Editorial Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Diabetes. Jun 10, 2015; 6(5): 673-678
Published online Jun 10, 2015. doi: 10.4239/wjd.v6.i5.673
Update on type 2 diabetes-related osteoporosis
Kannikar Wongdee, Narattaphol Charoenphandhu, Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok 10400, Thailand
Kannikar Wongdee, Office of Academic Management, Faculty of Allied Health Sciences, Burapha University, Chonburi 20131, Thailand
Narattaphol Charoenphandhu, Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
Author contributions: Wongdee K and Charoenphandhu N contributed equally for literature review, data analysis and preparation of the manuscript.
Supported by Grants from the Cluster and Program Management Office (CPMO), National Science and Technology Development Agency (P-11-00639); the Thailand Research Fund (TRF)-Mahidol University through the TRF Senior Research Scholar Grant (RTA5780001 to NC); the Faculty of Allied Health Sciences, Burapha University and Thailand Research Fund through TRF Research Career Development Grant (RSA5780041 to KW); the Research and Development Fund Burapha University (05/2557 to KW); the Faculty of Allied Health Sciences, Burapha University Research Grant of Fiscal Year 2015 (AHS05/2558 to KW).
Conflict-of-interest: Kannikar Wongdee and Narattaphol Charoenphandhu declare no conflicts of interest.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Narattaphol Charoenphandhu, MD, PhD, Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Bangkok 10400, Thailand. naratt@narattsys.com
Telephone: +66-2-3547154 Fax: +66-2-3547154
Received: January 11, 2015
Peer-review started: January 15, 2015
First decision: February 7, 2015
Revised: February 12, 2015
Accepted: March 5, 2015
Article in press: March 9, 2015
Published online: June 10, 2015

Abstract

It was previously understood that body weight gain and obesity observed in type 2 diabetes mellitus (T2DM) could be beneficial since body weight increase elevated bone mineral density and thus helped maintain the skeletal framework. However, a number of recent findings in humans and rodents have revealed that T2DM is not only associated with trabecular defects but also increases cortical porosity, and compromised bone cell function and bone mechanical properties. Hyperglycemia and insulin resistance in T2DM may further induce osteoblast apoptosis and uncoupling bone turnover. Prolonged accumulation of advanced glycation end products and diminished activity of lysyl oxidase, an essential enzyme for collagen cross-link, can lead to structural abnormalities of bone collagen fibrils, brittle matrix, and fragility fractures. Our studies in T2DM rats showed that dyslipidemia, which often occurs in T2DM, could obscure the T2DM-associated changes in bone microstructure and osteopenia. Longitudinal bone growth regulated by the growth plate chondrocytes is also impaired by T2DM since differentiation of growth plate chondrocytes is arrested and retained in the resting state while only a small number of cells undergo hypertrophic differentiation. Such a delayed chondrocyte differentiation may have also resulted from premature apoptosis of the growth plate chondrocytes. Nevertheless, the underlying cellular and molecular mechanisms of insulin resistance in osteoblasts, osteoclasts, osteocytes, and growth plate chondrocytes remain to be investigated.

Key Words: Advanced glycation end products, Chondrocyte apoptosis, Collagen, Dyslipidemia, Fracture, Growth plate, Type 2 diabetes mellitus, Osteoporosis

Core tip: Type 2 diabetes mellitus (T2DM) negatively affects bone density and strength by inducing cellular and extracellular matrix failures. Insulin resistance in T2DM deteriorates osteoblast proliferation and activity, but enhances osteoclast activity, leading to uncoupled bone remodeling. Hyperglycemia also aggravates osteoblast dysfunction, thus contributing to cellular failure. Extracellular matrix failure is caused by abnormal collagen synthesis and aberrant collagen structure and alignment, the latter of which results, in part, from advanced glycation end products (AGEs). With hyperglycemia and AGEs, impaired bone strength may occur despite high bone mineral density. It is, therefore, concluded that T2DM can be considered a cause of osteoporosis and/or poor bone mechanical properties.



INTRODUCTION

Although type 1 diabetes mellitus (T1DM) is known to compromise bone microstructure[1,2], how type 2 diabetes mellitus (T2DM) affects bone metabolism has long been debate for decades. It was previously believed that T2DM could be protective against osteoporosis since a number of clinical studies and meta-analyses revealed an increase in bone mineral density (BMD) in T2DM patients[1,2]. However, several recent lines of evidence in both humans and rodents have corroborated that T2DM is indeed detrimental to bone, leading to impaired osteoblast-mediated bone formation, accelerated bone resorption, microstructural defect, and poor bone quality. The previous controversial data stem from the use of low-resolution X-ray-based techniques, including measurement of areal BMD by dual energy X-ray absorptiometry (DXA), rather than evaluation of high-resolution bone microstructure or bone mechanical properties. This article has updated the recent findings of T2DM-related osteopenia and osteoporosis, as summarized in Table 1.

Table 1 Recent studies on the type 2 diabetes mellitus-related osteoporosis in humans (2010-2014).
Site of boneTechniqueSubjectsMain findings in T2DM groupRef.
Distal radiusHR-pQCT60 postmenopausal womenLow cortical thickness and low trabecular number[3]
Distal tibiaHR-pQCTLower cortical thickness
Mid-shaft tibiaMicroindentationPoor bone quality (low BMS)
Distal radiusDXA, HR-pQCT80 postmenopausal women with or without fracturesHigher cortical porosity in T2DM with fractures[4]
Ultradistal radiusDXA, HR-pQCTHigher cortical porosity in T2DM with fractures
Distal tibiaDXA, HR-pQCTLower cortical porosity in T2DM with fractures
Ultradistal tibiaDXA, HR-pQCTLower BMD and cortical porosity in T2DM with fractures
Distal radiusMRI60 postmenopausal womenGreater trabecular network holes[5]
Distal radiusHR-pQCT38 postmenopausal womenHigher cortical porosity[6]
TibiaHR-pQCTHigher volumetric BMD and trabecular thickness
Mid-shaft radiuspQCT1171 men (≥ 65 yr)Lower bone bending strength[7]
Mid-shaft tibiapQCTLower bone bending strength
T2DM INCREASES BONE POROSITY AND DECREASES MECHANICAL BONE STRENGTH

Previous investigations in human mostly focus on trabecular bone changes in T2DM, but recent investigations have moved to the study of cortical changes and mechanical properties. By using more advanced techniques, such as high-resolution 3-dimensional computed tomography and microindentation, it was found that T2DM negatively affected bone strength despite the presence of relatively high BMD. Several cross-sectional studies in T2DM patients using high-resolution peripheral quantitative computed tomography (HR-pQCT) and magnetic resonance imaging (MRI) consistently revealed quality defects in both cortical and trabecular networks that would increase fracture risk[3-6]. For instance, Farr et al[3] by assessing bone quality with HR-pQCT in 30 postmenopausal T2DM patients at distal radius and distal tibia, found lower cortical thickness in T2DM was lower than normal non-diabetic controls, while bone microindentation testing showed lower bone material strength (BMS) in T2DM patients. Moreover, the radius quality evaluated by MRI, showed trabecular network holes being approximately 10% larger in postmenopausal T2DM patients than normal controls[5]. The cortical part was similarly affected by T2DM[4]. Patsch et al[4] investigated changes in bone microarchitecture in postmenopausal T2DM patients with or without fractures at radius and tibia by using DXA and HR-pQCT. Interestingly, they found that T2DM patients with fractures had higher pore-related deficits, i.e., greater cortical pore volume, cortical porosity, and endocortical bone surface, than diabetic patients without fractures[4], consistent with the previous report in the radii of T2DM patients that having greater cortical pore volume (approximately 150%) and cortical porosity (approximately 125%) than normal individuals[6]. These cortical defects were often accompanied by impaired mechanical properties, such as increased failure load and low bone bending strength, that led to reduction in overall bone strength and increase in fracture risk[4,7].

