Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 388
Filtrar
1.
Int J Mol Sci ; 22(22)2021 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-34830274

RESUMEN

The imbalance between bone resorption and bone formation in favor of resorption results in bone loss and deterioration of bone architecture. Osteoblast differentiation is a sequential event accompanying biogenesis of matrix vesicles and mineralization of collagen matrix with hydroxyapatite crystals. Considerable efforts have been made in developing naturally-occurring plant compounds, preventing bone pathologies, or enhancing bone regeneration. Coumarin aesculetin inhibits osteoporosis through hampering the ruffled border formation of mature osteoclasts. However, little is known regarding the effects of aesculetin on the impairment of matrix vesicle biogenesis. MC3T3-E1 cells were cultured in differentiation media with 1-10 µM aesculetin for up to 21 days. Aesculetin boosted the bone morphogenetic protein-2 expression, and alkaline phosphatase activation of differentiating MC3T3-E1 cells. The presence of aesculetin strengthened the expression of collagen type 1 and osteoprotegerin and transcription of Runt-related transcription factor 2 in differentiating osteoblasts for 9 days. When ≥1-5 µM aesculetin was added to differentiating cells for 15-18 days, the induction of non-collagenous proteins of bone sialoprotein II, osteopontin, osteocalcin, and osteonectin was markedly enhanced, facilitating the formation of hydroxyapatite crystals and mineralized collagen matrix. The induction of annexin V and PHOSPHO 1 was further augmented in ≥5 µM aesculetin-treated differentiating osteoblasts for 21 days. In addition, the levels of tissue-nonspecific alkaline phosphatase and collagen type 1 were further enhanced within the extracellular space and on matrix vesicles of mature osteoblasts treated with aesculetin, indicating matrix vesicle-mediated bone mineralization. Finally, aesculetin markedly accelerated the production of thrombospondin-1 and tenascin C in mature osteoblasts, leading to their adhesion to preformed collagen matrix. Therefore, aesculetin enhanced osteoblast differentiation, and matrix vesicle biogenesis and mineralization. These findings suggest that aesculetin may be a potential osteo-inductive agent preventing bone pathologies or enhancing bone regeneration.


Asunto(s)
Matriz Ósea/metabolismo , Calcificación Fisiológica/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Vesículas Extracelulares/metabolismo , Osteoblastos/citología , Umbeliferonas/farmacología , Animales , Matriz Ósea/efectos de los fármacos , Línea Celular , Colágeno Tipo I/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Vesículas Extracelulares/efectos de los fármacos , Sialoproteína de Unión a Integrina/metabolismo , Ratones , Osteoblastos/efectos de los fármacos , Osteocalcina/metabolismo , Osteogénesis/efectos de los fármacos , Osteonectina/metabolismo , Osteopontina/metabolismo , Osteoprotegerina/metabolismo , Transducción de Señal/efectos de los fármacos
2.
Tissue Cell ; 73: 101618, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34391938

RESUMEN

OBJECTIVE: The aim of this study was to evaluate the effects of caffeic acid phenethyl ester (CAPE) on osteoblast-like cell cultures (SAOS-2). METHODS: SAOS-2 were exposed to CAPE at 1 nM, 10 nM, 100 nM, 1 µM, and 10 µM. Non-exposed cultures were used as control. The following parameters were assayed: 1) cell viability at 1, 3, and 7 days; 2) alkaline phosphatase (ALP) activity at 5 and 10 days; 3) matrix mineralization at 14 days; and 4) Runt-related transcription factor 2 (RUNX2), ALP, osteopontin (SPP1), and osteocalcin (BGLAP) gene expression at 5 and 10 days. The data were analyzed by ANOVA two-way or Kruskal-Wallis (α = 5%). RESULTS: At day 1, cell viability was similar among all groups (p > 0.05). At days 3 and 7, cultures exposed to CAPE at 10 µM exhibited a significant reduction in cell viability compared with the others groups (p < 0.05). At day 5, ALP activity was similar among all experimental groups; at day 10, however, the stain intensity was higher in cultures exposed to CAPE at 100 nM and 10 nM in comparison with the other groups (p < 0.05). At days 5 and 10, RUNX2, ALP, SPP1, and BGLAP gene expression was greater in cultures exposed to CAPE in comparison with the control (p < 0.05). At day 14, matrix mineralization was similar in cultures exposed to CAPE at 1 nM and 10 nM (p > 0.05), but superior to those ones observed in the other experimental groups (p < 0.05). CONCLUSION: CAPE at low concentrations can positively module the osteogenesis in vitro.


