Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Appl Phys Rev ; 10(1): 011410, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36915902

RESUMO

Osteocalcin (OC), an abundant non-collagenous protein in bone extracellular matrix, plays a vital role in both its biological and mechanical function. OC undergoes post-translational modification, such as glycation; however, it remains unknown whether glycation of OC affects bone's resistance to fracture. Here, for the first time, we demonstrate the formation of pentosidine, an advanced glycation end-product (AGE) cross-link on mouse OC analyzed by ultra-performance liquid chromatography. Next, we establish that the presence of OC in mouse bone matrix is associated with lower interlamellar separation (distance) and thicker bridges spanning the lamellae, both of which are critical for maintaining bone's structural integrity. Furthermore, to determine the impact of modification of OC by glycation on bone toughness, we glycated bone samples in vitro from wild-type (WT) and osteocalcin deficient (Oc-/-) mice, and compared the differences in total fluorescent AGEs and fracture toughness between the Oc -/- glycated and control mouse bones and the WT glycated and control mouse bones. We determined that glycation resulted in significantly higher AGEs in WT compared to Oc-/- mouse bones (delta-WT > delta-OC, p = 0.025). This observed change corresponded to a significant decrease in fracture toughness between WT and Oc-/- mice (delta-WT vs delta-OC, p = 0.018). Thus, we propose a molecular deformation and fracture mechanics model that corroborates our experimental findings and provides evidence to support a 37%-90% loss in energy dissipation of OC due to formation of pentosidine cross-link by glycation. We anticipate that our study will aid in elucidating the effects of a major non-collagenous bone matrix protein, osteocalcin, and its modifications on bone fragility and help identify potential therapeutic targets for maintaining skeletal health.

2.
Curr Opin Endocrinol Diabetes Obes ; 29(4): 333-342, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35777968

RESUMO

PURPOSE OF REVIEW: Multiple biochemical and biophysical approaches have been broadly used for detection and quantitation of posttranslational protein modifications associated with diabetic bone, yet these techniques present a variety of challenges. In this review, we discuss recent advancements and complementary roles of analytical (UPLC/UPLC-MS/MS and ELISA) and biophysical (Raman and FTIR) techniques used for characterization of glycation products, measured from bone matrix and serum, and provide recommendations regarding the selection of a technique for specific study of diabetic bone. RECENT FINDINGS: Hyperglycemia and oxidative stress in diabetes contribute to the formation of a large subgroup of advanced glycation end products (AGEs) known as glycoxidation end products (AGOEs). AGEs/AGOEs have various adverse effects on bone health. Commonly, accumulation of AGEs/AGOEs leads to increased bone fragility. For example, recent studies show that carboxymethyllysine (CML) and pentosidine (PEN) are formed in bone at higher levels in certain diseases and metabolic conditions, in particular, in diabetes and aging. Detection and quantitation of AGEs/AGOEs in rare and/or precious samples is feasible because of a number of technological advancements of the past decade. SUMMARY: Recent technological advancements have led to a significant improvement of several key analytical biochemistry and biophysics techniques used for detection and characterization of AGEs/AGOEs in bone and serum. Their principles and applications to skeletal tissue studies as well as limitations are discussed in this review.


Assuntos
Doenças Ósseas , Diabetes Mellitus , Cromatografia Líquida , Produtos Finais de Glicação Avançada/metabolismo , Humanos , Espectrometria de Massas em Tandem
3.
JBMR Plus ; 6(2): e10583, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35229063

RESUMO

Obesity is a common comorbidity of type 2 diabetes (T2D). Therefore, increased risk of fragility fractures in T2D is often confounded by the effects of obesity. This study was conducted to elucidate the mechanistic basis by which T2D alone leads to skeletal fragility. We hypothesized that obesity independent T2D would deteriorate bone's material quality by accumulating defects in the mineral matrix and undesired modifications in its organic matrix associated with increased oxidative stress and hyperglycemia. To test this hypothesis, we used 15-week-old male non-obese mice with engineered muscle creatine kinase promoter/human dominant negative insulin growth factor 1 (IGF-I) receptor (MKR) and FVB/N wild-type (WT) controls (n = 12/group). MKR mice exhibit reduced insulin production and loss of glycemic control leading to diabetic hyperglycemia, verified by fasting blood glucose measurements (>250 mg/dL), without an increase in body weight. MKR mice showed a significant decrease in femoral radial geometry (cortical area, moment of inertia, cortical thickness, endosteal diameter, and periosteal diameter). Bone mineral density (BMD), as assessed by micro-computed tomography (µCT), remained unchanged; however, the quality of bone mineral was altered. In contrast to controls, MKR mice had significantly increased hydroxyapatite crystal thickness, measured by small-angle X-ray scattering, and elongated c-axis length of the crystals evaluated by confocal Raman spectroscopy. There was an increase in changes in the organic matrix of MKR mice, associated with enhanced glycoxidation (carboxymethyl-lysine [CML] and pentosidine) and overall glycation (fluorescent advanced glycation end products), both of which were associated with various measures of bone fragility. Moreover, increased CML formation positively correlated with elongated mineral crystal length, supporting the role of this negatively charged side chain to attract calcium ions, promote growth of hydroxyapatite, and build a physical link between mineral and collagen. Collectively, our results show, for the first time, changes in bone matrix in a non-obese T2D model in which skeletal fragility is attributable to alterations in the mineral quality and undesired organic matrix modifications. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

