Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Adv Mater ; 36(6): e2307639, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38009631

RESUMEN

Treating bone infections and ensuring bone repair is one of the greatest global challenges of modern orthopedics, made complex by antimicrobial resistance (AMR) risks due to long-term antibiotic treatment and debilitating large bone defects following infected tissue removal. An ideal multi-faceted solution would will eradicate bacterial infection without long-term antibiotic use, simultaneously stimulating osteogenesis and angiogenesis. Here, a multifunctional collagen-based scaffold that addresses these needs by leveraging the potential of antibiotic-free antimicrobial nanoparticles (copper-doped bioactive glass, CuBG) to combat infection without contributing to AMR in conjunction with microRNA-based gene therapy (utilizing an inhibitor of microRNA-138) to stimulate both osteogenesis and angiogenesis, is developed. CuBG scaffolds reduce the attachment of gram-positive bacteria by over 80%, showcasing antimicrobial functionality. The antagomiR-138 nanoparticles induce osteogenesis of human mesenchymal stem cells in vitro and heal a large load-bearing defect in a rat femur when delivered on the scaffold. Combining both promising technologies results in a multifunctional antagomiR-138-activated CuBG scaffold inducing hMSC-mediated osteogenesis and stimulating vasculogenesis in an in vivo chick chorioallantoic membrane model. Overall, this multifunctional scaffold catalyzes killing mechanisms in bacteria while inducing bone repair through osteogenic and angiogenic coupling, making this platform a promising multi-functional strategy for treating and repairing complex bone infections.


Asunto(s)
MicroARNs , Nanopartículas , Humanos , Ratas , Animales , Andamios del Tejido , Regeneración Ósea , MicroARNs/genética , Antagomirs/farmacología , Osteogénesis , Antibacterianos/farmacología , Antibacterianos/uso terapéutico
2.
Curr Osteoporos Rep ; 21(6): 806-814, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38001387

RESUMEN

PURPOSE OF REVIEW: This review aims to explore the potential of biomimetic hydrogels as an alternative to bone cement in vertebral body stenting (VBS), a minimally invasive treatment for vertebral compression fractures. RECENT FINDINGS: The use of bone cement in VBS procedures can lead to complications such as incomplete fracture reduction and cement leakage. Biomimetic hydrogels have gained significant attention as potential biomaterial alternatives for VBS due to their unique properties, including tuneable therapeutic and mechanical properties. Over the past decade, there has been significant advancements in the development of biomimetic hydrogels for bone regeneration, employing a wide range of approaches to enhance the structural and functional properties of hydrogels. Biomimetic hydrogels hold significant promise as safer and reparative alternatives to bone cement for VBS procedures. However, further research and development in this field are necessary to explore the full potential of hydrogel-based systems for vertebral bone repair.


Asunto(s)
Fracturas por Compresión , Fracturas Osteoporóticas , Fracturas de la Columna Vertebral , Humanos , Fracturas de la Columna Vertebral/cirugía , Fracturas por Compresión/tratamiento farmacológico , Fracturas por Compresión/cirugía , Cementos para Huesos/uso terapéutico , Cuerpo Vertebral , Hidrogeles/uso terapéutico , Biomimética , Estudios Retrospectivos , Resultado del Tratamiento , Fracturas Osteoporóticas/cirugía
3.
Gels ; 9(10)2023 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-37888382

RESUMEN

Injectable hydrogels were discovered as attractive materials for bone tissue engineering applications given their outstanding biocompatibility, high water content, and versatile fabrication platforms into materials with different physiochemical properties. However, traditional hydrogels suffer from weak mechanical strength, limiting their use in heavy load-bearing areas. Thus, the fabrication of mechanically robust injectable hydrogels that are suitable for load-bearing environments is of great interest. Successful material design for bone tissue engineering requires an understanding of the composition and structure of the material chosen, as well as the appropriate selection of biomimetic natural or synthetic materials. This review focuses on recent advancements in materials-design considerations and approaches to prepare mechanically robust injectable hydrogels for bone tissue engineering applications. We outline the materials-design approaches through a selection of materials and fabrication methods. Finally, we discuss unmet needs and current challenges in the development of ideal materials for bone tissue regeneration and highlight emerging strategies in the field.

4.
Biochim Biophys Acta Gen Subj ; 1867(6): 130361, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37019341

RESUMEN

Breast cancer is the most common ailment among women. In 2020, it had the highest incidence of any type of cancer. Many Phase II and III anti-cancer drugs fail due to efficacy, durability, and side effects. Thus, accelerated drug screening models must be accurate. In-vivo models have been used for a long time, but delays, inconsistent results, and a greater sense of responsibility among scientists toward wildlife have led to the search for in-vitro alternatives. Stromal components support breast cancer growth and survival. Multi-compartment Transwell models may be handy instruments. Co-culturing breast cancer cells with endothelium and fibroblasts improves modelling. The extracellular matrix (ECM) supports native 3D hydrogels in natural and polymeric forms. 3D Transwell cultured tumor spheroids mimicked in-vivo pathological conditions. Tumor invasion, migration, Trans-endothelial migration, angiogenesis, and spread are studied using comprehensive models. Transwell models can create a cancer niche and conduct high-throughput drug screening, promising future applications. Our comprehensive shows how 3D in-vitro multi compartmental models may be useful in producing breast cancer stroma in Transwell culture.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Modelos Epidemiológicos , Técnicas de Cocultivo , Matriz Extracelular
5.
ACS Biomater Sci Eng ; 9(3): 1243-1250, 2023 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-36749897

RESUMEN

Materials that are evaluated for bioengineering purposes are carefully tested to evaluate cellular interactions with respect to biocompatibility and in some cases cell differentiation. A key perspective that is often considered is the ability for decellularized synthetic or natural based matrices to facilitate cell migration or tissue ingrowth. Current methods of measuring cell migration range from simple scratch assays to Boyden chamber inserts and fluorescent imaging of seeded spheroids. Many of these methods require tissue processing for histological analysis and fixing and staining for imaging, which can be difficult and dependent on the stability of the hydrogel subject. Herein we present a simple platform that can be manufactured using 3D printing and easily applied to in vitro cell culture, allowing the researcher to image live cellular migration into a cellular materials. We found this to be an adaptable, cheap, and replicable technique to evaluate cellular interaction that has applications in the research and development of hydrogels for tissue engineering purposes.


Asunto(s)
Hidrogeles , Ingeniería de Tejidos , Ingeniería de Tejidos/métodos , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular
6.
Acta Biomater ; 150: 22-33, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35914694

RESUMEN

Scarring is a major clinical issue that affects a considerable number of patients. The associated problems go beyond the loss of skin functionality, as scars bring aesthetic, psychological, and social difficulties. Therefore, new strategies are required to improve the process of healing and minimize scar formation. Research has highlighted the important role of mechanical forces in the process of skin tissue repair and scar formation, in addition to the chemical signalling. A more complete understanding of how engineered biomaterials can modulate these mechanical stimuli and modify the mechanotransduction signals in the wound microenvironment is expected to enable scar tissue reduction. The present review aims to provide an overview of our current understanding of skin biomechanics and mechanobiology underlying wound healing and scar formation, with an emphasis on the development of novel mechanomodulatory wound dressings with the capacity to offload mechanical tension in the wound environment. Furthermore, a broad overview of current challenges and future perspectives of promising mechanomodulatory biomaterials for this application are provided. STATEMENT OF SIGNIFICANCE: Scarring still is one of the biggest challenges in cutaneous wound healing. Beyond the loss of skin functionality, pathological scars, like keloids and hypertrophic, are associated to aesthetic, psychological, and social distress. Nonetheless, the understanding of the pathophysiology behind the formation of those scars remains elusive, which has in fact hindered the development of effective therapeutics. Therefore, in this review we provide an overview of our current understanding of skin biomechanics and mechanobiology underlying wound healing and scar formation, with an emphasis on the development of novel mechanomodulatory wound dressings with the capacity to offload mechanical tension in the wound environment.


Asunto(s)
Cicatriz Hipertrófica , Queloide , Materiales Biocompatibles/uso terapéutico , Cicatriz Hipertrófica/patología , Humanos , Queloide/patología , Queloide/prevención & control , Mecanotransducción Celular , Piel/patología , Cicatrización de Heridas/fisiología
7.
Mater Sci Eng C Mater Biol Appl ; 128: 112340, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34474890

RESUMEN

Mechanical robustness is an essential consideration in the development of hydrogel platforms for bone regeneration, and despite significant advances in the field of injectable hydrogels, many fail in this regard. Inspired by the mechanical properties of carboxylated single wall carbon nanotubes (COOH-SWCNTs) and the biological advantages of natural polymers, COOH-SWCNTs were integrated into chitosan and collagen to formulate mechanically robust, injectable and thermoresponsive hydrogels with interconnected molecular structure for load-bearing applications. This study presents a complete characterisation of the structural and biological properties, and mechanism of gelation of these novel formulated hydrogels. Results demonstrate that ß-glycerophosphate (ß-GP) and temperature play important roles in attaining gelation at physiological conditions, and the integration with COOH-SWCNTs significantly changed the structural morphology of the hydrogels to a more porous and aligned network. This led to a crystalline structure and significantly increased the mechanical strength of the hydrogels from kPa to MPa, which is closer to the mechanical strength of the bone. Moreover, increased osteoblast proliferation and rapid adsorption of hydroxyapatite on the surface of the hydrogels indicates increased bioactivity with addition of COOH-SWCNTs. Therefore, these nano-engineered hydrogels are expected to have wide utility in the area of bone tissue engineering and regenerative medicine.


Asunto(s)
Quitosano , Nanotubos de Carbono , Colágeno , Hidrogeles , Ingeniería de Tejidos
8.
Polymers (Basel) ; 13(15)2021 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-34372114

RESUMEN

Impaired skin wound healing due to severe injury often leads to dysfunctional scar tissue formation as a result of excessive and persistent myofibroblast activation, characterised by the increased expression of α-smooth muscle actin (αSMA) and extracellular matrix (ECM) proteins. Yet, despite extensive research on impaired wound healing and the advancement in tissue-engineered skin substitutes, scar formation remains a significant clinical challenge. This study aimed to first investigate the effect of methacrylate gelatin (GelMA) biomaterial stiffness on human dermal fibroblast behaviour in order to then design a range of 3D-printed GelMA scaffolds with tuneable structural and mechanical properties and understand whether the introduction of pores and porosity would support fibroblast activity, while inhibiting myofibroblast-related gene and protein expression. Results demonstrated that increasing GelMA stiffness promotes myofibroblast activation through increased fibrosis-related gene and protein expression. However, the introduction of a porous architecture by 3D printing facilitated healthy fibroblast activity, while inhibiting myofibroblast activation. A significant reduction was observed in the gene and protein production of αSMA and the expression of ECM-related proteins, including fibronectin I and collagen III, across the range of porous 3D-printed GelMA scaffolds. These results show that the 3D-printed GelMA scaffolds have the potential to improve dermal skin healing, whilst inhibiting fibrosis and scar formation, therefore potentially offering a new treatment for skin repair.

10.
J Mech Behav Biomed Mater ; 114: 104174, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33191173

RESUMEN

Tissue engineering products, like collagen-glycosaminoglycan scaffolds, have been successfully applied to chondrogenic defects. Inducible Pluripotent Stem cell (iPS) technology allows reprograming of somatic cells into an embryonic-like state, allowing for redifferentiation. We postulated that a fibroblast cell line (BJ cells - 'pre-iPSF') cycled through iPS reprogramming and redifferentiated into fibroblasts (post-iPSF) could lubricate collagen-glycosaminoglycan scaffolds; fibroblasts are known to produce lubricating molecules (e.g., lubricin) in the synovium. Herein, we quantified the coefficient of friction (CoF) of collagen-glycosaminoglycan scaffolds seeded with post-iPSF; tested whether cell-free scaffolds made of post-iPSF derived extracellular matrix had reduced friction vs. pre-iPSF; and assessed lubricin quantity as a possible protein responsible for lubrication. Post-iPSF seeded CG had 6- to 10-fold lower CoF versus pre-iPSF. Scaffolds consisting of a collagen and pre-/post-iPSF extracellular matrix blend outperformed these cell-seeded scaffolds (~5-fold lower CoF), yielding excellent CoF values close to synovial fluid. Staining revealed an increased presence of lubricin within post-iPSF scaffolds (confirmed by western blotting) and on the surface of iPSF-seeded collagen-glycosaminoglycan scaffolds. Interestingly, when primary cells from patient biopsy-derived fibroblasts were used, iPS reprogramming did not further reduce the already low CoF of these cells and no lubricin expression was found. We conclude that iPS reprogramming activates lubricating properties in iPS-derived cells in a source cell-specific manner. Additionally, lubricin appears to play a lubricating role, yet other proteins also contribute to lubrication. This work constitutes an important step for understanding post-iPSF lubrication of scaffolds and its potential for cartilage tissue engineering.


Asunto(s)
Condrogénesis , Colágeno , Células Madre Pluripotentes , Andamios del Tejido , Cartílago , Fibroblastos , Humanos
11.
Adv Drug Deliv Rev ; 160: 212-233, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33122088

RESUMEN

Disrupted bone metabolism can lead to delayed fracture healing or non-union, often requiring intervention to correct. Although the current clinical gold standard bone graft implants and commercial bone graft substitutes are effective, they possess inherent drawbacks and are limited in their therapeutic capacity for delayed union and non-union repair. Research into advanced biomaterials and therapeutic biomolecules has shown great potential for driving bone regeneration, although few have achieved commercial success or clinical translation. There are a number of therapeutics, which influence bone remodelling, currently licensed for clinical use. Providing an alternative local delivery context for these therapies, can enhance their efficacy and is an emerging trend in bone regenerative therapeutic strategies. This review aims to provide an overview of how biomaterial design has advanced from currently available commercial bone graft substitutes to accommodate previously licensed therapeutics that target local bone restoration and healing in a synergistic manner, and the challenges faced in progressing this research towards clinical reality.


Asunto(s)
Materiales Biocompatibles/farmacología , Remodelación Ósea/fisiología , Sustitutos de Huesos/administración & dosificación , Sustitutos de Huesos/farmacología , Curación de Fractura/fisiología , Envejecimiento/fisiología , Materiales Biocompatibles/administración & dosificación , Remodelación Ósea/efectos de los fármacos , Curación de Fractura/efectos de los fármacos , Conductas Relacionadas con la Salud , Humanos , Estilo de Vida , Andamios del Tejido
12.
Artículo en Inglés | MEDLINE | ID: mdl-31921799

RESUMEN

Porous collagen-glycosaminoglycan (collagen-GAG) scaffolds have shown promising clinical results for wound healing; however, these scaffolds do not replace the dermal and epidermal layer simultaneously and rely on local endogenous signaling to direct healing. Functionalizing collagen-GAG scaffolds with signaling factors, and/or additional matrix molecules, could help overcome these challenges. An ideal candidate for this is platelet-rich plasma (PRP) as it is a natural reservoir of growth factors, can be activated to form a fibrin gel, and is available intraoperatively. We tested the factors released from PRP (PRPr) and found that at specific concentrations, PRPr enhanced cell proliferation and migration and induced angiogenesis to a greater extent than fetal bovine serum (FBS) controls. This motivated us to develop a strategy to successfully incorporate PRP homogeneously within the pores of the collagen-GAG scaffolds. The composite scaffold released key growth factors for wound healing (FGF, TGFß) and vascularization (VEGF, PDGF) for up to 14 days. In addition, the composite scaffold had enhanced mechanical properties (when compared to PRP gel alone), while providing a continuous upper surface of extracellular matrix (ECM) for keratinocyte seeding. The levels of the factors released from the composite scaffold were sufficient to sustain proliferation of key cells involved in wound healing, including human endothelial cells, mesenchymal stromal cells, fibroblasts, and keratinocytes; even in the absence of FBS supplementation. In functional in vitro and in vivo vascularization assays, our composite scaffold demonstrated increased angiogenic and vascularization potential, which is known to lead to enhanced wound healing. Upon pro-inflammatory induction, macrophages released lower levels of the pro-inflammatory marker MIP-1α when treated with PRPr; and released higher levels of the anti-inflammatory marker IL1-ra upon both pro- and anti-inflammatory induction when treated with the composite scaffold. Finally, our composite scaffold supported a co-culture system of human fibroblasts and keratinocytes that resulted in an epidermal-like layer, with keratinocytes constrained to the surface of the scaffold; by contrast, keratinocytes were observed infiltrating the PRP-free scaffold. This novel composite scaffold has the potential for rapid translation to the clinic by isolating PRP from a patient intraoperatively and combining it with regulatory approved scaffolds to enhance wound repair.

13.
J Biomed Mater Res B Appl Biomater ; 105(1): 136-144, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-26435360

RESUMEN

Cathepsin K inhibitors (CKIs) are an emerging class of drugs that are potent antagonists of osteoclastic activity. We speculated that they may be beneficial in bone tissue engineering, where a stress shielded environment can lead to rapid resorption of new bone. Most CKIs require frequent dosing, so to achieve a sustained release we manufactured polymer nanoparticles encapsulating the CKI L006235 (CKI/nP). CKI/nP and the collagen matrices that were used to deliver them were characterized by electron microscopy and fluorescent confocal microscopy, and data indicated that the particles were evenly distributed throughout the collagen. Elution studies indicated a linear release of the inhibitor from the CKI/nP, with approximately 2% of the drug being released per day. In an in vivo study, mice were implanted with collagen scaffolds containing rhBMP-2 that were loaded with the CKI/nP. Measurement of bone volume (BV) by microCT showed no significant increase with CKI/nP incorporation, and other parameters similarly showed no statistical differences. Cell culture studies confirmed the activity of the drug, even at low concentrations. These data indicate that polymer nanoparticles are an effective method for sustained drug delivery of a CKI, however, this may not be readily translatable to substantively improved bone tissue engineering outcomes. © 2015 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 136-144, 2017.


Asunto(s)
Benzamidas , Proteína Morfogenética Ósea 2 , Catepsina K/antagonistas & inhibidores , Sistemas de Liberación de Medicamentos/métodos , Nanosferas/química , Osteoclastos/metabolismo , Poliglactina 910 , Tiazoles , Animales , Benzamidas/química , Benzamidas/farmacología , Proteína Morfogenética Ósea 2/química , Proteína Morfogenética Ósea 2/farmacología , Humanos , Ratones , Poliglactina 910/química , Poliglactina 910/farmacología , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacología , Tiazoles/química , Tiazoles/farmacología
14.
J Biomed Mater Res A ; 104(1): 291-304, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26386362

RESUMEN

We have previously examined osteoblast behavior on porous collagen-glycosaminoglycan (CG) scaffolds with a range of mean pore sizes demonstrating superior cell attachment and migration in scaffolds with the largest pores (325 µm). Scaffolds provide a framework for construct development; therefore, it is crucial to identify the optimal pore size for augmented tissue formation. Utilizing the same range of scaffolds (85 µm - 325 µm), this study aimed to examine the effects of mean pore size on subsequent osteoblast differentiation and matrix mineralization, and to understand the mechanism by which pore size influences behavior of different cell types. Consequently, primary mesenchymal stem cells (MSCs) were assessed and their behavior compared to osteoblasts. Results demonstrated that scaffolds with the largest pore size (325 µm) facilitated improved osteoblast infiltration, earlier expression of mature bone markers osteopontin (OPN) and osteocalcin (OCN), and increased mineralization. MSCs responded similarly to osteoblasts whereby cell attachment and scaffold infiltration improved with increasing pore size. However, MSCs showed reduced cell motility, proliferation, and scaffold infiltration compared to osteoblasts. This was associated with differences in the profile of integrin subunits (α2) and collagen receptors (CD44), indicating that osteoblasts have a stronger affinity for CG scaffolds compared to MSCs. In summary, these results reveal how larger pores promote improved cell infiltration, essential for construct development, however the optimal scaffold pore size can be cell type specific. As such, this study highlights a necessity to tailor both scaffold micro-architecture and cell-type when designing constructs for successful bone tissue engineering applications.


Asunto(s)
Calcificación Fisiológica/efectos de los fármacos , Colágeno/farmacología , Matriz Extracelular/metabolismo , Glicosaminoglicanos/farmacología , Células Madre Mesenquimatosas/citología , Andamios del Tejido/química , Animales , Bovinos , Adhesión Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Citometría de Flujo , Integrinas/metabolismo , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteopontina/metabolismo , Porosidad , Ratas Wistar , Tiburones
15.
J Tissue Eng ; 6: 2041731415609448, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26668709

RESUMEN

Sucrose acetate isobutyrate (SAIB) is a sugar-based carrier. We have previously applied SAIB as a minimally invasive system for the co-delivery of recombinant human bone morphogenetic protein-2 (rhBMP-2) and found synergy when co-delivering zoledronic acid (ZA) and hydroxyapatite (HA) nanoparticles. Alternative bioceramics were investigated in a murine SAIB/rhBMP-2 injection model. Neither beta-tricalcium phosphate (TCP) nor Bioglass (BG) 45S5 had a significant effect on bone volume (BV) alone or in combination with the ZA. (14)C-labelled ZA binding assays showed particle size and ceramic composition affected binding with nano-HA > micro-HA > TCP > BG. Micro-HA and nano-HA increased BV in a rat model of rhBMP-2/SAIB injection (+278% and +337%), and BV was further increased with ZA-adsorbed micro-HA and nano-HA (+530% and +889%). These data support the use of ZA-adsorbed nanoparticle-sized HA as an optimal additive for the SAIB/rhBMP-2 injectable system for bone tissue engineering.

16.
J Bone Miner Res ; 30(6): 1022-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25484198

RESUMEN

Bisphosphonates (BP) are antiresorptive drugs with a high affinity for bone. Despite the therapeutic success in treating osteoporosis and metabolic bone diseases, chronic BP usage has been associated with reduced repair of microdamage and atypical femoral fracture (AFF). The latter has a poor prognosis, and although anabolic interventions such as teriparatide (PTH(1-34) ) have been suggested as treatment options, there is a limited evidence base in support of their efficacy. Because PTH(1-34) acts to increase bone turnover, we hypothesized that it may be able to increase BP in turnover in the skeleton, which, in turn, may improve bone healing. To test this, we employed a mixture of fluorescent Alexa647-labelled pamidronate (Pam) and radiolabeled (14) C-ZA (zoledronic acid). These traceable BPs were dosed to Wistar rats in models of normal growth and closed fracture repair. Rats were cotreated with saline or 25 µg/kg/d PTH(1-34) , and the effects on BP liberation and bone healing were examined by X-ray, micro-CT, autoradiography, and fluorescent confocal microscopy. Consistent with increased BP remobilization with PTH(1-34) , there was a significant decrease in fluorescence in both the long bones and in the fracture callus in treated animals compared with controls. This was further confirmed by autoradiography for (14) C-ZA. In this model of acute BP treatment, callus bone volume (BV) was significantly increased in fractured limbs, and although we noted significant decreases in callus-bound BP with PTH(1-34) , these were not sufficient to alter this BV. However, increased intracellular BP was noted in resorbing osteoclasts, confirming that, in principle, PTH(1-34) increases bone turnover as well as BP turnover.


Asunto(s)
Difosfonatos , Fracturas del Fémur , Curación de Fractura/efectos de los fármacos , Imidazoles , Hormona Paratiroidea , Animales , Isótopos de Carbono/farmacocinética , Isótopos de Carbono/farmacología , Difosfonatos/farmacocinética , Difosfonatos/farmacología , Fracturas del Fémur/tratamiento farmacológico , Fracturas del Fémur/metabolismo , Imidazoles/farmacocinética , Imidazoles/farmacología , Marcaje Isotópico , Masculino , Pamidronato , Hormona Paratiroidea/farmacocinética , Hormona Paratiroidea/farmacología , Ratas , Ratas Wistar , Ácido Zoledrónico
17.
J Orthop Res ; 32(12): 1549-56, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25224138

RESUMEN

Activated Protein C (APC) is an anticoagulant with strong cytoprotective properties that has been shown to promote wound healing. In this study APC was investigated for its potential orthopedic application using a Bone Morphogenetic Protein 2 (rhBMP-2) induced ectopic bone formation model. Local co-administration of 10 µg rhBMP-2 with 10 µg or 25 µg APC increased bone volume at 3 weeks by 32% (N.S.) and 74% (p<0.01) compared to rhBMP-2 alone. This was associated with a significant increase in CD31+ and TRAP+ cells in tissue sections of ectopic bone, consistent with enhanced vascularity and bone turnover. The actions of APC are largely mediated by its receptors endothelial protein C receptor (EPCR) and protease-activated receptors (PARs). Cultured pre-osteoblasts and bone nodule tissue sections were shown to express PAR1/2 and EPCR. When pre-osteoblasts were treated with APC, cell viability and phosphorylation of ERK1/2, Akt, and p38 were increased. Inhibition with PAR1 and sometimes PAR2 antagonists, but not with EPCR blocking antibodies, ameliorated the effects of APC on cell viability and kinase phosphorylation. These data indicate that APC can affect osteoblast viability and signaling, and may have in vivo applications with rhBMP-2 for bone repair.


Asunto(s)
Huesos/efectos de los fármacos , Proteína C/farmacología , Receptor PAR-1/fisiología , Receptor PAR-2/fisiología , Animales , Proteína Morfogenética Ósea 2/farmacología , Resorción Ósea/prevención & control , Huesos/metabolismo , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Osteoblastos/metabolismo , Osteogénesis/efectos de los fármacos , Proteína C/metabolismo
18.
Int Orthop ; 38(7): 1527-33, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24389948

RESUMEN

PURPOSE: Legg-Calve-Perthes disease is a paediatric condition encompassing idiopathic osteonecrosis of the femoral head (ONFH). Preventing collapse and the need for subsequent joint replacement remains the major goal of clinical management. This exploratory study utilises a porcine model of surgically induced ONFH. METHODS: rhBMP-2 with and without zoledronic acid (ZA) was delivered by intra-osseous injection in the phase-transitioning sucrose acetate isobutyrate (SAIB) in an attempt to prevent femoral head collapse. Epiphyseal quotient (EQ) at eight weeks post-surgery was the primary outcome measure. Heterotopic ossification in the joint capsule and bisphosphonate retention in the femoral head were key secondary outcomes. RESULTS: Femoral heads with ONFH and no treatment all collapsed (3/3, EQ < 0.4, P < 0.05 compared to no ONFH). Local delivery of rhBMP-2/SAIB into the femoral head prevented collapse by EQ measurement one of four samples; however, this specimen still showed evidence of significant collapse. In contrast, the combination of local rhBMP-2 and local ZA prevented collapse in two of four samples. Confocal fluorescence microscopy showed locally dosed bisphosphonate entered and was retained in the femoral head. This group also showed strong Calcein signal, indicating new bone formation. Treatment with rhBMP-2 was associated with a limited amount of heterotrophic ossification in the joint capsules in some specimens. CONCLUSIONS: Operators reported SAIB to be an efficient way to deliver rhBMP-2 to the femoral head. These data suggest that rhBMP-2 is ineffective for preventing femoral head collapse without the addition of bisphosphonate. Further research will be required to validate the clinical efficacy of a combined local rhBMP-2/bisphosphonate approach.


Asunto(s)
Conservadores de la Densidad Ósea/administración & dosificación , Proteína Morfogenética Ósea 2/administración & dosificación , Difosfonatos/administración & dosificación , Necrosis de la Cabeza Femoral/prevención & control , Imidazoles/administración & dosificación , Enfermedad de Legg-Calve-Perthes/tratamiento farmacológico , Factor de Crecimiento Transformador beta/administración & dosificación , Animales , Portadores de Fármacos , Necrosis de la Cabeza Femoral/etiología , Inyecciones , Enfermedad de Legg-Calve-Perthes/complicaciones , Proyectos Piloto , Proteínas Recombinantes/administración & dosificación , Sacarosa/análogos & derivados , Porcinos , Ácido Zoledrónico
19.
Acta Biomater ; 10(5): 2250-8, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24456759

RESUMEN

An emerging paradigm in orthopedics is that a bone-healing outcome is the product of the anabolic (bone-forming) and catabolic (bone-resorbing) outcomes. Recently, surgical and tissue engineering strategies have emerged that combine recombinant human bone morphogenetic proteins (rhBMPs) and bisphosphonates (BPs) in order to maximize anabolism and minimize catabolism. Collagen-based scaffolds that are the current surgical standard can bind rhBMPs, but not BPs. We hypothesized that a biomimetic collagen-hydroxyapatite (CHA) scaffold would bind both agents and produce superior in vivo outcomes. Consistent with this concept, in vitro elution studies utilizing rhBMP-2 ELISA assays and scintillation counting of (14)C-radiolabeled zoledronic acid (ZA) confirmed delayed release of both agents from the CHA scaffold. Next, scaffolds were tested for their capacity to form ectopic bone after surgical implantation into the rat hind limb. Using CHA, a significant 6-fold increase in bone volume was seen in rhBMP-2/ZA groups compared to rhBMP-2 alone, confirming the ability of ZA to enhance rhBMP-2 bone formation. CHA scaffolds were found to be capable of generating mineralized tissue in the absence of rhBMP-2. This study has implications for future clinical treatments of critical bone defects. It demonstrates the relative advantages of co-delivering anabolic and anti-catabolic agents using a multicomponent scaffold system.


Asunto(s)
Proteínas Morfogenéticas Óseas/farmacología , Colágeno/química , Difosfonatos/farmacología , Sistemas de Liberación de Medicamentos , Durapatita/química , Proteínas Recombinantes/farmacología , Fosfatasa Ácida/metabolismo , Animales , Huesos/diagnóstico por imagen , Huesos/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Colorantes Fluorescentes/metabolismo , Humanos , Isoenzimas/metabolismo , Masculino , Ratones , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteogénesis/efectos de los fármacos , Porosidad , Implantación de Prótesis , Ratas , Ratas Wistar , Fosfatasa Ácida Tartratorresistente , Andamios del Tejido , Microtomografía por Rayos X
20.
Eur Cell Mater ; 26: 120-32, 2013 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-24052425

RESUMEN

Bone tissue engineering has emerged as one of the leading fields in tissue engineering and regenerative medicine. The success of bone tissue engineering relies on understanding the interplay between progenitor cells, regulatory signals, and the biomaterials/scaffolds used to deliver them--otherwise known as the tissue engineering triad. This review will discuss the roles of these fundamental components with a specific focus on the interaction between cell behaviour and scaffold structural properties. In terms of scaffold architecture, recent work has shown that pore size can affect both cell attachment and cellular invasion. Moreover, different materials can exert different biomechanical forces, which can profoundly affect cellular differentiation and migration in a cell type specific manner. Understanding these interactions will be critical for enhancing the progress of bone tissue engineering towards clinical applications.


Asunto(s)
Regeneración Ósea , Osteoblastos/metabolismo , Células Madre/metabolismo , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Materiales Biocompatibles/farmacología , Humanos , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteogénesis , Células Madre/citología , Células Madre/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...