Asunto(s)
Ácidos Cafeicos/farmacología , Osteogénesis/efectos de los fármacos , Alcohol Feniletílico/análogos & derivados , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Matriz Ósea/efectos de los fármacos , Matriz Ósea/metabolismo , Calcificación Fisiológica/efectos de los fármacos , Calcificación Fisiológica/genética , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Osteocalcina/genética , Osteocalcina/metabolismo , Osteogénesis/genética , Osteopontina/genética , Osteopontina/metabolismo , Alcohol Feniletílico/farmacología
3.
Int J Mol Sci ; 22(16)2021 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-34445788

RESUMEN

Bone formation and growth are crucial for treating bone fractures. Improving bone-reconstruction methods using autologous bone and synthetic implants can reduce the recovery time. Here, we investigated three treatments using two different materials, a bone-derived decellularized extracellular matrix (bdECM) and ß-tricalcium phosphate (ß-TCP), individually and in combination, as osteogenic promoter between bone and 3D-printed polycaprolactone scaffold (6-mm diameter) in rat calvarial defects (8-mm critical diameter). The materials were tested with a human pre-osteoblast cell line (MG63) to determine the effects of the osteogenic promoter on bone formation in vitro. A polycaprolactone (PCL) scaffold with a porous structure was placed at the center of the in vivo rat calvarial defects. The gap between the defective bone and PCL scaffold was filled with each material. Animals were sacrificed four weeks post-implantation, and skull samples were preserved for analysis. The preserved samples were scanned by micro-computed tomography and analyzed histologically to examine the clinical benefits of the materials. The bdECM-ß-TCP mixture showed faster bone formation and a lower inflammatory response in the rats. Therefore, our results imply that a bdECM-ß-TCP mixture is an ideal osteogenic promoter for treating fractures.


Asunto(s)
Fosfatos de Calcio/farmacología , Matriz Extracelular/efectos de los fármacos , Fracturas Óseas/tratamiento farmacológico , Hidrogeles/farmacología , Osteogénesis/efectos de los fármacos , Poliésteres/farmacología , Andamios del Tejido/química , Animales , Matriz Ósea/efectos de los fármacos , Regeneración Ósea/efectos de los fármacos , Células Cultivadas , Humanos , Osteoblastos/efectos de los fármacos , Impresión Tridimensional , Ratas , Ratas Sprague-Dawley , Ingeniería de Tejidos/métodos
4.
Biomed Res Int ; 2021: 3582342, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33834063

RESUMEN

In the field of biology and medicine, one hears often about stem cells and their potential. The dental implant new surfaces, subjected to specific treatments, perform better and allow for quicker healing times and better clinical performance. The purpose of this study is to evaluate from a biological point of view the interaction and cytotoxicity between stem cells derived from dental pulp (DPSCs) and titanium surfaces. Through the creation of complex cells/implant, this study is aimed at analyzing the cytotoxicity of dental implant surfaces (Myth (Maipek Manufacturer Industrial Care, Naples, Italy)) and the adhesion capacity of cells on them and at considering the essential factors for implant healing such as osteoinduction and vasculogenesis. These parameters are pointed out through histology (3D cell culture), immunofluorescence, proliferation assays, scanning electron microscopy, and PCR investigations. The results of the dental implant surface and its interaction with the DPSCs are encouraging, obtaining results increasing the mineralization of the tissues. The knowledge of this type of interaction, highlighting its chemical and biological features, is certainly also an excellent starting point for the development of even more performing surfaces for having better healing in the oral surgical procedures related to dental implant positioning.


Asunto(s)
Implantes Dentales , Pulpa Dental/citología , Células Madre/citología , Matriz Ósea/efectos de los fármacos , Matriz Ósea/metabolismo , Adhesión Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Humanos , Neovascularización Fisiológica/efectos de los fármacos , Oseointegración/efectos de los fármacos , Osteocalcina/metabolismo , Células Madre/efectos de los fármacos , Células Madre/ultraestructura , Propiedades de Superficie , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
Arch Toxicol ; 95(3): 1023-1037, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33426622

RESUMEN

Uranium is widely spread in the environment due to its natural and anthropogenic occurrences, hence the importance of understanding its impact on human health. The skeleton is the main site of long-term accumulation of this actinide. However, interactions of this metal with biological processes involving the mineralized extracellular matrix and bone cells are still poorly understood. To get a better insight into these interactions, we developed new biomimetic bone matrices containing low doses of natural uranium (up to 0.85 µg of uranium per cm2). These models were characterized by spectroscopic and microscopic approaches before being used as a support for the culture and differentiation of pre-osteoclastic cells. In doing so, we demonstrate that uranium can exert opposite effects on osteoclast resorption depending on its concentration in the bone microenvironment. Our results also provide evidence for the first time that resorption contributes to the remobilization of bone matrix-bound uranium. In agreement with this, we identified, by HRTEM, uranium phosphate internalized in vesicles of resorbing osteoclasts. Thanks to the biomimetic matrices we developed, this study highlights the complex mutual effects between osteoclasts and uranium. This demonstrates the relevance of these 3D models to further study the cellular mechanisms at play in response to uranium storage in bone tissue, and thus better understand the impact of environmental exposure to uranium on human bone health.


