Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Bone Miner Res ; 38(8): 1154-1174, 2023 08.
Article in English | MEDLINE | ID: mdl-37221143

ABSTRACT

The gut microbiome impacts bone mass, which implies a disruption to bone homeostasis. However, it is not yet clear how the gut microbiome affects the regulation of bone mass and bone quality. We hypothesized that germ-free (GF) mice have increased bone mass and decreased bone toughness compared with conventionally housed mice. We tested this hypothesis using adult (20- to 21-week-old) C57BL/6J GF and conventionally raised female and male mice (n = 6-10/group). Trabecular microarchitecture and cortical geometry were measured from micro-CT of the femur distal metaphysis and cortical midshaft. Whole-femur strength and estimated material properties were measured using three-point bending and notched fracture toughness. Bone matrix properties were measured for the cortical femur by quantitative back-scattered electron imaging and nanoindentation, and, for the humerus, by Raman spectroscopy and fluorescent advanced glycation end product (fAGE) assay. Shifts in cortical tissue metabolism were measured from the contralateral humerus. GF mice had reduced bone resorption, increased trabecular bone microarchitecture, increased tissue strength and decreased whole-bone strength that was not explained by differences in bone size, increased tissue mineralization and fAGEs, and altered collagen structure that did not decrease fracture toughness. We observed several sex differences in GF mice, most notably for bone tissue metabolism. Male GF mice had a greater signature of amino acid metabolism, and female GF mice had a greater signature of lipid metabolism, exceeding the metabolic sex differences of the conventional mice. Together, these data demonstrate that the GF state in C57BL/6J mice alters bone mass and matrix properties but does not decrease bone fracture resistance. © 2023 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Bone and Bones , Fractures, Bone , Female , Male , Mice , Animals , Mice, Inbred C57BL , Bone and Bones/diagnostic imaging , Bone and Bones/metabolism , Bone Density/physiology , Bone Matrix/metabolism , Fractures, Bone/metabolism
2.
J Steroid Biochem Mol Biol ; 231: 106314, 2023 07.
Article in English | MEDLINE | ID: mdl-37088440

ABSTRACT

Vitamin D contributes to the development and maintenance of bone. Evidence suggests vitamin D status can also alter energy balance and gut health. In young animals, vitamin D deficiency (VDD) negatively affects bone mineral density (BMD) and bone microarchitecture, and these effects may also occur due to chronic ethanol intake. However, evidence is limited in mature models, and addressing this was a goal of the current study. Seven-month-old female C57BL/6 mice (n = 40) were weight-matched and randomized to one of four ad libitum diets: control, alcohol (Alc), vitamin D deficient (0 IU/d), or Alc+VDD for 8 weeks. A purified (AIN-93) diet was provided with water or alcohol (10 %) ad libitum. Body weight and food intake were recorded weekly, and feces were collected at 0, 4, and 8 weeks. At the age of 9 months, intestinal permeability was assessed by oral gavage of fluorescein isothiocyanate-dextran. Thereafter, bone mineral density (BMD) was measured by dual-energy X-ray absorptiometry. The microarchitecture of the distal femur was assessed by micro-computed tomography and biomechanical properties were evaluated by cyclic reference point indentation. VDD did not affect BMD or most bone microarchitecture parameters, however, the polar moment of inertia (p < 0.05) was higher in the VDD groups compared to vitamin D sufficient groups. VDD mice also had lower whole bone water content (p < 0.05) and a greater average unloading slope (p < 0.01), and energy dissipated (p < 0.01), indicating the femur displayed a brittle phenotype. In addition, VDD caused a greater increase in energy intake (p < 0.05), weight gain (p < 0.05), and a trend for higher intestinal permeability (p = 0.08). The gut microbiota of the VDD group had a reduction in alpha diversity (p < 0.05) and a lower abundance of ASVs from Rikenellaceae, Clostridia_UCG-014, Oscillospiraceae, and Lachnospiraceae (p < 0.01). There was little to no effect of alcohol supplementation on outcomes. Overall, these findings suggest that vitamin D deficiency causes excess weight gain and reduces the biomechanical strength of the femur as indicated by the higher average unloading slope and energy dissipated without an effect on BMD in a mature murine model.


