptide 1 receptors agonists and dipeptidyl peptidase-4 inhibitors) [38,39,40] remain to be confirmed in specifically designed studies. Conversely, the long-term use of thiazolidinediones has been independently associated with fracture risk [41], and sodium-glucose cotransporter-2 inhibitors could also have this effect [42,43]. Remarkably, both insulin and sulfonylureas significantly increase fall-related fractures due to episodes of hypoglycemia [44]. In this vein, other prevalent factors in T2DM (i.e., visual impairment, peripheral neuropathy, autonomic dysfunction/postural hypotension, foot ulcers/amputation, and sarcopenia) also lead to an increased risk of fall-related fractures [31,45].

4. Bone Density and Fracture Risk Prediction in T2DM

Despite skeletal fragility and fracture risk being greater in subjects with T2DM, this condition is usually associated with normal or even increased BMD measured by DXA [46]. Thus, women with T2DM in the Women’s Health Initiative Observational Study presented higher hip and spine BMD scores compared to those without T2DM [47]. Similarly, in a cross-sectional study including two Swedish cohorts, both men and women exhibited a progressively higher hip BMD according to normal fasting plasma glucose/impaired fasting plasma glucose/T2DM subgroups [48]. In the prospective population-based cohort from the Rotterdam Study, inadequate glycemic control was associated with both higher BMD and increased fracture risk in participants with T2DM [27]. Furthermore, a meta-analysis of 15 observational studies (3473 subjects with T2DM and 19,139 healthy controls) showed that participants with T2DM had significantly higher BMD at the femoral neck, hip, and spine [49].
It is noteworthy that these results contrast with those reported by studies assessing BMD in type 1 diabetes mellitus (T1DM), in which BMD is generally low [50]. Although the mechanisms involved in the association between T2DM and normal/high BMD are not fully understood, some data suggest that these findings might be related to chronic hyperinsulinemia and insulin resistance [51], as well as the effect of some adipokines, such as leptin, on bone metabolism [52]. Excess weight/obesity, which are often encountered in patients with T2DM, could also play a role in increased BMD, although some studies have reported that this relationship remains after adjusting for the body mass index (BMI) [49]. Since T2DM is associated with increased fracture risk, regardless of whether there is a normal/high BMD, a fact known as “the diabetic paradox of bone fragility” [53], the diagnosis of osteoporosis based on BMD measured by DXA, should be cautiously considered [21].
On the other hand, the Fracture Risk Assessment Tool (FRAX), which is widely used to estimate 10-year absolute fracture risk, has been demonstrated to underestimate the risk for both hip and major osteoporotic fractures in patients with T2DM [54]. These results are influenced, in part, by the higher BMD observed in patients with T2DM [49]. Indeed, contrary to T1DM, T2DM is not included in the FRAX tool as a secondary cause of osteoporosis [55]. In this regard, some authors have proposed a correction factor with the use of glycated hemoglobin in order to improve the predictive ability of this algorithm for fracture risk [56]. Recently, adjustment of FRAX for T2DM has been suggested in order to create a useful alternative [57,58], although further research is warranted to confirm these results. Alternatively, certain methods (i.e., inputting rheumatoid arthritis, adjusting FRAX by TBS, reducing the femoral T-score by 0.5, and increasing the age by 10 years) have been proposed to improve the performance of FRAX in T2DM, although no single method appears to be optimal in all settings [59]. In light of the above, new approaches to the evaluation of bone fragility in patients with T2DM are needed.

5. Bone Microstructure in T2DM

As previously discussed, patients with T2DM have normal or elevated BMD; however, bone microarchitecture alterations may be present in this group, resulting in an increased fracture risk [60]. In this context, the trabecular bone score, high-resolution peripheral quantitative computed tomography, and microindentation are useful techniques for the evaluation of the bone microstructure in T2DM.

5.1. Trabecular Bone Score

The TBS is a non-invasive, indirect index of trabecular microarchitecture [61]. It is derived from experimental variograms of the projected two-dimensional lumbar spine DXA image and can assess pixel gray-level variations of this area, which translate into a bone microstructure-related score [61]. Accordingly, a high TBS is related to numerous, well-connected and less sparse trabeculae (i.e., normal bone microarchitecture), whereas a low TBS indicates a reduced number of trabeculae and less connectivity, as well as trabecular separation (i.e., altered bone microarchitecture) [61], as shown in Figure 2. In this regard, the proposed TBS cut-off values are as follows: TBS > 1.31 (normal microarchitecture), TBS between 1.23 and 1.31 (partially degraded microarchitecture), and TBS < 1.23 (degraded microarchitecture) [62].
TBS has been demonstrated to be an independent predictor for osteoporotic fractures [62,63,64]. In addition to this, TBS can detect differences between DXA images with similar BMDs [61] and helps to improve the performance of BMD assessed by DXA in the prediction of osteoporotic fractures [65,66]. Indeed, TBS has been incorporated into the FRAX algorithm (FRAX adjusted for TBS), although the clinical impact of this adjustment is yet to be properly evaluated [62].
In patients with T2DM, TBS has been reported to be significantly decreased compared to subjects without diabetes, which suggests that this index could be a useful tool for the diagnosis of bone fragility in this population [67]. TBS may be decreased even in prediabetes, indicating that the degradation of bone microarchitecture may occur in early stages of the disease [68]. Interestingly, in a recent cross-sectional study including 137 patients with T2DM aged 49–85 and 300 healthy controls, the presence of T2DM was associated with significantly lower TBS values despite higher lumbar spine BMD; adiposity (estimated by the relative fat mass) and insulin resistance could play a role in these results [69]. Accordingly, visceral fat reduction may increase TBS values [70]. Furthermore, higher glycated hemoglobin levels and a longer disease duration in patients with T2DM are related to lower TBS values, although the interference of abdominal soft tissue thickness should be considered when interpreting these findings [68,71,72,73]. Moreover, diabetic microvascular disease may be linked to lower TBS [74].
Notably, several studies have shown that TBS can predict incident/prevalent osteoporotic fractures independent of BMD [75,76,77,78] (Table 1). In a retrospective cohort study from the Manitoba Bone Density Program (29,407 women ≥ 50 years, 2356 with diagnosed T2DM), lumbar spine TBS was a BMD-independent predictor of major osteoporotic fractures in both participants with and without T2DM [75]. In a study including 206 postmenopausal women with/without T2DM, TBS values ≤1.130 presented an adequate diagnostic accuracy for vertebral fractures in the former [76], whereas, in a cross-sectional study conducted on 548 patients with T2DM, TBS correlated with prevalent vertebral fractures [77]. Finally, in a study including 285 postmenopausal women with T2DM, TBS had the strongest association with vertebral fractures [78]. Considering all these findings together, TBS may constitute a useful approach for the diagnosis of bone fragility and the evaluation of fracture risk in T2DM, although further prospective studies are needed to corroborate these data.

5.2. High-Resolution, Peripheral, Quantitative Computed Tomography

HR-pQCT is a non-invasive three-dimensional imaging modality that permits the assessment of bone microarchitecture, including the measurement of volumetric cortical and trabecular bone mineral density (vBMD), cortical thickness/porosity, bone strength, and other parameters in the appendicular skeleton (i.e., distal radius and tibia) [79]. In recent years, HR-pQCT has emerged as a promising technique that could become widely used for the diagnosis of osteoporosis and for clinical fragility fracture prediction [80,81].
In a pilot study conducted on 19 postmenopausal women with T2DM matched to 19 controls, Burghardt et al. showed for the first time that T2DM may be associated with bone microarchitecture alterations, as assessed by HR-pQCT [82]. It was observed that, although participants with T2DM had higher trabecular vBMD and trabecular thickness, they also presented higher cortical porosity and impaired bone strength, measured by micro-finite element analysis [82]. Similarly, Patsh et al. reported increased cortical porosity at the ultradistal and distal radio and tibia in 80 postmenopausal women with T2DM [83], while Yu and colleagues also found defects in cortical bone microarchitecture (i.e., higher cortical porosity and lower cortical vBMD) in African American women with T2DM compared to healthy controls [84]. Data from the Framingham Study (a total of 1069 subjects underwent HR-pQCT, 129 subjects with T2DM) showed that patients with T2DM had lower vBMD and higher cortical porosity compared to controls [85]. Interestingly, in a prospective exploratory study that involved postmenopausal women with T2DM with/without a history of fragility fractures and controls, patients with T2DM and a history of fractures exhibited the highest cortical porosity [86]. Cortical porosity increased over time similarly in the three groups, although patients with T2DM and a history of fractures presented the greatest decreases in bone strength indices in the follow-up period, a fact that suggests that cortical porosity may develop early, followed by small increases in this parameter along with significant material strength impairment [86]. Of note, cortical bone deficits assessed by HR-pQCT in T2DM may be driven by the presence of microvascular disease and/or poor metabolic control [87,88].
Conversely, other studies did not find significant differences in bone microarchitecture determined by HR-pQCT between subjects with and without T2DM [89]. Intriguingly, in a population-based sample of women aged 75–80 (99 women with T2DM and 954 controls), T2DM was associated with better bone microarchitecture (including higher trabecular and cortical vBMD in several regions and lower cortical porosity) [90]. In this context, large-scale clinical studies on the topic are required to evaluate the role of HR-pQCT in the diagnosis of bone fragility in T2DM. Moreover, the impacts of cortical porosity and other parameters, as estimated by HR-pQCT, on the prediction of fractures in T2DM are yet to be elucidated.