4.
Bone ; 156: 116302, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34952229

RESUMO

Poor bone quality is associated with Type 2 Diabetes (T2D), with patients having a higher risk of fracture despite normal to high bone mineral density (BMD). Diabetes contributes to modifications of the mineral and organic matrix of bone. Hyperglycemia has been linked to the formation of advanced glycation end-products (AGEs) which increase the risk for skeletal fragility fractures. To this end, we investigated diabetes-induced skeletal fragility using a high-fat diet (HFD) mouse model and evaluated the efficacy of phenacyl thiazolium chloride (PTC) for in vitro removal of glycation products to rescue bone toughness. Ten-week-old C57BL/6 J male mice (n = 6/group) were fed a HFD or low-fat diet (LFD) for 22 weeks. Mice given a HFD developed T2D and increased body mass compared to LFD-fed mice. MicroCT results showed that diabetic mice had altered microarchitecture and increased mineralization as determined by volumetric BMD and increased mineral crystal size as determined by X-ray Diffraction (XRD). Diabetic mice demonstrated loss of initiation and maximum toughness, which represent estimates of the stress intensity factor at a notch tip using yield force and ultimate force, respectively. Diabetic mice also showed higher accumulation of AGEs measured by biochemical assay (total fAGEs) and confocal Raman spectroscopy (Pentosidine (PEN), Carboxymethyl-lysine (CML)). Regression analyses confirmed the association between increased glycoxidation (CML, PEN) and loss of fracture toughness. Within the diabetic group, CML was the most significant predictor of initiation toughness while PEN predicted maximum toughness as determined by stepwise linear regression (i.e., stepAIC). Contralateral femora from HFD group were harvested and treated with PTC in vitro. PTC-treated samples showed total fAGEs decreased by 41.2%. PTC treatment partially restored bone toughness as, compared to T2D controls, maximum toughness increased by 35%. Collectively, our results demonstrate that matrix modifications in diet-induced T2D, particularly AGEs, induce bone fragility and their removal from bone matrix partially rescues T2D associated bone fragility.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Fraturas Ósseas , Animais , Densidade Óssea , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Tipo 2/complicações , Dieta Hiperlipídica , Modelos Animais de Doenças , Produtos Finais de Glicação Avançada/análise , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Minerais
5.
JBMR Plus ; 5(11): e10548, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34761150

RESUMO

It has been a challenge to establish a link between specific advanced glycation end products (AGEs) as causal agents of different pathologies and age-related diseases, primarily because of the lack of suitable in vitro experimental strategies facilitating increased formation of a specific AGE, here carboxymethyllysine (CML), over other AGEs under controlled conditions. CML is of considerable importance to various oxidative stress-related diseases, because in vivo formation of this AGE is connected with cellular oxidative/carbonyl metabolism. The mechanistic implications of CML accumulation in bone remain to be elucidated. To facilitate such studies, we developed a new in vitro strategy that allows preferential generation of CML in bone matrix over other AGEs. Using bone samples from human donors of different age (young, middle-age, and elderly), we show successful in vitro generation of the desired levels of CML and show that they mimic those observed in vivo in several bone disorders. Formation of such physiologically relevant CML levels was achieved by selecting two oxidative/carbonyl stress compounds naturally produced in the human body, glyoxal and glyoxylic acid. Kinetic studies using the two compounds revealed differences not only between their reaction rates but also in the progression and enhanced formation of CML over other AGEs (measured by their collective fluorescence as fluorescent AGEs [fAGEs]) Consequently, through the regulation of reaction time, the levels of CML and fAGEs could be controlled and separated. Given that the developed approach does not fully eliminate the formation of other uncharacterized glycation products, this could be considered as the study limitation. We expect that the concepts of our experimental approach can be used to develop diverse strategies facilitating production of the desired levels of selected AGEs in bone and other tissues, and thus, opens new avenues for investigating the role and mechanistic aspects of specific AGEs, here CML, in bone. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