Subject(s)
Bone Density , Vitamin D Deficiency , Animals , Female , Mice , Diet , Ethanol/pharmacology , Mice, Inbred C57BL , Vitamin D/pharmacology , Vitamins/pharmacology , Weight Gain , X-Ray Microtomography
3.
Bone Rep ; 17: 101634, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36389626

ABSTRACT

Chemical crosslinks known as advanced glycation end-products (AGEs) are associated with increased bone fracture risk and deteriorated bone mechanical properties. However, measurement of bone AGEs via ex vivo and in vitro methods has been limited to quantification of bulk fluorescent AGEs (fAGEs) and pentosidine only, which is a crosslinking fluorescent AGE. However, a non-crosslinking and non-fluorescent AGE such as carboxymethyl-lysine (CML) is found to be 40-100 times higher in quantity than pentosidine, but only one previous study has reported it in cortical bone, and one study reported it in trabecular bone. In our study, we wanted to investigate if accumulation of CML differs in cortical and trabecular compartments and if they are more strongly associated with bone mechanical properties than with fAGEs. We hypothesized that CML and fAGEs level would be higher in the trabecular compartment and show negative correlations to mechanical properties in cortical and trabecular bone. We obtained human cadaveric cortical and trabecular bone specimens, induced the formation of AGEs via the established in vitro ribosylation method, imaged specimens by microcomputed tomography to assess specimen geometry and microarchitecture, and mechanically tested cortical specimens by cyclic reference point indentation and fracture toughness tests and trabecular specimens by compression tests, followed by measurement of fAGEs and CML. fAGEs were 22 % higher in cortical bone (687 ± 44.8 ng Q/mg collagen) compared to trabecular bone (859 ± 317.1 ng Q/mg collagen), whereas CML levels were found to be 148 % higher in trabecular bone (6189.9 ± 866 ng/mg of protein) compared to cortical bone (924.6 ± 576.3 ng/mg of protein). Pooling the specimens from both the control and ribose groups, Spearman correlation analysis indicated that CML levels, but not fAGEs, are moderately associated with cortical porosity (r = +0.505, p ≤ 0.05) and mechanical properties such indentation depth (r = +0.460, p ≤ 0.05), total indentation depth (r = +0.440, p ≤ 0.05), and average energy dissipated (r = +0.465, p ≤ 0.05) in cortical bone. fAGEs showed a trend towards negative association with crack propagation toughness in cortical bone (r = -0.365, p = 0.055). No significant correlations were observed between CML and microarchitecture or mechanical properties in trabecular bone. CML levels were also associated with fAGEs in cortical bone (r = +0.596, p ≤ 0.05) but not in trabecular bone. Our preliminary findings indicate that CML, a non-crosslinking AGE, may affect bone material and mechanical properties differently than bulk fluorescent AGEs, given the higher accumulation of CML in each bone compartment. This study provides direction to future studies to quantify crosslinking and non-crosslinking AGEs separately as their effect on material and mechanical properties may be different and it would help identify better biomarkers for bone strength prediction.

4.
Bone Rep ; 15: 101137, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34660852

ABSTRACT

As both anabolic and anti-catabolic osteoporosis drugs affect bone formation and resorption processes, they may contribute to bone's overall mechanical behavior by altering the quality of the bone matrix. We used an ovariectomized rat model and a novel fracture mechanics approach to investigate whether treatment with an anabolic (parathyroid hormone) or anti-catabolic (alendronate) osteoporosis drugs will alter the organic and mineral matrix components and consequently cortical bone fracture toughness. Ovariectomized (at 5 months age) rats were treated with either parathyroid hormone or alendronate at low and high doses for 6 months (age 6-12 months). Specifically, treatment groups included untreated ovariectomized controls (n = 9), high-dose alendronate (n = 10), low-dose alendronate (n = 9), high-dose parathyroid hormone (n = 10), and low-dose parathyroid hormone (n = 9). After euthanasia, cortical microbeams from the lateral quadrant were extracted, notched, and tested in 3-point bending to measure fracture toughness. Portions of the bone were used to measure changes in the 1) organic matrix through quantification of advanced glycation end-products (AGEs) and non-collagenous proteins, and 2) mineral matrix through assessment of mineral crystallinity. Compared to the ovariectomized group, rats treated with high doses of parathyroid hormone and alendronate had significantly increased cortical bone fracture toughness, which corresponded primarily to increased non-collagenous proteins while there was no change in AGEs. Additionally, low-dose PTH treatment increased matrix crystallinity and decreased AGE levels. In summary, ovariectomized rats treated with pharmaceutical drugs had increased non-collagenous matrix proteins and improved fracture toughness compared to controls. Further investigation is required for different doses and longer treatment periods.

5.
J Biomech ; 125: 110600, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34246065

ABSTRACT

Diabetes is associated with increased fracture risk in human bone, especially in the elderly population. In the present study, we investigate how simulated advanced glycation end-products (AGEs) and materials heterogeneity affect crack growth trajectory in human cortical bone. We used a phase field fracture framework on 2D models of cortical microstructure created from human tibias to analyze crack propagation. The increased AGEs level results in a higher rate of crack formation. The simulations also indicate that the mismatch between the fracture properties (e.g., critical energy release rate) of osteons and interstitial tissue can alter the post-yielding behavior. The results show that if the critical energy release rate of cement lines is lower than that of osteons and the surrounding interstitial matrix, cracks can be arrested by cement lines. Additionally, activation of toughening mechanisms such as crack merging and branching depends on bone microstructural morphology (i.e., osteons geometrical parameters, canals, and lacunae porosities). In conclusion, the present findings suggest that materials heterogeneity of microstructural features and the crack-microstructure interactions can play important roles in bone fragility.