5.3. Microindentation

Microindentation is an invasive technique that enables percutaneous evaluation of the resistance of bone to indentation in vivo [91]. By indenting a probe tip through the skin covering the tibia and measuring the depth that it penetrates the bone after the generation of an impact force, impact microindentation measurement directly assesses the mechanical characteristics of cortical bone, which are estimated by the bone material strength index (BMSi) [92]. This technique may be particularly useful in populations presenting discrepancies between BMD and increased fracture risk, such as those with T2DM [93]. Accordingly, some studies have reported decreased BMSi in postmenopausal women with T2DM [89,90,94]. Moreover, altered matrix bone properties evaluated by microindentation were confirmed in this population, even though BMD assessed by DXA and/or bone microarchitecture assessed by HR-pQCT showed no differences between subjects with T2DM and healthy controls [89,90]. Remarkably, in a cross-sectional study including 340 men aged 33–96, participants with T2DM exhibited lower mean BMSi compared to subjects with normoglycemia/impaired fasting glucose [95]. However, it should be noted that further work is needed with regard to this technique for the assessment of bone fragility in patients with T2DM.

6. Bone Quality in T2DM: The Role of Biomarkers of Bone Fragility

In addition to bone mineralization and microarchitecture, skeletal material properties are also influenced by bone turnover and the quality of collagen, which may be affected by the accumulation of AGEs, leading to the alteration of collagen crosslinks and function as discussed in previous sections [23]. In this regard, it has been stated that bone turnover is decreased in T2DM, which results in reduced serum levels of bone remodeling markers [23,96,97,98]. However, it remains unknown whether these biochemical markers may be helpful for the diagnosis of bone fragility or the prediction of fracture risk in patients with T2DM. On the one hand, decreased circulating levels of parathyroid hormone (PTH) along with osteocalcin were shown to be associated with a higher risk of vertebral fracture in postmenopausal women with T2DM [99]. On the contrary, in a recent study, Napoli et al. showed that serum bone turnover markers (terminal telopeptide of type 1 collagen-CTX, osteocalcin, and procollagen type 1 N-terminal propeptide-P1NP) were not able to predict fracture risk in T2DM [100].
On the other hand, AGES related to collagen, such as pentosidine and N-carboxymethyl lysine (CML), are increased in bone biopsy specimens from subjects with T2DM [60,101,102]. Therefore, circulating/urinary levels of these AGEs may become attractive surrogate markers of bone quality in subjects with T2DM. Besides this, other novel biomarkers could play a role in the evaluation of bone fragility in T2DM.

6.1. Pentosidine

Pentosidine is a well-characterized AGE derived from the non-enzymatic reaction of pentoses with lysine and arginine residues [103]. Pentosidine levels are increased in T2DM [104]; moreover, circulating levels of pentosidine appear to be higher in patients with T2DM and poor metabolic control, and they are also related to T2DM-associated cardiovascular disease and microvascular complications [104,105,106].
Higher concentrations of pentosidine can also be found in the cancellous bone of patients with T2DM, and this accumulation may be associated with bone fragility via reduced post-yield strain and toughness due to alterations of the bone matrix [60,107,108]. These disturbances may be related to a decreased bone turnover induced by this AGE [109]. Of note, serum/urinary levels of pentosidine may also be applicable markers of bone fragility in T2DM. Thus, serum levels of pentosidine have been reported to be linked to the presence of vertebral fractures in postmenopausal women with T2DM, who presented similar BMD values/bone turnover markers to controls [110]. Furthermore, in a cross-sectional study, urine pentosidine levels were higher in patients with T2DM and vertebral fractures, and were negatively correlated with TBS [111]. In an observational cohort study (501 participants with T2DM and 427 without T2DM), Schwartz et al. showed that urine pentosidine was able to predict incident clinical fractures only in adults with T2DM, while prevalent vertebral fractures were also associated with urine pentosidine in this population [112].

6.2. N-carboxymethyl Lysine

The AGE N-carboxymethyl lysine (CML) may also play an important role in bone fragility in patients with T2DM [102]. In this regard, CML content in human cortical bone has been reported to be higher in subjects with T2DM, which may affect collagen properties [102]. In a large cohort from the Cardiovascular Health Study (3373 participants), serum levels of CML were associated with increased risk of incident hip fracture, independent of the BMD, with no differences in the hazard ratio between participants with and without T2DM [113]. Recently, in a cohort study including 712 participants with T2DM and 2332 subjects without, Dhaliwal et al. showed that circulating levels of CML were higher in patients with T2DM, and higher levels of this AGE were related to an increased risk of incident clinical fractures in this group, independent of the BMD [114]. Indeed, in this study, no relationship was found between hip BMD and CML, which reinforces the notion that bone quality is a major determinant of the pathophysiology of increased fracture risk in T2DM [114].

6.3. Sclerostin

Sclerostin is an inhibitor of the pro-osteogenic Wnt signaling pathway, which results in decreased bone turnover [23,115]. Hence, some studies have found that higher levels of this protein could be associated with a higher risk of osteoporotic fractures [116,117].
Increased circulating levels of sclerostin have been observed in patients with T2DM and may be involved in low bone turnover and a greater risk of fracture found in this population [118]. Thus, higher serum levels of sclerostin have been reported in postmenopausal women with T2DM and fragility fractures, compared to those without fragility fractures [119,120]. In addition to this, in a cross-sectional study including postmenopausal women and men aged >50 years with T2DM, elevated sclerostin levels correlated with the presence of vertebral fractures [121].

6.4. MicroRNAs

MicroRNAs (miRNAs) are epigenetic regulators of different cellular processes, including bone development, homeostasis, and healing [122]. Although evidence regarding the role of these elements in bone fragility in T2DM is still limited, some studies have shed light on their potential utility [123,124,125]. In a study conducted on 168 postmenopausal women with T2DM, three different miRNAs, including senescent miR-31-5p, were significantly associated with incident fragility fractures [123]. In previous analyses, Heilmeier et al. also reported that individual miRNAs or miRNA combinations were able to discriminate the fracture status in postmenopausal women with T2DM [124]. Chen et al. also described several miRNAs with potential implications for fracture prediction in postmenopausal women with T2DM [125].

6.5. Other Biomarkers

Aside from in the serum and urine, AGE deposition can be measured in other tissues, such as the skin. Therefore, skin autofluorescence (SAF), which is based on the non-invasive measurement of AGE accumulation in the human skin, has emerged as a promising technique [126]. However, little evidence is available concerning bone fragility/fracture risk estimation through this tool. In two cross-sectional studies, SAF was inversely correlated with BMSi in patients with T2DM [94,127]. Interestingly, SAF was associated with prevalent vertebral and major osteoporotic fractures in participants from the Rotterdam Study [128]. However, these data must be assessed specifically in individuals with T2DM.
In another area, the fingernail quality may serve as a non-invasive marker of the bone quality in T2DM [129,130]. Nevertheless, further investigation is needed.

7. Conclusions

Since traditional methods for the evaluation of BMD and fracture risk in individuals with T2DM can lead to significant errors, additional techniques are needed. TBS may be considered as a useful non-invasive index of bone microarchitecture, which is often altered in patients with T2DM. Since TBS is derived from DXA images, it may represent an applicable tool for the diagnosis of bone fragility in T2DM. In addition, it could facilitate follow-up and the evaluation of response to treatment in these patients, and may help to unravel the role of certain glucose-lowering agents in bone fragility. HR-pQCT also permits the evaluation of bone microstructure; however, this technique involves significant costs and exposure to radiation, which should be considered. Future opportunities in this area include the evaluation of bone microstructure by DXA-3D, which has shown remarkable results in several conditions other than T2DM and may provide accurate estimations of bone structure and strength, thus offering additional information with regard to fracture risk. Despite the fact that microindentation is a promising method for the evaluation of bone matrix properties, it requires an invasive procedure, which may limit its application in clinical practice. On the other hand, some biochemical markers may represent interesting non-invasive alternatives for the evaluation of skeletal fragility/fracture risk prediction in patients with T2DM, although it is noteworthy that the current evidence regarding some of these alternatives is still limited; therefore, further research (e.g., validation studies) is needed before these biomarkers may be included in routine practice. Further large-scale, long-term prospective studies are needed in the evaluation of quality and bone microstructure alterations in patients with T2DM.