6.
FASEB J ; 35(9): e21786, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34411349

RESUMO

Biological functions, including glycemic control and bone metabolism, are highly influenced by the body's internal clock. Circadian rhythms are biological rhythms that run with a period close to 24 hours and receive input from environmental stimuli, such as the light/dark cycle. We investigated the effects of circadian rhythm disruption (CRD), through alteration of the light/dark schedule, on glycemic control and bone quality of mice. Ten-week-old male mice (C57/BL6, n = 48) were given a low-fat diet (LFD) or a high-fat diet (HFD) and kept on a dayshift or altered schedule (RSS3) for 22 weeks. Mice were divided into four experimental groups (n = 12/group): Dayshift/LFD, Dayshift/HFD, RSS3/LFD, and RSS3/HFD. CRD in growing mice fed a HFD resulted in a diabetic state, with a 36.2% increase in fasting glucose levels compared to the Dayshift/LFD group. Micro-CT scans of femora revealed a reduction in inner and outer surface expansion for mice on a HFD and altered light schedule. Cancellous bone demonstrated deterioration of bone quality as trabecular number and thickness decreased while trabecular separation increased. While HFD increased cortical bone mineral density, its combination with CRD reduced this phenomenon. The growth of mineral crystals, determined by small angle X-ray scattering, showed HFD led to smaller crystals. Considering modifications of the organic matrix, regardless of diet, CRD exacerbated the accumulation of fluorescent advanced glycation end-products (fAGEs) in collagen. Strength testing of tibiae showed that CRD mitigated the higher strength in the HFD group and increased brittleness indicated by lower post-yield deflection and work-to-fracture. Consistent with accumulation of fAGEs, various measures of toughness were lowered with CRD, but combination of CRD with HFD protected against this decrease. Differences between strength and toughness results represent different contributions of structural and material properties of bone to energy dissipation. Collectively, these results demonstrate that combination of CRD with HFD impairs glycemic control and have complex effects on bone quality.


Assuntos
Glicemia/metabolismo , Osso e Ossos/fisiologia , Ritmo Circadiano , Dieta Hiperlipídica/efeitos adversos , Animais , Glicemia/efeitos dos fármacos , Densidade Óssea/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Osso Esponjoso/efeitos dos fármacos , Osso Esponjoso/fisiologia , Fêmur/efeitos dos fármacos , Fêmur/fisiologia , Masculino , Camundongos
7.
Elife ; 92020 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-33295868

RESUMO

Protein phosphorylation, critical for cellular regulatory mechanisms, is implicated in various diseases. However, it remains unknown whether heterogeneity in phosphorylation of key structural proteins alters tissue integrity and organ function. Here, osteopontin phosphorylation level declined in hypo- and hyper- phosphatemia mouse models exhibiting skeletal deformities. Phosphorylation increased cohesion between osteopontin polymers, and adhesion of osteopontin to hydroxyapatite, enhancing energy dissipation. Fracture toughness, a measure of bone's mechanical competence, increased with ex-vivo phosphorylation of wildtype mouse bones and declined with ex-vivo dephosphorylation. In osteopontin-deficient mice, global matrix phosphorylation level was not associated with toughness. Our findings suggest that phosphorylated osteopontin promotes fracture toughness in a dose-dependent manner through increased interfacial bond formation. In the absence of osteopontin, phosphorylation increases electrostatic repulsion, and likely protein alignment and interfilament distance leading to decreased fracture resistance. These mechanisms may be of importance in other connective tissues, and the key to unraveling cell-matrix interactions in diseases.