Asunto(s)
Matriz Ósea/efectos de los fármacos , Modelos Biológicos , Osteoclastos/efectos de los fármacos , Uranio/metabolismo , Animales , Biomimética , Matriz Ósea/metabolismo , Resorción Ósea/metabolismo , Línea Celular Tumoral , Humanos , Ratones , Osteoclastos/metabolismo , Células RAW 264.7 , Distribución Tisular , Uranio/administración & dosificación
6.
Macromol Biosci ; 21(3): e2000336, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33346401

RESUMEN

Bone defects remains a challenge for surgeons. Bone graft scaffold can fill the defect and enhance the bone regeneration. Demineralized bone matrix (DBM) is an allogeneic bone graft substitute, which can only be used as a filling material rather than a structural bone graft. Coating of the scaffolds with nanoscale DBM may enhance the osteoinductivity or osteoconductivity. Herein the lyophilization method is presented to coat the nano-DBM on surface of the porous polycaprolactone (PCL)/ß-tricalcium phosphate (ß-TCP) scaffolds fabricated by 3D printing technology. The morphology, elastic modulus, in vitro cell biocompatibility, and in vivo performance are investigated. Scanning electron microscope (SEM) shows DBM particle clusters with size of 200-500 nm are observed on scaffolds fibers after coating. MC3T3-E1 cells on nano-DBM coated PCL/ß-TCP scaffold show better activity than on PCL/ß-TCP scaffold. In vivo tests show better infiltration of new bone tissue in nano-DBM coated PCL/ß-TCP scaffold than PCL/ß-TCP scaffold via the interface. These results show the presence of nano-DBM coating on PCL/ß-TCP scaffold could enhance the attachment, proliferation, and viability of cells and benefit for the new bone formation surrounding and deep inside the scaffolds. Nano-DBM could potentially be used as a new kind of biomaterial for bone defect treatment.


Asunto(s)
Matriz Ósea/fisiología , Regeneración Ósea/efectos de los fármacos , Fosfatos de Calcio/farmacología , Nanopartículas/química , Poliésteres/farmacología , Andamios del Tejido/química , Animales , Matriz Ósea/efectos de los fármacos , Línea Celular , Ratones , Nanopartículas/ultraestructura , Tamaño de los Órganos/efectos de los fármacos , Implantación de Prótesis , Microtomografía por Rayos X
7.
Int J Mol Sci ; 23(1)2021 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-35008800

RESUMEN

Osseointegration is a prerequisite for the long-term success of implants. Titanium implants are preferred for their biocompatibility and mechanical properties. Nonetheless, the need for early and immediate loading requires enhancing these properties by adding bioactive coatings. In this preclinical study, extracellular matrix properties and cellular balance at the implant/bone interface was examined. Polyelectrolyte multilayers of chitosan and gelatin or with chitosan and Hyaluronic acid fabricated on titanium alloy using a layer-by-layer self-assembly process were compared with native titanium alloy. The study aimed to histologically evaluate bone parameters that correlate to the biomechanical anchorage enhancement resulted from bioactive coatings of titanium implants in a rat animal model. Superior collagen fiber arrangements and an increased number of active osteocytes reflected a significant improvement of bone matrix quality at the bone interface of the chitosan/gelatin-coated titan implants over chitosan/hyaluronic acid-coated and native implants. Furthermore, the numbers and localization of osteoblasts and osteoclasts in the reparative and remodeling phases suggested a better cellular balance in the chitosan/Gel-coated group over the other two groups. Investigating the micro-mechanical properties of bone tissue at the interface can elucidate detailed discrepancies between different promising bioactive coatings of titanium alloys to maximize their benefit in future medical applications.


Asunto(s)
Matriz Ósea/patología , Interfase Hueso-Implante/fisiología , Materiales Biocompatibles Revestidos/farmacología , Osteocitos/patología , Tibia/fisiología , Titanio/farmacología , Animales , Fenómenos Biomecánicos/efectos de los fármacos , Matriz Ósea/efectos de los fármacos , Calcificación Fisiológica/efectos de los fármacos , Colágenos Fibrilares/metabolismo , Masculino , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteocitos/efectos de los fármacos , Ratas Sprague-Dawley , Tibia/efectos de los fármacos
8.
Matrix Biol ; 94: 95-109, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33002580

RESUMEN

Recent in vitro evidence shows that glycosaminoglycans (GAGs) and proteoglycans (PGs) in bone matrix may functionally be involved in the tissue-level toughness of bone. In this study, we showed the effect of biglycan (Bgn), a small leucine-rich proteoglycan enriched in extracellular matrix of bone and the associated GAG subtype, chondroitin sulfate (CS), on the toughness of bone in vivo, using wild-type (WT) and Bgn deficient mice. The amount of total GAGs and CS in the mineralized compartment of Bgn KO mouse bone matrix decreased significantly, associated with the reduction of the toughness of bone, in comparison with those of WT mice. However, such differences between WT and Bgn KO mice diminished once the bound water was removed from bone matrix. In addition, CS was identified as the major subtype in bone matrix. We then supplemented CS to both WT and Bgn KO mice to test whether supplemental GAGs could improve the tissue-level toughness of bone. After intradermal administration of CS, the toughness of WT bone was greatly improved, with the GAGs and bound water amount in the bone matrix increased, while such improvement was not observed in Bgn KO mice or with supplementation of dermatan sulfate (DS). Moreover, CS supplemented WT mice exhibited higher bone mineral density and reduced osteoclastogenesis. Interestingly, Bgn KO bone did not show such differences irrespective of the intradermal administration of CS. In summary, the results of this study suggest that Bgn and CS in bone matrix play a pivotal role in imparting the toughness to bone most likely via retaining bound water in bone matrix. Moreover, supplementation of CS improves the toughness of bone in mouse models.