Author Contributions

Conceptualization, J.I.M.-M. and M.M.-T.; writing—original draft preparation, J.I.M.-M.; writing—review and editing, J.I.M.-M., B.G.-F., C.G.-F. and M.M.-T.; supervision, B.G.-F., C.G.-F. and M.M.-T.; funding acquisition, B.G.-F., C.G.-F. and M.M.-T. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Instituto de Salud Carlos III grants (PI18–00803, PI21–01069, and PI18–01235), co-funded by the European Regional Development Fund (FEDER) and by a Junta de Andalucía grant (PI-0268–2019). In addition, C.G-F. was funded by a postdoctoral fellowship from the Instituto de Salud Carlos III (CD20/00022).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The images in Figure 1 and Figure 2 were obtained from smart.servier.com (accessed on 26 February 2022).

Conflicts of Interest

The authors declare no conflict of interest. The funders played no role in the writing of the manuscript or in the decision to publish the results.

References

  1. Zheng, Y.; Ley, S.H.; Hu, F.B. Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nat. Rev. Endocrinol. 2018, 14, 88–98. [Google Scholar] [CrossRef] [PubMed]
  2. Dal Canto, E.; Ceriello, A.; Rydén, L.; Ferrini, M.; Hansen, T.B.; Schnell, O.; Standl, E.; Beulens, J.W. Diabetes as a cardiovascular risk factor: An overview of global trends of macro and micro vascular complications. Eur. J. Prev. Cardiol. 2019, 26 (Suppl. 2), 25–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Alaofè, H.; Amoussa Hounkpatin, W.; Djrolo, F.; Ehiri, J.; Rosales, C. Factors Associated with Quality of Life in Patients with Type 2 Diabetes of South Benin: A Cross-Sectional Study. Int. J. Environ. Res. Public Health 2022, 19, 2360. [Google Scholar] [CrossRef] [PubMed]
  4. Fenwick, E.K.; Pesudovs, K.; Khadka, J.; Dirani, M.; Rees, G.; Wong, T.Y.; Lamoureux, E.L. The impact of diabetic retinopathy on quality of life: Qualitative findings from an item bank development project. Qual. Life Res. 2012, 21, 1771–1782. [Google Scholar] [CrossRef] [PubMed]
  5. Degu, H.; Wondimagegnehu, A.; Yifru, Y.M.; Belachew, A. Is health related quality of life influenced by diabetic neuropathic pain among type II diabetes mellitus patients in Ethiopia? PLoS ONE 2019, 14, e0211449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Sinjari, B.; Feragalli, B.; Cornelli, U.; Belcaro, G.; Vitacolonna, E.; Santilli, M.; Rexhepi, I.; D’Addazio, G.; Zuccari, F.; Caputi, S. Artificial Saliva in Diabetic Xerostomia (ASDIX): Double Blind Trial of Aldiamed® Versus Placebo. J. Clin. Med. 2020, 9, 2196. [Google Scholar] [CrossRef]
  7. Khosla, S.; Samakkarnthai, P.; Monroe, D.G.; Farr, J.N. Update on the pathogenesis and treatment of skeletal fragility in type 2 diabetes mellitus. Nat. Rev. Endocrinol. 2021, 17, 685–697. [Google Scholar] [CrossRef]
  8. Schousboe, J.T.; Morin, S.N.; Kline, G.A.; Lix, L.M.; Leslie, W.D. Differential risk of fracture attributable to type 2 diabetes mellitus according to skeletal site. Bone 2021, 154, 116220. [Google Scholar] [CrossRef]
  9. Wang, B.; Wang, Z.; Poundarik, A.A.; Zaki, M.J.; Bockman, R.S.; Glicksberg, B.S.; Nadkarni, G.N.; Vashishth, D. Unmasking Fracture Risk in Type 2 Diabetes: The Association of Longitudinal Glycemic Hemoglobin Level and Medications. J. Clin. Endocrinol. Metab. 2021, 107, e1390–e1401. [Google Scholar] [CrossRef]
  10. Schwartz, A.V. Epidemiology of fractures in type 2 diabetes. Bone 2016, 82, 2–8. [Google Scholar] [CrossRef]
  11. Koromani, F.; Ghatan, S.; van Hoek, M.; Zillikens, M.C.; Oei, E.H.G.; Rivadeneira, F.; Oei, L. Type 2 Diabetes Mellitus and Vertebral Fracture Risk. Curr. Osteoporos. Rep. 2021, 19, 50–57. [Google Scholar] [CrossRef] [PubMed]
  12. Koromani, F.; Oei, L.; Shevroja, E.; Trajanoska, K.; Schoufour, J.; Muka, T.; Franco, O.H.; Ikram, M.A.; Zillikens, M.C.; Uitterlinden, A.G.; et al. Vertebral Fractures in Individuals with Type 2 Diabetes: More Than Skeletal Complications Alone. Diabetes Care 2020, 43, 137–144. [Google Scholar] [CrossRef] [PubMed]
  13. Vilaca, T.; Schini, M.; Harnan, S.; Sutton, A.; Poku, E.; Allen, I.E.; Cummings, S.R.; Eastell, R. The risk of hip and non-vertebral fractures in type 1 and type 2 diabetes: A systematic review and meta-analysis update. Bone 2020, 137, 115457. [Google Scholar] [CrossRef] [PubMed]
  14. Janghorbani, M.; van Dam, R.M.; Willett, W.C.; Hu, F.B. Systematic Review of Type 1 and Type 2 Diabetes Mellitus and Risk of Fracture. Am. J. Epidemiol. 2007, 166, 495–505. [Google Scholar] [CrossRef]
  15. Ammarullah, M.I.; Afif, I.Y.; Maula, M.I.; Winarni, T.I.; Tauviqirrahman, M.; Akbar, I.; Basri, H.; van der Heide, E.; Jamari, J. Tresca Stress Simulation of Metal-on-Metal Total Hip Arthroplasty during Normal Walking Activity. Materials 2021, 14, 7554. [Google Scholar] [CrossRef]
  16. Jamari, J.; Ammarullah, M.; Saad, A.P.M.; Syahrom, A.; Uddin, M.; van der Heide, E.; Basri, H. The Effect of Bottom Profile Dimples on the Femoral Head on Wear in Metal-on-Metal Total Hip Arthroplasty. J. Funct. Biomater. 2021, 12, 38. [Google Scholar] [CrossRef]
  17. Sato, M.; Ye, W.; Sugihara, T.; Isaka, Y. Fracture risk and healthcare resource utilization and costs among osteoporosis patients with type 2 diabetes mellitus and without diabetes mellitus in Japan: Retrospective analysis of a hospital claims database. BMC Musculoskelet. Disord. 2016, 17, 489. [Google Scholar] [CrossRef] [Green Version]
  18. Shah, A.; Wu, F.; Jones, G.; Cicuttini, F.; Toh, L.S.; Laslett, L.L. The association between incident vertebral deformities, health-related quality of life and functional impairment: A 10.7-year cohort study. Osteoporos. Int. 2021, 32, 2247–2255. [Google Scholar] [CrossRef]
  19. Peeters, C.M.M.; Visser, E.; Van de Ree, C.L.P.; Gosens, T.; Den Oudsten, B.L.; De Vries, J. Quality of life after hip fracture in the elderly: A systematic literature review. Injury 2016, 47, 1369–1382. [Google Scholar] [CrossRef]
  20. Ferrari, S.L.; Abrahamsen, B.; Napoli, N.; Akesson, K.; Chandran, M.; Eastell, R.; El-Hajj Fuleihan, G.; Josse, R.; Kendler, D.L.; Kraenzlin, M.; et al. Diagnosis and management of bone fragility in diabetes: An emerging challenge. Osteoporos. Int. 2018, 29, 2585–2596. [Google Scholar] [CrossRef] [Green Version]
  21. de Waard, E.A.C.; van Geel, T.A.C.M.; Savelberg, H.H.C.M.; Koster, A.; Geusens, P.P.M.M.; van den Bergh, J.P.W. Increased fracture risk in patients with type 2 diabetes mellitus: An overview of the underlying mechanisms and the usefulness of imaging modalities and fracture risk assessment tools. Maturitas 2014, 79, 265–274. [Google Scholar] [CrossRef] [PubMed]
  22. Walsh, J.S.; Vilaca, T. Obesity, Type 2 Diabetes and Bone in Adults. Calcif. Tissue Int. 2017, 100, 528–535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Hofbauer, L.C.; Busse, B.; Eastell, R.; Ferrari, S.; Frost, M.; Müller, R.; Burden, A.M.; Rivadeneira, F.; Napoli, N.; Rauner, M. Bone fragility in diabetes: Novel concepts and clinical implications. Lancet Diabetes Endocrinol. 2022, 10, 207–220. [Google Scholar] [CrossRef]
  24. Majumdar, S.R.; Leslie, W.D.; Lix, L.M.; Morin, S.N.; Johansson, H.; Oden, A.; McCloskey, E.V.; Kanis, J.A. Longer Duration of Diabetes Strongly Impacts Fracture Risk Assessment: The Manitoba BMD Cohort. J. Clin. Endocrinol. Metab. 2016, 101, 4489–4496. [Google Scholar] [CrossRef] [PubMed]
  25. Dufour, A.B.; Kiel, D.P.; Williams, S.A.; Weiss, R.J.; Samelson, E.J. Risk Factors for Incident Fracture in Older Adults with Type 2 Diabetes: The Framingham Heart Study. Diabetes Care 2021, 44, 1547–1555. [Google Scholar] [CrossRef] [PubMed]
  26. Li, C.-I.; Liu, C.-S.; Lin, W.-Y.; Meng, N.-H.; Chen, C.-C.; Yang, S.-Y.; Chen, H.-J.; Lin, C.-C.; Li, T.-C. Glycated Hemoglobin Level and Risk of Hip Fracture in Older People with Type 2 Diabetes: A Competing Risk Analysis of Taiwan Diabetes Cohort Study. J. Bone Miner. Res. 2015, 30, 1338–1346. [Google Scholar] [CrossRef] [Green Version]
  27. Oei, L.; Zillikens, M.C.; Dehghan, A.; Buitendijk, G.H.S.; Castaño-Betancourt, M.C.; Estrada, K.; Stolk, L.; Oei, E.H.G.; van Meurs, J.B.J.; Janssen, J.A.M.J.L.; et al. High Bone Mineral Density and Fracture Risk in Type 2 Diabetes as Skeletal Complications of Inadequate Glucose Control: The Rotterdam Study. Diabetes Care 2013, 36, 1619–1628. [Google Scholar] [CrossRef] [Green Version]
  28. Romero-Díaz, C.; Duarte-Montero, D.; Gutiérrez-Romero, S.A.; Mendivil, C.O. Diabetes and Bone Fragility. Diabetes Ther. 2020, 12, 71–86. [Google Scholar] [CrossRef]
  29. Starup-Linde, J.; Vestergaard, P. Biochemical bone turnover markers in diabetes mellitus—A systematic review. Bone 2016, 82, 69–78. [Google Scholar] [CrossRef]
  30. Tonks, K.T.; White, C.; Center, J.R.; Samocha-Bonet, D.; Greenfield, J. Bone Turnover Is Suppressed in Insulin Resistance, Independent of Adiposity. J. Clin. Endocrinol. Metab. 2017, 102, 1112–1121. [Google Scholar] [CrossRef] [Green Version]
  31. Napoli, N.; Chandran, M.; Pierroz, D.D.; Abrahamsen, B.; Schwartz, A.V.; Ferrari, S.L. Mechanisms of diabetes mellitus-induced bone fragility. Nat. Rev. Endocrinol. 2017, 13, 208–219. [Google Scholar] [CrossRef] [PubMed]
  32. Nuche-Berenguer, B.; Portal-Núñez, S.; Moreno, P.; González, N.; Acitores, A.; López-Herradón, A.; Esbrit, P.; Valverde, I.; Villanueva-Peñacarrillo, M.L. Presence of a functional receptor for GLP-1 in osteoblastic cells, independent of the cAMP-linked GLP-1 receptor. J. Cell. Physiol. 2010, 225, 585–592. [Google Scholar] [CrossRef] [PubMed]
  33. Muñoz-Torres, M.; Reyes-García, R.; García-Martin, A.; Jiménez-Moleón, J.J.; Gonzalez-Ramírez, A.R.; Lara-Villoslada, M.J.; Moreno, P.R. Ischemic heart disease is associated with vertebral fractures in patients with type 2 diabetes mellitus. J. Diabetes Investig. 2013, 4, 310–315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Mitri, J.; Pittas, A.G. Vitamin D and Diabetes. Endocrinol. Metab. Clin. N. Am. 2014, 43, 205–232. [Google Scholar] [CrossRef] [Green Version]
  35. Knudsen, J.K.; Leutscher, P.; Sørensen, S. Gut Microbiota in Bone Health and Diabetes. Curr. Osteoporos. Rep. 2021, 19, 462–479. [Google Scholar] [CrossRef]
  36. Shanbhogue, V.V.; Mitchell, D.M.; Rosen, C.J.; Bouxsein, M.L. Type 2 diabetes and the skeleton: New insights into sweet bones. Lancet Diabetes Endocrinol. 2016, 4, 159–173. [Google Scholar] [CrossRef]
  37. Rozas-Moreno, P.; Reyes-García, R.; Jódar-Gimeno, E.; Varsavsky, M.; Luque-Fernández, I.; Cortés-Berdonces, M.; Muñoz-Torres, M. Recomendaciones sobre el efecto de los fármacos antidiabéticos en el hueso. Endocrinol. Diabetes Nutr. 2017, 64, 1–6. [Google Scholar] [CrossRef]
  38. Molinuevo, M.S.; Schurman, L.; McCarthy, A.D.; Cortizo, A.M.; Tolosa, M.J.; Gangoiti, M.V.; Arnol, V.; Sedlinsky, C. Effect of metformin on bone marrow progenitor cell differentiation: In vivo and in vitro studies. J. Bone Miner. Res. 2010, 25, 211–221. [Google Scholar] [CrossRef] [Green Version]
  39. 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, 2474–2476. [Google Scholar] [CrossRef] [Green Version]
  40. Su, B.; Sheng, H.; Zhang, M.; Bu, L.; Yang, P.; Li, L.; Li, F.; Sheng, C.; Han, Y.; Qu, S.; et al. Risk of bone fractures associated with glucagon-like peptide-1 receptor agonists’ treatment: A meta-analysis of randomized controlled trials. Endocrine 2014, 48, 107–115. [Google Scholar] [CrossRef]
  41. Zhu, Z.-N.; Jiang, Y.-F.; Ding, T. Risk of fracture with thiazolidinediones: An updated meta-analysis of randomized clinical trials. Bone 2014, 68, 115–123. [Google Scholar] [CrossRef] [PubMed]
  42. Kohan, D.E.; Fioretto, P.; Tang, W.; List, J.F. Long-term study of patients with type 2 diabetes and moderate renal impairment shows that dapagliflozin reduces weight and blood pressure but does not improve glycemic control. Kidney Int. 2014, 85, 962–971. [Google Scholar] [CrossRef] [Green Version]
  43. Neal, B.; Perkovic, V.; Mahaffey, K.W.; de Zeeuw, D.; Fulcher, G.; Erondu, N.; Shaw, W.; Law, G.; Desai, M.; Matthews, D.R.; et al. Canagliflozin and Cardiovascular and Renal Events in Type 2 Diabetes. N. Engl. J. Med. 2017, 377, 644–657. [Google Scholar] [CrossRef] [PubMed]
  44. Johnston, S.S.; Conner, C.; Aagren, M.; Ruiz, K.; Bouchard, J. Association between hypoglycaemic events and fall-related fractures in Medicare-covered patients with type 2 diabetes. Diabetes Obes. Metab. 2012, 14, 634–643. [Google Scholar] [CrossRef] [PubMed]
  45. Mayne, D.; Stout, N.R.; Aspray, T.J. Diabetes, falls and fractures. Age Ageing 2010, 39, 522–525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Dennison, E.M.; Syddall, H.E.; Aihie Sayer, A.; Craighead, S.; Phillips, D.I.W.; Cooper, C. Type 2 diabetes mellitus is associated with increased axial bone density in men and women from the Hertfordshire Cohort Study: Evidence for an indirect effect of insulin resistance? Diabetologia 2004, 47, 1963–1968. [Google Scholar] [CrossRef]
  47. Bonds, D.E.; Larson, J.C.; Schwartz, A.V.; Strotmeyer, E.S.; Robbins, J.; Rodriguez, B.L.; Johnson, K.C.; Margolis, K. Risk of Fracture in Women with Type 2 Diabetes: The Women’s Health Initiative Observational Study. J. Clin. Endocrinol. Metab. 2006, 91, 3404–3410. [Google Scholar] [CrossRef] [Green Version]
  48. Mitchell, A.; Fall, T.; Melhus, H.; Wolk, A.; Michaëlsson, K.; Byberg, L. Type 2 Diabetes in Relation to Hip Bone Density, Area, and Bone Turnover in Swedish Men and Women: A Cross-Sectional Study. Calcif. Tissue Int. 2018, 103, 501–511. [Google Scholar] [CrossRef] [Green Version]
  49. Ma, L.; Oei, L.; Jiang, L.; Estrada, K.; Chen, H.; Wang, Z.; Yu, Q.; Zillikens, M.C.; Gao, X.; Rivadeneira, F. Association between bone mineral density and type 2 diabetes mellitus: A meta-analysis of observational studies. Eur. J. Epidemiol. 2012, 27, 319–332. [Google Scholar] [CrossRef] [Green Version]
  50. Pan, H.; Wu, N.; Yang, T.; He, W. Association between bone mineral density and type 1 diabetes mellitus: A meta-analysis of cross-sectional studies. Diabetes Metab. Res. Rev. 2014, 30, 531–542. [Google Scholar] [CrossRef]
  51. Srikanthan, P.; Crandall, C.J.; Miller-Martinez, D.; Seeman, T.E.; Greendale, G.A.; Binkley, N.; Karlamangla, A.S. Insulin Resistance and Bone Strength: Findings From the Study of Midlife in the United States. J. Bone Miner. Res. 2014, 29, 796–803. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Upadhyay, J.; Farr, O.M.; Mantzoros, C.S. The role of leptin in regulating bone metabolism. Metabolism 2015, 64, 105–113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Botella Martínez, S.; Varo Cenarruzabeitia, N.; Escalada San Martin, J.; Calleja Canelas, A. The diabetic paradox: Bone mineral density and fracture in type 2 diabetes. Endocrinol. Nutr. 2016, 63, 495–501. [Google Scholar] [CrossRef] [PubMed]
  54. Giangregorio, L.M.; Leslie, W.D.; Lix, L.M.; Johansson, H.; Oden, A.; McCloskey, E.; Kanis, J.A. FRAX underestimates fracture risk in patients with diabetes. J. Bone Miner. Res. 2012, 27, 301–308. [Google Scholar] [CrossRef] [PubMed]
  55. El Miedany, Y. FRAX: Re-adjust or re-think. Arch. Osteoporos. 2020, 15, 150. [Google Scholar] [CrossRef]
  56. Valentini, A.; Cianfarani, M.A.; De Meo, L.; Morabito, P.; Romanello, D.; Tarantino, U.; Federici, M.; Bertoli, A. FRAX tool in type 2 diabetic subjects: The use of HbA1c in estimating fracture risk. Acta Diabetol. 2018, 55, 1043–1050. [Google Scholar] [CrossRef]
  57. Wen, Z.; Ding, N.; Chen, R.; Liu, S.; Wang, Q.; Sheng, Z.; Liu, H. Comparison of methods to improve fracture risk assessment in chinese diabetic postmenopausal women: A case-control study. Endocrine 2021, 73, 209–216. [Google Scholar] [CrossRef]
  58. Hu, L.; Li, T.; Zou, Y.; Yin, X.-L.; Gan, H. The Clinical Value of the RA-Adjusted Fracture Risk Assessment Tool in the Fracture Risk Prediction of Patients with Type 2 Diabetes Mellitus in China. Int. J. Gen. Med. 2021, 14, 327–333. [Google Scholar] [CrossRef]
  59. Leslie, W.D.; Johansson, H.; McCloskey, E.V.; Harvey, N.C.; Kanis, J.A.; Hans, D. Comparison of Methods for Improving Fracture Risk Assessment in Diabetes: The Manitoba BMD Registry. J. Bone Miner. Res. 2018, 33, 1923–1930. [Google Scholar] [CrossRef] [Green Version]
  60. Hunt, H.B.; Torres, A.M.; Palomino, P.M.; Marty, E.; Saiyed, R.; Cohn, M.; Jo, J.; Warner, S.; Sroga, G.E.; King, K.B.; et al. Altered Tissue Composition, Microarchitecture, and Mechanical Performance in Cancellous Bone From Men With Type 2 Diabetes Mellitus. J. Bone Miner. Res. 2019, 34, 1191–1206. [Google Scholar] [CrossRef]
  61. Silva, B.C.; Leslie, W.D.; Resch, H.; Lamy, O.; Lesnyak, O.; Binkley, N.; McCloskey, E.V.; Kanis, J.A.; Bilezikian, J.P. Trabecular Bone Score: A Noninvasive Analytical Method Based Upon the DXA Image. J. Bone Miner. Res. 2014, 29, 518–530. [Google Scholar] [CrossRef] [PubMed]
  62. McCloskey, E.V.; Oden, A.; Harvey, N.C.; Leslie, W.D.; Hans, D.; Johansson, H.; Barkmann, R.; Boutroy, S.; Brown, J.; Chapurlat, R.; et al. A meta-analysis of trabecular bone score in fracture risk prediction and its relationship to FRAX. J. Bone Miner. Res. 2016, 31, 940–948. [Google Scholar] [CrossRef] [PubMed]
  63. Hans, D.; Goertzen, A.L.; Krieg, M.-A.; Leslie, W.D. Bone microarchitecture assessed by TBS predicts osteoporotic fractures independent of bone density: The Manitoba study. J. Bone Miner. Res. 2011, 26, 2762–2769. [Google Scholar] [CrossRef] [PubMed]
  64. Leslie, W.D.; Aubry-Rozier, B.; Lix, L.M.; Morin, S.N.; Majumdar, S.R.; Hans, D. Spine bone texture assessed by trabecular bone score (TBS) predicts osteoporotic fractures in men: The Manitoba Bone Density Program. Bone 2014, 67, 10–14. [Google Scholar] [CrossRef] [PubMed]
  65. Briot, K.; Paternotte, S.; Kolta, S.; Eastell, R.; Reid, D.M.; Felsenberg, D.; Glüer, C.C.; Roux, C. Added value of trabecular bone score to bone mineral density for prediction of osteoporotic fractures in postmenopausal women: The OPUS study. Bone 2013, 57, 232–236. [Google Scholar] [CrossRef]
  66. Boutroy, S.; Hans, D.; Sornay-Rendu, E.; Vilayphiou, N.; Winzenrieth, R.; Chapurlat, R. Trabecular bone score improves fracture risk prediction in non-osteoporotic women: The OFELY study. Osteoporos. Int. 2013, 24, 77–85. [Google Scholar] [CrossRef]
  67. Ho-Pham, L.T.; Nguyen, T.V. Association between trabecular bone score and type 2 diabetes: A quantitative update of evidence. Osteoporos. Int. 2019, 30, 2079–2085. [Google Scholar] [CrossRef]
  68. Ho-Pham, L.T.; Tran, B.; Do, A.T.; Nguyen, T.V. Association between pre-diabetes, type 2 diabetes and trabecular bone score: The Vietnam Osteoporosis Study. Diabetes Res. Clin. Pract. 2019, 155, 107790. [Google Scholar] [CrossRef]
  69. Hayón-Ponce, M.; García-Fontana, B.; Avilés-Pérez, M.D.; González-Salvatierra, S.; Andújar-Vera, F.; Moratalla-Aranda, E.; Muñoz-Torres, M. Lower trabecular bone score in type 2 diabetes mellitus: A role for fat mass and insulin resistance beyond hyperglycaemia. Diabetes Metab. 2021, 47, 101276. [Google Scholar] [CrossRef]
  70. Moon, H.U.; Lee, N.; Chung, Y.-S.; Choi, Y.J. Reduction of visceral fat could be related to the improvement of TBS in diabetes mellitus. J. Bone Miner. Metab. 2020, 38, 702–709. [Google Scholar] [CrossRef]
  71. Palomo, T.; Dreyer, P.; Muszkat, P.; Weiler, F.G.; Bonansea, T.C.P.; Domingues, F.C.; Vieira, J.G.H.; Silva, B.C.; Brandão, C.M.A. Effect of soft tissue noise on trabecular bone score in postmenopausal women with diabetes: A cross sectional study. Bone 2022, 157, 116339. [Google Scholar] [CrossRef] [PubMed]
  72. Depczynski, B.; Liew, P.Y.; White, C. Association of glycaemic variables with trabecular bone score in post-menopausal women with type 2 diabetes mellitus. Diabet. Med. 2020, 37, 1545–1552. [Google Scholar] [CrossRef] [PubMed]
  73. Iki, M.; Fujita, Y.; Kouda, K.; Yura, A.; Tachiki, T.; Tamaki, J.; Winzenrieth, R.; Sato, Y.; Moon, J.-S.; Okamoto, N.; et al. Hyperglycemia is associated with increased bone mineral density and decreased trabecular bone score in elderly Japanese men: The Fujiwara-kyo osteoporosis risk in men (FORMEN) study. Bone 2017, 105, 18–25. [Google Scholar] [CrossRef] [PubMed]