Assuntos
Osso e Ossos/fisiopatologia , Matriz Extracelular/fisiologia , Fraturas Ósseas/fisiopatologia , Osteopontina/metabolismo , Animais , Fraturas Ósseas/metabolismo , Camundongos , Fosforilação , Estresse Mecânico
8.
Bone ; 140: 115556, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32730921

RESUMO

Type 2 diabetes mellitus (T2DM), a metabolic disease on the rise, is associated with substantial increase in bone fracture risk. Because individuals with T2DM have normal or high bone mineral density (BMD), osteodensitometric measurements of BMD do not predict fracture risk with T2DM. Here, we aim to identify the underlying mechanism of the diabetes-induced fracture risk using a high-resolution multi-scale analysis of human cortical bone with special emphasis on osseous cellular activity. Specifically, we show increased cortical porosity in a subgroup of T2DM individuals accompanied by changed mineralization patterns and glycoxidative damage to bone protein, caused by non-enzymatic glycation of bone by reducing sugar. Furthermore, the high porosity T2DM subgroup presents with higher regional mineralization heterogeneity and lower mineral maturity, whereas in the T2DM subgroup regional higher mineral-to-matrix ratio was observed. Both T2DM groups show significantly higher carboxymethyl-lysine accumulation. Our results show a dimorphic pattern of cortical bone reorganization in individuals afflicted with T2DM and hence provide new insight into the diabetic bone disease leading to increased fracture risk.


Assuntos
Diabetes Mellitus Tipo 2 , Fraturas Ósseas , Densidade Óssea , Osso Cortical/diagnóstico por imagem , Fêmur/diagnóstico por imagem , Humanos
9.
J Bone Miner Res ; 34(7): 1191-1206, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30866111

RESUMO

People with type 2 diabetes mellitus (T2DM) have normal-to-high BMDs, but, counterintuitively, have greater fracture risks than people without T2DM, even after accounting for potential confounders like BMI and falls. Therefore, T2DM may alter aspects of bone quality, including material properties or microarchitecture, that increase fragility independently of bone mass. Our objective was to elucidate the factors that influence fragility in T2DM by comparing the material properties, microarchitecture, and mechanical performance of cancellous bone in a clinical population of men with and without T2DM. Cancellous specimens from the femoral neck were collected during total hip arthroplasty (T2DM: n = 31, age = 65 ± 8 years, HbA1c = 7.1 ± 0.9%; non-DM: n = 34, age = 62 ± 9 years, HbA1c = 5.5 ± 0.4%). The T2DM specimens had greater concentrations of the advanced glycation endproduct pentosidine (+ 36%, P < 0.05) and sugars bound to the collagen matrix (+ 42%, P < 0.05) than the non-DM specimens. The T2DM specimens trended toward a greater bone volume fraction (BV/TV) (+ 24%, NS, P = 0.13) and had greater mineral content (+ 7%, P < 0.05) than the non-DM specimens. Regression modeling of the mechanical outcomes revealed competing effects of T2DM on bone mechanical behavior. The trend of higher BV/TV values and the greater mineral content observed in the T2DM specimens increased strength, whereas the greater values of pentosidine in the T2DM group decreased postyield strain and toughness. The long-term medical management and presence of osteoarthritis in these patients may influence these outcomes. Nevertheless, our data indicate a beneficial effect of T2DM on cancellous microarchitecture, but a deleterious effect of T2DM on the collagen matrix. These data suggest that high concentrations of advanced glycation endproducts can increase fragility by reducing the ability of bone to absorb energy before failure, especially for the subset of T2DM patients with low BV/TV. © 2019 American Society for Bone and Mineral Research.


Assuntos
Osso Esponjoso/patologia , Osso Esponjoso/fisiopatologia , Diabetes Mellitus Tipo 2/fisiopatologia , Fenômenos Biomecânicos , Densidade Óssea , Osso Esponjoso/diagnóstico por imagem , Estudos de Coortes , Diabetes Mellitus Tipo 2/diagnóstico por imagem , Módulo de Elasticidade , Hemoglobinas Glicadas/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Análise de Regressão , Fatores de Risco , Microtomografia por Raio-X
10.
J Bone Miner Res ; 33(12): 2214-2229, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30001467

RESUMO

Phosphorylation of bone matrix proteins is of fundamental importance to all vertebrates including humans. However, it is currently unknown whether increase or decline of total protein phosphorylation levels, particularly in hypophosphatemia-related osteoporosis, osteomalacia, and rickets, contribute to bone fracture. To address this gap, we combined biochemical measurements with mechanical evaluation of bone to discern fracture characteristics associated with age-related development of skeletal fragility in relation to total phosphorylation levels of bone matrix proteins and one of the key representatives of bone matrix phosphoproteins, osteopontin (OPN). Here for the first time, we report that as people age the total phosphorylation level declines by approximately 20% for bone matrix proteins and approximately 30% for OPN in the ninth decade of human life. Moreover, our results suggest that the decline of total protein phosphorylation of extracellular matrix (ECM) contributes to bone fragility, but less pronouncedly than glycation. We theorize that the separation of two sources of OPN negative charges, acidic backbone amino acids and phosphorylation, would be nature's means of assuring that OPN functions in both energy dissipation and biomineralization. We propose that total phosphorylation decline could be an important contributor to the development of osteoporosis, increased fracture risk and skeletal fragility. Targeting the enzymes kinase FamC20 and bone alkaline phosphatase involved in the regulation of matrix proteins' phosphorylation could be a means for the development of suitable therapeutic treatments. © 2018 American Society for Bone and Mineral Research.


Assuntos
Matriz Óssea/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Adulto , Idoso de 80 Anos ou mais , Envelhecimento/metabolismo , Fenômenos Biomecânicos , Raquitismo Hipofosfatêmico Familiar/metabolismo , Raquitismo Hipofosfatêmico Familiar/patologia , Raquitismo Hipofosfatêmico Familiar/fisiopatologia , Feminino , Fraturas Ósseas/metabolismo , Fraturas Ósseas/patologia , Produtos Finais de Glicação Avançada/metabolismo , Humanos , Modelos Lineares , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Fosforilação
11.
Bone ; 110: 128-133, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29408699

RESUMO

Advanced glycation end-products (AGEs) are a category of post translational modification associated with the degradation of the structural properties of multiple different types of tissues. Typically, AGEs are the result of a series of post-translational modification reactions between sugars and proteins through a process known as non-enzymatic glycation (NEG). Increases in the rate of NEG of bone tissue are associated with type 2 diabetes and skeletal fragility. Current methods of assessing NEG and its impact on bone fracture risk involve measurement of pentosidine or total fluorescent AGEs (fAGEs). However, pentosidine represents only a small fraction of possible fAGEs present in bone, and neither pentosidine nor total fAGE measurement accounts for non-fluorescent AGEs, which are known to form in significant amounts in skin and other collagenous tissues. Carboxymethyl-lysine (CML) is a non-fluorescent AGE that is often measured and has been shown to accumulate in tissues such as skin, heart, arteries, and intervertebral disks, but is currently not assessed in bone. Here we show the localization of CML to collagen I using mass spectrometry for the first time in human bone. We then present a new method using demineralization followed by heating and trypsin digestion to measure CML content in human bone and demonstrate that CML in bone is 40-100 times greater than pentosidine (the current most commonly used marker of AGEs in bone). We then establish the viability of CML as a measurable AGE in bone by showing that levels of CML, obtained from bone using this technique, increase with age (p<0.05) and are correlated with previously reported measures of bone toughness. Thus, CML is a viable non-fluorescent AGE target to assess AGE accumulation and fragility in bone. The method developed here to extract and measure CML from human bone could facilitate the development of a new diagnostic assay to evaluate fracture risk and potentially lead to new therapeutic approaches to address bone fragility.


Assuntos
Osso Cortical/metabolismo , Lisina/análogos & derivados , Adulto , Idoso , Idoso de 80 Anos ou mais , Arginina/análogos & derivados , Arginina/metabolismo , Osso e Ossos , Sobrevivência Celular , Colágeno/metabolismo , Feminino , Fraturas Ósseas/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Humanos , Hidroxiprolina/metabolismo , Modelos Lineares , Lisina/metabolismo , Masculino , Espectrometria de Massas , Pessoa de Meia-Idade , Risco , Adulto Jovem
12.
J Bone Miner Res ; 33(3): 437-448, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29160901

RESUMO

Back pain is a leading cause of global disability that can arise from vertebral fracture and osteoporosis. Although poor general health and obesity are among the strongest risk factors for back pain, there is remarkably little known about how diet influences spinal diseases. Advanced glycation end-products (AGEs) are implicated in increased fracture risk, yet no studies investigated how dietary AGEs affect spinal health. We tested the hypothesis that high dietary AGE ingestion will diminish vertebral structure and function in a sex- and age-dependent manner. Female and male mice were fed low-AGE (L-AGE) or high-AGE (H-AGE) isocaloric diets for 6 and 18 months and multiple measurements of bone structure and function were taken. AGE levels in serum and cortical vertebrae were increased only for 6-month-old H-AGE female mice while blood glucose and body weight remained normal for all animals. When fed an H-AGE diet, 6-month-old female mice had inferior vertebral trabecular structure with decreased bone mineral density (BMD) and bone volume fraction. Biomechanical testing and analytical modeling further showed functional deterioration in 6-month-old H-AGE females with reduced shear and compression moduli, and maximum load to failure. At 18 months, H-AGE mice of both sexes had significant but small decreases in cortical BMD and thickness, yet functional biomechanical behaviors were not distinguishable from other aging changes. We conclude that an H-AGE diet, without diabetic or overweight conditions, diminished vertebral microstructure, mechanical behaviors, and fracture resistance in young female mice in a manner suggesting accelerated bone aging. © 2017 American Society for Bone and Mineral Research.


Assuntos
Envelhecimento/patologia , Dieta/efeitos adversos , Produtos Finais de Glicação Avançada/efeitos adversos , Caracteres Sexuais , Coluna Vertebral/patologia , Coluna Vertebral/fisiopatologia , Análise de Variância , Animais , Fenômenos Biomecânicos , Osso Esponjoso/diagnóstico por imagem , Osso Esponjoso/patologia , Osso Esponjoso/fisiopatologia , Osso Cortical/diagnóstico por imagem , Osso Cortical/patologia , Osso Cortical/fisiopatologia , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/fisiopatologia , Módulo de Elasticidade , Feminino , Produtos Finais de Glicação Avançada/sangue , Masculino , Camundongos Endogâmicos C57BL , Sobrepeso/sangue , Sobrepeso/patologia , Coluna Vertebral/diagnóstico por imagem , Microtomografia por Raio-X
14.
Anal Biochem ; 499: 85-89, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26851341

RESUMO

Current studies of protein phosphorylation focus primarily on the importance of specific phosphoproteins and their landscapes of phosphorylation in the regulation of different cellular functions. However, global changes in phosphorylation of extracellular matrix phosphoproteins measured "in bulk" are equally important. For example, correct global phosphorylation of different bone matrix proteins is critical to healthy tissue biomineralization. To study changes of bone matrix global phosphorylation, we developed a strategy that combines a procedure for in vitro phosphorylation/dephosphorylation of fully mineralized bone in addition to quantitation of the global phosphorylation levels of bone matrix proteins. For the first time, we show that it is possible to enzymatically phosphorylate/dephosphorylate fully mineralized bone originating from either cadaveric human donors or laboratory animals (mice). Using our strategy, we detected the difference in the global phosphorylation levels of matrix proteins isolated from wild-type and osteopontin knockout mice. We also observed that the global phosphorylation levels of matrix proteins isolated from human cortical bone were lower than those isolated from trabecular bone. The developed strategy has the potential to open new avenues for studies on the global phosphorylation of bone matrix proteins and their role in biomineralization as well for other tissues/cells and protein-based materials.


Assuntos
Matriz Óssea/química , Proteínas da Matriz Extracelular/química , Matriz Extracelular/química , Fosfoproteínas/análise , Fosfoproteínas/química , Idoso , Idoso de 80 Anos ou mais , Animais , Matriz Óssea/metabolismo , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/isolamento & purificação , Proteínas da Matriz Extracelular/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Osteopontina/química , Osteopontina/deficiência , Osteopontina/metabolismo , Fosforilação
15.
J Mech Behav Biomed Mater ; 52: 120-130, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26530231

RESUMO

Non-enzymatic glycation (NEG) is an age-related process accelerated by diseases like diabetes, and causes the accumulation of advanced glycation end-products (AGEs). NEG-mediated modification of bone's organic matrix, principally collagen type-I, has been implicated in impairing skeletal physiology and mechanics. Here, we present evidence, from in vitro and in vivo models, and establish a causal relationship between collagen glycation and alterations in bone fracture at multiple length scales. Through atomic force spectroscopy, we established that NEG impairs collagen's ability to dissipate energy. Mechanical testing of in vitro glycated human bone specimen revealed that AGE accumulation due to NEG dramatically reduces the capacity of organic and mineralized matrix to creep and caused bone to fracture under impact at low levels of strain (3000-5000 µstrain) typically associated with fall. Fracture mechanics tests of NEG modified human cortical bone of varying ages, and their age-matched controls revealed that NEG disrupted microcracking based toughening mechanisms and reduced bone propagation and initiation fracture toughness across all age groups. A comprehensive mechanistic model, based on experimental and modeling data, was developed to explain how NEG and AGEs are causal to, and predictive of bone fragility. Furthermore, fracture mechanics and indentation testing on diabetic mice bones revealed that diabetes mediated NEG severely disrupts bone matrix quality in vivo. Finally, we show that AGEs are predictive of bone quality in aging humans and have diagnostic applications in fracture risk.