Asunto(s)
Biglicano/genética , Matriz Ósea/crecimiento & desarrollo , Glicosaminoglicanos/metabolismo , Proteoglicanos/metabolismo , Animales , Densidad Ósea/efectos de los fármacos , Matriz Ósea/efectos de los fármacos , Matriz Ósea/metabolismo , Huesos/efectos de los fármacos , Huesos/metabolismo , Sulfatos de Condroitina/farmacología , Dermatán Sulfato/farmacología , Matriz Extracelular/genética , Glicosaminoglicanos/genética , Humanos , Ratones , Ratones Noqueados , Proteoglicanos/genética , Agua
9.
Phytomedicine ; 79: 153351, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32987362

RESUMEN

BACKGROUND: Since enhanced bone resorption due to osteoclast differentiation and activation cause skeletal diseases, there is a growing need in therapeutics for combating bone-resorbing osteoclasts. Botanical antioxidants are being increasingly investigated for their health-promoting effects on bone. Edible Cirsium setidens contains various polyphenols of linarin, pectolinarin, and apigenin with antioxidant and hepatoprotective effects. PURPOSE: This study aimed to determine whether linarin present in Cirsium setidens water extracts (CSE) and its aglycone acacetin inhibited osteoclastogenesis of RANKL-exposed RAW 264.7 murine macrophages for 5 days. METHODS: This study assessed the osteoprotective effects of CSE, linarin and acacetin on RANKL-induced differentiation and activation of osteoclasts by using MTT assay, TRAP staining, Western blot analysis, bone resorption assay actin ring staining, adhesion assay and immunocytochemical assay. This study explored the underlying mechanisms of their osteoprotection, and identified major components present in CSE by HPLC analysis. RESULTS: Linarin and pectolinarin were identified as major components of CSE. Nontoxic linarin and acacetin as well as CSE, but not pectolinarin attenuated the RANKL-induced macrophage differentiation into multinucleated osteoclasts, and curtailed osteoclastic bone resorption through reducing lacunar acidification and bone matrix degradation in the osteoclast-bone interface. Linarin and acacetin in CSE reduced the transmigration and focal contact of osteoclasts to bone matrix-mimicking RGD peptide. Such reduction was accomplished by inhibiting the induction of integrins, integrin-associated proteins of paxillin and gelsolin, cdc42 and CD44 involved in the formation of actin rings. The inhibition of integrin-mediated actin ring formation by linarin and acacetin entailed the disruption of TRAF6-c-Src-PI3K signaling of bone-resorbing osteoclasts. The functional inhibition of c-Src was involved in the loss of F-actin-enriched podosome core protein cortactin-mediated actin assembly due to linarin and acacetin. CONCLUSION: These observations demonstrate that CSE, linarin and acacetin were effective in retarding osteoclast function of focal adhesion to bone matrix and active bone resorption via inhibition of diffuse cloud-associated αvß3 integrin and core-linked CD44.


Asunto(s)
Resorción Ósea/tratamiento farmacológico , Flavonas/farmacología , Adhesiones Focales/efectos de los fármacos , Glicósidos/farmacología , Osteoclastos/efectos de los fármacos , Actinas/metabolismo , Animales , Matriz Ósea/efectos de los fármacos , Matriz Ósea/metabolismo , Resorción Ósea/metabolismo , Cirsium/química , Adhesiones Focales/metabolismo , Receptores de Hialuranos/metabolismo , Integrina alfaVbeta3/metabolismo , Ratones , Osteoclastos/metabolismo , Osteogénesis/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Extractos Vegetales/farmacología , Células RAW 264.7
10.
J Mater Chem B ; 8(15): 3064-3075, 2020 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-32202266

RESUMEN

The simulation of the native bone matrix formation process is crucial for the construction of the cellular microenvironment for bone regeneration. However, it is still challenging to design bioactive materials that simultaneously mimic the composition and dynamic mineralization process of the bone matrix, let alone realize osteoinduction by a biomimetic dynamic microenvironment. In this study, we prepared a biomimetic mineralizable collagen hydrogel (CAV) and explored the effects of a dynamic mineralized matrix on the osteogenesis of stem cells both in vitro and in vivo. We showed the feasibility of the biomimetic CAV hydrogel to induce mineralization in simulated media including simulated body fluid (SBF), glycerol phosphate calcium salt hydrate (CaGP) solution and cell co-cultured systems. The participation of cells in the mineralization process is more likely to induce matrix remodeling due to the synergistic effects of CAV mineralization and cellular secretion, resulting in higher matrix strength. We also demonstrated that the biomimetic mineralized hydrogel could up-regulate osteogenic genes and protein expression of bone marrow mesenchymal stem cells (BMSCs), thus enhancing osteogenesis in vivo. The interactions between the mineralizable hydrogel and cells play an important role in regulating dynamic matrix mineralization and osteogenesis. Our findings prove that the biomimetic mineralizable hydrogel is a promising candidate for implantable orthopedic applications and provides essential implications for the future design of materials for bone regeneration.


Asunto(s)
Materiales Biomiméticos/farmacología , Matriz Ósea/efectos de los fármacos , Colágeno/farmacología , Hidrogeles/farmacología , Osteogénesis/efectos de los fármacos , Animales , Materiales Biomiméticos/síntesis química , Materiales Biomiméticos/química , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Microambiente Celular/efectos de los fármacos , Colágeno/síntesis química , Colágeno/química , Femenino , Hidrogeles/síntesis química , Hidrogeles/química , Células Madre Mesenquimatosas/efectos de los fármacos , Estructura Molecular , Tamaño de la Partícula , Ratas , Ratas Sprague-Dawley , Propiedades de Superficie
11.
Biomed Res Int ; 2020: 2087475, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32083125

RESUMEN

Poly-ε-caprolactone (PCL) is a promising synthetic material in bone tissue engineering (BTE). Particularly, the introduction of rapid prototyping (RP) represents the possibility of manufacturing PCL scaffolds with customized appearances and structures. Bio-Oss is a natural bone mineral matrix with significant osteogenic effects; however, it has limitations in being constructed and maintained into specific shapes and sites. In this study, we used RP and fabricated a hollow-structured cage-shaped PCL scaffold loaded with Bio-Oss to form a hybrid scaffold for BTE. Moreover, we adopted NaOH surface treatment to improve PCL hydrophilicity and enhance cell adhesion. The results showed that the NaOH-treated hybrid scaffold could enhance the osteogenesis of human bone marrow-derived mesenchymal stem cells (hBMMSCs) both in vitro and in vivo. Altogether, we reveal a novel hybrid scaffold that not only possesses osteoinductive function to promote bone formation but can also be fabricated into specific forms. This scaffold design may have great application potential in bone tissue engineering.


Asunto(s)
Huesos/efectos de los fármacos , Caproatos/química , Caproatos/farmacología , Lactonas/química , Lactonas/farmacología , Animales , Matriz Ósea/efectos de los fármacos , Regeneración Ósea/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Humanos , Interacciones Hidrofóbicas e Hidrofílicas/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Minerales/farmacología , Osteogénesis/efectos de los fármacos , Impresión Tridimensional , Ingeniería de Tejidos/métodos , Andamios del Tejido
12.
Q J Nucl Med Mol Imaging ; 63(2): 98-111, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31298015

RESUMEN

Bone metastases remain a common feature of advanced cancers and are associated with significant morbidity and mortality. Recent research has identified promising novel treatment targets to improve current treatment strategies for bone metastatic disease. This review summarizes the well-known and recently discovered molecular biology pathways in bone that govern normal physiological remodeling or drive the pathophysiological changes observed when bone metastases are present. In the rapidly changing world of targeted cancer treatments, it is important to recognize the specific treatment effects induced in bone by these agents and the potential impact on common imaging strategies. The osteoclastic targets (bisphosphonates, LGR4, RANKL, mTOR, MET-VEGFR, cathepsin K, Src, Dock 5) and the osteoblastic targets (Wnt and endothelin) are discussed, and the emerging field of osteo-immunity is introduced as potential future therapeutic target. Finally, a summary is provided of available trial data for agents that target these pathways and that have been assessed in patients. The ultimate goal of research into novel pathways and targets involved in the tumor-bone microenvironment is to tackle one of the great remaining unmet needs in oncology, that is finding a cure for bone metastatic disease.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/secundario , Terapia Molecular Dirigida/métodos , Animales , Matriz Ósea/efectos de los fármacos , Matriz Ósea/metabolismo , Matriz Ósea/patología , Neoplasias Óseas/inmunología , Neoplasias Óseas/patología , Humanos , Inmunidad Innata/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteoclastos/patología , Microambiente Tumoral/efectos de los fármacos
13.
Front Immunol ; 10: 709, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31024546

RESUMEN

The release of the prototypic DAMP High Mobility Group Box 1 (HMGB1) into extracellular environment and its binding to the Receptor for Advanced Glycation End Products (RAGE) has been described to trigger sterile inflammation and regulate healing outcome. However, their role on host response to Ti-based biomaterials and in the subsequent osseointegration remains unexplored. In this study, HMGB1 and RAGE inhibition in the Ti-mediated osseointegration were investigated in C57Bl/6 mice. C57Bl/6 mice received a Ti-device implantation (Ti-screw in the edentulous alveolar crest and a Ti-disc in the subcutaneous tissue) and were evaluated by microscopic (microCT [bone] and histology [bone and subcutaneous]) and molecular methods (ELISA, PCR array) during 3, 7, 14, and 21 days. Mice were divided into 4 groups: Control (no treatment); GZA (IP injection of Glycyrrhizic Acid for HMGB1 inhibition, 4 mg/Kg/day); RAP (IP injection of RAGE Antagonistic Peptide, 4 mg/Kg/day), and vehicle controls (1.5% DMSO solution for GZA and 0.9% saline solution for RAP); treatments were given at all experimental time points, starting 1 day before surgeries. HMGB1 was detected in the Ti-implantation sites, adsorbed to the screws/discs. In Control and vehicle groups, osseointegration was characterized by a slight inflammatory response at early time points, followed by a gradual bone apposition and matrix maturation at late time points. The inhibition of HMGB1 or RAGE impaired the osseointegration, affecting the dynamics of mineralized and organic bone matrix, and resulting in a foreign body reaction, with persistence of macrophages, necrotic bone, and foreign body giant cells until later time points. While Control samples were characterized by a balance between M1 and M2-type response in bone and subcutaneous sites of implantation, and also MSC markers, the inhibition of HMGB1 or RAGE caused a higher expression M1 markers and pro-inflammatory cytokines, as well chemokines and receptors for macrophage migration until later time points. In conclusion, HMGB1 and RAGE have a marked role in the osseointegration, evidenced by their influence on host inflammatory immune response, which includes macrophages migration and M1/M2 response, MSC markers expression, which collectively modulate bone matrix deposition and osseointegration outcome.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Artroplastia/métodos , Materiales Biocompatibles/metabolismo , Proteínas HMGB/metabolismo , Inflamación/inmunología , Macrófagos/inmunología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Titanio/metabolismo , Animales , Materiales Biocompatibles/química , Biomarcadores/metabolismo , Matriz Ósea/efectos de los fármacos , Movimiento Celular , Ácido Glicirrínico/administración & dosificación , Proteínas HMGB/antagonistas & inhibidores , Humanos , Inmunomodulación , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Oseointegración , Péptidos/administración & dosificación , Titanio/química
14.
Biomed Res Int ; 2019: 5678548, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30800672

