Delayed bone regeneration and low bone mass in a rat model of insulin-resistant type 2 ... · 2017....

9
Delayed bone regeneration and low bone mass in a rat model of insulin-resistant type 2 diabetes mellitus is due to impaired osteoblast function Christine Hamann,* 1 Claudia Goettsch,* 2 Jan Mettelsiefen, 1 Veit Henkenjohann, 1 Martina Rauner, 2 Ute Hempel, 3 Ricardo Bernhardt, 4 Nadja Fratzl-Zelman, 5 Paul Roschger, 5 Stefan Rammelt, 6,7 Klaus-Peter Günther, 1,7 and Lorenz C. Hofbauer 2,7 Departments of 1 Orthopedics and 6 Trauma and Reconstructive Surgery and 2 Division of Endocrinology, Diabetes, and Metabolic Bone Diseases, Department of Medicine III, Dresden Technical University Medical Center, Dresden; 3 Institute of Physiological Chemistry and 4 Max Bergmann Center of Biomaterials, Technical University, Dresden; 7 Center for Regenerative Therapies Dresden, Germany; and 5 Ludwig-Boltzmann-Institute of Osteology at the Hanusch Hospital of Vienna Health Insurance Fund and Austrian Workers’ Compensation Board Trauma Centre Meidling, 1st Medical Department, Hanusch Hospital, Vienna, Austria Submitted 22 July 2011; accepted in final form 31 August 2011 Hamann C, Goettsch C, Mettelsiefen J, Henkenjohann V, Rauner M, Hempel U, Bernhardt R, Fratzl-Zelman N, Roschger P, Rammelt S, Günther KP, Hofbauer LC. Delayed bone regener- ation 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 301: E1220 –E1228, 2011. First published Septem- ber 6, 2011; doi:10.1152/ajpendo.00378.2011.—Patients with diabe- tes mellitus have an impaired bone metabolism; however, the underlying mechanisms are poorly understood. Here, we analyzed the impact of type 2 diabetes mellitus on bone physiology and regeneration using Zucker diabetic fatty (ZDF) rats, an established rat model of insulin-resistant type 2 diabetes mellitus. ZDF rats develop diabetes with vascular complica- tions when fed a Western diet. In 21-wk-old diabetic rats, bone mineral density (BMD) was 22.5% (total) and 54.6% (trabecular) lower at the distal femur and 17.2% (total) and 20.4% (trabecular) lower at the lumbar spine, respectively, compared with nondiabetic animals. BMD distribu- tion measured by backscattered electron imaging postmortem was not different between diabetic and nondiabetic rats, but evaluation of histo- morphometric indexes revealed lower mineralized bone volume/tissue volume, trabecular thickness, and trabecular number. Osteoblast differ- entiation of diabetic rats was impaired based on lower alkaline phospha- tase activity (20%) and mineralized matrix formation (55%). In addition, the expression of the osteoblast-specific genes bone morphoge- netic protein-2, RUNX2, osteocalcin, and osteopontin was reduced by 40 – 80%. Osteoclast biology was not affected based on tartrate-resistant acidic phosphatase staining, pit formation assay, and gene profiling. To validate the implications of these molecular and cellular findings in a clinically relevant model, a subcritical bone defect of 3 mm was created at the left femur after stabilization with a four-hole plate, and bone regeneration was monitored by X-ray and microcomputed tomography analyses over 12 wk. While nondiabetic rats filled the defects by 57%, diabetic rats showed delayed bone regeneration with only 21% defect filling. In conclusion, we identified suppressed osteoblastogenesis as a cause and mechanism for low bone mass and impaired bone regeneration in a rat model of type 2 diabetes mellitus. bone defect; bone matrix mineralization; bone regeneration; type 2 diabetes mellitus; osteoblast; osteoclast DIABETES MELLITUS TYPE 2 AND the associated metabolic syn- drome have become epidemic clinical and economic health problems (1). Morbidity and mortality of diabetes mellitus are determined by vascular complications, including cardiovascu- lar disease, retinopathy, nephropathy, and polyneuropathy (11). Skeletal sequelae of long-standing diabetes mellitus include Charcot neuroarthropathy and the diabetic foot syndrome, which may require amputation. More recently, osteoporosis with an increased risk of fragility fractures has emerged as a complication in patients with long-standing or poorly con- trolled type 1 and type 2 diabetes mellitus (8). Several factors may contribute to poor bone health in patients with diabetes mellitus, including hypercalciuria in periods of glucosuria in type 1 and 2 diabetes mellitus (20), delayed bone mass accrual and a lower peak bone mass in type 1 diabetes mellitus (8), poor bone quality despite higher bone mineral density (BMD) in type 2 diabetes mellitus (26), and a higher propensity to falls and osteoporotic fractures in long-standing type 1 and 2 dia- betes, in particular when neuropathy, retinopathy, or nephrop- athy are present (3). After sustaining fractures, patients with diabetes type 2 have a higher risk of wound infection, delayed fracture healing, a higher incidence of nonunion, and impaired osseointegration (22), which translate in prolonged hospital- ization and decreased mobility. Despite a large body of clinical and epidemiological evi- dence linking diabetes mellitus to poor bone health, there are limited data on the mechanisms whereby diabetes mellitus affects bone mass and bone quality. This may be, at least in part, due to the lack of appropriate rodent models of type 2 diabetes mellitus, which accounts for over 90% of diabetes mellitus in humans. Recently, two groups (17, 21) have re- ported on the skeletal phenotype of Zucker diabetic fatty (ZDF) rats. Male ZDF rats start to develop obesity and type 2 diabetes mellitus at 9 wk of age after exposure to a high-fat and high-carbohydrate diet. The phenotype is characterized by progressive insulin resistance with hyperglycemia and initial hyperinsulinemia and results in the development of the full spectrum of the metabolic syndrome, including dyslipidemia and diabetic vascular complications, which is reminiscent of the metabolic syndrome in humans. Skeletal abnormalities of these ZDF rats include a low bone mass and low bone forma- tion (17). However, the biological basis for these observations and the functional consequences of these alterations has re- mained poorly defined. * C. Hamann and C. Goettsch contributed equally to this work. Address for reprint requests and other correspondence: L. C. Hofbauer, Division of Endocrinology, Diabetes and Metabolic Bone Diseases, Dept. of Medicine III, Dresden Technical Univ. Medical Center, Fetscherstraße 74, 01307 Dresden, Germany (e-mail: [email protected]). Am J Physiol Endocrinol Metab 301: E1220–E1228, 2011. First published September 6, 2011; doi:10.1152/ajpendo.00378.2011. 0193-1849/11 Copyright © 2011 the American Physiological Society http://www.ajpendo.org E1220 by 10.220.33.1 on May 1, 2017 http://ajpendo.physiology.org/ Downloaded from

Transcript of Delayed bone regeneration and low bone mass in a rat model of insulin-resistant type 2 ... · 2017....

Page 1: Delayed bone regeneration and low bone mass in a rat model of insulin-resistant type 2 ... · 2017. 5. 1. · Delayed bone regeneration and low bone mass in a rat model of insulin-resistant

Delayed bone regeneration and low bone mass in a rat modelof insulin-resistant type 2 diabetes mellitus is due to impaired osteoblast function

Christine Hamann,*1 Claudia Goettsch,*2 Jan Mettelsiefen,1 Veit Henkenjohann,1 Martina Rauner,2

Ute Hempel,3 Ricardo Bernhardt,4 Nadja Fratzl-Zelman,5 Paul Roschger,5 Stefan Rammelt,6,7

Klaus-Peter Günther,1,7 and Lorenz C. Hofbauer2,7

Departments of 1Orthopedics and 6Trauma and Reconstructive Surgery and 2Division of Endocrinology, Diabetes, andMetabolic Bone Diseases, Department of Medicine III, Dresden Technical University Medical Center, Dresden; 3Institute ofPhysiological Chemistry and 4Max Bergmann Center of Biomaterials, Technical University, Dresden; 7Center forRegenerative Therapies Dresden, Germany; and 5Ludwig-Boltzmann-Institute of Osteology at the Hanusch Hospital of ViennaHealth Insurance Fund and Austrian Workers’ Compensation Board Trauma Centre Meidling, 1st Medical Department,Hanusch Hospital, Vienna, Austria

Submitted 22 July 2011; accepted in final form 31 August 2011

Hamann C, Goettsch C, Mettelsiefen J, Henkenjohann V,Rauner M, Hempel U, Bernhardt R, Fratzl-Zelman N, RoschgerP, Rammelt S, Günther KP, Hofbauer LC. Delayed bone regener-ation and low bone mass in a rat model of insulin-resistant type 2diabetes mellitus is due to impaired osteoblast function. Am J PhysiolEndocrinol Metab 301: E1220–E1228, 2011. First published Septem-ber 6, 2011; doi:10.1152/ajpendo.00378.2011.—Patients with diabe-tes mellitus have an impaired bone metabolism; however, the underlyingmechanisms are poorly understood. Here, we analyzed the impact of type2 diabetes mellitus on bone physiology and regeneration using Zuckerdiabetic fatty (ZDF) rats, an established rat model of insulin-resistant type2 diabetes mellitus. ZDF rats develop diabetes with vascular complica-tions when fed a Western diet. In 21-wk-old diabetic rats, bone mineraldensity (BMD) was 22.5% (total) and 54.6% (trabecular) lower at thedistal femur and 17.2% (total) and 20.4% (trabecular) lower at the lumbarspine, respectively, compared with nondiabetic animals. BMD distribu-tion measured by backscattered electron imaging postmortem was notdifferent between diabetic and nondiabetic rats, but evaluation of histo-morphometric indexes revealed lower mineralized bone volume/tissuevolume, trabecular thickness, and trabecular number. Osteoblast differ-entiation of diabetic rats was impaired based on lower alkaline phospha-tase activity (�20%) and mineralized matrix formation (�55%). Inaddition, the expression of the osteoblast-specific genes bone morphoge-netic protein-2, RUNX2, osteocalcin, and osteopontin was reduced by40–80%. Osteoclast biology was not affected based on tartrate-resistantacidic phosphatase staining, pit formation assay, and gene profiling. Tovalidate the implications of these molecular and cellular findings in aclinically relevant model, a subcritical bone defect of 3 mm was createdat the left femur after stabilization with a four-hole plate, and boneregeneration was monitored by X-ray and microcomputed tomographyanalyses over 12 wk. While nondiabetic rats filled the defects by 57%,diabetic rats showed delayed bone regeneration with only 21% defectfilling. In conclusion, we identified suppressed osteoblastogenesis as acause and mechanism for low bone mass and impaired bone regenerationin a rat model of type 2 diabetes mellitus.

bone defect; bone matrix mineralization; bone regeneration; type 2diabetes mellitus; osteoblast; osteoclast

DIABETES MELLITUS TYPE 2 AND the associated metabolic syn-drome have become epidemic clinical and economic health

problems (1). Morbidity and mortality of diabetes mellitus aredetermined by vascular complications, including cardiovascu-lar disease, retinopathy, nephropathy, and polyneuropathy (11).Skeletal sequelae of long-standing diabetes mellitus includeCharcot neuroarthropathy and the diabetic foot syndrome,which may require amputation. More recently, osteoporosiswith an increased risk of fragility fractures has emerged as acomplication in patients with long-standing or poorly con-trolled type 1 and type 2 diabetes mellitus (8). Several factorsmay contribute to poor bone health in patients with diabetesmellitus, including hypercalciuria in periods of glucosuria intype 1 and 2 diabetes mellitus (20), delayed bone mass accrualand a lower peak bone mass in type 1 diabetes mellitus (8),poor bone quality despite higher bone mineral density (BMD)in type 2 diabetes mellitus (26), and a higher propensity to fallsand osteoporotic fractures in long-standing type 1 and 2 dia-betes, in particular when neuropathy, retinopathy, or nephrop-athy are present (3). After sustaining fractures, patients withdiabetes type 2 have a higher risk of wound infection, delayedfracture healing, a higher incidence of nonunion, and impairedosseointegration (22), which translate in prolonged hospital-ization and decreased mobility.

Despite a large body of clinical and epidemiological evi-dence linking diabetes mellitus to poor bone health, there arelimited data on the mechanisms whereby diabetes mellitusaffects bone mass and bone quality. This may be, at least inpart, due to the lack of appropriate rodent models of type 2diabetes mellitus, which accounts for over 90% of diabetesmellitus in humans. Recently, two groups (17, 21) have re-ported on the skeletal phenotype of Zucker diabetic fatty (ZDF)rats. Male ZDF rats start to develop obesity and type 2 diabetesmellitus at 9 wk of age after exposure to a high-fat andhigh-carbohydrate diet. The phenotype is characterized byprogressive insulin resistance with hyperglycemia and initialhyperinsulinemia and results in the development of the fullspectrum of the metabolic syndrome, including dyslipidemiaand diabetic vascular complications, which is reminiscent ofthe metabolic syndrome in humans. Skeletal abnormalities ofthese ZDF rats include a low bone mass and low bone forma-tion (17). However, the biological basis for these observationsand the functional consequences of these alterations has re-mained poorly defined.

* C. Hamann and C. Goettsch contributed equally to this work.Address for reprint requests and other correspondence: L. C. Hofbauer,

Division of Endocrinology, Diabetes and Metabolic Bone Diseases, Dept. ofMedicine III, Dresden Technical Univ. Medical Center, Fetscherstraße 74,01307 Dresden, Germany (e-mail: [email protected]).

Am J Physiol Endocrinol Metab 301: E1220–E1228, 2011.First published September 6, 2011; doi:10.1152/ajpendo.00378.2011.

0193-1849/11 Copyright © 2011 the American Physiological Society http://www.ajpendo.orgE1220

by 10.220.33.1 on May 1, 2017

http://ajpendo.physiology.org/D

ownloaded from

Page 2: Delayed bone regeneration and low bone mass in a rat model of insulin-resistant type 2 ... · 2017. 5. 1. · Delayed bone regeneration and low bone mass in a rat model of insulin-resistant

To analyze the biological basis for abnormal bone mass ininsulin-resistant type 2 diabetes mellitus, we elected the ZDFrat model, since it recapitulates most of the metabolic andmicrovascular features of type 2 diabetes mellitus in humans.We hypothesized that osteoblast function is impaired in thismodel that should result in an altered bone matrix mineraliza-tion and abnormal bone regeneration in vivo using a subcriticalfemoral defect model. Our study analyzes BMD, bone matrixmineralization, bone cell function, and bone defect regenera-tion in a rat model of type 2 diabetes mellitus.

MATERIALS AND METHODS

Animal maintenance. All research protocols were approved by theInstitutional Animal Care Committee (registration no. 24D-9168.11–1/2008–30). Male ZDF rats (ZDF Gmi/fa/fa) and male Zucker leanrats (ZDF Gmifa/fa) were purchased from Charles River Laboratories(Sulzfeld, Germany) at the age of 9 wk. Male ZDF fa/fa rats sponta-neously develop type 2 diabetes mellitus between the age of weeks 9and 11. The Zucker lean rats (ZDF�/�) are nondiabetic and served asthe control. The animals were housed under standard laboratoryconditions with a 12:12-h light-dark cycle and a controlled roomtemperature of 20–21°C. The animals had free access to water, andboth groups were fed a high-fat, high-carbohydrate chow (Purina5008).

Critical size defect model. At the age of 9 wk, the left femur wasshaved and disinfected. Under general anesthesia with intraperitonealinjection of 90 mg/kg ketamine and 10 mg/kg xylazine, a four-holeplate (Stryker, Hamburg, Germany) was fixed with four screws (1.5 �5 mm) on the left femur. A 3-mm cross-sectional subcritical defectwas created at the midshaft using an oscillating saw between thesecond and the third screw. The wound was closed, and a postoper-ative X-ray was performed. The animals were returned to free cageactivity. During surgery and 24 h later, all animals received subcuta-neous injections of antibiotics (sulfadoxin and trimethoprin, 5:1, 15mg/kg) to minimize the risk of infections. After surgery, both groupswere maintained on a high-fat, high-carbohydrate chow (Purina5008). Twelve weeks after surgery, the animals were killed undergeneral anesthesia, blood samples were collected, and the bones wereremoved for further analysis. To verify the diabetic state of theanimals at 3, 8, 10, and 12 wk after surgery, plasma glucose levels ofthe animals were measured.

Serum analysis. Serum measurements of glucose, calcium, phos-phate, creatinine, alkaline phosphatase (AP), and bone-specific APwere performed using standard methods. Serum parathyroid hormone(PTH) [intra-assay coefficient of variation (CV): 3.1%; interassay CV:5.1%] and insulin (intra-assay CV: 3.9%; interassay CV: 9.3%) levelswere determined using immunoassays from DGR Diagnostics (Mar-burg, Germany). Serum procollagen 1 NH2-terminal peptide (P1NP)(intra-assay CV: 2.1%; interassay CV: 7.4%), osteocalcin (intra-assayCV: 2.2%; interassay CV: 5.5%), and COOH-terminal collagen crosslinks (CTX) (intra-assay CV: 2.9%; interassay CV: 5.6%) levels weremeasured with ELISAs from Immunodiagnostic Systems (Frankfurt,Germany).

X-ray, peripheral quantitative computed, and microcomputed to-mography analysis. X-rays were performed after surgery to controlthe defect size and the position of the plate and screws as well as 3,8, 10, and 12 wk after surgery to document the progress of defecthealing. Defect areas were quantified from the X-ray images usingImageJ software (National Institutes of Health). Peripheral quantita-tive computed tomography (pQCT) of the right femur and the lumbarspine was performed before surgery in vivo and 12 wk later ex vivousing a Stratec XCT Research M� pQCT (Stratec Medical Systems).pQCT scans were performed at the nonoperated right femur at the metaph-yseal and diaphyseal area and the fourth lumbar vertebra. The bones weremeasured in plastic tubes filled with 70% ethanol. The regions of interests

were predefined, and contour mode 1 and peel mode 20 were used.The percentage of trabecular bone was set to 50% of the cross-sectional area in the proximal tibia. A threshold of 710 mg/cm3 wasused for calculation of cortical BMD. For nondestructive analysis ofnewly formed bone inside the defect zone, a Scanco vivaCT 75 system(Scanco Medical, Brüttisellen, Switzerland) and a Synchroton micro-computed tomography (SR�CT) device at BESSY II (Berlin, Ger-many) were used. Femur explants with metallic plates were measuredwith a voxel resolution of 20 �m (SR�CT 9 �m) and X-ray energy of70 keV (SR�CT 55 keV, monochromatic). A fixed constant thresholdwas applied to all samples. The digital volumes were aligned to thelong bone axis, and a reference area for stationary bone was measuredat the top and bottom slices of the callus volume. The relative amountof newly formed bone within the callus was calculated based on themedium bone amount for all slices along the axis divided by thereference values using ImageJ. Standard deviation of the average boneamount for each sample was calculated using individual slices alongcallus in the �CT volumes.

Quantitative backscattered electron imaging. To assess the mineralcontent of bone at the material level, i.e., bone mineralization densitydistribution (BMDD), quantitative backscattered electron imaging(qBEI) was performed at the distal femur. The right (nonoperated)distal femurs of diabetic (n � 6) and nondiabetic (n � 6) animals werefixed and stored in 70% ethanol before embedding in polymetham-ethylacrylate. Longitudinal bone sections containing cancellous andcortical bone were processed. Trabecular and cortical BMDD wasdetermined by qBEI using a digital scanning electron microscope(DSM 962; Zeiss, Oberkochen, Germany) equipped with a four-quadrant semiconductor backscattered electron detector as describedpreviously (23). The accelerating voltage of the electron beam wasadjusted to 20 kV, the probe current to 110 pA, and the workingdistance to 15 mm. The cancellous and cortical bone areas wereimaged at 100-fold nominal magnification (corresponding to a pixelresolution of 2 �m/pixel). From these digital images, gray levelhistograms were deduced, displaying the percentage of bone areaoccupied by pixels of a certain gray level. The transformation of theseinto calcium weight percent (wt/100 wt) histograms led to a bin widthof 0.17 wt/100 wt calcium. A technical precision of 0.3% wasachieved. BMDD parameters like the mean (weighted mean, CaMean)and mode (CaPeak, peak position of the BMDD) calcium concentra-tion in the sample, the width of the distribution (CaWidth, full widthat half-maximum) reflecting the heterogeneity in matrix mineraliza-tion, the fraction of low mineralized bone (CaLow), which is thepercentage of the area below 17.7 wt/100 wt Ca (corresponding to the5th percentile of human reference BMDD), and the fraction of highmineralized bone (CaHigh), which is the percentage of the area above25.3 wt/100 wt Ca (corresponding to the 75th percentile of humanreference BMDD), were derived from the histogram. CaHigh wasonly determined for metaphyseal bone.

Structural histomorphometric parameters [mineralized bone vol-ume/tissue volume (mBV/TV), trabecular thickness (TbTh) and tra-becular number (TbN)] were determined by analyzing a tissue areabetween 0.5 and 1.5 mm below the growth plate of metaphyseal boneat the distal femurs from nondiabetic vs. diabetic rats (n � 6) usingqBEI.

Assessment of osteoblast and osteoclast functions. Bone marrowcells were rinsed with DMEM containing 10% FCS. Colony-formingunits (CFU)-fibroblast were determined by seeding cells at a densityof 200,000/cm2 and culturing them in DMEM with 10% FCS and 1%penicillin/streptomycin for 14 days. Thereafter, cells were stainedwith 1% crystal violet (Sigma), and formed colonies were counted. Inaddition, the specifically bound dye was eluted with 0.2% TritonX-100 and measured using a spectrophotometer at 590 nm. Cellvitality was determined using Cell Titer Blue (Promega). Bone mar-row stromal cells were differentiated toward osteoblasts using DMEMcontaining 25 mM of glucose supplemented with 10% FCS, 1%penicillin/streptomycin, 100 �M ascorbate phosphate, 5 mM �-glyc-

E1221BONE REGENERATION IN DIABETES MELLITUS

AJP-Endocrinol Metab • VOL 301 • DECEMBER 2011 • www.ajpendo.org

by 10.220.33.1 on May 1, 2017

http://ajpendo.physiology.org/D

ownloaded from

Page 3: Delayed bone regeneration and low bone mass in a rat model of insulin-resistant type 2 ... · 2017. 5. 1. · Delayed bone regeneration and low bone mass in a rat model of insulin-resistant

erol phosphate, and 10 nM dexamethasone. CFU-osteoblasts wasassessed by seeding 200,000 cells/cm2 and culturing them for 21 days.Osteoblastic colonies were quantified by counting alizarin red S-stained colonies and subsequently eluting the dye with 100 mMcetylpyridinium chloride (Sigma). For all other experiments, cellswere seeded at a density of 1 � 106/cm2. AP activity was assessedafter 14 days. Therefore, cells were lysed and centrifuged for 30 minat 25,000 g at 4°C. Aliquots of each sample were incubated with 100�l AP substrate buffer for 30 min at 37°C. The reaction was stoppedwith 100 mM NaOH. Optical density was measured at a wavelengthof 405 nm and normalized to the total protein content determined bythe BCA method. After 21 days of culture, matrix formation wasanalyzed by alizarin red S staining. Osteoblasts were fixed in 10%paraformaldehyde for 30 min and stained with 1% alizarin red S (pH5.5; Sigma) for 10 min. Excess dye was removed with distilled water,and the amount of incorporated dye was eluted with 100 mM ce-tylpyridinium chloride (Sigma) for 30 min at room temperature.Aliquots were taken and measured spectrophotometrically at 450 nmin triplicates.

For osteoclast assessment, bone marrow cells were collected andplated at a density of 2 � 106 cells/cm2 in �-minimal essentialmedium containing 5.5 mM of glucose, 10% FCS, and 1% penicillin/streptomycin. Following attachment, medium was replaced with basalmedium supplemented with macrophage colony-stimulating factor(M-CSF, 25 ng/ml; R & D Systems) for 2 days. Thereafter, cells werecultured for 6 days in differentiation medium containing M-CSF (25ng/ml) and receptor activator of NF-�B ligand (50 ng/ml; R & DSystems). Tartrate-resistant acidic phosphatase (TRAP) staining wasperformed using the leukocyte acid phosphatase kit (Sigma). Toassess resorption activity, osteoclast precursor cells were plated in24-well-plates coated with osteoblast-derived matrix as reported else-where (19). After 8 days, bone matrix was stained with von Kossa,and the resulting resorption pits were quantified by densitometry.

RNA isolation and real-time PCR analysis. Total RNA was isolatedusing the High Pure RNA Isolation Kit (Roche). In reverse transcrip-tase reactions, equal amounts of total RNA (500 ng) were incubatedfor 3 min at 70°C and subsequently reverse-transcribed into cDNAusing random hexamer primers for 1 h at 42°C. For real-time PCRreactions, the following primer pairs for amplification were used:RUNX2 (sense: 5=-CCCACGGCCCTCCCTGAACT-3=; antisense:5=-CGTGTGGAAGACAGCGGCGT-3=), osteopontin (sense: 5=-gag-gagaaggcgcattacag-3=; antisense: 5=-atggctttcattggagttgc-3=), osteo-calcin (5=-GAGGGCAGTAAGGTGGTGAA-3=; antisense: 5=-GTC-CGCTAGCTCGTCACAAT-3=), bone morphogenetic protein (BMP)-2(sense: 5=-ACATCCACTCCACAAACGAG-3=; antisense: 5=-GTCAT-TCCACCCCACATCAC-3=), and TRAP5b (sense: 5=-gagaacggtgtgggc-tatgt-3=; antisense: 5=-gtgaagccacccagagagtc-3=). The mRNA expression wasnormalized to �-actin.

Statistical analysis. Results are presented as means SE. Allexperiments were repeated at least three times. Single-group compar-isons were performed using a Student’s t-test. P values of 0.05 wereconsidered statistically significant.

RESULTS

Characteristics and laboratory findings. At baseline at anage of 9 wk, i.e., immediately before surgery, mean serumglucose levels were 9.0 0.62 mmol/l for nondiabetic rats and30.8 0.60, mmol/l for diabetic rats (P 0.01). Moreover, thebody weight of ZDF rats was higher (367 4.6 g) for diabeticrats than for nondiabetic rats (296 5.6 g) (P 0.01). Total,cortical/subcortical, and trabecular BMD at the distal femur,total BMD, and cortical thickness of the femoral shaft were notstatistically different at 9 wk between diabetic and nondiabeticZDF rats as measured in vivo by pQCT.

When exposed to a diet with a high degree of saturated fatand carbohydrate (Purina 5008) for 12 wk, serum glucoselevels of diabetic ZDF rats (21 wk old) increased to values of40 mmol/l (Table 1) while serum glucose levels of nondiabeticZDF rats fed Purina 5008 rose to 19 mmol/l. In 21-wk-old rats,serum insulin levels were similar in nondiabetic and diabeticZDF rats, a finding that has been reported previously (27). Ofnote, the body weight was similar between the two groups,presumably because of the persistent hyperglycemia and glu-cosuria of diabetic rats (27). The clinical and laboratory find-ings of 21-wk-old nondiabetic and diabetic ZDF rats aresummarized in Table 1. Diabetic ZDF rats showed significanthigher blood glucose levels (P 0.01). Serum calcium levelswere increased markedly in diabetic rats compared with non-diabetic rats (P 0.01), and there was a trend toward higherserum PTH levels, whereas serum phosphate levels were notdifferent. Serum urea levels, but not creatinine levels, wereelevated in the diabetic animals, indicating renal impairment(Table 1). Serum levels of P1NP (P � 0.016) and osteocalcin(P � 0.001), two markers of bone formation, were lower indiabetic rats compared with nondiabetic rats, whereas serumCTX levels, a bone resorption marker, were threefold higher indiabetic compared with nondiabetic ZDF rats (P � 0.013)(Table 1).

Lower BMD at all skeletal sites in diabetic rats. At 21 wk ofage, detailed pQCT analysis revealed significantly lower totalBMD at the distal femur, the femoral shaft, and the lumbarspine in diabetic rats compared with nondiabetic rats (Fig. 1).At the distal femur, total BMD (Fig. 1A), trabecular BMD (Fig.1B), and cortical/subcortical BMD (Fig. 1C) were 22.5% (P 0.01), 54.6% (P 0.01), and 15.9% (P 0.05) lower,respectively, in diabetic rats compared with nondiabetic rats.At the femoral shaft, cortical BMD and cortical thickness were2.3% (P 0.05) and 17.7% (P 0.01) lower in diabetic rats,respectively (Fig. 1, D and E). At the fourth lumbar vertebra,total BMD (�17.2%, P 0.01), trabecular BMD (�20.4%,P 0.01), and cortical/subcortical BMD (�15.8%, P 0.01)were significantly lower in diabetic rats compared with nondi-abetic rats (Fig. 1, F–H).

Lack of alteration in bone matrix mineralization of diabeticrats. BMDD of trabecular and cortical bone was very similarbetween diabetic and nondiabetic rats (Table 2). Both groups

Table 1. Body weight and laboratory characteristics of21-wk-old nondiabetic (�/�) and diabetic (fa/fa) ZDF rats

ParameterNondiabetic ZDF�/�

RatsDiabetic ZDFfa/fa

RatsP

Value

No. of animals 15 15Body wt, g 409 7.80 423 8.40 0.265Blood glucose levels, mmol/l 18.7 1.18 39.5 0.72 0.001Serum insulin levels, ng/l 0.35 0.07 0.43 0.07 0.249Serum calcium levels, mmol/l 2.50 0.06 2.70 0.03 0.006Serum phosphate levels, mmol/l 2.39 0.12 2.43 0.12 0.784Serum creatinine levels, �mol/l 35.3 2.37 38.3 1.50 0.724Serum urea levels, mmol/l 7.14 0.21 8.49 0.40 0.006Serum PTH levels, pg/ml 783 110 962 120 0.283Serum P1NP levels, ng/ml 15.5 1.46 11.0 0.93 0.016Serum osteocalcin levels, pg/ml 458 49.4 175 21.9 0.001Serum CTX levels, ng/ml 35.9 2.6 96.3 17.0 0.013

Values are means SE. ZDF, Zucker diabetic fatty; PTH, parathyroidhormone; P1NP, procollagen 1 NH2-terminal peptide; CTX, COOH-terminalcollagen cross links.

E1222 BONE REGENERATION IN DIABETES MELLITUS

AJP-Endocrinol Metab • VOL 301 • DECEMBER 2011 • www.ajpendo.org

by 10.220.33.1 on May 1, 2017

http://ajpendo.physiology.org/D

ownloaded from

Page 4: Delayed bone regeneration and low bone mass in a rat model of insulin-resistant type 2 ... · 2017. 5. 1. · Delayed bone regeneration and low bone mass in a rat model of insulin-resistant

displayed characteristic modifications in BMDD dependent onbone tissue site: a shift of the BMDD curves toward highermatrix mineralization (to the right) was observed from metaph-yseal to epiphyseal and to cortical bone as reflected by an

increase in the mean calcium content (CaMean) and the peakposition of the BMDD (CaPeak). At the same time, the heter-ogeneity in matrix mineralization (CaWidth) became narrower,and the fraction of low mineralized bone matrix (CaLow)

Fig. 1. Lower spinal and femoral bone mineraldensity (BMD) in diabetic rats. VolumetricBMD from nondiabetic (�/�) and diabetic (fa/fa) Zucker diabetic fatty (ZDF) rats was mea-sured by peripheral quantitative computed to-mography. A-C: distal femur. Total BMD (A),trabecular BMD (B), and cortical/subcorticalBMD (C) of the distal femur, n � 3–4 rats ineach group. D and E: femoral shaft. CorticalBMD (D) and cortical thickness (E) of thefemoral shaft, n � 3–4. F-H: lumbar spine.Total BMD (F), trabecular BMD (G), and cor-tical/subcortical BMD (H) of the 4th lumbarvertebra, n � 10–12. Data represent means SE. aP 0.05, �/� vs. fa/fa.

Table 2. BMDD parameters of metaphyseal, epiphyseal, and cortical midshaft bone of distal femurs from 21-wk-oldnondiabetic vs. diabetic rats

BMDD Parameters

Metaphyseal Epiphyseal Cortical

Nondiabetic Diabetic Nondiabetic Diabetic Nondiabetic Diabetic

CaMean, wt/100 wt Ca 23.7 0.24 24.24 0.17 24.12 0.32 24.6 0.15 25.2 0.20 25.5 0.20CaPeak, wt/100 wt Ca 24.0 0.23 24.4 [24.26; 24.87] 24.70 0.27 25.1 0.14 25.6 0.21 25.7 0.21CaWidth, Dwt/100 wt Ca 3.32 0.08 4.07* [3.38; 4.33] 2.95 [2.77; 2.95] 2.95 0.06 3.12 [3.03;3.21] 3.06 0.12CaLow, %BAr 4.00 0.24 4.56 0.22 2.83 0.31 2.93 0.19 1.59 0.06 1.73 0.04CaHigh, %BAr 25.0 3.24 38.3 3.02* NM NM NM NM

Values represent means SE or the median with the 25th and 75th percentiles in square brackets if data did not pass the normality test (n � 6). BMDD, bonemineralization density distribution; BAr, bone ares; NM, not measured. *P 0.05 between controls and diabetic animals using unpaired t-tests or Mann-Whitneyrank sums tests, if data were not normally distributed. For abbreviations of the BMDD parameters, please see MATERIALS AND METHODS.

E1223BONE REGENERATION IN DIABETES MELLITUS

AJP-Endocrinol Metab • VOL 301 • DECEMBER 2011 • www.ajpendo.org

by 10.220.33.1 on May 1, 2017

http://ajpendo.physiology.org/D

ownloaded from

Page 5: Delayed bone regeneration and low bone mass in a rat model of insulin-resistant type 2 ... · 2017. 5. 1. · Delayed bone regeneration and low bone mass in a rat model of insulin-resistant

decreased from metaphysis to epiphysis and to cortex (Fig. 2,A and B, and Table 2).

Moreover, BMDD of metaphyseal bone displayed a specificcontribution in the highly mineralized range (Fig. 2C). Theobserved relative increased fraction of residual mineralizedcartilage motifs in the trabecular bone matrix was responsiblefor this increase in CaWidth (�25%, P � 0.041) and CaHigh(�53.5%, P � 0.013) in diabetic vs. nondiabetic animals (Fig.2C and Table 2). Structural histomorphometric parametersobtained from qBEI indicated a 48% lower mineralized BV/TVin diabetic compared with nondiabetic rats and a significantreduction in TbTh and TbN (Table 3).

Impaired osteoblastic functions in diabetic rats. To assessthe underlying cellular mechanism for the observed decrease inBMD in type 2 diabetes mellitus, osteoblast and osteoclast

biology was analyzed. Bone marrow cells from diabetic andnondiabetic rats displayed a similar vitality and contained anequal number of precursor cells as assessed by colony-formingassays (data not shown), indicating that there was a similarsupply of osteoblastic precursor cells. Osteoblast differentia-tion was impaired by type 2 diabetes mellitus based on a 55%lower mineralized matrix formation in cells from diabeticcompared with nondiabetic rats after 21 days of differentiation(Fig. 3A). In addition, AP activity of osteoblasts was reducedby 20% (Fig. 3B). Furthermore, osteoblast-specific transcrip-tion factors, growth factors, and extracellular matrix proteinssuch as RUNX2, osteopontin, BMP-2, and osteocalcin werereduced by 40–80% after osteogenic differentiation for 7 and21 days, respectively (Fig. 3, C–F).

By contrast, osteoclast functions did not differ betweendiabetic and nondiabetic rats (Fig. 4). Specifically, the numberof TRAP-positive osteoclasts (Fig. 4A), osteoclast resorptivecapacity as determined by a pit formation assay (Fig. 4B), andTRAP mRNA steady-state levels expressed by osteoclasts(Fig. 4C) were comparable.

Delayed defect regeneration in diabetic vs. nondiabetic rats.To validate the functional implications of these observations ina translational rodent model, a bone defect of 3 mm wascreated by microsurgical techniques at the left femur afterstabilizing the femur with a four-hole plate (Fig. 5A). Defectregeneration was monitored in vivo by X-ray over the course of12 wk and with �CT analysis postmortem. While nondiabeticrats partially filled the defects by a mean of 57% after 12 wk,diabetic rats showed delayed new bone defect filling, resultingin only 21% filling of the defect (P 0.01, Fig. 5, B and C).Longitudinal X-ray analysis was consistent with these data andindicated that decreased defect healing in diabetic rats wasmainly due to delayed bone regeneration in the first 8 wkpostoperatively, after which the bone defects of diabetic andnondiabetic rats healed at a similar rate (Fig. 5D).

DISCUSSION

Using a well-established rat model of type 2 diabetes mel-litus, we show that bone formation is impaired by type 2diabetes due to a specific osteoblast defect that leads to de-creased bone mass and delayed bone regeneration.

Diabetes mellitus has been increasingly recognized as a riskfactor for osteoporosis and fragility fractures in humans. Poorglycemic control over a long time and the presence of vascularcomplications have been identified as potential determinants oflow bone mass and increased risk of fractures (8). Clinically,patients with diabetes mellitus carry a higher risk of delayed

Fig. 2. Bone mineralization density distribution (BMDD) of intact femurs indiabetic rats. A: backscattered electron image (BEI) overview of distal femurfrom a nondiabetic rat (longitudinal section). The regions for BMDD analysesare indicated (areas within yellow line). Ms, cancellous bone of metaphysealregion; Es, cancellous bone of epiphyseal region; Ct, cortical bone at midshaftregion. B: example of BMDD as obtained from these three bone sites. Forcomparison, the BMDD curve of human normal reference bone is represented(gray curve, dotted line). C: comparison of BMDD from metaphyseal cancel-lous bone between diabetic and nondiabetic rats. The fraction of highlymineralized bone area (CaHigh) is increased in the diabetic rat compared withthe control because of the existence of residual mineralized cartilage that ishigher mineralized than bone. Please note the corresponding shoulder at thehigh-mineralized end of the curve.

Table 3. Structural histomorphometric parameters ofmetaphyseal bone of distal femurs from nondiabetic vs.diabetic rats

Structural Parameters Nondiabetic Diabetic

BV/TV, % 26.5 2.73 13.8 1.40*TbTh, �m 86.3 5.71 60.7 3.03*TbN, 1/mm 3.04 0.18 2.25 0.15*

Values were derived from quantitative backscattered electron imaging of atissue area between 0.5 to 1.5 mm below the growth plate and represent themeans SE; n � 6 rats in each group. BV/TV, bone volume/total volume;TbTh, trabecular thickness; TbN, trabecular number. *P 0.01 betweencontrols and diabetic animals using unpaired t-tests.

E1224 BONE REGENERATION IN DIABETES MELLITUS

AJP-Endocrinol Metab • VOL 301 • DECEMBER 2011 • www.ajpendo.org

by 10.220.33.1 on May 1, 2017

http://ajpendo.physiology.org/D

ownloaded from

Page 6: Delayed bone regeneration and low bone mass in a rat model of insulin-resistant type 2 ... · 2017. 5. 1. · Delayed bone regeneration and low bone mass in a rat model of insulin-resistant

fracture healing as well as developing nonunion and impairedosseointegration (3, 22). Various individual skeletal and ex-traskeletal factors have been implicated to contribute to poorbone health (8, 20, 26) in diabetes, however, they remain

poorly defined at the cellular and molecular level. Studies ofthe complex relationship between bone metabolism in type 2diabetes mellitus were limited by the lack of appropriate rodentmodels. The ZDF fa/fa rats represent a suitable, diet-induced

Fig. 3. Impaired osteoblast functions in diabetic rats.Osteoblasts from nondiabetic (�/�) and diabetic (fa/fa)ZDF rats were obtained by differentiating bone marrowstromal cells using dexamethasone, �-glycerol phos-phate, and ascorbic acid as indicated. A: alizarin red Sstaining of the mineralized matrix was performed after7 and 21 days, n � 3–5. B: alkaline phosphatase (ALP)activity was measured after 14 days and normalized tothe total protein content, n � 3–5. C-E: RNA wasextracted at day 7, reverse transcribed, and subjected toreal-time PCR analysis. Gene expression of RUNX2(C), osteopontin (D), and bone morphogenetic protein(BMP)-2 (E) was normalized to �-actin, n � 4–6.F: RNA was extracted at day 21, reverse transcribed,and subjected to real-time PCR analysis. Gene expres-sion of osteoclacin was normalized to �-actin, n �4–6. Data are presented as means SE. aP 0.05,�/� vs. fa/fa.

Fig. 4. Osteoclast function is not altered in diabetic rats.Osteoclasts from nondiabetic (�/�) and diabetic (fa/fa) ZDFrats were generated by differentiating bone marrow cells usingmacrophage colony-stimulating factor and receptor activator ofNF-�B ligand for 7 days. Osteoclast differentiation was as-sessed by counting tartrate-resistant acidic phosphatase(TRAP)-positive multinucleated cells (A, n � 6), by perform-ing resorption pit assays (B, n � 3; left, representative pit image;right, quantitative analysis of pit size), and by analyzing TRAPmRNA levels (C, n � 6). Data are represented as means SE.

E1225BONE REGENERATION IN DIABETES MELLITUS

AJP-Endocrinol Metab • VOL 301 • DECEMBER 2011 • www.ajpendo.org

by 10.220.33.1 on May 1, 2017

http://ajpendo.physiology.org/D

ownloaded from

Page 7: Delayed bone regeneration and low bone mass in a rat model of insulin-resistant type 2 ... · 2017. 5. 1. · Delayed bone regeneration and low bone mass in a rat model of insulin-resistant

rodent model of type 2 diabetes mellitus, since they recapitu-late most features of poorly controlled type 2 diabetes mellitusin humans (17, 21). Although hyperglycemia was consistentlydocumented in diabetic rats, serum insulin levels after 21 wkwere comparable as reported (27). Thus, this reflects a latestage of long-lasting type 2 diabetes mellitus at which there isa relative �-cell failure secondary to prolonged hyperinsulin-emia and insulin resistance. In addition, mild renal impairmentwas already present (27). Unexpectedly, diabetic rats displayedmild hypercalcemia, which was not fully explained by PTHvariation and was not associated with phosphate abnormalities.These findings have no correlate in humans with type 2diabetes mellitus where hypercalcemia in diabetic individualsis mainly due to comorbidity (e.g., primary hyperparathyroid-ism) or comedication (e.g., thiazide diuretics).

Diabetic ZDF rats display reduced BMD at different skeletalsites, lower histomorphometric indexes of BV/TV, and lowerbone formation markers osteocalcin and P1NP in vivo that areparalleled by impaired osteoblast functions in vitro, includinglower expression of osteoblastic transcription factors andgenes, less efficient differentiation, and markedly reducedformation of mineralized matrix. Of note, our study showedthat trabecular BMD was particularly decreased in ZDF rats.Interestingly, the suppressive effect of the diabetic microenvi-ronment on osteogenic differentiation persists even after cul-ture for up to 3 wk ex vivo. Our data are in accordance withearlier findings obtained in ZDF rats that demonstrated im-paired bone mass and bone strength in diabetic ZDF rats (17,21) and provide a cellular explanation for these observations,

which identify a predominant defect of osteoblastogenesis.This is in line with previous in vitro (9, 7) and in vivo studiesinvolving rodent models of type 1 diabetes mellitus (2, 18).Our findings are also consistent with the observations made inspontaneously diabetic Torii rats, a model of nonobese type 2diabetes mellitus that displayed a lower bone formation rateand decreased gene expression of AP and osteocalcin (6).These changes were reversed by insulin treatment.

We did not observe altered osteoclast functions ex vivo, incontrast to other reports, which suggested that a diabeticmicroenvironment tends to inhibit osteoclast differentiationand function (30, 16, 29, 4, 12). However, none of these studiesused an in vivo model of rats with type 2 diabetes mellitus. Apotential limitation of our bone cell differentiation assays isthat heterogeneous cell populations are obtained from the bonemarrow harboring self-renewing stem cells and committedprogenitor cells. This may also explain the higher susceptibilityof osteoblasts to the inhibitory effects of diabetes mellitus ifone considers that a significant proportion of cells, rangingfrom stem cells to bipotent progenitor cells, may be primed toenter the adipogenic differentiation pathway instead of theosteogenic pathway and that this lack of osteogenic precursorcells cannot be compensated during the culture period. In fact,recent studies indicate that several established oral antidiabeticdrugs may specifically modulate osteoblast differentiation.While metformin stimulates osteoblast differentiation throughthe transactivation of RUNX2 via the AMPK/USF-1/SHPregulatory cascade (10), glitazones activate the peroxisomeproliferator-activating receptor-� and may direct bone marrow

Fig. 5. Delayed bone regeneration in diabetic rats. A: thesubcritical defect model was generated by fixing a 4-holeplate (a and b) to the left femur and removing a 3-mmbone specimen using an oscillating saw between the 2ndand the 3rd screw (b and c). a: 4-hole plate with screws;b: postoperative X-ray with plate and defect; c: bonespecimen. B: representative Synchroton microcomputedtomography image of the femoral bone defect from non-diabetic (�/�) and diabetic (fa/fa) ZDF rats. C: quanti-tative analysis of bone defect filling after 12 wk, n �7–10. aP 0.01. D: analysis of defect size over time.Defect zone of the X-ray images was quantified usingImageJ, n � 8. aP 0.05, �/� vs. fa/fa on one timepoint. Data represent means SE.

E1226 BONE REGENERATION IN DIABETES MELLITUS

AJP-Endocrinol Metab • VOL 301 • DECEMBER 2011 • www.ajpendo.org

by 10.220.33.1 on May 1, 2017

http://ajpendo.physiology.org/D

ownloaded from

Page 8: Delayed bone regeneration and low bone mass in a rat model of insulin-resistant type 2 ... · 2017. 5. 1. · Delayed bone regeneration and low bone mass in a rat model of insulin-resistant

stromal cells toward the adipocytic lineage at the cost ofosteoblasts (13).

The lack of significant differences in BMDD between dia-betic and nondiabetic rats with the exception of metaphysealbone suggests that the bone mineralization at the material levelis not affected in diabetic animals. Thus, lower BMD indiabetic rats is most likely caused by a decrease in bonevolume/mass alone but not by impaired matrix mineralizationas evidenced by qBEI. The variations in BMDD found betweenmetaphyseal, epiphyseal, and cortical bone are due to thedifferences in average tissue age at these specific skeletal sites.Younger bone tissue is less mineralized than older bone tissue(24). Thus, in the investigated femurs of the diabetic andnondiabetic rats, the metaphyseal bone tissue is the leastmineralized and therefore the youngest because of ongoing(re)modeling and the cortical bone the highest mineralized andtherefore the oldest one because cortical bone is less (re)mod-eled. These differences in mineralization are consistent withprevious observations in mouse femurs (5). Of note, the in-creased CaWidth and CaHigh found in metaphyseal bone fordiabetic rats suggests that the diabetic rats have a disturbedendochrondral ossification in the subchrondal region of thegrowth plate, probably related to the diminished bone forma-tion, leading to a higher portion of residual mineralized carti-lage in the metaphyseal bone. Thus, bone mineralization indiabetic rats appears to be intact and does not account for theirlow BMD.

While nondiabetic rats partially filled the bone defects,diabetic ZDF rats showed delayed bone regeneration of thesubcritical bone defect. These findings are consistent with theex vivo studies demonstrating markedly reduced osteoblasticfunction in diabetic animals. Bone regeneration that occurs inareas of bone defects is mainly driven by osteoblastic boneformation. Our data indicate that this process is particularlyimpaired in diabetic rats within the first 6 wk of the bonedefect, which may indicate altered recruitment of osteoblasticprecursor cells under diabetic conditions. In fact, Lu et al. (18)reported in a type 1 mouse model that delayed bone formationwas due to inadequately low expression of genes that controlosteoblast differentiation such as RUNX-2 and Dlx5, whereasimmature mesenchymal tissue with putative osteoblastic pre-cursors was sufficient. Impaired bone healing with reducedbone formation parameters was also observed in a bone histo-morphometric study of GK rats with type 2 diabetes mellituswith titanium implants placed in the femora (28). Thus, basedon these findings, osteoinductive biomaterials or bone-anabolictherapies that stimulate osteoblastic differentiation and func-tion locally or systemically may be promising strategies totarget impaired bone formation. This should be formally as-sessed in patients with type 2 diabetes mellitus.

Our study has several limitations. First, we used relativelyyoung rats that underwent surgery at 9 wk and were studiedafter 21 wk of age. Therefore, our observations cannot beextrapolated to older rats. However, in light of the severity ofdiabetes mellitus, we minimized the risk of increased periop-erative infections and lethality that was observed in olderanimals with advanced diabetic complications (data notshown). Second, we left the diabetes mellitus untreated for 12wk in the ZDF rats and did not attempt to alter the course ofdisease using dietary intervention, oral antidiabetic drugs, orinsulin. Certainly, the rapid, rather than insidious, onset of type

2 diabetes mellitus with persistently elevated serum glucoselevels of 40 mmol/l for 8 wk represents an extreme metabolicsituation that cannot fully translate into clinical medicine.However, despite these inherent differences, this model mayrecapitulate some of the endocrine and vascular consequencesof poorly controlled type 2 diabetes mellitus that may bediagnosed 5 to 10 years after its onset and is frequently onlyrecognized through cardiovascular complications. Third, wedid not assess chondrocyte functions and involved pathways(14, 15) or the effect of altered enzymatic and nonenzymaticcollagen cross-linking on bone strength (25), two aspects thatmay contribute to poor bone quality in type 2 diabetes. Fourth,we employed whole bone marrow rather than more highlyenriched precursors for the in vitro differentiation assays.Despite these limitations, most findings of our study may berelevant also for skeletal health in patients with diabetesmellitus.

In conclusion, our study identified suppressed osteoblasto-genesis as a key mechanism for low bone mass and impairedbone regeneration in a rat model of type 2 diabetes mellituswhile bone matrix mineralization is intact. Local or systemicstrategies that specifically target this mechanism may improvebone health and bone regeneration in diabetes.

ACKNOWLEDGMENTS

We thank Nicole Pacyna for technical assistance and Gerda Dinst, PhaedraMessmer, Daniela Gabriel, Petra Keplinger, and Sonja Lueger for carefulsample preparations and qBEI measurements.

GRANTS

This work was supported by grants from the Elsbeth Bonhoff Foundation(C. Hamann, C. Goettsch, and L. C. Hofbauer), a Center for RegenerativeTherapies Dresden seed grant (C. Hamann, R. Bernhardt, K.-P. Günther, andL. C. Hofbauer), by grants from Deutsche Forschungsgemeinschaft Transre-gio-67 (U. Hempel, R. Bernhardt, S. Rammelt, and L. C. Hofbauer), RA1923/1-1 (M. Rauner), GO 1801/4-1 (C. Goettsch), and HO 1875/8-1 (L. C.Hofbauer), and by the Austrian Workers’ Compensation Board and the ViennaHealth Insurance Fund (N. Fratzl-Zelman and P. Roschger).

DISCLOSURES

The authors have no conflict of interest.

REFERENCES

1. American Diabetes Association. Economic Costs of Diabetes in the U.S.in 2007. Diabetes Care 31: 596–615, 2008.

2. Botolin S, Faugere MC, Malluche H, Orth M, Meyer R, McCabe LR.Increased bone adiposity and peroxisomal proliferator-activated receptor-gamma2 expression in type I diabetic mice. Endocrinology 146: 3622–3631, 2005.

3. Chaudhary SB, Liporace FA, Gandhi A, Donley BG, Pinzur MS, LinSS. Complications of ankle fracture in patients with diabetes. J Am AcadOrthop Surg 16: 159–170, 2008.

4. Dienelt A, Zur Nieden NI. Hyperglycemia impairs skeletogenesis fromembryonic stem cells by affecting osteoblast and osteoclast differentiation.Stem Cells Dev 20: 465–474, 2011.

5. Fratzl-Zelman N, Morello R, Lee B, Rauch F, Glorieux FH, MisofBM, Klaushofer K, Roschger P. CRTAP deficiency leads to abnormallyhigh bone matrix mineralization in a murine model and in children withosteogenesis imperfecta type VII. Bone 46: 820–826, 2010.

6. Fujii H, Hamada Y, Fukagawa M. Bone formation in spontaneouslydiabetic Torii-newly established model of non-obese type 2 diabetes rats.Bone 42: 372–379, 2008.

7. Gopalakrishnan V, Vignesh RC, Arunakaran J, Aruldhas MM, Srini-vasan N. Effects of glucose and its modulation by insulin and estradiol onBMSC differentiation into osteoblastic lineages. Biochem Cell Biol 84:93–101, 2006.

E1227BONE REGENERATION IN DIABETES MELLITUS

AJP-Endocrinol Metab • VOL 301 • DECEMBER 2011 • www.ajpendo.org

by 10.220.33.1 on May 1, 2017

http://ajpendo.physiology.org/D

ownloaded from

Page 9: Delayed bone regeneration and low bone mass in a rat model of insulin-resistant type 2 ... · 2017. 5. 1. · Delayed bone regeneration and low bone mass in a rat model of insulin-resistant

8. Hofbauer LC, Brueck CC, Singh SK, Dobnig H. Osteoporosis inpatients with diabetes mellitus. J Bone Miner Res 22: 1317–1328, 2007.

9. Inaba M, Terada M, Koyama H, Yoshida O, Ishimura E, KawagishiT, Okuno Y, Nishizawa Y, Otani S, Morii H. Influence of high glucoseon 1,25-dihydroxyvitamin D3-induced effect on human osteoblast-likeMG-63 cells. J Bone Miner Res 10: 1050–1056, 1995.

10. Jang WG, Kim EJ, Bae IH, Lee KN, Kim YD, Kim DK, Kim SH, LeeCH, Franceschi RT, Choi HS, Koh JT. Metformin induces osteoblastdifferentiation via orphan nuclear receptor SHP-mediated transactivationof Runx2. Bone 48: 885–893, 2010.

11. Jeffcoate WJ, Rasmussen LM, Hofbauer LC, Game FL. Medial arterialcalcification in diabetes and its relationship to neuropathy. Diabetologia52: 2478–2488, 2009.

12. Kasahara T, Imai S, Kojima H, Katagi M, Kimura H, Chan L,Matsusue Y. Malfunction of bone marrow-derived osteoclasts and thedelay of bone fracture healing in diabetic mice. Bone 47: 617–625, 2010.

13. Kawai M, Sousa KM, MacDougald OA, Rosen CJ. The many facets ofPPARgamma: novel insights for the skeleton. Am J Physiol EndocrinolMetab 299: E3–E9, 2010.

14. Kayal RA, Alblowi J, McKenzie E, Krothapalli N, Silkman L, Ger-stenfeld L, Einhorn TA, Graves DT. Diabetes causes the accelerated lossof cartilage during fracture repair which is reversed by insulin treatment.Bone 44: 357–363, 2009.

15. Kayal RA, Siqueira M, Alblowi J, McLean J, Krothapalli N, FaibishD, Einhorn TA, Gerstenfeld LC, Graves DT. TNF-alpha mediatesdiabetes-enhanced chondrocyte apoptosis during fracture healing andstimulates chondrocyte apoptosis through FOXO1. J Bone Miner Res 25:1604–1615, 2010.

16. Kim JM, Jeong D, Kang HK, Jung SY, Kang SS, Min BM. Osteoclastprecursors display dynamic metabolic shifts toward accelerated glucosemetabolism at an early stage of RANKL-stimulated osteoclast differenti-ation. Cell Physiol Biochem 20: 935–946, 2007.

17. Liu Z, Aronson J, Wahl EC, Liu L, Perrien DS, Kern PA, Fowlkes JL,Thrailkill KM, Bunn RC, Cockrell GE, Skinner RA, Lumpkin CK Jr.A novel rat model for the study of deficits in bone formation in type-2diabetes. Acta Orthop 78: 46–55, 2007.

18. Lu H, Kraut D, Gerstenfeld LC, Graves DT. Diabetes interferes withthe bone formation by affecting the expression of transcription factors thatregulate osteoblast differentiation. Endocrinology 144: 346–352, 2003.

19. Lutter AH, Hempel U, Wolf-Brandstetter C, Garbe AI, Goettsch C,Hofbauer LC, Jessberger R, Dieter P. A novel resorption assay forosteoclast functionality based on an osteoblast-derived native extracellularmatrix. J Cell Biochem 109: 1025–1032, 2010.

20. Nagasaka S, Murakami T, Uchikawa T, Ishikawa SE, Saito T. Effectof glycemic control on calcium and phosphorus handling and parathyroidhormone level in patients with non-insulin-dependent diabetes mellitus.Endocr J 42: 377–383, 1995.

21. Reinwald S, Peterson RG, Allen MR, Burr DB. Skeletal changesassociated with the onset of type 2 diabetes in the ZDF and ZDSD rodentmodels. Am J Physiol Endocrinol Metab 296: E765–E774, 2009.

22. Retzepi M, Donos N. The effect of diabetes mellitus on osseous healing.Clin Oral Implants Res 21: 673–681, 2010.

23. Roschger P, Paschalis EP, Fratzl P, Klaushofer K. Bone mineralizationdensity distribution in health and disease. Bone 42: 456–466, 2008.

24. Ruffoni D, Fratzl P, Roschger P, Phipps R, Klaushofer K, WeinkamerR. Effect of temporal changes in bone turnover on the bone mineralizationdensity distribution: a computer simulation study. J Bone Miner Res 23:1905–1914, 2008.

25. Saito M, Fujii K, Mori Y, Marumo K. Role of collagen enzymatic andglycation induced cross-links as a determinant of bone quality in sponta-neously diabetic WBN/Kob rats. Osteoporos Int 17: 1514–1523, 2006.

26. Saito M, Marumo K. Collagen cross-links as a determinant of bonequality: a possible explanation for bone fragility in aging, osteoporosis,and diabetes mellitus. Osteoporos Int 21: 195–214, 2010.

27. Vora JP, Zimsen SM, Houghton DC, Anderson S. Evolution of meta-bolic and renal changes in the ZDF/Drt-fa rat model of type II diabetes. JAm Soc Nephrol 7: 113–117, 1996.

28. Wang F, Song YL, Li DH, Li CX, Wang Y, Zhang N, Wang BG. Type2 diabetes mellitus impairs bone healing of dental implants in GK rats.Diabetes Res Clin Pract 88: 7–9, 2010.

29. Williams JP, Blair HC, McDonald JM, McKenna MA, Jordan SE,Williford J, Hardy RW. Regulation of osteoclastic bone resorption byglucose. Biochem Biophys Res Commun 235: 646–651, 1997.

30. Wittrant Y, Gorin Y, Woodruff K, Horn D, Abboud HE, Mohan S,Abboud-Werner SL. High d(�)glucose concentration inhibits RANKL-induced osteoclastogenesis. Bone 42: 1122–1130, 2008.

E1228 BONE REGENERATION IN DIABETES MELLITUS

AJP-Endocrinol Metab • VOL 301 • DECEMBER 2011 • www.ajpendo.org

by 10.220.33.1 on May 1, 2017

http://ajpendo.physiology.org/D

ownloaded from