  74. El Asri, M.M.; Rodrigo, E.P.; de la Flor, S.D.-S.; Valdivieso, S.P.; Barrón, M.C.R.; Martínez, J.M.O.; Hernández, J.L.H. Índice trabecular óseo y niveles de 25-hidroxivitamina D en las complicaciones microvasculares de la diabetes mellitus tipo 2. Med. Clin. 2021, in press. [Google Scholar] [CrossRef]
  75. Leslie, W.D.; Aubry-Rozier, B.; Lamy, O.; Hans, D.; Manitoba Bone Density Program. TBS (Trabecular Bone Score) and Diabetes-Related Fracture Risk. J. Clin. Endocrinol. Metab. 2013, 98, 602–609. [Google Scholar] [CrossRef] [Green Version]
  76. Zhukouskaya, V.V.; Ellen-Vainicher, C.; Gaudio, A.; Privitera, F.; Cairoli, E.; Ulivieri, F.M.; Palmieri, S.; Morelli, V.; Grancini, V.; Orsi, E.; et al. The utility of lumbar spine trabecular bone score and femoral neck bone mineral density for identifying asymptomatic vertebral fractures in well-compensated type 2 diabetic patients. Osteoporos. Int. 2016, 27, 49–56. [Google Scholar] [CrossRef]
  77. Yamamoto, M.; Yamauchi, M.; Sugimoto, T. Prevalent vertebral fracture is dominantly associated with spinal microstructural deterioration rather than bone mineral density in patients with type 2 diabetes mellitus. PLoS ONE 2019, 14, e0222571. [Google Scholar] [CrossRef]
  78. Lin, Y.-C.; Wu, J.; Kuo, S.-F.; Cheung, Y.-C.; Sung, C.-M.; Fan, C.-M.; Chen, F.-P.; Mhuircheartaigh, J.N. Vertebral Fractures in Type 2 Diabetes Patients: Utility of Trabecular Bone Score and Relationship with Serum Bone Turnover Biomarkers. J. Clin. Densitom. 2020, 23, 37–43. [Google Scholar] [CrossRef]
  79. Nishiyama, K.K.; Shane, E. Clinical Imaging of Bone Microarchitecture with HR-pQCT. Curr. Osteoporos. Rep. 2013, 11, 147–155. [Google Scholar] [CrossRef]
  80. Mikolajewicz, N.; Bishop, N.; Burghardt, A.J.; Folkestad, L.; Hall, A.; Kozloff, K.M.; Lukey, P.T.; Molloy-Bland, M.; Morin, S.N.; Offiah, A.; et al. HR-pQCT Measures of Bone Microarchitecture Predict Fracture: Systematic Review and Meta-Analysis. J. Bone Miner. Res. 2019, 35, 446–459. [Google Scholar] [CrossRef]
  81. Cheung, W.; Hung, V.W.; Cheuk, K.; Chau, W.; Tsoi, K.K.; Wong, R.M.; Chow, S.K.; Lam, T.; Yung, P.S.; Law, S.; et al. Best Performance Parameters of HR-pQCT to Predict Fragility Fracture: Systematic Review and Meta-Analysis. J. Bone Miner. Res. 2021, 36, 2381–2398. [Google Scholar] [CrossRef] [PubMed]
  82. Burghardt, A.J.; Issever, A.S.; Schwartz, A.V.; Davis, K.A.; Masharani, U.; Majumdar, S.; Link, T.M. 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, 5045–5055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Patsch, J.M.; Burghardt, A.J.; Yap, S.P.; Baum, T.; Schwartz, A.V.; Joseph, G.B.; Link, T.M. Increased cortical porosity in type 2 diabetic postmenopausal women with fragility fractures. J. Bone Miner. Res. 2013, 28, 313–324. [Google Scholar] [CrossRef] [PubMed]
  84. Yu, E.W.; Putman, M.S.; Derrico, N.; Abrishamanian-Garcia, G.; Finkelstein, J.S.; Bouxsein, M.L. Defects in cortical microarchitecture among African-American women with type 2 diabetes. Osteoporos. Int. 2015, 26, 673–679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Samelson, E.J.; Demissie, S.; Cupples, L.A.; Zhang, X.; Xu, H.; Liu, C.-T.; Boyd, S.K.; McLean, R.R.; Broe, K.E.; Kiel, D.P.; et al. Diabetes and Deficits in Cortical Bone Density, Microarchitecture, and Bone Size: Framingham HR-pQCT Study. J. Bone Miner. Res. 2018, 33, 54–62. [Google Scholar] [CrossRef]
  86. Heilmeier, U.; Joseph, G.B.; Pasco, C.; Dinh, N.; Torabi, S.; Darakananda, K.; Youm, J.; Carballido-Gamio, J.; Burghardt, A.J.; Link, T.M.; et al. Longitudinal Evolution of Bone Microarchitecture and Bone Strength in Type 2 Diabetic Postmenopausal Women with and without History of Fragility Fractures—A 5-Year Follow-Up Study Using High Resolution Peripheral Quantitative Computed Tomography. Front. Endocrinol. 2021, 12, 599316. [Google Scholar] [CrossRef]
  87. Shanbhogue, V.V.; Hansen, S.; Frost, M.; Jørgensen, N.R.; Hermann, A.P.; Henriksen, J.E.; Brixen, K. Compromised cortical bone compartment in type 2 diabetes mellitus patients with microvascular disease. Eur. J. Endocrinol. 2016, 174, 115–124. [Google Scholar] [CrossRef]
  88. De Waard, E.A.C.; De Jong, J.J.A.; Koster, A.; Savelberg, H.H.C.M.; Van Geel, T.A.; Houben, A.J.H.M.; Schram, M.T.; Dagnelie, P.C.; Van Der Kallen, C.J.; Sep, S.J.S.; et al. The association between diabetes status, HbA1c, diabetes duration, microvascular disease, and bone quality of the distal radius and tibia as measured with high-resolution peripheral quantitative computed tomography—The Maastricht Study. Osteoporos. Int. 2018, 29, 2725–2738. [Google Scholar] [CrossRef] [Green Version]
  89. Farr, J.N.; Drake, M.T.; Amin, S.; Melton, L.J., 3rd; McCready, L.K.; Khosla, S. In Vivo Assessment of Bone Quality in Postmenopausal Women with Type 2 Diabetes. J. Bone Miner. Res. 2014, 29, 787–795. [Google Scholar] [CrossRef]
  90. Nilsson, A.G.; Sundh, D.; Johansson, L.; Nilsson, M.; Mellström, D.; Rudäng, R.; Zoulakis, M.; Wallander, M.; Darelid, A.; Lorentzon, M. Type 2 Diabetes Mellitus Is Associated with Better Bone Microarchitecture But Lower Bone Material Strength and Poorer Physical Function in Elderly Women: A Population-Based Study. J. Bone Miner. Res. 2017, 32, 1062–1071. [Google Scholar] [CrossRef] [Green Version]
  91. Randall, C.; Bridges, D.; Guerri, R.; Nogues, X.; Puig, L.; Torres, E.; Mellibovsky, L.; Hoffseth, K.; Stalbaum, T.; Srikanth, A.; et al. Applications of a New Handheld Reference Point Indentation Instrument Measuring Bone Material Strength. J. Med. Devices 2013, 7, 041005. [Google Scholar] [CrossRef] [PubMed]
  92. Bridges, D.; Randall, C.; Hansma, P.K. A new device for performing reference point indentation without a reference probe. Rev. Sci. Instrum. 2012, 83, 044301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Herrera, S.; Diez-Perez, A. Clinical experience with microindentation in vivo in humans. Bone 2017, 95, 175–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Furst, J.R.; Bandeira, L.C.; Fan, W.-W.; Agarwal, S.; Nishiyama, K.K.; McMahon, D.J.; Dworakowski, E.; Jiang, H.; Silverberg, S.J.; Rubin, M.R. Advanced Glycation Endproducts and Bone Material Strength in Type 2 Diabetes. J. Clin. Endocrinol. Metab. 2016, 101, 2502–2510. [Google Scholar] [CrossRef] [Green Version]
  95. Holloway-Kew, K.L.; Betson, A.; Rufus-Membere, P.G.; Gaston, J.; Diez-Perez, A.; Kotowicz, M.A.; Pasco, J.A. Impact microindentation in men with impaired fasting glucose and type 2 diabetes. Bone 2021, 142, 115685. [Google Scholar] [CrossRef]
  96. Starup-Linde, J.; Lykkeboe, S.; Handberg, A.; Vestergaard, P.; Høyem, P.; Fleischer, J.; Hansen, T.K.; Poulsen, P.L.; Laugesen, E. Glucose variability and low bone turnover in people with type 2 diabetes. Bone 2021, 153, 116159. [Google Scholar] [CrossRef]
  97. Starup-Linde, J.; Eriksen, S.A.; Lykkeboe, S.; Handberg, A.; Vestergaard, P. Biochemical markers of bone turnover in diabetes patients—A meta-analysis, and a methodological study on the effects of glucose on bone markers. Osteoporos. Int. 2014, 25, 1697–1708. [Google Scholar] [CrossRef]
  98. Reyes-Garcia, R.; Rozas-Moreno, P.; López-Gallardo, G.; Garcia-Martin, A.; Varsavsky, M.; Avilés-Pérez, M.D.; Muñoz-Torres, M. Serum levels of bone resorption markers are decreased in patients with type 2 diabetes. Acta Diabetol. 2013, 50, 47–52. [Google Scholar] [CrossRef]
  99. Yamamoto, M.; Yamaguchi, T.; Nawata, K.; Yamauchi, M.; Sugimoto, T. Decreased PTH Levels Accompanied by Low Bone Formation Are Associated with Vertebral Fractures in Postmenopausal Women with Type 2 Diabetes. J. Clin. Endocrinol. Metab. 2012, 97, 1277–1284. [Google Scholar] [CrossRef]
  100. Napoli, N.; Conte, C.; Eastell, R.; Ewing, S.K.; Bauer, D.C.; Strotmeyer, E.S.; Black, D.M.; Samelson, E.J.; Vittinghoff, E.; Schwartz, A.V. Bone Turnover Markers Do Not Predict Fracture Risk in Type 2 Diabetes. J. Bone Miner. Res. 2020, 35, 2363–2371. [Google Scholar] [CrossRef]
  101. Karim, L.; Moulton, J.; Van Vliet, M.; Velie, K.; Robbins, A.; Malekipour, F.; Abdeen, A.; Ayres, D.; Bouxsein, M.L. Bone microarchitecture, biomechanical properties, and advanced glycation end-products in the proximal femur of adults with type 2 diabetes. Bone 2018, 114, 32–39. [Google Scholar] [CrossRef] [PubMed]
  102. Wölfel, E.M.; Jähn-Rickert, K.; Schmidt, F.N.; Wulff, B.; Mushumba, H.; Sroga, G.E.; Püschel, K.; Milovanovic, P.; Amling, M.; Campbell, G.M.; et al. Individuals with type 2 diabetes mellitus show dimorphic and heterogeneous patterns of loss in femoral bone quality. Bone 2020, 140, 115556. [Google Scholar] [CrossRef] [PubMed]
  103. Sell, D.R.; Monnier, V.M. Structure elucidation of a senescence cross-link from human extracellular matrix. Implication of pen-toses in the aging process. J. Biol. Chem. 1989, 264, 21597–21602. [Google Scholar] [CrossRef]
  104. Sugiyama, S.; Miyata, T.; Ueda, Y.; Tanaka, H.; Maeda, K.; Kawashima, S.; Strihou, C.V.Y.D.; Kurokawa, K. Plasma levels of pentosidine in diabetic patients: An advanced glycation end product. J. Am. Soc. Nephrol. 1998, 9, 1681–1688. [Google Scholar] [CrossRef] [PubMed]
  105. Yoshida, N.; Okumura, K.-I.; Aso, Y. High serum pentosidine concentrations are associated with increased arterial stiffness and thickness in patients with type 2 diabetes. Metabolism 2005, 54, 345–350. [Google Scholar] [CrossRef] [PubMed]
  106. Kerkeni, M.; Saïdi, A.; Bouzidi, H.; Letaief, A.; Ben Yahia, S.; Hammami, M. Pentosidine as a biomarker for microvascular complications in type 2 diabetic patients. Diabetes Vasc. Dis. Res. 2013, 10, 239–245. [Google Scholar] [CrossRef] [PubMed]
  107. Yamamoto, M.; Sugimoto, T. Advanced Glycation End Products, Diabetes, and Bone Strength. Curr. Osteoporos. Rep. 2016, 14, 320–326. [Google Scholar] [CrossRef] [Green Version]
  108. Viguet-Carrin, S.; Roux, J.P.; Arlot, M.E.; Merabet, Z.; Leeming, D.; Byrjalsen, I.; Delmas, P.D.; Bouxsein, M.L. Contribution of the advanced glycation end product pentosidine and of maturation of type I collagen to compressive biomechanical properties of human lumbar vertebrae. Bone 2006, 39, 1073–1079. [Google Scholar] [CrossRef]
  109. Valcourt, U.; Merle, B.; Gineyts, E.; Viguet-Carrin, S.; Delmas, P.D.; Garnero, P. Non-enzymatic Glycation of Bone Collagen Modifies Osteoclastic Activity and Differentiation. J. Biol. Chem. 2007, 282, 5691–5703. [Google Scholar] [CrossRef] [Green Version]
  110. Yamamoto, M.; Yamaguchi, T.; Yamauchi, M.; Yano, S.; Sugimoto, T. Serum Pentosidine Levels Are Positively Associated with the Presence of Vertebral Fractures in Postmenopausal Women with Type 2 Diabetes. J. Clin. Endocrinol. Metab. 2008, 93, 1013–1019. [Google Scholar] [CrossRef] [Green Version]
  111. Choi, Y.J.; Ock, S.Y.; Jin, Y.; Lee, J.S.; Kim, S.H.; Chung, Y.-S. Urinary Pentosidine levels negatively associates with trabecular bone scores in patients with type 2 diabetes mellitus. Osteoporos. Int. 2018, 29, 907–915. [Google Scholar] [CrossRef] [PubMed]
  112. Schwartz, A.V.; Garnero, P.; Hillier, T.A.; Sellmeyer, D.E.; Strotmeyer, E.S.; Feingold, K.R.; Resnick, H.E.; Tylavsky, F.A.; Black, D.M.; Cummings, S.R.; et al. Pentosidine and Increased Fracture Risk in Older Adults with Type 2 Diabetes. J. Clin. Endocrinol. Metab. 2009, 94, 2380–2386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Barzilay, J.I.; Bůžková, P.; Zieman, S.J.; Kizer, J.R.; Djoussé, L.; Ix, J.H.; Tracy, R.P.; Siscovick, D.S.; Cauley, J.A.; Mukamal, K.J. Circulating Levels of Carboxy-Methyl-Lysine (CML) Are Associated with Hip Fracture Risk: The Cardiovascular Health Study. J. Bone Miner. Res. 2014, 29, 1061–1066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Dhaliwal, R.; Ewing, S.K.; Vashishth, D.; Semba, R.D.; Schwartz, A.V. Greater Carboxy-Methyl-Lysine Is Associated with Increased Fracture Risk in Type 2 Diabetes. J. Bone Miner. Res. 2022, 37, 265–272. [Google Scholar] [CrossRef] [PubMed]
  115. Delgado-Calle, J.; Sato, A.Y.; Bellido, T. Role and mechanism of action of sclerostin in bone. Bone 2017, 96, 29–37. [Google Scholar] [CrossRef] [Green Version]
  116. Ardawi, M.-S.M.; Rouzi, A.A.; Al-Sibiani, S.A.; Al-Senani, N.S.; Qari, M.H.; Mousa, S.A. High serum sclerostin predicts the occurrence of osteoporotic fractures in postmenopausal women: The center of excellence for osteoporosis research study. J. Bone Miner. Res. 2012, 27, 2592–2602. [Google Scholar] [CrossRef]
  117. Arasu, A.; Cawthon, P.M.; Lui, L.-Y.; Do, T.P.; Arora, P.S.; Cauley, J.A.; Ensrud, K.E.; Cummings, S.R. The Study of Osteoporotic Fractures Research Group Serum Sclerostin and Risk of Hip Fracture in Older Caucasian Women. J. Clin. Endocrinol. Metab. 2012, 97, 2027–2032. [Google Scholar] [CrossRef] [Green Version]
  118. García-Martín, A.; Rozas-Moreno, P.; Reyes-Garcia, R.; Morales-Santana, S.; García-Fontana, B.; Garcia-Salcedo, J.A.; Muñoz-Torres, M. Circulating Levels of Sclerostin Are Increased in Patients with Type 2 Diabetes Mellitus. J. Clin. Endocrinol. Metab. 2012, 97, 234–241. [Google Scholar] [CrossRef] [Green Version]
  119. Heilmeier, U.; Carpenter, D.R.; Patsch, J.M.; Harnish, R.; Joseph, G.B.; Burghardt, A.J.; Baum, T.; Schwartz, A.V.; Lang, T.F.; Link, T.M. Volumetric femoral BMD, bone geometry, and serum sclerostin levels differ between type 2 diabetic postmenopausal women with and without fragility fractures. Osteoporos. Int. 2015, 26, 1283–1293. [Google Scholar] [CrossRef] [Green Version]
  120. Ardawi, M.-S.M.; Akhbar, D.H.; AlShaikh, A.; Ahmed, M.M.; Qari, M.H.; Rouzi, A.A.; Ali, A.Y.; Abdulrafee, A.A.; Saeda, M.Y. Increased serum sclerostin and decreased serum IGF-1 are associated with vertebral fractures among postmenopausal women with type-2 diabetes. Bone 2013, 56, 355–362. [Google Scholar] [CrossRef]
  121. Yamamoto, M.; Yamauchi, M.; Sugimoto, T. Elevated Sclerostin Levels Are Associated with Vertebral Fractures in Patients with Type 2 Diabetes Mellitus. J. Clin. Endocrinol. Metab. 2013, 98, 4030–4037. [Google Scholar] [CrossRef] [PubMed]
  122. Hensley, A.P.; McAlinden, A. The role of microRNAs in bone development. Bone 2021, 143, 115760. [Google Scholar] [CrossRef] [PubMed]
  123. Heilmeier, U.; Hackl, M.; Schroeder, F.; Torabi, S.; Kapoor, P.; Vierlinger, K.; Eiriksdottir, G.; Gudmundsson, E.F.; Harris, T.B.; Gudnason, V.; et al. Circulating serum microRNAs including senescent miR-31-5p are associated with incident fragility fractures in older postmenopausal women with type 2 diabetes mellitus. Bone 2022, 158, 116308. [Google Scholar] [CrossRef] [PubMed]
  124. Heilmeier, U.; Hackl, M.; Skalicky, S.; Weilner, S.; Schroeder, F.; Vierlinger, K.; Patsch, J.M.; Baum, T.; Oberbauer, E.; Lobach, I.; et al. Serum miRNA Signatures Are Indicative of Skeletal Fractures in Postmenopausal Women with and without Type 2 Diabetes and Influence Osteogenic and Adipogenic Differentiation of Adipose Tissue-Derived Mesenchymal Stem Cells In Vitro. J. Bone Miner. Res. 2016, 31, 2173–2192. [Google Scholar] [CrossRef] [Green Version]
  125. Chen, Y.-S.; Kang, X.-R.; Zhou, Z.-H.; Yang, J.; Xin, Q.; Ying, C.-T.; Zhang, Y.-P.; Tao, J. MiR-1908/EXO1 and MiR-203a/FOS, regulated by scd1, are associated with fracture risk and bone health in postmenopausal diabetic women. Aging 2020, 12, 9549–9584. [Google Scholar] [CrossRef]
  126. Mulder, D.J.; Van De Water, T.; Lutgers, H.L.; Graaff, R.; Gans, R.O.; Zijlstra, F.; Smit, A.J. Skin Autofluorescence, a Novel Marker for Glycemic and Oxidative Stress-Derived Advanced Glycation Endproducts: An Overview of Current Clinical Studies, Evidence, and Limitations. Diabetes Technol. Ther. 2006, 8, 523–535. [Google Scholar] [CrossRef] [Green Version]
  127. Samakkarnthai, P.; Sfeir, J.G.; Atkinson, E.J.; Achenbach, S.J.; Wennberg, P.W.; Dyck, P.J.; Tweed, A.J.; Volkman, T.L.; Amin, S.; Farr, J.N.; et al. Determinants of Bone Material Strength and Cortical Porosity in Patients with Type 2 Diabetes Mellitus. J. Clin. Endocrinol. Metab. 2020, 105, e3718–e3729. [Google Scholar] [CrossRef]
  128. Waqas, K.; Chen, J.; Koromani, F.; Trajanoska, K.; Van Der Eerden, B.C.J.; Uitterlinden, A.G.; Rivadeneira, F.; Zillikens, M.C. Skin Autofluorescence, a Noninvasive Biomarker for Advanced Glycation End-Products, Is Associated with Prevalent Vertebral and Major Osteoporotic Fractures: The Rotterdam Study. J. Bone Miner. Res. 2020, 35, 1904–1913. [Google Scholar] [CrossRef]
  129. Sihota, P.; Pal, R.; Yadav, R.N.; Neradi, D.; Karn, S.; Goni, V.G.; Sharma, S.; Mehandia, V.; Bhadada, S.K.; Kumar, N.; et al. Can fingernail quality predict bone damage in Type 2 diabetes mellitus? A pilot study. PLoS ONE 2021, 16, e0257955. [Google Scholar] [CrossRef]
  130. Sihota, P.; Yadav, R.N.; Dhiman, V.; Bhadada, S.K.; Mehandia, V.; Kumar, N. Investigation of diabetic patient’s fingernail quality to monitor type 2 diabetes induced tissue damage. Sci. Rep. 2019, 9, 3193. [Google Scholar] [CrossRef]
Figure 1. Determinants of bone fragility and increased fracture risk in type 2 diabetes. AGEs, advanced glycation end products.
Figure 1. Determinants of bone fragility and increased fracture risk in type 2 diabetes. AGEs, advanced glycation end products.
Jcm 11 02206 g001
Figure 2. Trabecular bone score (TBS) as a useful technology for the assessment of the trabecular microarchitecture. TBS > 1.31 (left) denotes a normal microarchitecture, whereas TBS < 1.23 (right) indicates an altered microarchitecture. TBS can detect differences between similar values of lumbar spine bone mineral density (BMD) estimated by dual-energy X-ray absorptiometry (DXA) (g/cm2).
Figure 2. Trabecular bone score (TBS) as a useful technology for the assessment of the trabecular microarchitecture. TBS > 1.31 (left) denotes a normal microarchitecture, whereas TBS < 1.23 (right) indicates an altered microarchitecture. TBS can detect differences between similar values of lumbar spine bone mineral density (BMD) estimated by dual-energy X-ray absorptiometry (DXA) (g/cm2).
Jcm 11 02206 g002
Table 1. Clinical studies showing an independent association between the trabecular bone score (TBS) and osteoporotic fractures in patients with type 2 diabetes mellitus.
Table 1. Clinical studies showing an independent association between the trabecular bone score (TBS) and osteoporotic fractures in patients with type 2 diabetes mellitus.
StudyDesignStudy PopulationResults
Leslie et al., 2013 [75]Retrospective cohort (mean follow-up 4.7 years)29,407 women ≥ 50 years (2356 with diagnosed T2DM)TBS predicted major osteoporotic fractures (hip, spine, forearm and humerus) in T2DM (HR 1.27, CI 1.10–1.46)
Zhukouskaya et al., 2015 [76]Cross-sectional99 postmenopausal women with T2DM/107 healthy controlsTBS was associated with VF (AUC 0.69, cut-off value 1.130 in ROC curve analysis)
Yamamoto et al., 2019 [77]Cross-sectional584 patients with T2DM (257 postmenopausal women and 291 men > 50 years)TBS correlated with prevalent VF in multivariate logistic regression analysis
Lin et al., 2019 [78]Cross-sectional285 postmenopausal women with T2DMTBS had the strongest association with VF (AUC 0.775)
T2DM, type 2 diabetes mellitus; TBS, trabecular bone score; VF, vertebral fractures; HR, hazard ratio; CI, confidence interval; ROC, receiver operating characteristic; AUC, area under the curve.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Martínez-Montoro, J.I.; García-Fontana, B.; García-Fontana, C.; Muñoz-Torres, M. Evaluation of Quality and Bone Microstructure Alterations in Patients with Type 2 Diabetes: A Narrative Review. J. Clin. Med. 2022, 11, 2206. https://doi.org/10.3390/jcm11082206

AMA Style

Martínez-Montoro JI, García-Fontana B, García-Fontana C, Muñoz-Torres M. Evaluation of Quality and Bone Microstructure Alterations in Patients with Type 2 Diabetes: A Narrative Review. Journal of Clinical Medicine. 2022; 11(8):2206. https://doi.org/10.3390/jcm11082206

Chicago/Turabian Style

Martínez-Montoro, José Ignacio, Beatriz García-Fontana, Cristina García-Fontana, and Manuel Muñoz-Torres. 2022. "Evaluation of Quality and Bone Microstructure Alterations in Patients with Type 2 Diabetes: A Narrative Review" Journal of Clinical Medicine 11, no. 8: 2206. https://doi.org/10.3390/jcm11082206

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop