Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Nat Mater ; 14(12): 1269-77, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26366848

RESUMO

The effectiveness of stem cell therapies has been hampered by cell death and limited control over fate. These problems can be partially circumvented by using macroporous biomaterials that improve the survival of transplanted stem cells and provide molecular cues to direct cell phenotype. Stem cell behaviour can also be controlled in vitro by manipulating the elasticity of both porous and non-porous materials, yet translation to therapeutic processes in vivo remains elusive. Here, by developing injectable, void-forming hydrogels that decouple pore formation from elasticity, we show that mesenchymal stem cell (MSC) osteogenesis in vitro, and cell deployment in vitro and in vivo, can be controlled by modifying, respectively, the hydrogel's elastic modulus or its chemistry. When the hydrogels were used to transplant MSCs, the hydrogel's elasticity regulated bone regeneration, with optimal bone formation at 60 kPa. Our findings show that biophysical cues can be harnessed to direct therapeutic stem cell behaviours in situ.


Assuntos
Desenvolvimento Ósseo , Matriz Extracelular/fisiologia , Hidrogéis , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Materiais Biocompatíveis , Elasticidade
2.
Biomacromolecules ; 17(7): 2329-36, 2016 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-27285121

RESUMO

Osteonecrosis of the femoral head (ONFH) is a debilitating hip disorder, which often produces a permanent femoral head deformity and osteoarthritis. The local delivery of biological agents capable of stimulating bone healing offer potential new treatment options for patients with ONFH. Previous studies from our laboratory have shown that a local intraosseous infusion of bone morphogenic protein-2 (BMP-2) was effective in stimulating new bone formation in a piglet model of ischemic ONFH. However, infusion of BMP-2 solution was associated with unwanted dissemination of BMP-2 out of the femoral head and produced heterotopic ossification in the hip capsule. Injectable hydrogels offer a potential method to control the dissemination of biological molecules in vivo. In the present study, we evaluated the potential of a peptide-based, self-assembling hydrogel called RADA16 to transition from a solution to a gel following infusion into the femoral head, thereby preventing backflow, as well as its potential use as a delivery vehicle for BMP-2. Cadaver pig femoral heads were used to study the backflow and the distribution of RADA16 following an intraosseous infusion. Microcomputed tomography analysis following the infusion of RADA16 mixed with a radiocontrast agent revealed a significant decrease in the amount of back flow of radiocontrast agent down the needle track compared to the soluble infusion of radiocontrast without RADA16. Furthermore, RADA16 mixed with radiocontrast agent showed good distribution within the femoral head. In addition, in vitro experiments revealed that higher concentrations of RADA16 decreased the rate of BMP-2 dissemination out of the hydrogel. The BMP-2 that was released from RADA16 maintains its biological activity, inducing the phosphorylation of SMAD1/5/8 in pig primary bone marrow stromal cells. Lastly, pig primary bone marrow stromal cells showed significantly increased in vitro proliferation on RADA16 hydrogels over 1 week compared to tissue culture plastic, suggesting that it is a suitable matrix for supporting cellular proliferation. In conclusion, RADA16 showed potential for use as a drug delivery vehicle to control the delivery of BMP-2 within the femoral head. This novel therapy may be able to provide benefits to patients suffering from debilitating conditions such as osteonecrosis of the femoral head.


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Sistemas de Liberação de Medicamentos , Cabeça do Fêmur/efeitos dos fármacos , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Osteogênese/efeitos dos fármacos , Peptídeos/administração & dosagem , Animais , Proteína Morfogenética Óssea 2/administração & dosagem , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cabeça do Fêmur/metabolismo , Cabeça do Fêmur/patologia , Hidrogel de Polietilenoglicol-Dimetacrilato/administração & dosagem , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Peptídeos/química , Fosforilação/efeitos dos fármacos , Proteínas Smad/metabolismo , Suínos , Microtomografia por Raio-X
3.
Biomacromolecules ; 15(2): 445-55, 2014 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-24400664