DYSLIPIDEMIA MIGHT OBSCURE T2DM-INDUCED OSTEOPOROSIS

Previously it was believed that greater body weight or obesity associated with T2DM could be beneficial to the skeletal system through increasing BMD and bone mass[7,8]. However, our group recently reported the possible masking effects of dyslipidemia on diabetic bone in rats[9]. In our study, we determined the effects of dyslipidemia on bone microstructure were determined in Goto-Kakizaki (GK) diabetic rats treated with high cholesterol diet compared those fed with normal diet. The GK rats-a non-obese T2DM rat model without obesity-induced bone gain-were found to manifest stable fasting hyperglycemia and insulin resistance, while cholesterol-fed GK rats exhibited hypercholesterolemia, hypertriglyceridemia and hyperglycemia without significant weight gain[10]. Bone histomorphometry revealed that GK rats with T2DM manifested several signs of suppressed osteoblast function, such as decreases in osteoblast surface and bone formation rate, whereas the osteoclast-mediated bone resorption was markedly enhanced. It was noted that, these microstructural changes disappeared after 16-wk of high cholesterol consumption, suggesting that high cholesterol diet and perhaps the resultant dyslipidemia could obscure the T2DM-associated osteopenia and changes in bone microstructural defect[9]. Thus, the difficulty in detecting bone deterioration in T2DM rats with dyslipidemia could explain, in part, why osteopenia was not observed in some T2DM studies.

T2DM AND LONGITUDINAL BONE GROWTH

Up until now, few studies have investigated relationship between T2DM and longitudinal bone growth. Generally, longitudinal bone growth is controlled by proliferation and differentiation of chondrocytes in the growth plate, which is histologically divided into 3 zones, i.e., resting zone (RZ), proliferative zone (PZ) and hypertrophic zone (HZ). The RZ consists of low mitotic activity stem-like cells that gradually migrate to the PZ where chondrocytes proliferate and align into vertical columns and eventually reach the mature state in HZ. Thereafter, the hypertrophic chondrocytes in HZ undergo apoptosis and are replaced by capillaries and osteoblasts, which later use cartilaginous scaffold as a template for bone formation and bone elongation[11,12]. Since several investigations reported reduced bone length in diabetic rats compared with normal rats[9,13], T2DM may be a cause of aberrant growth plate function. Lapmanee et al[9] examined changes in the growth plate of diabetic GK rats and found impairment of chondrocyte differentiation as indicated by increased RZ height and decreased HZ height. It was possible that differentiation of chondrocyte precursors in T2DM rats were arrested and cells remained in the resting state, with only a small number of proliferating cells undergoing differentiation into hypertrophic chondrocytes[9].

Aiemlapa et al[14] further demonstrated the underlying mechanism of delayed growth plate chondrocyte differentiation. By using terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay for apoptosis, they found premature apoptosis of chondrocytes in the HZ and chondro-osseous junction of GK rats. The massive loss of growth plate chondrocytes was accompanied by an increase in serum IGF-1 level, and overexpression of parathyroid hormone related protein (PTHrP), runt-related transcription factor (Runx2) and vascular endothelial growth factor (VEGF) in the growth plate, all of which might have been compensatory responses to mitigate excessive loss of chondrocytes due to premature apoptosis[14]. However, it was possible that the effects of T2DM on the growth plate might be dependent on animal strain and model of DM induction. For instance, Wu et al[15] found acceleration of longitudinal bone growth as indicated by bone elongation and increased heights of PZ and HZ in insulin resistant mice induced by high fat diet. In this model, insulin might remain to have a stimulatory effect on bone growth in vivo similar to its reported stimulatory effect on metatarsal linear growth in vitro[15]. Furthermore, high fat diet-induced dyslipidemia could complicate the matter since 7α-hydroxycholesterol and oxidized low-density lipoprotein (LDL) have been shown to modulate osteoblast and osteoclast functions, which, in turn, could have effects on bone elongation[16,17].

POSSIBLE CELLULAR MECHANISMS OF T2DM-RELATED FRAGILITY FRACTURES

The pathogenesis of T2DM-related fragility fracture can be looked upon from 2 aspects, i.e., cellular failure and extracellular matrix failure. At cellular level, T2DM was associated with diminished activities of osteoblasts, osteoclasts and osteocytes, and increased apoptosis of bone cells[9,18-21]. A decrease in osteocyte density (number of osteocyte-occupied lacunae per unit area) was conspicuously observed in streptozotocin-induced diabetic rats[21]. Hyperglycemia-induced insulin resistance is another important factor that cause both osteoblast and osteoclast malfunctions[22]. Since insulin is a suppressor of osteoclast-mediated bone resorption[23], T2DM-associated insulin resistance could enhance bone resorption. Moreover, high plasma glucose concentration can induce glucotoxicity in cells including osteoblasts, leading to osteoblast apoptosis[24]. In vivo experiment in transgenic liver-specific S503A CEACAM1 mutant (L-SACC1) mice, a model of impaired insulin clearance in the liver causing hyperinsulinemia and insulin resistance, suggested that the abnormally high bone mass in these mice might have resulted from low bone turnover as indicated by decreases in double-labeled surface (as determined by bone histomorphometry) and TRAP-positive osteoclasts, which represent activities of osteoblast-mediated bone formation and osteoclast-mediated bone resorption, respectively[22]. In other words, insulin resistance in this model was associated with a slowdown in bone turnover, which could eventually result in inadequate healing of microcracks, poor bone quality and increased fracture risk[22]. In addition, the experiment in high fat diet-fed Zucker diabetic fatty (ZDF) rats also showed impaired osteoblast function as indicated by downregulation of the expression of osteoblast-specific genes, e.g., bone morphogenetic protein-2 (BMP-2), Runx2, osteocalcin and osteopontin. Suppression of osteoblastogenesis in these ZDF rats possibly compromised bone regeneration capacity. Subcritical bone defect regeneration study further showed that nondiabetic rats filled the defect by 57%, whereas diabetic rats could fill only 21% of bone defect in 12 wk[18].

T2DM not only caused deterioration of bone cell functions (cellular failure), but it also damaged bone extracellular matrix. Most studies suggested that T2DM caused abnormality in the structure of collagen, which is the most abundant protein in organic bone matrix. García-Hernández et al[25] reported that high glucose concentration indeed increased biomineralization in human alveolar bone-derived osteoblasts, but the mineral quality was lower than that in low glucose-exposed group. Determination of mineral quality in term of calcium/phosphate (Ca/Pi) ratio in the mineralized extracellular matrix nodules by energy-dispersive X-ray microanalysis (EDX) showed that high concentration of glucose significantly decreased Ca/Pi ratio on day 7 and 14 of treatment. Hammond et al[26] further studied nanoscale morphology of type I collagen in tibiae of ZDF rats by Raman spectroscopy and reference point indentation (RPI), the latter of which applied a force to determine bone mechanical properties by measuring the relative displacement reference position[26,27]. RPI analysis revealed that bone matrix of ZDF diabetic rats was more resistant to plastic deformation, which might have resulted from abnormal formation of non-enzymatic collagen cross-link, toughening of the matrix, or the presence of advanced glycation end products (AGEs).

AGEs are non-enzymatic carbohydrate modifications of extracellular and intracellular proteins accumulated in long-lived tissues, such as skin and bone, and are often present in the plasma proteins of patients with DM and renal failure[28,29]. A number of investigations have revealed that AGEs are considered a factor that provokes fragility fractures in T2DM by inducing abnormal arrangement of collagen[26,28,30]. By using scanning electron microscope (SEM) and transmission electron microscope (TEM), Aoki et al[28] provided evidence that the rats subjected to adenine-induced renal failure exhibited AGEs accumulation and suppression of osteoblast function, similar to that observed in T2DM. SEM showed irregularity in collagen fibril alignment, while TEM revealed a wider diameter of collagen fibril in adenine-treated rats with renal osteodystrophy[28]. Immunohistochemistry also showed greater accumulation of AGEs in peritrabecular osteoblasts of adenine-treated rats than control rats. Further in vitro study in AGEs-treated MC3T3-E1 osteoblast-like cells showed a decrease in protein expression of secreted phosphoprotein 1 and lysyl oxidase, a mature osteoblast marker and essential enzyme for collagen cross-link, respectively. It was thus suggested that suppressed osteoblast differentiation and decreased lysyl oxidase production caused structural abnormalities of bone collagen fibrils leading to bone fragility[28].

Collagen is the most abundant protein in bone organic matrix, and it undergoes intra- and extracellular post-translational modifications[31]. To stabilize collagen fibrils, lysyl oxidase catalyzes intra- and intermolecular cross-link between collagen molecules essential for bone strength[31]. It was reported that glycation of collagen caused abnormal arrangement of collagen leading to brittle matrix and fragile bone[26,28,30], but little is known whether a decrease in lysyl oxidase-dependent collagen cross-link contributes to diabetic bone fragility and osteoporosis. The underlying mechanism of AGEs-attenuated lysyl oxidase activity was explored in mouse and rat primary osteoblasts and it was found that the carboxymethylated collagen, a form of AGEs, was not able to promote lysyl oxidase-mediated cross-linking due to failure of binding between abnormal collagen and discoidin domain receptor-2[30].

CONCLUSION

Currently, it can be concluded that T2DM compromises bone microstructure by inducing aberrant bone cell function (cellular failure) and abnormal matrix structure (matrix failure). Regarding the cellular effect, T2DM is associated with increased osteoblast apoptosis, diminished osteoblast differentiation, and enhanced osteoclast-mediated bone resorption, which, in part, resulted from hyperglycemia and insulin resistance. Prolonged accumulation of AGEs coexisting with a decrease in lysyl oxidase activity causes abnormal structure and alignment of collagen, leading to bone fragility. Several confounding factors in T2DM, particularly body weight gain, obesity, and dyslipidemia, are able to mask the detrimental effects of T2DM, and may delay diagnosis of diabetic osteoporosis. In other words, bone is already damaged in T2DM despite a relatively high BMD. Although deleterious effects of T2DM on bone have been elucidated, the underlying cellular and molecular mechanisms remain unclear. For example, how does insulin resistance occur in osteoblasts and how do phosphorylation of insulin-receptor substrate isoforms (IRSs) and resultant insulin resistance in osteoblasts, osteoclasts and perhaps osteocytes contribute to diabetic bone loss? Indeed, osteocytes residing inside lacunae play an important role in bone remodeling in health and disease since they are responsible for inducing bone loss under certain conditions, such as during lactation[32,33]. Further investigation is required to demonstrate whether osteocytic dysfunction does exist in T2DM.

ACKNOWLEDGMENTS

The authors thank Professor Nateetip Krishnamra for valuable comments on the manuscript.

Footnotes

P- Reviewer: Grieco A, Hiwatashi A S- Editor: Ji FF L- Editor: A E- Editor: Zhang DN

References
1.  Wongdee K, Charoenphandhu N. Osteoporosis in diabetes mellitus: Possible cellular and molecular mechanisms. World J Diabetes. 2011;2:41-48.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Hofbauer LC, Brueck CC, Singh SK, Dobnig H. Osteoporosis in patients with diabetes mellitus. J Bone Miner Res. 2007;22:1317-1328.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Farr JN, Drake MT, Amin S, Melton LJ, McCready LK, Khosla S. In vivo assessment of bone quality in postmenopausal women with type 2 diabetes. J Bone Miner Res. 2014;29:787-795.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Patsch JM, Burghardt AJ, Yap SP, Baum T, Schwartz AV, Joseph GB, Link TM. Increased cortical porosity in type 2 diabetic postmenopausal women with fragility fractures. J Bone Miner Res. 2013;28:313-324.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Pritchard JM, Giangregorio LM, Atkinson SA, Beattie KA, Inglis D, Ioannidis G, Punthakee Z, Adachi JD, Papaioannou A. Association of larger holes in the trabecular bone at the distal radius in postmenopausal women with type 2 diabetes mellitus compared to controls. Arthritis Care Res (Hoboken). 2012;64:83-91.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Burghardt AJ, Issever AS, Schwartz AV, Davis KA, Masharani U, Majumdar S, Link TM. 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.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Petit MA, Paudel ML, Taylor BC, Hughes JM, Strotmeyer ES, Schwartz AV, Cauley JA, Zmuda JM, Hoffman AR, Ensrud KE. Bone mass and strength in older men with type 2 diabetes: the Osteoporotic Fractures in Men Study. J Bone Miner Res. 2010;25:285-291.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Yamaguchi T, Kanazawa I, Yamamoto M, Kurioka S, Yamauchi M, Yano S, Sugimoto T. Associations between components of the metabolic syndrome versus bone mineral density and vertebral fractures in patients with type 2 diabetes. Bone. 2009;45:174-179.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Lapmanee S, Charoenphandhu N, Aeimlapa R, Suntornsaratoon P, Wongdee K, Tiyasatkulkovit W, Kengkoom K, Chaimongkolnukul K, Seriwatanachai D, Krishnamra N. High dietary cholesterol masks type 2 diabetes-induced osteopenia and changes in bone microstructure in rats. Lipids. 2014;49:975-986.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Kengkoom K, Klinkhamhom A, Sirimontaporn A, Singha O, Ketjareon T, Panavechkijkul Y, Seriwatanachai D, Ukong S, Ampawong S. Effects on high cholesterol-fed to liver, retina, hippocampus, and Harderian gland in Goto-Kakizaki rat. Int J Clin Exp Pathol. 2013;6:639-649.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Wongdee K, Krishnamra N, Charoenphandhu N. Endochondral bone growth, bone calcium accretion, and bone mineral density: how are they related? J Physiol Sci. 2012;62:299-307.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Mackie EJ, Ahmed YA, Tatarczuch L, Chen KS, Mirams M. Endochondral ossification: how cartilage is converted into bone in the developing skeleton. Int J Biochem Cell Biol. 2008;40:46-62.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Zhang L, Liu Y, Wang D, Zhao X, Qiu Z, Ji H, Rong H. Bone biomechanical and histomorphometrical investment in type 2 diabetic Goto-Kakizaki rats. Acta Diabetol. 2009;46:119-126.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Aeimlapa R, Wongdee K, Charoenphandhu N, Suntornsaratoon P, Krishnamra N. Premature chondrocyte apoptosis and compensatory upregulation of chondroregulatory protein expression in the growth plate of Goto-Kakizaki diabetic rats. Biochem Biophys Res Commun. 2014;452:395-401.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Wu S, Aguilar AL, Ostrow V, De Luca F. Insulin resistance secondary to a high-fat diet stimulates longitudinal bone growth and growth plate chondrogenesis in mice. Endocrinology. 2011;152:468-475.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Hamel P, Abed E, Brissette L, Moreau R. Characterization of oxidized low-density lipoprotein-induced hormesis-like effects in osteoblastic cells. Am J Physiol Cell Physiol. 2008;294:C1021-C1033.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Luegmayr E, Glantschnig H, Wesolowski GA, Gentile MA, Fisher JE, Rodan GA, Reszka AA. Osteoclast formation, survival and morphology are highly dependent on exogenous cholesterol/lipoproteins. Cell Death Differ. 2004;11 Suppl 1:S108-S118.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Hamann C, Goettsch C, Mettelsiefen J, Henkenjohann V, Rauner M, Hempel U, Bernhardt R, Fratzl-Zelman N, Roschger P, Rammelt S. Delayed bone regeneration and low bone mass in a rat model of insulin-resistant type 2 diabetes mellitus is due to impaired osteoblast function. Am J Physiol Endocrinol Metab. 2011;301:E1220-E1228.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Kawashima Y, Fritton JC, Yakar S, Epstein S, Schaffler MB, Jepsen KJ, LeRoith D. Type 2 diabetic mice demonstrate slender long bones with increased fragility secondary to increased osteoclastogenesis. Bone. 2009;44:648-655.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Weinberg E, Maymon T, Weinreb M. AGEs induce caspase-mediated apoptosis of rat BMSCs via TNFα production and oxidative stress. J Mol Endocrinol. 2014;52:67-76.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Villarino ME, Sánchez LM, Bozal CB, Ubios AM. Influence of short-term diabetes on osteocytic lacunae of alveolar bone. A histomorphometric study. Acta Odontol Latinoam. 2006;19:23-28.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Huang S, Kaw M, Harris MT, Ebraheim N, McInerney MF, Najjar SM, Lecka-Czernik B. Decreased osteoclastogenesis and high bone mass in mice with impaired insulin clearance due to liver-specific inactivation to CEACAM1. Bone. 2010;46:1138-1145.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Thomas DM, Udagawa N, Hards DK, Quinn JM, Moseley JM, Findlay DM, Best JD. Insulin receptor expression in primary and cultured osteoclast-like cells. Bone. 1998;23:181-186.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Zhen D, Chen Y, Tang X. Metformin reverses the deleterious effects of high glucose on osteoblast function. J Diabetes Complications. 2010;24:334-344.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  García-Hernández A, Arzate H, Gil-Chavarría I, Rojo R, Moreno-Fierros L. High glucose concentrations alter the biomineralization process in human osteoblastic cells. Bone. 2012;50:276-288.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Hammond MA, Gallant MA, Burr DB, Wallace JM. Nanoscale changes in collagen are reflected in physical and mechanical properties of bone at the microscale in diabetic rats. Bone. 2014;60:26-32.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Gallant MA, Brown DM, Organ JM, Allen MR, Burr DB. Reference-point indentation correlates with bone toughness assessed using whole-bone traditional mechanical testing. Bone. 2013;53:301-305.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Aoki C, Uto K, Honda K, Kato Y, Oda H. Advanced glycation end products suppress lysyl oxidase and induce bone collagen degradation in a rat model of renal osteodystrophy. Lab Invest. 2013;93:1170-1183.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  McCarthy AD, Molinuevo MS, Cortizo AM. Ages and bone ageing in diabetes mellitus. J Diabetes Metab. 2013;4:276.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Khosravi R, Sodek KL, Faibish M, Trackman PC. Collagen advanced glycation inhibits its Discoidin Domain Receptor 2 (DDR2)-mediated induction of lysyl oxidase in osteoblasts. Bone. 2014;58:33-41.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Saito M, Kida Y, Kato S, Marumo K. Diabetes, collagen, and bone quality. Curr Osteoporos Rep. 2014;12:181-188.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Qing H, Ardeshirpour L, Pajevic PD, Dusevich V, Jähn K, Kato S, Wysolmerski J, Bonewald LF. Demonstration of osteocytic perilacunar/canalicular remodeling in mice during lactation. J Bone Miner Res. 2012;27:1018-1029.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Suntornsaratoon P, Kraidith K, Teerapornpuntakit J, Dorkkam N, Wongdee K, Krishnamra N, Charoenphandhu N. Pre-suckling calcium supplementation effectively prevents lactation-induced osteopenia in rats. Am J Physiol Endocrinol Metab. 2014;306:E177-E188.  [PubMed]  [DOI]  [Cited in This Article: ]