Subject(s)
Fractures, Bone , Models, Biological , Aged , Bone and Bones , Cortical Bone , Haversian System , Humans
6.
Proc Natl Acad Sci U S A ; 117(22): 12029-12040, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32404427

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is a uniformly fatal condition that is especially prevalent in skin, cardiovascular, and musculoskeletal systems. A wide gap exists between our knowledge of the disease and a promising treatment or cure. The aim of this study was to first characterize the musculoskeletal phenotype of the homozygous G608G BAC-transgenic progeria mouse model, and to determine the phenotype changes of HGPS mice after a five-arm preclinical trial of different treatment combinations with lonafarnib, pravastatin, and zoledronic acid. Microcomputed tomography and CT-based rigidity analyses were performed to assess cortical and trabecular bone structure, density, and rigidity. Bones were loaded to failure with three-point bending to assess strength. Contrast-enhanced µCT imaging of mouse femurs was performed to measure glycosaminoglycan content, thickness, and volume of the femoral head articular cartilage. Advanced glycation end products were assessed with a fluorometric assay. The changes demonstrated in the cortical bone structure, rigidity, stiffness, and modulus of the HGPS G608G mouse model may increase the risk for bending and deformation, which could result in the skeletal dysplasia characteristic of HGPS. Cartilage abnormalities seen in this HGPS model resemble changes observed in the age-matched WT controls, including early loss of glycosaminoglycans, and decreased cartilage thickness and volume. Such changes might mimic prevalent degenerative joint diseases in the elderly. Lonafarnib monotherapy did not improve bone or cartilage parameters, but treatment combinations with pravastatin and zoledronic acid significantly improved bone structure and mechanical properties and cartilage structural parameters, which ameliorate the musculoskeletal phenotype of the disease.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Disease Models, Animal , Lamin Type A/genetics , Progeria , Aging/drug effects , Aging/pathology , Animals , Bone and Bones/drug effects , Bone and Bones/pathology , Cartilage/drug effects , Cartilage/pathology , Femur/drug effects , Femur/pathology , Glycosaminoglycans/analysis , Joints/drug effects , Joints/pathology , Lamin Type A/metabolism , Mice , Mice, Transgenic , Mutation , Osteoarthritis/drug therapy , Osteoarthritis/pathology , Phenotype , Piperidines/therapeutic use , Pravastatin/therapeutic use , Progeria/drug therapy , Progeria/genetics , Protein Processing, Post-Translational/drug effects , Pyridines/therapeutic use , X-Ray Microtomography , Zoledronic Acid/therapeutic use
7.
Bone ; 136: 115369, 2020 07.
Article in English | MEDLINE | ID: mdl-32320892

ABSTRACT

Elevation of bone fluoride levels due to drinking beverages with high fluoride content or other means such as inhalation can result in skeletal fluorosis and lead to increased joint pain, skeletal deformities, and fracture. Because skeletal fluorosis alters bone's mineral composition, it is likely to affect bone's tissue-level mechanical properties with consequent effects on whole bone mechanical behavior. To investigate this, we determined whether incubation with in vitro sodium fluoride (NaF) altered bone's mechanical behavior at both the tissue- and whole bone-levels using cyclic reference point indentation (cRPI) and traditional 3-point bending, respectively. Forty-two ulnas from female adult rats (5-6 months) were randomly divided into 5 groups (vehicle, 0.05 M NaF, 0.25 M NaF, 0.75 M NaF, and 1.5 M NaF). Bones were washed in a detergent solution to remove organic barriers to ion exchange and incubated in respective treatment solutions (12 h, 23 °C). Cortical tissue mineral density (TMD) and geometry at the mid-diaphysis were determined by microCT. cRPI was performed on the distal diaphysis (9 N, 2 Hz, 10 cycles), and then bones were tested in 3-point bending to assess whole bone mechanical properties. The incubations in vehicle (0 M) up to 1.5 M in vitro NaF concentrations achieved bone fluoride levels ranging from approximately 0.70 to 15.8 ppm. NaF-incubated bones had significantly greater indentation distances, higher displacement-to-maximum force, and lower estimated elastic modulus, ultimate stress, and bending rigidity with increasing NaF concentration compared to vehicle-incubated bones. cRPI variables were moderately correlated to whole bone mechanical properties such that higher indentation distances were associated with lower estimated elastic modulus, ultimate stress, and bending rigidity. In conclusion, in vitro NaF incubation mostly has a deleterious effect on bone mechanical behavior with increasing NaF levels that is independent of bone turnover and reflected, in part, by less resistance of the tissue to cRPI-based indentation.


Subject(s)
Fluorides , Fractures, Bone , Animals , Bone Remodeling , Bone and Bones/diagnostic imaging , Female , Rats , Sodium Fluoride
8.
J Orthop Res ; 38(5): 972-983, 2020 05.
Article in English | MEDLINE | ID: mdl-31793028

ABSTRACT

Advanced glycation end-products (AGEs) have been suggested to contribute to bone fragility in type 2 diabetes (T2D). AGEs can be induced through in vitro sugar incubations but there is limited data on the effect of total fluorescent AGEs on mechanical properties of human cortical bone, which may have altered characteristics in T2D. Thus, to examine the effect of AGEs on bone directly in T2D patients with uncontrolled sugar levels, it is essential to first understand the fundamental mechanisms by studying the effects of controlled in vitro-induced AGEs on cortical bone mechanical behavior. Here, human cortical bone specimens from female cadaveric tibias (ages 57-87) were incubated in an in vitro 0.6 M ribose or vehicle solution (n = 20/group) for 10 days at 37°C, their mechanical properties were assessed by microindentation and fracture toughness tests, and induced AGE levels were quantified through a fluorometric assay. Results indicated that ribose-incubated bone had significantly more AGEs (+81%, p ≤ 0.005), lower elastic modulus assessed by traditional microindentation, and lower fracture toughness compared with vehicle controls. Furthermore, based on pooled data, increased AGEs were significantly correlated with deteriorated mechanical properties. The findings presented here show that the accumulation of AGEs allows for lower stiffness and increased ability to initiate a crack in human cortical bone. Statement of clinical significance: High sugar levels as in T2D results in deteriorated bone quality via AGE accumulation with a consequent weakening in bone's mechanical integrity. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:972-983, 2020.


Subject(s)
Bone and Bones/drug effects , Elastic Modulus/drug effects , Glycation End Products, Advanced/metabolism , Ribose/toxicity , Aged , Aged, 80 and over , Bone and Bones/metabolism , Bone and Bones/ultrastructure , Female , Humans , Middle Aged
9.
Curr Osteoporos Rep ; 17(5): 291-300, 2019 10.
Article in English | MEDLINE | ID: mdl-31392668

ABSTRACT

PURPOSE OF REVIEW: There is ample evidence that patients with type 2 diabetes (T2D) have increased risk of fracture even though they have normal or high bone mineral density. As a result, poor bone quality is suggested to contribute to skeletal fragility in this population. Thus, our goal was to conduct a comprehensive literature review to understand how bone quality components are altered in T2D and their effects on bone biomechanics and fracture risk. RECENT FINDINGS: T2D does affect bone quality via alterations in bone microarchitecture, organic matrix, and cellular behavior. Further, studies indicate that bone biomechanical properties are generally deteriorated in T2D, but there are few reports in patients. Additional work is needed to better understand molecular and cellular mechanisms that contribute to skeletal fragility in T2D. This knowledge can contribute to the development of improved diagnostic tools and drug targets to for improved quality of life for those with T2D.


Subject(s)
Diabetes Mellitus, Type 2/complications , Fractures, Bone/etiology , Biomechanical Phenomena , Bone Density , Humans , Risk Factors
10.
Bone ; 114: 32-39, 2018 09.
Article in English | MEDLINE | ID: mdl-29857063

ABSTRACT

Skeletal fragility is a major complication of type 2 diabetes mellitus (T2D), but there is a poor understanding of mechanisms underlying T2D skeletal fragility. The increased fracture risk has been suggested to result from deteriorated bone microarchitecture or poor bone quality due to accumulation of advanced glycation end-products (AGEs). We conducted a clinical study to determine whether: 1) bone microarchitecture, AGEs, and bone biomechanical properties are altered in T2D bone, 2) bone AGEs are related to bone biomechanical properties, and 3) serum AGE levels reflect those in bone. To do so, we collected serum and proximal femur specimens from T2D (n = 20) and non-diabetic (n = 33) subjects undergoing total hip replacement surgery. A section from the femoral neck was imaged by microcomputed tomography (microCT), tested by cyclic reference point indentation, and quantified for AGE content. A trabecular core taken from the femoral head was imaged by microCT and subjected to uniaxial unconfined compression tests. T2D subjects had greater HbA1c (+23%, p ≤ 0.0001), but no difference in cortical tissue mineral density, cortical porosity, or trabecular microarchitecture compared to non-diabetics. Cyclic reference point indentation revealed that creep indentation distance (+18%, p ≤ 0.05) and indentation distance increase (+20%, p ≤ 0.05) were greater in cortical bone from T2D than in non-diabetics, but no other indentation variables differed. Trabecular bone mechanical properties were similar in both groups, except for yield stress, which tended to be lower in T2D than in non-diabetics. Neither serum pentosidine nor serum total AGEs were different between groups. Cortical, but not trabecular, bone AGEs tended to be higher in T2D subjects (21%, p = 0.09). Serum AGEs and pentosidine were positively correlated with cortical and trabecular bone AGEs. Our study presents new data on biomechanical properties and AGEs in adults with T2D, which are needed to better understand mechanisms contributing to diabetic skeletal fragility.


Subject(s)
Bone Density/physiology , Diabetes Mellitus, Type 2/diagnostic imaging , Diabetes Mellitus, Type 2/metabolism , Femur Neck/diagnostic imaging , Femur Neck/metabolism , Glycation End Products, Advanced/metabolism , Adult , Aged , Arthroplasty, Replacement, Hip/trends , Biomechanical Phenomena/physiology , Female , Glycation End Products, Advanced/analysis , Humans , Male , Middle Aged , X-Ray Microtomography/methods
11.
Bone ; 106: 90-95, 2018 Jan.
Article in English | MEDLINE | ID: mdl-25862290

ABSTRACT

Although low bone mineral density (BMD) is strongly associated with increased fracture risk, up to 50% of those who suffer fractures are not detected as high-risk patients by BMD testing. Thus, new approaches may improve identification of those at increased risk for fracture by in vivo assessment of altered bone tissue properties, which may contribute to skeletal fragility. Recently developed reference point indentation (RPI) allows for assessment of cortical bone indentation properties in vivo using devices that apply cyclic loading or impact loading, but there is little information available to assist with interpretation of RPI measurements. Our goals were to use human cadaveric tibia to determine: 1) the associations between RPI variables, cortical bone density, and morphology; 2) the association between variables obtained from RPI systems using cyclic, slow loading versus a single impact load; and 3) age-related differences in RPI variables. We obtained 20 human tibia and femur pairs from female donors (53-97years), measured total hip BMD using dual-energy X-ray absorptiometry, assessed tibial cortical microarchitecture using high-resolution peripheral quantitative computed tomography (HR-pQCT), and assessed cortical bone indentation properties at the mid-tibial diaphysis using both the cyclic and impact-based RPI systems (Biodent and Osteoprobe, respectively, Active Life Scientific, Santa Barbara, CA). We found a few weak associations between RPI variables, BMD, and cortical geometry; a few weak associations between measurements obtained by the two RPI systems; and no age-related differences in RPI variables. Our findings indicate that in cadaveric tibia from older women RPI measurements are largely independent of age, femoral BMD, and cortical geometry. Furthermore, measurements from the cyclic and impact loading RPI devices are weakly related to each other, indicating that each device reflects different aspects of cortical bone indentation properties.


Subject(s)
Femur Neck/diagnostic imaging , Tibia/diagnostic imaging , Absorptiometry, Photon , Aged , Aged, 80 and over , Bone Density/physiology , Female , Femur Neck/physiology , Fractures, Bone/diagnostic imaging , Fractures, Bone/physiopathology , Humans , Middle Aged , Tibia/physiology
12.
J Bone Miner Res ; 32(4): 814-820, 2017 04.
Article in English | MEDLINE | ID: mdl-27925290

ABSTRACT

Bisphosphonates are being increasingly used to treat pediatric patients with skeletal disorders. However, the effects of long-term bisphosphonate therapy and cessation of therapy during growth are unclear. Thus, studies were undertaken to determine the effects of alendronate discontinuation after treatment of C57Bl/6 mice during the period of rapid skeletal growth. Compared with vehicle-treated mice, 16 weeks of alendronate treatment starting at age 18 days resulted in a 3.7-fold increase in trabecular bone in the setting of suppressed bone formation. Alendronate therapy for 8 weeks followed by 8 weeks of vehicle treatment resulted in a more pronounced increase in trabecular bone compared with mice treated with alendronate for 16 weeks (1.7-fold) and to vehicle-treated controls (6.5-fold). Mice that received alendronate for 8 weeks followed by 8 weeks of vehicle exhibited increased osteoblast surface (2.5-fold), mineralizing surface (5.7-fold), and bone formation rate (5.1-fold) compared with mice treated continuously with alendronate. However, these parameters were not restored to the levels observed in the vehicle-treated mice. Thus, partial resumption of bone formation upon cessation of bisphosphonate therapy leads to a greater increase in trabecular bone than that found when bisphosphonates are administered continuously to growing mice. These data suggest that intermittent administration of bisphosphonates may optimize their beneficial effects on the growing skeleton. © 2017 American Society for Bone and Mineral Research.


Subject(s)
Bone Resorption , Diphosphonates/pharmacology , Osteoblasts , Osteogenesis/drug effects , Animals , Bone Resorption/drug therapy , Bone Resorption/metabolism , Bone Resorption/pathology , Male , Mice , Osteoblasts/metabolism , Osteoblasts/pathology
13.
Injury ; 47 Suppl 2: S11-20, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27338221

ABSTRACT

This review will define the role of collagen and within-bone heterogeneity and elaborate the importance of trabecular and cortical architecture with regard to their effect on the mechanical strength of bone. For each of these factors, the changes seen with osteoporosis and ageing will be described and how they can compromise strength and eventually lead to bone fragility.


Subject(s)
Bone Remodeling/physiology , Collagen/metabolism , Osteocytes/physiology , Osteoporosis/physiopathology , Aging , Biomechanical Phenomena , Bone Density , Humans , Weight-Bearing
14.
Bone ; 82: 21-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26211993

ABSTRACT

There is clear evidence that patients with type 2 diabetes mellitus (T2D) have increased fracture risk, despite having high bone mineral density (BMD) and body mass index (BMI). Thus, poor bone quality has been implicated as a mechanism contributing to diabetic skeletal fragility. Poor bone quality in T2D may result from the accumulation of advanced glycation end-products (AGEs), which are post-translational modifications of collagen resulting from a spontaneous reaction between extracellular sugars and amino acid residues on collagen fibers. This review discusses what is known and what is not known regarding AGE accumulation and diabetic skeletal fragility, examining evidence from in vitro experiments to simulate a diabetic state, ex vivo studies in normal and diabetic human bone, and diabetic animal models. Key findings in the literature are that AGEs increase with age, affect bone cell behavior, and are altered with changes in bone turnover. Further, they affect bone mechanical properties and microdamage accumulation, and can be inhibited in vitro by various inhibitors and breakers (e.g. aminoguanidine, N-Phenacylthiazolium Bromide, vitamin B6). While a few studies report higher AGEs in diabetic animal models, there is little evidence of AGE accumulation in bone from diabetic patients. There are several limitations and inconsistencies in the literature that should be noted and studied in greater depth including understanding the discrepancies between glycation levels across reported studies, clarifying differences in AGEs in cortical versus cancellous bone, and improving the very limited data available regarding glycation content in diabetic animal and human bone, and its corresponding effect on bone material properties in T2D.


Subject(s)
Bone Density/physiology , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/metabolism , Glycation End Products, Advanced/metabolism , Animals , Biomechanical Phenomena/physiology , Diabetes Mellitus, Type 2/diagnosis , Fractures, Bone/diagnosis , Fractures, Bone/epidemiology , Fractures, Bone/metabolism , Humans , Osteoporosis/diagnosis , Osteoporosis/epidemiology , Osteoporosis/metabolism
15.
Osteoporos Int ; 26(3): 865-873, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25326375

ABSTRACT

UNLABELLED: To better understand the association between different components of bone quality, we investigated the relationship among in vivo generated non-enzymatic glycation, resorption, and microdamage. The results showed negative correlation between advanced glycation end-products (AGEs) and resorption independent of age highlighting the interaction between these parameters that may lead to bone fragility. INTRODUCTION: Changes in the quality of bone material contribute significantly to bone fragility. In order to establish a better understanding of the interaction of the different components of bone quality and their influence on bone fragility, we investigated the relationship between non-enzymatic glycation, resorption, and microdamage generated in vivo in cortical bone using bone specimens from the same donors. METHODS: Total fluorescent advanced glycation end-products (AGEs) were measured in 96 human cortical bone samples from 83 donors. Resorption pit density, average resorption pit area, and percent resorption area were quantified in samples from 48 common donors with AGE measurements. Linear microcrack density and diffuse damage were measured in 21 common donors with AGE and resorption measurements. Correlation analyses were performed between all measured variables to establish the relationships among them and their variation with age. RESULTS: We found that average resorption pit area and percent resorption area decreased with increasing AGEs independently of age. Resorption pit density and percent resorption area demonstrated negative age-adjusted correlation with diffuse damage. Furthermore, average resorption pit area, resorption pit density, and percent resorption area were found to decrease significantly with age. CONCLUSIONS: The current study demonstrated the in vivo interrelationship between the organic constituents, remodeling, and damage formation in cortical bone. In addition to the age-related reduction in resorption, there is a negative correlation between AGEs and resorption independent of age. This inverse relationship indicates that AGEs alter the resorption process and/or accumulate in the tissue as a result of reduced resorption and may lead to bone fragility by adversely affecting fracture resistance through altered bone matrix properties.


Subject(s)
Bone Resorption/metabolism , Glycation End Products, Advanced/metabolism , Tibia/metabolism , Adult , Aged , Aged, 80 and over , Aging/pathology , Aging/physiology , Bone Resorption/pathology , Bone Resorption/physiopathology , Female , Humans , Male , Middle Aged , Tibia/injuries , Tibia/pathology , Tibia/physiopathology , Young Adult
16.
J Bone Miner Res ; 29(5): 1025-40, 2014.
Article in English | MEDLINE | ID: mdl-24585709

ABSTRACT

Evidence indicating that adult type 2 diabetes (T2D) is associated with increased fracture risk continues to mount. Unlike osteoporosis, diabetic fractures are associated with obesity and normal to high bone mineral density, two factors that are typically associated with reduced fracture risk. Animal models will likely play a critical role in efforts to identify the underlying mechanisms of skeletal fragility in T2D and to develop preventative treatments. In this review we critically examine the ability of current rodent models of T2D to mimic the skeletal characteristics of human T2D. We report that although there are numerous rodent models of T2D, few have undergone thorough assessments of bone metabolism and strength. Further, we find that many of the available rodent models of T2D have limitations for studies of skeletal fragility in T2D because the onset of diabetes is often prior to skeletal maturation and bone mass is low, in contrast to what is seen in adult humans. There is an urgent need to characterize the skeletal phenotype of existing models of T2D, and to develop new models that more closely mimic the skeletal effects seen in adult-onset T2D in humans.


Subject(s)
Bone Diseases/metabolism , Diabetes Complications/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Animals , Bone Diseases/pathology , Diabetes Complications/pathology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/pathology , Humans , Mice , Rats
17.
J Bone Miner Metab ; 32(5): 476-83, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24114195

ABSTRACT

Charged particle radiation such as iron ions and their secondary fragmentation products are of particular concern to the skeleton due to their high charge and energy deposition. However, little is known about the long-term effects of these particles on trabecular and cortical bone morphology when applied at relatively low levels. We hypothesized that even a 4.4 cGy dose of a complex secondary iron ion radiation field will compromise skeletal quantity and architecture in adult mice. One year after radiation exposure and compared to age-matched controls, 4.4 cGy irradiated mice had 51 % more trabecular bone, 56 % greater trabecular bone volume fraction, 16 % greater trabecular number, and 17 % less trabecular separation in the distal metaphysis of the femur. Similar to the metaphysis, trabecular bone of the distal femoral epiphysis in 4.4 cGy mice had 33 % more trabecular bone, 31 % greater trabecular bone volume fraction, and a 33 % smaller structural model index. Cortical bone morphology, whole bone mechanical properties, and lower leg muscle mass were unaffected. When compared to two additional groups, irradiated at either 8.9 or 17.8 cGy, a (negative) dose response relationship was observed for trabecular bone in the metaphysis but not in the epiphysis. In contrast to our original hypothesis, these data indicated that a secondary field of low-level, high-linear energy transfer iron radiation may cause long-term augmentation, rather than deterioration, of trabecular bone in the femoral metaphysis and epiphysis of mice.


Subject(s)
Bone and Bones/anatomy & histology , Bone and Bones/radiation effects , Radiation, Ionizing , Animals , Bone and Bones/diagnostic imaging , Epiphyses/diagnostic imaging , Epiphyses/radiation effects , Male , Mice, Inbred C57BL , X-Ray Microtomography
18.
J Clin Endocrinol Metab ; 98(5): 1923-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23509107

ABSTRACT

CONTEXT: Adolescents with anorexia nervosa (AN) have low areal bone mineral density (aBMD) at both cortical and trabecular sites, and recent data show impaired trabecular microarchitecture independent of aBMD. However, data are lacking regarding both cortical microarchitecture and bone strength assessment by finite element analysis (FEA) in adolescents with AN. Because microarchitectural abnormalities and FEA may predict fracture risk independent of aBMD, these data are important to obtain. OBJECTIVE: Our objective was to compare both cortical and trabecular bone microarchitecture and FEA estimates of bone strength in adolescent girls with AN vs normal-weight controls. DESIGN, SETTING, AND SUBJECTS: We conducted a cross-sectional study at a clinical research center that included 44 adolescent girls (21 with AN and 23 normal-weight controls) 14 to 22 years old. MAIN OUTCOME MEASURES: We evaluated 1) aBMD (dual-energy x-ray absorptiometry) at the distal radius, lumbar spine, and hip, 2) cortical and trabecular microarchitecture at the ultradistal radius (high-resolution peripheral quantitative computed tomography), and 3) FEA-derived estimates of failure load at the ultradistal radius. RESULTS: aBMD was lower in girls with AN vs controls at the lumbar spine and hip but not at the distal radius. Girls with AN had lower total (P < .0001) and trabecular volumetric BMD (P = .02) and higher cortical porosity (P = .03) and trabecular separation (P = .04). Despite comparable total cross-sectional area, trabecular area was higher in girls with AN (P = .04), and cortical area and thickness were lower (P = .002 and .02, respectively). FEA-estimated failure load was lower in girls with AN (P = .004), even after controlling for distal radius aBMD. CONCLUSIONS: Both cortical and trabecular microarchitecture are altered in adolescent girls with AN. FEA-estimated failure load is decreased, indicative of reduced bone strength. The finding of reduced cortical bone area in girls with AN is consistent with impaired cortical bone formation at the endosteum as a mechanism underlying these findings.