RESUMO

Many strategies for controlling the fate of transplanted stem cells rely on the concurrent delivery of soluble growth factors that have the potential to produce undesirable secondary effects in surrounding tissue. Such off target effects could be eliminated by locally presenting growth factor peptide mimics from biomaterial scaffolds to control stem cell fate. Peptide mimics of bone morphogenetic protein 2 (BMP-2) were synthesized by solid phase Fmoc-peptide synthesis and covalently bound to alginate hydrogels via either carbodiimide or sulfhydryl-based coupling strategies. Successful peptide conjugation was confirmed by (1)H NMR spectroscopy and quantified by fluorescently labeling the peptides. Peptides derived from the knuckle epitope of BMP-2, presented from both 2D surfaces and 3D alginate hydrogels, were shown to increase alkaline phosphatase activity in clonally derived murine osteoblasts. Furthermore, when presented in 3D hydrogels, these peptides were shown to initiate Smad signaling, upregulate osteopontin production, and increase mineral deposition with clonally derived murine mesenchymal stem cells. These data suggest that these peptide-conjugated hydrogels may be effective alternatives to local BMP-2 release in directly and spatially eliciting osteogenesis from transplanted or host osteoprogenitors in the future.


Assuntos
Materiais Biomiméticos/farmacologia , Proteína Morfogenética Óssea 2/química , Hidrogéis/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Peptídeos/farmacologia , Materiais Biomiméticos/síntese química , Materiais Biomiméticos/química , Células Cultivadas , Humanos , Hidrogéis/síntese química , Hidrogéis/química , Espectroscopia de Ressonância Magnética , Células-Tronco Mesenquimais/citologia , Estrutura Molecular , Osteoblastos/citologia , Peptídeos/síntese química , Peptídeos/química , Proteínas Recombinantes/química
4.
Eur Cell Mater ; 24: 358-71; discussion 371, 2012 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-23124984

RESUMO

This study addresses the hypothesis that callus formation, patterning, and mineralisation are impaired during the early phase of critical sized bone defect healing, and may relate to inter-fragmentary tissue strains within the bone defect area. Twenty four 12 week old Sprague Dawley rats were used for this study. They were divided into two groups defined by the femur bone defect size: (i) 1 mm resulting in normal healing (NH), and (ii) a large sized 5 mm defect resulting in critical healing (CH). Callus formation, patterning, and mineralisation kinetics in both groups were examined in the periosteal and osteotomy gap regions using a novel longitudinal study setup. Finite element analyses on µCT generated tomograms were used to determine inter-fragmentary tissue strain patterns and compared to callus formation and patterning over the course of time. Using a novel longitudinal study technique with µCT, in vivo tracking and computer simulation approaches, this study demonstrates that: (i) periosteal bone formation and patterning are significantly influenced by bone defect size as early as 2 weeks; (ii) osteotomy gap callus formation and patterning are influenced by bone defect size, and adapt towards a non-union in critical cases by deviating into a medullary formation route as early as 2 weeks after osteotomy; (iii) the new bone formation in the osteotomy gap enclosing the medullary cavity in the CH group is highly mineralised; (iv) inter-fragmentary strain patterns predicted during the very early soft callus tissue phase (less than 2 weeks) are concurrent with callus formation and patterning at later stages. In conclusion, bone defect size influences early onset of critical healing patterns.


Assuntos
Calo Ósseo/fisiologia , Fêmur/fisiologia , Cicatrização , Animais , Feminino , Fêmur/diagnóstico por imagem , Fêmur/cirurgia , Osteotomia , Periósteo/fisiologia , Ratos , Ratos Sprague-Dawley , Estresse Mecânico , Microtomografia por Raio-X
5.
Clin Orthop Relat Res ; 469(11): 3102-10, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21590486

RESUMO

BACKGROUND: Gender and stability of fixation independently influence bone regeneration but their combined effects are unclear. QUESTIONS/PURPOSES: In a pilot study we determined the combined influence of gender and fixation stability on the callus of middle-aged rats regarding (1) biomechanical properties; (2) bridging over time; (3) callus formation; and (4) callus size, geometry, mineralization, and microstructure. METHODS: We osteotomized the left femur of 32 Sprague-Dawley rats (12 months old). Femurs were externally fixed with a gap of 1.5 mm in four groups of eight animals each: female semirigid, male semirigid, female rigid, and male rigid. Qualitative and quantitative in vivo radiographic analyses were performed twice weekly. Six weeks postoperatively, harvested femora were evaluated using micro-CT and biomechanical testing. RESULTS: Torsional stiffness and maximum torque at failure were higher in male and in semirigidly fixed fractures. Radiographic analysis revealed earlier bridging and callus formation in both male groups. Micro-CT analysis showed a larger callus size, altered geometry, and microstructure in males and semirigidly fixed animals, whereas mineralization was similar in all animals. CONCLUSION: Our data suggest female gender represents an independent risk factor for bone healing in middle-aged rats. Although healing in females was delayed compared with males, they exhibited a similar response (superior callus properties) to a more semirigid fixation. CLINICAL RELEVANCE: While female gender appears to reflect a risk for impaired bone healing in middle-aged female rats, clinical studies would be required to confirm the finding in humans.