RESUMEN

Bone fragility and associated fracture risk are major problems in aging. Oxidative stress and mitochondrial dysfunction play a key role in the development of bone fragility. Mitochondrial dysfunction is closely associated with excessive production of reactive oxygen species (ROS). L-Carnitine (L-C), a fundamental cofactor in lipid metabolism, has an important antioxidant property. Several studies have shown how L-C enhances osteoblastic proliferation and activity. In the current study, we investigated the potential effects of L-C on mitochondrial activity, ROS production, and gene expression involved in osteoblastic differentiation using osteoblast-like cells (hOBs) derived from elderly patients. The effect of 5mM L-C treatment on mitochondrial activity and L-C antioxidant activity was studied by ROS production evaluation and cell-based antioxidant activity assay. The possible effects of L-C on hOBs differentiation were assessed by analyzing gene and protein expression by Real Time PCR and western blotting, respectively. L-C enhanced mitochondrial activity and improved antioxidant defense of hOBs. Furthermore, L-C increased the phosphorylation of Ca2+/calmodulin-dependent protein kinase II. Additionally, L-C induced the phosphorylation of ERK1/2 and AKT and the main kinases involved in osteoblastic differentiation and upregulated the expression of osteogenic related genes, RUNX2, osterix (OSX), bone sialoprotein (BSP), and osteopontin (OPN) as well as OPN protein synthesis, suggesting that L-C exerts a positive modulation of key osteogenic factors. In conclusion, L-C supplementation could represent a possible adjuvant in the treatment of bone fragility, counteracting oxidative phenomena and promoting bone quality maintenance.


Asunto(s)
Matriz Ósea/efectos de los fármacos , Carnitina/farmacología , Diferenciación Celular/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Anciano , Anciano de 80 o más Años , Antioxidantes/metabolismo , Matriz Ósea/metabolismo , Calcificación Fisiológica/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Células Cultivadas , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Femenino , Humanos , Sialoproteína de Unión a Integrina/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Osteoblastos/metabolismo , Osteogénesis/efectos de los fármacos , Osteopontina/metabolismo , Oxidación-Reducción , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción Sp7/metabolismo , Regulación hacia Arriba/efectos de los fármacos
15.
J Biomed Mater Res A ; 107(7): 1476-1490, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30786151

RESUMEN

Due to the similar collagen composition and closely physiological relationship with soft connective tissues, demineralized bone matrices (DBMs) were used to repair the injured tendon or ligament. However, the osteoinductivity of DBMs would be a huge barrier of these applications. Hydrogen peroxide (H2 O2 ) has been proved to reduce the osteoinductivity of DBMs. Nevertheless, the biological properties of H2 O2 -treated DBMs have not been evaluated completely, while the potential mechanism of H2 O2 compromising osteoinductivity is also unclear. Hence, the purpose of this study was to characterize the biological properties of H2 O2 -treated DBMs and search for the proof that H2 O2 could compromise osteoinductivity of DBMs. Decellularized and demineralized bone matrices (DCDBMs) were washed by 3% H2 O2 for 12 h to fabricate the H2 O2 -treated DCDBMs (HPTBMs). Similar biological properties including collagen, biomechanics, and biocompatibility were observed between DCDBMs and HPTBMs. The immunohistochemistry staining of bone morphogenetic protein 2 (BMP-2) was negative in HPTBMs. Furthermore, HPTBMs exhibited significantly reduced osteoinductivity both in vitro and in vivo. Taken together, these findings suggest that the BMP-2 in DCDBMs could be the target of H2 O2 . HPTBMs could be expected to be used as a promising scaffold for tissue engineering. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A, 2019.


Asunto(s)
Matriz Ósea/fisiología , Calcificación Fisiológica/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Oseointegración/efectos de los fármacos , Animales , Matriz Ósea/efectos de los fármacos , Matriz Ósea/ultraestructura , Bovinos , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Células 3T3 NIH , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Ratas Sprague-Dawley
16.
J Bone Miner Metab ; 37(1): 28-35, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29392472

RESUMEN

Parathyroid hormone (1-34, PTH) combined ß-tricalcium phosphate (ß-TCP) achieves stable bone regeneration without cell transplantation in previous studies. Recently, with the development of tissue engineering slow release technology, PTH used locally to promote bone defect healing become possible. This study by virtue of collagen with a combination of drugs and has a slow release properties, and investigated bone regeneration by ß-TCP/collagen (ß-TCP/COL) with the single local administration of PTH. After the creation of a rodent critical-sized femoral metaphyseal bone defect, ß-TCP/COL was prepared by mixing sieved granules of ß-TCP and atelocollagen for medical use, then ß-TCP/COL with dripped PTH solution (1.0 µg) was implanted into the defect of OVX rats until death at 4 and 8 weeks. The defected area in distal femurs of rats was harvested for evaluation by histology, micro-CT, and biomechanics. The results of our study show that single-dose local administration of PTH combined local usage of ß-TCP/COL can increase the healing of defects in OVX rats. Furthermore, treatments with single-dose local administration of PTH and ß-TCP/COL showed a stronger effect on accelerating the local bone formation than ß-TCP/COL used alone. The results from our study demonstrate that combination of single-dose local administration of PTH and ß-TCP/COL had an additive effect on local bone formation in osteoporosis rats.


Asunto(s)
Fosfatos de Calcio/farmacología , Colágeno/farmacología , Fémur/patología , Ovariectomía , Hormona Paratiroidea/administración & dosificación , Cicatrización de Heridas/efectos de los fármacos , Animales , Materiales Biocompatibles/farmacología , Fenómenos Biomecánicos/efectos de los fármacos , Matriz Ósea/efectos de los fármacos , Matriz Ósea/metabolismo , Femenino , Fémur/diagnóstico por imagen , Fémur/efectos de los fármacos , Imagenología Tridimensional , Ratas Sprague-Dawley
17.
J Bone Miner Metab ; 37(4): 658-667, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30357566

RESUMEN

Antioxidant properties of several nutrients may influence bone metabolism, affording protection against damaging effects caused by oxidative stress. Thus, we hypothesized that lycopene may benefit bone tissue metabolism and functional activity of osteoblastic cells from bone marrow of osteoporotic female rats. Wistar rats were ovariectomized and paired with sham animals. In vitro evaluations were performed after 60 days of surgery, when cells were cultured in osteogenic medium and divided in control (C), ovariectomized (OVX) and ovariectomized + 1 µmol/L lycopene (OVXL) groups. Besides, in vivo studies were carried out to evaluate femur bone remodeling by histological and histomorphometric analyses after daily intake of 10 mg/kg of lycopene for 30 and 60 days after ovariectomy. Cell proliferation was significantly higher in OVX and OVXL groups after 10 days of culture. Alkaline phosphatase activity (ALP) was higher in OVXL group in later periods of cell culture, whereas its in situ detection was higher for this group in all experimental periods; nevertheless, mineralization did not show significant differences among the groups. There was a significant upregulation of genes Sp7, Runx2 and Bsp after 3 days and genes Runx2 and Bglap after 10 days from OVXL when compared to OVX. In vivo results demonstrated that daily intake of 10 mg/kg of lycopene for 60 days decreased bone loss in femur epiphysis in ovariectomized rats by maintaining trabecular bone similar to controls. Data obtained suggest that lycopene might benefit the functional activity of osteoblastic cells from ovariectomized rats, as well as avoid further bone resorption.


Asunto(s)
Resorción Ósea/tratamiento farmacológico , Resorción Ósea/prevención & control , Fémur/patología , Licopeno/uso terapéutico , Osteoblastos/metabolismo , Osteoporosis/tratamiento farmacológico , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Densidad Ósea/efectos de los fármacos , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Matriz Ósea/efectos de los fármacos , Matriz Ósea/metabolismo , Resorción Ósea/patología , Resorción Ósea/fisiopatología , Calcificación Fisiológica/efectos de los fármacos , Hueso Esponjoso/efectos de los fármacos , Hueso Esponjoso/patología , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Fémur/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Licopeno/farmacología , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteoporosis/patología , Osteoporosis/fisiopatología , Ovariectomía , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Wistar
18.
Sci Rep ; 8(1): 15824, 2018 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-30361649

RESUMEN

Transplantation of engineered three-dimensional (3D) bone tissue may provide therapeutic benefits to patients with various bone diseases. To achieve this goal, appropriate 3D scaffolds and cells are required. In the present study, we devised a novel nanogel tectonic material for artificial 3D scaffold, namely the nanogel-cross-linked porous (NanoCliP)-freeze-dried (FD) gel, and estimated its potential as a 3D scaffold for bone tissue engineering. As the osteoblasts, directly converted osteoblasts (dOBs) were used, because a large number of highly functional osteoblasts could be induced from fibroblasts that can be collected from patients with a minimally invasive procedure. The NanoCliP-FD gel was highly porous, and fibronectin coating of the gel allowed efficient adhesion of the dOBs, so that the cells occupied the almost entire surface of the walls of the pores after culturing for 7 days. The dOBs massively produced calcified bone matrix, and the culture could be continued for at least 28 days. The NanoCliP-FD gel with dOBs remarkably promoted bone regeneration in vivo after having been grafted to bone defect lesions that were artificially created in mice. The present findings suggest that the combination of the NanoCliP-FD gel and dOBs may provide a feasible therapeutic modality for bone diseases.


Asunto(s)
Regeneración Ósea/efectos de los fármacos , Reprogramación Celular/efectos de los fármacos , Fibroblastos/citología , Osteoblastos/citología , Polietilenglicoles/farmacología , Polietileneimina/farmacología , Andamios del Tejido/química , Animales , Matriz Ósea/efectos de los fármacos , Matriz Ósea/metabolismo , Adhesión Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Reactivos de Enlaces Cruzados/química , Fibroblastos/efectos de los fármacos , Fibronectinas/farmacología , Liofilización , Humanos , Ratones , Nanogeles , Osteoblastos/efectos de los fármacos , Porosidad , Cicatrización de Heridas/efectos de los fármacos
19.
Bone ; 117: 130-137, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30261327

RESUMEN

The lacunar-canaliculi system is a network of channels that is created and maintained by osteocytes as they are embedded throughout cortical bone. As osteocytes modify their lacuna space, the local tissue composition and tissue strength are subject to change. Although continual exposure to parathyroid hormone (PTH) can induce adaptation at the lacunar wall, the impact of intermittent PTH treatment on perilacunar adaptation remains unclear. Therefore, the primary objective of this study was to establish how intermittent PTH(1-34) treatment influences perilacunar adaptation with respect to changes in tissue composition. We hypothesized that local changes in tissue composition following PTH(1-34) are associated with corresponding gains in tissue strength and resistance to microdamage at the whole bone level. Adult male C57BL/6J mice were treated daily with PTH(1-34) or vehicle for 3 weeks. In response to PTH(1-34), Raman spectroscopy revealed a significant decrease in the carbonate-to-phosphate ratio and crystallinity across the entire tissue, while the mineral-to-matrix ratio demonstrated a significant decrease in just the perilacunar region. The shift in perilacunar composition largely explained the corresponding increase in tissue strength, while the degree of new tissue added at the endosteum and periosteum did not produce any significant changes in cortical area or moment of inertia that would explain the increase in tissue strength. Furthermore, fatigue testing revealed a greater resistance to crack formation within the existing tissue following PTH(1-34) treatment. As a result, the shift in perilacunar composition presents a unique mechanism by which PTH(1-34) produces localized differences in tissue quality that allow more energy to be dissipated under loading, thereby increasing tissue strength and resistance to microdamage. In addition, our findings demonstrate the potential for PTH(1-34) to amplify osteocytes' mechanotransduction by producing a more compliant tissue. Overall, the present study demonstrates that changes in tissue composition localized at the lacuna wall contribute to the strength and fatigue resistance of cortical bone gained in response to intermittent PTH(1-34) treatment.


Asunto(s)
Huesos/fisiología , Hormona Paratiroidea/farmacología , Animales , Fenómenos Biomecánicos/efectos de los fármacos , Matriz Ósea/efectos de los fármacos , Matriz Ósea/metabolismo , Huesos/anatomía & histología , Huesos/efectos de los fármacos , Huesos/patología , Masculino , Ratones Endogámicos C57BL , Minerales/metabolismo , Tamaño de los Órganos/efectos de los fármacos , Espectrometría Raman
20.
PLoS One ; 13(9): e0202833, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30180177

RESUMEN

Bone is a highly organized tissue in which each structural level influences the macroscopic and microscopic mechanical behavior. In particular, the quantity, quality, and distribution of the different bone components, i.e. collagen matrix and hydroxyapatite crystals, are associated with bone strength or fragility. Common spectroscopic techniques used to assess bone composition have resolutions limited to the micrometer range. In this study, our aims were two-fold: i) to develop and validate the AFM-IR methodology for skeletal tissues and ii) to apply the methodology to sheep cancellous bone with the objective to obtain novel findings on the composition and structure of trabecular packets.To develop the methodology, we assessed spatial and temporal reproducibility using a known homogeneous material (polymethylmethacrylate, PMMA). We verified that the major peak positions were similar and not shifted when compared to traditional Fourier Transform Infrared imaging (FTIRI). When AFM-IR was applied to sheep cancellous bone, the mineral-to-matrix ratio increased and the acid phosphate substitution ratio decreased as a function of tissue maturity. The resolution of the technique enabled visualization of different stages of the bone maturation process, particularly newly-formed osteoid prior to mineralization. We also observed alternating patterns of IR parameters in line and imaging measurements, suggesting the apposition of layers of alternating structure and / or composition that were not visible with traditional spectroscopic methods. In conclusion, nanoscale IR spectroscopy demonstrates novel compositional and structural changes within trabecular packets in cancellous bone. Based on these results, AFM-IR is a valuable tool to investigate cancellous bone at the nanoscale and, more generally, to analyze small dynamic areas that are invisible to traditional spectroscopic methods.


Asunto(s)
Hueso Esponjoso/química , Hueso Esponjoso/diagnóstico por imagen , Nanotecnología/métodos , Espectroscopía Infrarroja por Transformada de Fourier/métodos , Animales , Matriz Ósea/química , Matriz Ósea/diagnóstico por imagen , Matriz Ósea/efectos de los fármacos , Calcificación Fisiológica/efectos de los fármacos , Hueso Esponjoso/efectos de los fármacos , Colágeno/química , Durapatita/química , Energía Filtrada en la Transmisión por Microscopía Electrónica , Polimetil Metacrilato/química , Polimetil Metacrilato/farmacología , Reproducibilidad de los Resultados , Ovinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...