Assuntos
Colágeno/metabolismo , Fraturas Ósseas/metabolismo , Animais , Fenômenos Biomecânicos , Matriz Óssea/metabolismo , Calcificação Fisiológica , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Diabetes Mellitus/fisiopatologia , Metabolismo Energético , Feminino , Fêmur/lesões , Fêmur/metabolismo , Fêmur/fisiopatologia , Fraturas Ósseas/fisiopatologia , Glicólise , Humanos , Camundongos , Pessoa de Meia-Idade
16.
PLoS One ; 10(2): e0117240, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25679213

RESUMO

To better understand some aspects of bone matrix glycation, we used an in vitro glycation approach. Within two weeks, our glycation procedures led to the formation of advanced glycation end products (AGEs) at the levels that corresponded to approx. 25-30 years of the natural in vivo glycation. Cortical and cancellous bones from human tibias were glycated in vitro using either glucose (glucosylation) or ribose (ribosylation). Both glucosylation and ribosylation led to the formation of higher levels of AGEs and pentosidine (PEN) in cancellous than cortical bone dissected from all tested donors (young, middle-age and elderly men and women). More efficient glycation of bone matrix proteins in cancellous bone most likely depended on the higher porosity of this tissue, which facilitated better accessibility of the sugars to the matrix proteins. Notably, glycation of cortical bone from older donors led to much higher AGEs levels as compared to young donors. Such efficient in vitro glycation of older cortical bone could result from aging-related increase in porosity caused by the loss of mineral content. In addition, more pronounced glycation in vivo would be driven by elevated oxidation processes. Interestingly, the levels of PEN formation differed pronouncedly between glucosylation and ribosylation. Ribosylation generated very high levels of PEN (approx. 6- vs. 2.5-fold higher PEN level than in glucosylated samples). Kinetic studies of AGEs and PEN formation in human cortical and cancellous bone matrix confirmed higher accumulation of fluorescent crosslinks for ribosylation. Our results suggest that in vitro glycation of bone using glucose leads to the formation of lower levels of AGEs including PEN, whereas ribosylation appears to support a pathway toward PEN formation. Our studies may help to understand differences in the progression of bone pathologies related to protein glycation by different sugars, and raise awareness for excessive sugar supplementation in food and drinks.


Assuntos
Glucose/metabolismo , Reação de Maillard , Ribose/metabolismo , Tíbia/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Arginina/análogos & derivados , Arginina/biossíntese , Progressão da Doença , Feminino , Produtos Finais de Glicação Avançada/metabolismo , Glicosilação , Humanos , Cinética , Lisina/análogos & derivados , Lisina/biossíntese , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Osteocalcina/metabolismo , Conformação Proteica
17.
PLoS One ; 10(1): e0117046, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25629402

RESUMO

Despite our extensive knowledge of insulin-like growth factor 1 (IGF1) action on the growing skeleton, its role in skeletal homeostasis during aging and age-related development of certain diseases is still unclear. Advanced glycation end products (AGEs) derived from glucose are implicated in osteoporosis and a number of diabetic complications. We hypothesized that because in humans and rodents IGF1 stimulates uptake of glucose (a glycation substrate) from the bloodstream in a dose-dependent manner, the decline of IGF1 could be associated with the accumulation of glycation products and the decreasing resistance of bone to fracture. To test the aforementioned hypotheses, we used human tibial posterior cortex bone samples to perform biochemical (measurement of IGF1, fluorescent AGEs and pentosidine (PEN) contents) and mechanical tests (crack initiation and propagation using compact tension specimens). Our results for the first time show a significant, age-independent association between the levels of IGF1 and AGEs. Furthermore, AGEs (fAGEs, PEN) predict propensity of bone to fracture (initiation and propagation) independently of age in human cortical bone. Based on these results we propose a model of IGF1-based regulation of bone fracture. Because IGF1 level increases postnatally up to the juvenile developmental phase and decreases thereafter with aging, we propose that IGF1 may play a protective role in young skeleton and its age-related decline leads to bone fragility and an increased fracture risk. Our results may also have important implications for current understanding of osteoporosis- and diabetes-related bone fragility as well as in the development of new diagnostic tools to screen for fragile bones.


Assuntos
Envelhecimento/metabolismo , Densidade Óssea/fisiologia , Fraturas Ósseas/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Tíbia/metabolismo , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Feminino , Produtos Finais de Glicação Avançada/metabolismo , Humanos , Pessoa de Meia-Idade , Modelos Biológicos , Osteoporose/metabolismo , Medição de Risco , Estresse Mecânico , Adulto Jovem
18.
Proc Natl Acad Sci U S A ; 109(47): 19178-83, 2012 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-23129653

RESUMO

Toughening in hierarchically structured materials like bone arises from the arrangement of constituent material elements and their interactions. Unlike microcracking, which entails micrometer-level separation, there is no known evidence of fracture at the level of bone's nanostructure. Here, we show that the initiation of fracture occurs in bone at the nanometer scale by dilatational bands. Through fatigue and indentation tests and laser confocal, scanning electron, and atomic force microscopies on human and bovine bone specimens, we established that dilatational bands of the order of 100 nm form as ellipsoidal voids in between fused mineral aggregates and two adjacent proteins, osteocalcin (OC) and osteopontin (OPN). Laser microdissection and ELISA of bone microdamage support our claim that OC and OPN colocalize with dilatational bands. Fracture tests on bones from OC and/or OPN knockout mice (OC(-/-), OPN(-/-), OC-OPN(-/-;-/-)) confirm that these two proteins regulate dilatational band formation and bone matrix toughness. On the basis of these observations, we propose molecular deformation and fracture mechanics models, illustrating the role of OC and OPN in dilatational band formation, and predict that the nanometer scale of tissue organization, associated with dilatational bands, affects fracture at higher scales and determines fracture toughness of bone.


Assuntos
Osso e Ossos/patologia , Fraturas Ósseas/patologia , Animais , Matriz Óssea/metabolismo , Matriz Óssea/patologia , Matriz Óssea/ultraestrutura , Osso e Ossos/ultraestrutura , Bovinos , Ensaio de Imunoadsorção Enzimática , Dureza , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Microscopia de Força Atômica , Microscopia Confocal , Osteocalcina/metabolismo , Osteopontina/metabolismo
19.
J Phys Chem C Nanomater Interfaces ; 116(10): 6320-6331, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22822414

RESUMO

Age-related bone fragility fractures present a significant problem for public health. Measures of bone quality are increasingly recognized to complement the conventional bone mineral density (BMD) based assessment of fracture risk. The ability to probe and understand bone quality at the molecular level is desirable in order to unravel how the structure of organic matrix and its association with mineral contribute to the overall mechanical properties. The (13)C{(31)P} REDOR MAS NMR (Rotational Echo Double Resonance Magic Angle Spinning Nuclear Magnetic Resonance) technique is uniquely suited for the study of the structure of the organic-mineral interface in bone. For the first time, we have applied it successfully to analyze the structure of intact (non-powdered) human cortical bone samples, from young healthy and old osteoporotic donors. Loading problems associated with the rapid rotation of intact bone were solved using a Finite Element Analysis (FEA) approach, and a method allowing osteoporotic samples to be balanced and spun reproducibly is described. REDOR NMR parameters were set to allow insight into the arrangement of the amino acids at the mineral interface to be accessed, and SVD (Singular Value Decomposition) was applied to enhance the signal to noise ratio and enable a better analysis of the data. From the REDOR data, it was found that carbon atoms belonging to citrate/glucosaminoglycans (GAGs) are closest to the mineral surface regardless of age or site. In contrast, the arrangement of the collagen backbone at the interface varied with site and age. The relative proximity of two of the main amino acids in bone matrix proteins, hydroxyproline and alanine, with respect to the mineral phase was analyzed in more detail, and discussed in view of glycation measurements which were carried out on the tissues. Overall, this work shows that the (13)C{(31)P} REDOR NMR approach could be used as a complementary technique to assess a novel aspect of bone quality, the organic-mineral interface structure.

20.
Curr Osteoporos Rep ; 10(2): 141-50, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22535528

RESUMO

Bone mineral density alone cannot reliably predict fracture risk in humans and laboratory animals. Therefore, other factors including the quality of organic bone matrix components and their interactions may be of crucial importance to understanding of fragility fractures. Emerging research evidence shows, that in addition to collagen, certain noncollagenous proteins (NCPs) play a significant role in the structural organization of bone and influence its mechanical properties. However, their contribution to bone strength still remains largely undefined. Collagen and NCPs undergo different post-translational modifications, which alter the quality of the extracellular matrix and the response of bone to mechanical load. The primary focus of this overview is on NCPs that, together with collagen, contribute to structural and mechanical properties of bone. Current information on several mechanisms through which some NCPs influence bone's resistance to fracture, including the role of nonenzymatic glycation, is also presented.


Assuntos
Matriz Óssea/fisiologia , Colágeno/fisiologia , Fraturas Ósseas/fisiopatologia , Osteoporose/fisiopatologia , Animais , Fenômenos Biomecânicos , Densidade Óssea/fisiologia , Fraturas Ósseas/epidemiologia , Humanos , Modelos Animais , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...