Assuntos
Fraturas do Fêmur/cirurgia , Fixação de Fratura/métodos , Consolidação da Fratura/fisiologia , Fatores Etários , Animais , Regeneração Óssea/fisiologia , Calo Ósseo/fisiologia , Modelos Animais de Doenças , Feminino , Fêmur/fisiologia , Fêmur/cirurgia , Masculino , Osteotomia/métodos , Projetos Piloto , Ratos , Ratos Sprague-Dawley , Fatores Sexuais
6.
Arch Orthop Trauma Surg ; 131(1): 121-9, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20652815

RESUMO

INTRODUCTION: The objectives of this study were to (1) establish a reproducible atrophic non-union model in rats by creation of a segmental femoral bone defect that allows, (2) in-depth characterization of impaired healing, and (3) contrast its healing patterns to the normal course. Hypothesis was that a 5-mm bone defect in male rats would deviate from uneventful healing patterns and result in an atrophic non-union. MATERIALS AND METHODS: A femoral osteotomy was performed in two groups of 12-week-old male rats (1 vs. 5 mm gap) stabilized with an external fixator. Bone healing in these models was evaluated by radiology, biomechanics, and histology at 6 or 8 weeks. The evaluation of the 5-mm group revealed in some cases a delayed rather than a non-union, and therefore, a group of female counterparts was included. RESULTS: The creation of a 5-mm defect in female rats resulted in a reproducible atrophic non-union characterized by sealing of the medullary canal, lack of cartilage formation, and negligible mechanical properties of the callus. In both gap size models, the male subjects showed advanced healing compared to females. DISCUSSION AND CONCLUSION: This study showed that even under uneventful healing conditions in terms of age and bone defect size, there is a sex-specific advanced healing in male compared to female subjects. Contrary to our initial hypothesis, only the creation of a 5-mm segmental femoral defect in female rats led to a reproducible atrophic non-union. It has been shown that an atrophic non-union exhibits different healing patterns compared to uneventful healing. A total lack of endochondral bone formation, soft tissue prolapse into the defect, and bony closure of the medullary cavity have been shown to occur in the non-union model.


Assuntos
Fraturas do Fêmur/cirurgia , Animais , Atrofia , Calo Ósseo/patologia , Modelos Animais de Doenças , Fixadores Externos , Feminino , Processamento de Imagem Assistida por Computador , Masculino , Ratos , Ratos Sprague-Dawley , Fatores Sexuais , Cicatrização
7.
Clin Orthop Relat Res ; 467(3): 850-4, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18820984

RESUMO

The use of cementless threaded cups in THA is a well-established treatment. Fractures of the cups are rare complications recorded in individual cases with material defects being discussed as the primary cause. We analyzed three cases of fractured cups. Although all three cups were well fixed to existing bone, we observed deficient osseous backing dorsocranially and abrasion particles. There were no signs of femoroacetabular impingement or infection. The cups showed corrosion debris. Scanning electron microscopic investigations showed characteristics of fretting and fretting-related corrosion. We concluded the fractures occurred because of fretting combined with inadequate bony support leading to fatigue of the material and subsequent fracture.


Assuntos
Artrite/cirurgia , Artroplastia de Quadril/instrumentação , Luxação Congênita de Quadril/cirurgia , Articulação do Quadril/cirurgia , Prótese de Quadril , Falha de Prótese , Adulto , Artrite/diagnóstico por imagem , Feminino , Luxação Congênita de Quadril/diagnóstico por imagem , Articulação do Quadril/diagnóstico por imagem , Humanos , Masculino , Desenho de Prótese , Radiografia , Reoperação , Estresse Mecânico , Propriedades de Superfície
8.
J Biomech ; 41(12): 2696-702, 2008 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-18644596

RESUMO

Mechanical conditions have a significant influence on the biological processes of bone healing. Small animal models that allow controlling the mechanical environment of fracture and bone defect healing are needed. The aim of this study was to develop a new animal model that allows to reliably control the mechanical environment in fracture and bone defect healing in rats using different implant materials. An external fixator was designed and mounted in vitro to rat femurs using four Kirschner-wires (titanium (T) or steel (S)) of 1.2 mm diameter. The specimens were distracted to a gap of 1.5mm. Axial and torsional stiffness of the device was tested increasing the offset (distance between bone and fixator crossbar) from 5 to 15 mm. In vivo performance (well-being, infection, breaking of wires and bone healing) was evaluated in four groups of 24 Sprague-Dawley rats varying in offset (7.5 and 15 mm) and implant material (S/T) over 6 weeks. Torsional and axial stiffness were higher in steel compared to titanium setups. A decrease in all configurations was observed by increasing the offset. The offset 7.5 mm showed a significantly higher torsional (S: p<0.01, T: p<0.001) and axial in vitro stiffness (S: p<0.001, T: p<0.001) compared to 15 mm offset of the fixator. Although in vitro designed to be different in mechanical stiffness, no difference was found between the groups regarding complication rate. The overall-complication rate was 5.2%. In conclusion, we were able to establish a small animal model for bone defect healing which allows modeling the mechanical conditions at the defect site in a defined manner.


Assuntos
Fraturas do Fêmur/fisiopatologia , Fraturas do Fêmur/terapia , Fixação de Fratura/instrumentação , Consolidação da Fratura/fisiologia , Animais , Fenômenos Biofísicos , Módulo de Elasticidade , Desenho de Equipamento , Análise de Falha de Equipamento , Feminino , Fixação de Fratura/métodos , Ratos , Ratos Sprague-Dawley , Estresse Mecânico
9.
Biosens Bioelectron ; 23(5): 728-34, 2007 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17888647

RESUMO

An integrated array of micron-dimension capacitors, originally developed for biometric applications (fingerprint identification), was engineered for detection of biological agents such as proteins and bacteria. This device consists of an array of 93,184 (256 x 364) individual capacitor-based sensing elements located underneath a thin (0.8 microm) layer of glass. This glass layer can be functionalized with organosilane-based monolayers to provide groups amenable for the immobilization of bioreceptors such as antibodies, enzymes, peptides, aptamers, and nucleotides. Upon functionalization with antibodies and in conjunction with signal amplification schemes that result in perturbation of the dielectric constant around the captured antigens, this system can be used as a detector of biological agents. Two signal amplification schemes were tested in this work: one consisted of 4 microm diameter latex immunobeads and a second one was based on colloidal gold catalyzed reduction of silver. These signal amplification approaches were demonstrated and show that this system is capable of specific detection of bacteria (Escherichia coli) and proteins (ovalbumin). The present work shows proof-of-principle demonstration that a simple fingerprint detector based on feedback capacitance measurements can be implemented as a biosensor. The approach presented could be easily expanded to simultaneously test for a large number of analytes and multiple samples given that this device has a large number of detectors. The device and required instrumentation is highly portable and does not require expensive and bulky instrumentation because it relies purely on electronic detection.


Assuntos
Bactérias/isolamento & purificação , Técnicas Biossensoriais , Capacitância Elétrica , Procedimentos Analíticos em Microchip , Proteínas/análise , Condutividade Elétrica , Microesferas , Soluções
10.
Acta Biomater ; 60: 50-63, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28739546

RESUMO

In-situ tissue regeneration aims to utilize the body's endogenous healing capacity through the recruitment of host stem or progenitor cells to an injury site. Stromal cell-derived factor-1α (SDF-1α) is widely discussed as a potent chemoattractant. Here we use a cell-free biomaterial-based approach to (i) deliver SDF-1α for the recruitment of endogenous bone marrow-derived stromal cells (BMSC) into a critical-sized segmental femoral defect in rats and to (ii) induce hydrogel stiffness-mediated osteogenic differentiation in-vivo. Ionically crosslinked alginate hydrogels with a stiffness optimized for osteogenic differentiation were used. Fast-degrading porogens were incorporated to impart a macroporous architecture that facilitates host cell invasion. Endogenous cell recruitment to the defect site was successfully triggered through the controlled release of SDF-1α. A trend for increased bone volume fraction (BV/TV) and a significantly higher bone mineral density (BMD) were observed for gels loaded with SDF-1α, compared to empty gels at two weeks. A trend was also observed, albeit not statistically significant, towards matrix stiffness influencing BV/TV and BMD at two weeks. However, over a six week time-frame, these effects were insufficient for bone bridging of a segmental femoral defect. While mechanical cues combined with ex-vivo cell encapsulation have been shown to have an effect in the regeneration of less demanding in-vivo models, such as cranial defects of nude rats, they are not sufficient for a SDF-1α mediated in-situ regeneration approach in segmental femoral defects of immunocompetent rats, suggesting that additional osteogenic cues may also be required. STATEMENT OF SIGNIFICANCE: Stromal cell-derived factor-1α (SDF-1α) is a chemoattractant used to recruit host cells for tissue regeneration. The concept that matrix stiffness can direct mesenchymal stromal cell (MSC) differentiation into various lineages was described a decade ago using in-vitro experiments. Recently, alginate hydrogels with an optimized stiffness and ex-vivo encapsulated MSCs were shown to have an effect in the regeneration of skull defects of nude rats. Here, we apply this material system, loaded with SDF-1α and without encapsulated MSCs, to (i) recruit endogenous cells and (ii) induce stiffness-mediated osteogenic differentiation in-vivo, using as model system a load-bearing femoral defect in immunocompetent rats. While a cell-free approach is of great interest from a translational perspective, the current limitations are described.


Assuntos
Células da Medula Óssea/metabolismo , Diferenciação Celular/efeitos dos fármacos , Quimiocina CXCL12 , Fêmur , Hidrogéis , Osteogênese/efeitos dos fármacos , Animais , Densidade Óssea/efeitos dos fármacos , Células da Medula Óssea/patologia , Quimiocina CXCL12/química , Quimiocina CXCL12/farmacocinética , Quimiocina CXCL12/farmacologia , Implantes de Medicamento/química , Implantes de Medicamento/farmacocinética , Implantes de Medicamento/farmacologia , Feminino , Fêmur/lesões , Fêmur/metabolismo , Fêmur/patologia , Hidrogéis/química , Hidrogéis/farmacocinética , Hidrogéis/farmacologia , Ratos , Ratos Sprague-Dawley , Células Estromais/metabolismo , Células Estromais/patologia
11.
Tissue Eng Part A ; 21(7-8): 1217-27, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25434326

RESUMO

Biomaterials may improve outcomes of endothelial progenitor-based therapies for the treatment of ischemic cardiovascular disease, due to their ability to direct cell behavior. We hypothesized that local, sustained delivery of exogenous vascular endothelial growth factor (VEGF) and stromal cell-derived factor (SDF) from alginate hydrogels could increase recruitment of systemically infused endothelial progenitors to ischemic tissue, and subsequent neovascularization. VEGF and SDF were found to enhance in vitro adhesion and migration of outgrowth endothelial cells (OECs) and circulating angiogenic cells (CACs), two populations of endothelial progenitors, by twofold to sixfold, and nearly doubled recruitment to both ischemic and nonischemic muscle tissue in vivo. Local delivery of VEGF and SDF to ischemic hind-limbs in combination with systemic CAC delivery significantly improved functional perfusion recovery over OEC delivery, or either treatment alone. Compared with OECs, CACs were more responsive to VEGF and SDF treatment, promoted in vitro endothelial sprout formation in a paracrine manner more potently, and demonstrated greater influence on infiltrating inflammatory cells in vivo. These studies demonstrate that accumulation of infused endothelial progenitors can be enriched using biomaterial-based delivery of VEGF and SDF, and emphasize the therapeutic benefit of using CACs for the treatment of ischemia.


Assuntos
Quimiocina CXCL12/farmacologia , Sistemas de Liberação de Medicamentos , Células Progenitoras Endoteliais/citologia , Células Progenitoras Endoteliais/transplante , Isquemia/terapia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Alginatos/farmacologia , Animais , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/crescimento & desenvolvimento , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Progenitoras Endoteliais/efeitos dos fármacos , Ácido Glucurônico/farmacologia , Ácidos Hexurônicos/farmacologia , Membro Posterior/irrigação sanguínea , Membro Posterior/patologia , Membro Posterior/cirurgia , Humanos , Hidrogéis/farmacologia , Inflamação/patologia , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/efeitos dos fármacos , Reperfusão
12.
J Biomed Mater Res A ; 103(11): 3516-25, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25953514

RESUMO

Interactions between cells and the extracellular matrix (ECM) are known to play critical roles in regulating cell phenotype. The identity of ECM ligands presented to mesenchymal stem cells (MSCs) has previously been shown to direct the cell fate commitment of these cells. To enhance osteogenic differentiation of MSCs, alginate hydrogels were prepared that present the DGEA ligand derived from collagen I. When presented from hydrogel surfaces in 2D, the DGEA ligand did not facilitate cell adhesion, while hydrogels presenting the RGD ligand derived from fibronectin did encourage cell adhesion and spreading. However, the osteogenic differentiation of MSCs encapsulated within alginate hydrogels presenting the DGEA ligand was enhanced when compared with unmodified alginate hydrogels and hydrogels presenting the RGD ligand. MSCs cultured in DGEA-presenting gels exhibited increased levels of osteocalcin production and mineral deposition. These data suggest that the presentation of the collagen I-derived DGEA ligand is a feasible approach for selectively inducing an osteogenic phenotype in encapsulated MSCs.


Assuntos
Colágeno Tipo I/farmacologia , Hidrogéis/farmacologia , Células-Tronco Mesenquimais/citologia , Oligopeptídeos/farmacologia , Osteogênese/efeitos dos fármacos , Peptídeos/farmacologia , Fosfatase Alcalina/metabolismo , Animais , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Fenótipo , Ratos
13.
ACS Nano ; 9(9): 9394-406, 2015 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-26284753

RESUMO

Uncontrolled bleeding from traumatic wounds is a major factor in deaths resulting from military conflict, accidents, disasters and crime. Self-assembling peptide nanofibers have shown superior hemostatic activity, and herein, we elucidate their mechanism by visualizing the formation of nanofiber-based clots that aggregate blood components with a similar morphology to fibrin-based clots. Furthermore, to enhance its direct application to a wound, we developed layer-by-layer assembled thin film coatings onto common materials used for wound dressings-gauze and gelatin sponges. We find these nanofibers elute upon hydration under physiological conditions and generate nanofiber-based clots with blood. After exposure to a range of harsh temperature conditions (-80 to 60 °C) for a week and even 5 months at 60 °C, these hemostatic bandages remain capable of releasing active nanofibers. In addition, the application of these nanofiber-based films from gauze bandages was found to accelerate hemostasis in porcine skin wounds as compared to plain gauze. The thermal robustness, in combination with the self-assembling peptide's potent hemostatic activity, biocompatibility, biodegradability, and low cost of production, makes this a promising approach for a cheap yet effective hemostatic bandage.


Assuntos
Bandagens , Hemorragia/tratamento farmacológico , Nanofibras/uso terapêutico , Técnicas de Fechamento de Ferimentos , Animais , Coagulação Sanguínea/efeitos dos fármacos , Modelos Animais de Doenças , Hemostáticos , Nanofibras/química , Peptídeos/química , Peptídeos/uso terapêutico , Suínos , Cicatrização
14.
Adv Healthc Mater ; 3(4): 500-7, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24030924

RESUMO

Electrically responsive hydrogels are created with interconnected macropores, which greatly enhance their ability to rapidly undergo volumetric collapse when subjected to moderate electric fields. When optimized, these electrogels are easily integrated into arrays capable of rapid configurational and chromatic optical modulations, and when loaded with drugs, are able to coordinate the delivery profile of multiple drugs.


Assuntos
Materiais Biocompatíveis/química , Hidrogéis/química , Eletroquímica , Tamanho da Partícula , Polímeros/química , Porosidade , Água/química
15.
Adv Healthc Mater ; 3(11): 1869-76, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24862232

RESUMO

Ferrogels are an attractive material for many biomedical applications due to their ability to deliver a wide variety of therapeutic drugs on-demand. However, typical ferrogels have yet to be optimized for use in cell-based therapies, as they possess limited ability to harbor and release viable cells. Previously, an active porous scaffold that exhibits large deformations and enhanced biological agent release under moderate magnetic fields has been demonstrated. Unfortunately, at small device sizes optimal for implantation (e.g., 2 mm thickness), these monophasic ferrogels no longer achieve significant deformation due to a reduced body force. A new biphasic ferrogel, containing an iron oxide gradient, capable of large deformations and triggered release even at small gel dimensions, is presented in this study. Biphasic ferrogels demonstrate increased porosity, enhanced mechanical properties, and potentially increased biocompatibility due to their reduced iron oxide content. With their ability to deliver drugs and cells on-demand, it is expected that these ferrogels will have wide utility in the fields of tissue engineering and regenerative medicine.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Compostos Férricos/química , Hidrogéis/química , Animais , Portadores de Fármacos/química , Feminino , Magnetismo , Camundongos , Camundongos Endogâmicos C57BL , Porosidade , Medicina Regenerativa/métodos , Engenharia Tecidual/métodos
16.
Dent Mater ; 30(7): e199-207, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24768062

RESUMO

OBJECTIVES: Calcium phosphate cement (CPC) is promising for dental and craniofacial applications due to its ability to be injected or filled into complex-shaped bone defects and molded for esthetics, and its resorbability and replacement by new bone. The objective of this study was to investigate bone regeneration via novel macroporous CPC containing absorbable fibers, hydrogel microbeads and growth factors in critical-sized cranial defects in rats. METHODS: Mannitol porogen and alginate hydrogel microbeads were incorporated into CPC. Absorbable fibers were used to provide mechanical reinforcement to CPC scaffolds. Six CPC groups were tested in rats: (1) control CPC without macropores and microbeads; (2) macroporous CPC+large fiber; (3) macroporous CPC+large fiber+nanofiber; (4) same as (3), but with rhBMP2 in CPC matrix; (5) same as (3), but with rhBMP2 in CPC matrix+rhTGF-ß1 in microbeads; (6) same as (3), but with rhBMP2 in CPC matrix+VEGF in microbeads. Rats were sacrificed at 4 and 24 weeks for histological and micro-CT analyses. RESULTS: The macroporous CPC scaffolds containing porogen, absorbable fibers and hydrogel microbeads had mechanical properties similar to cancellous bone. At 4 weeks, the new bone area fraction (mean±sd; n=5) in CPC control group was the lowest at (14.8±3.3)%, and that of group 6 (rhBMP2+VEGF) was (31.0±13.8)% (p<0.05). At 24 weeks, group 4 (rhBMP2) had the most new bone of (38.8±15.6)%, higher than (12.7±5.3)% of CPC control (p<0.05). Micro-CT revealed nearly complete bridging of the critical-sized defects with new bone for several macroporous CPC groups, compared to much less new bone formation for CPC control. SIGNIFICANCE: Macroporous CPC scaffolds containing porogen, fibers and microbeads with growth factors were investigated in rat cranial defects for the first time. Macroporous CPCs had new bone up to 2-fold that of traditional CPC control at 4 weeks, and 3-fold that of traditional CPC at 24 weeks, and hence may be useful for dental, craniofacial and orthopedic applications.


Assuntos
Regeneração Óssea , Fosfatos de Cálcio , Cimentos Dentários , Crânio/anormalidades , Alicerces Teciduais , Animais , Teste de Materiais , Porosidade , Ratos
17.
Biomaterials ; 34(33): 8042-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23896005

RESUMO

Although hydrogels now see widespread use in a host of applications, low fracture toughness and brittleness have limited their more broad use. As a recently described interpenetrating network (IPN) of alginate and polyacrylamide demonstrated a fracture toughness of ≈ 9000 J/m(2), we sought to explore the biocompatibility and maintenance of mechanical properties of these hydrogels in cell culture and in vivo conditions. These hydrogels can sustain a compressive strain of over 90% with minimal loss of Young's Modulus as well as minimal swelling for up to 50 days of soaking in culture conditions. Mouse mesenchymal stem cells exposed to the IPN gel-conditioned media maintain high viability, and although cells exposed to conditioned media demonstrate slight reductions in proliferation and metabolic activity (WST assay), these effects are abrogated in a dose-dependent manner. Implantation of these IPN hydrogels into subcutaneous tissue of rats for 8 weeks led to mild fibrotic encapsulation and minimal inflammatory response. These results suggest the further exploration of extremely tough alginate/PAAM IPN hydrogels as biomaterials.