Subject(s)
Adolescent Development , Anorexia Nervosa/pathology , Bone Development , Bone and Bones/pathology , Absorptiometry, Photon , Adolescent , Adult , Anorexia Nervosa/physiopathology , Body Mass Index , Bone Density , Bone and Bones/chemistry , Bone and Bones/diagnostic imaging , Boston/epidemiology , Chemical Phenomena , Cross-Sectional Studies , Diagnostic and Statistical Manual of Mental Disorders , Female , Finite Element Analysis , Fractures, Bone/epidemiology , Fractures, Bone/etiology , Humans , Imaging, Three-Dimensional , Porosity , Risk , Tomography, X-Ray Computed , Young Adult
19.
PLoS One ; 7(4): e35047, 2012.
Article in English | MEDLINE | ID: mdl-22514706

ABSTRACT

Non-enzymatic glycation (NEG) and enzymatic biochemical processes create crosslinks that modify the extracellular matrix (ECM) and affect the turnover of bone tissue. Because NEG affects turnover and turnover at the local level affects microarchitecture and formation and removal of microdamage, we hypothesized that NEG in cancellous bone is heterogeneous and accounts partly for the contribution of microarchitecture and microdamage on bone fragility. Human trabecular bone cores from 23 donors were subjected to compression tests. Mechanically tested cores as well as an additional 19 cores were stained with lead-uranyl acetate and imaged to determine microarchitecture and measure microdamage. Post-yield mechanical properties were measured and damaged trabeculae were extracted from a subset of specimens and characterized for the morphology of induced microdamage. Tested specimens and extracted trabeculae were quantified for enzymatic and non-enzymatic crosslink content using a colorimetric assay and Ultra-high Performance Liquid Chromatography (UPLC). Results show that an increase in enzymatic crosslinks was beneficial for bone where they were associated with increased toughness and decreased microdamage. Conversely, bone with increased NEG required less strain to reach failure and were less tough. NEG heterogeneously modified trabecular microarchitecture where high amounts of NEG crosslinks were found in trabecular rods and with the mechanically deleterious form of microdamage (linear microcracks). The extent of NEG in tibial cancellous bone was the dominant predictor of bone fragility and was associated with changes in microarchitecture and microdamage.


Subject(s)
Bone Density/physiology , Bone and Bones/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Amino Acids/metabolism , Arginine/analogs & derivatives , Arginine/metabolism , Biomechanical Phenomena/physiology , Female , Glycation End Products, Advanced/metabolism , Glycosylation , Humans , In Vitro Techniques , Lysine/analogs & derivatives , Lysine/metabolism , Male , Middle Aged , Stress, Mechanical , Tibia/metabolism , Tomography, X-Ray Computed , Young Adult
20.
Mol Cell Proteomics ; 10(9): M110.006718, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21606484

ABSTRACT

There is growing evidence supporting the need for a broad scale investigation of the proteins and protein modifications in the organic matrix of bone and the use of these measures to predict fragility fractures. However, limitations in sample availability and high heterogeneity of bone tissue cause unique experimental and/or diagnostic problems. We addressed these by an innovative combination of laser capture microscopy with our newly developed liquid chromatography separation methods, followed by gel electrophoresis and mass spectrometry analysis. Our strategy allows in-depth analysis of very limited amounts of bone material, and thus, can be important to medical sciences, biology, forensic, anthropology, and archaeology. The developed strategy permitted unprecedented biochemical analyses of bone-matrix proteins, including collagen modifications, using nearly nanoscale amounts of exceptionally homogenous bone tissue. Dissection of fully mineralized bone-tissue at such degree of homogeneity has not been achieved before. Application of our strategy established that: (1) collagen in older interstitial bone contains higher levels of an advanced glycation end product pentosidine then younger osteonal tissue, an observation contrary to the published data; (2) the levels of two enzymatic crosslinks (pyridinoline and deoxypiridinoline) were higher in osteonal than interstitial tissue and agreed with data reported by others; (3) younger osteonal bone has higher amount of osteopontin and osteocalcin then older interstitial bone and this has not been shown before. Taken together, these data show that the level of fluorescent crosslinks in collagen and the amount of two major noncollagenous bone matrix proteins differ at the level of osteonal and interstitial tissue. We propose that this may have important implications for bone remodeling processes and bone microdamage formation.


Subject(s)
Arginine/analogs & derivatives , Bone Matrix/metabolism , Chromatography, Liquid/methods , Dissection/methods , Haversian System/metabolism , Lysine/analogs & derivatives , Microscopy, Confocal/methods , Osteopontin/analysis , Proteomics/methods , Age Factors , Amino Acids/analysis , Amino Acids/chemistry , Anthropology/methods , Arginine/analysis , Arginine/chemistry , Bone Matrix/chemistry , Bone Remodeling , Calcification, Physiologic , Collagen/chemistry , Collagen/metabolism , Forensic Sciences/methods , Haversian System/chemistry , Humans , Lysine/analysis , Lysine/chemistry , Mass Spectrometry , Nanoparticles , Osteocalcin/chemistry , Osteocalcin/metabolism , Osteopontin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...