Assuntos
Resinas Acrílicas/química , Alginatos/química , Hidrogéis/química , Animais , Materiais Biocompatíveis/química , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Ácido Glucurônico/química , Ácidos Hexurônicos/química , Hidrogéis/efeitos adversos , Teste de Materiais , Camundongos , Próteses e Implantes
18.
Biomaterials ; 34(38): 9960-8, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24075478

RESUMO

The transplantation of autologous bone graft as a treatment for large bone defects has the limitation of harvesting co-morbidity and limited availability. This drives the orthopaedic research community to develop bone graft substitutes. Routinely, supra-physiological doses of bone morphogenetic proteins (BMPs) are applied perpetuating concerns over undesired side effects and cost of BMPs. We therefore aimed to design a composite scaffold that allows maintenance of protein bioactivity and enhances growth factor retention at the implantation site. Critical-sized defects in sheep tibiae were treated with the autograft and with two dosages of rhBMP-7, 3.5 mg and 1.75 mg, embedded in a slowly degradable medical grade poly(ε-caprolactone) (PCL) scaffold with ß-tricalcium phosphate microparticles (mPCL-TCP). Specimens were characterised by biomechanical testing, microcomputed tomography and histology. Bridging was observed within 3 months for the autograft and both rhBMP-7 treatments. No significant difference was observed between the low and high rhBMP-7 dosages or between any of the rhBMP-7 groups and autograft implantation. Scaffolds alone did not induce comparable levels of bone formation compared to the autograft and rhBMP-7 groups. In summary, the mPCL-TCP scaffold with the lower rhBMP-7 dose led to equivalent results to autograft transplantation or the high BMP dosage. Our data suggest a promising clinical future for BMP application in scaffold-based bone tissue engineering, lowering and optimising the amount of required BMP.


Assuntos
Poliésteres/química , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Proteínas Morfogenéticas Ósseas/química , Proteínas Morfogenéticas Ósseas/farmacologia , Osteogênese/efeitos dos fármacos , Ovinos , Tíbia/citologia
19.
Adv Drug Deliv Rev ; 64(12): 1257-76, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22626978

RESUMO

Complications in treatment of large bone defects using bone grafting still remain. Our understanding of the endogenous bone regeneration cascade has inspired the exploration of a wide variety of growth factors (GFs) in an effort to mimic the natural signaling that controls bone healing. Biomaterial-based delivery of single exogenous GFs has shown therapeutic efficacy, and this likely relates to its ability to recruit and promote replication of cells involved in tissue development and the healing process. However, as the natural bone healing cascade involves the action of multiple factors, each acting in a specific spatiotemporal pattern, strategies aiming to mimic the critical aspects of this process will likely benefit from the usage of multiple therapeutic agents. This article reviews the current status of approaches to deliver single GFs, as well as ongoing efforts to develop sophisticated delivery platforms to deliver multiple lineage-directing morphogens (multiple GFs) during bone healing.


Assuntos
Regeneração Óssea , Sistemas de Liberação de Medicamentos , Peptídeos e Proteínas de Sinalização Intercelular/administração & dosagem , Animais , Materiais Biocompatíveis/química , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Engenharia Tecidual/métodos
20.
Adv Drug Deliv Rev ; 64(12): 1142-51, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22664228

RESUMO

Bone is one of the few tissues in the human body with high endogenous healing capacity. However, failure of the healing process presents a significant clinical challenge; it is a tremendous burden for the individual and has related health and economic consequences. To overcome such healing deficits, various concepts for a local drug delivery to bone have been developed during the last decades. However, in many cases these concepts do not meet the specific requirements of either surgeons who must use these strategies or individual patients who might benefit from them. We describe currently available methods for local drug delivery and their limitations in therapy. Various solutions for drug delivery to bone focusing on clinical applications and intra-operative constraints are discussed and drug delivery by implant coating is highlighted. Finally, a new set of design and performance requirements for intra-operatively customized implant coatings for controlled drug delivery is proposed. In the future, these requirements may improve approaches for local and intra-operative treatment of patients.


Assuntos
Osso e Ossos/metabolismo , Sistemas de Liberação de Medicamentos , Implantes de Medicamento/administração & dosagem , Animais , Materiais Biocompatíveis/química , Regeneração Óssea , Preparações de Ação Retardada , Desenho de Equipamento , Equipamentos e Provisões , Humanos , Cuidados Intraoperatórios/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA