Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Environ Sci Pollut Res Int ; 30(34): 82199-82216, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37318735

RESUMEN

Activated carbon (AC) is an effective adsorbent for removing environmental pollutants. However, the traditional powder form of AC shows difficulty in handling during application which widely limits its utilization on the industrial scale. Herein, to avoid such limitation, traditional AC powder was encapsulated into calcium alginate (CA) microspheres. Calcium alginate/activated carbon (CAA) composite microspheres were prepared via cross-linking of sodium alginate/activated carbon composite solutions in a calcium chloride solution. Furthermore, in order to boost adsorption affinity of CAA composite microspheres toward elemental mercury (Hg°), ammonium iodide (NH4I)-treated calcium alginate/activated carbon (NCA) composite microspheres were obtained by a simple impregnation method using NH4I treatment. The morphological, structural, and textural properties of the microspheres were characterized and their Hg° adsorptive capacity was tested at different temperatures. Interestingly, the maximum adsorption capacity of NCA adsorbent composite microspheres was determined as 36,056.5 µg/g at a flow rate of 250 mL/min, temperature of 25 °C, and 500 µg/Nm3 of Hg° initial concentration. The Gibbs free energy (ΔG°) for NCA adsorbent composite microspheres varied from - 8.59 to - 10.54 kJ/mol indicating a spontaneous adsorption process with an exothermic nature. The experimental Hg° breakthrough curve correlated well with Yoon‒Nelson and Thomas models. The breakthrough time (tb) and equilibrium time (te) were found to be 7.5 days and 23 days, respectively. Collectively, the findings of this work indicate a good feasibility of using NCA composite microspheres as potential adsorbents for removing Hg° from natural gas.


Asunto(s)
Carbón Orgánico , Mercurio , Carbón Orgánico/química , Gas Natural , Mercurio/química , Alginatos/química , Microesferas , Polvos , Adsorción , Cinética
2.
Colloids Surf B Biointerfaces ; 217: 112650, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35763895

RESUMEN

Calcium phosphate cement (CPC) is a self-setting, biocompatible and osteoconductive bone cement, however its use as a bone substitute is still limited owing to its low bioactivity (i.e. its slow in vivo resorption and slow new bone formation rate) which is a challenging issue to be addressed. Herein, we report for the first time highly bioactive bone cement microspheres formulated from a cement paste containing α-tricalcium phosphate microparticles (α-TCP) and mesoporous calcium silicate bioactive glass nanoparticles (mesoporous BGn) using a water-in-oil emulsion method. Indeed, bioactive microspheres possess high potential as bone defect fillers for bone regeneration. The α-TCP microparticles were prepared by a solid state synthesis at 1400 ºC while mesoporous BGn were synthesized by template-assissted ultrasound-mediated sol-gel method. The particle size distribution of as-prepared cement microspheres was in the range of 200 - 450 µm with a sphericity index in the range of 0.92 - 0.94. The surface morphology of α-TCP microspheres revealed α-TCP micoparticles with smooth surfaces whereas α-TCP/BGn microspheres unveiled nano-roughened α-TCP microparticles. The as-prepared α-TCP/BGn cement microspheres exhibited larger specific surface area ca 18.6 m2/g, sustained release of soluble silicate (SiO44-) ions (118 ppm within a week) and high protein adsorption capacity (252 mg/g). Notably, the α-TCP/BGn cement microspheres showed excellent in vitro surface bioactivity via formation of massive amounts of bone-like hydroxyapatite spherules and aggregates on their surfaces after soaking in simulated body fluid. Importantly, the in vivo implantation of as-prepared α-TCP/BGn cement microspheres in rat calvarial critical size bone defects for 6 weeks unveiled high in vivo bioactivity in terms of substantial new bone ingrowth and significant new bone formation within the bone defect as evidenced by histological analyses, X-ray radiography and micro-computed tomography evaluations.


Asunto(s)
Cementos para Huesos , Nanopartículas , Animales , Materiales Biocompatibles/química , Cementos para Huesos/química , Cementos para Huesos/farmacología , Regeneración Ósea , Fosfatos de Calcio/química , Microesferas , Nanopartículas/química , Ratas , Microtomografía por Rayos X
3.
Transl Oncol ; 20: 101397, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35366536

RESUMEN

The utilization of bioactive glasses (BGs) in cancer therapy has recently become quite promising; herein, a series of Fe-doped mesoporous 45S5-based BGs (MBGs) were synthesized via the sol-gel method in the presence of Pluronic P123 as a soft template. The physico-chemical and biological properties of the prepared glasses were well-characterized through structural assessments, thermal analyses, and electron microscopic studies. Electrochemical analyses, including cyclic voltammetry (CV) and electrochemical impedance spectroscopy (EIS), were also performed to investigate the actual potential of the Fe2O3-containing MBGs in modulating the Fenton's reaction. The XRD results confirmed the glassy state of the Fe-doped samples before immersion in simulated body fluid (SBF). The prepared Fe-doped MBGs exhibited a particle size in the range of 11-86 nm, surface charge of 27-30 mV, SBET of 95-306 m2/g, and Ms of 0.08 to 0.2 emu/g. The incorporation of Fe2O3 led to a negligible decrease in the bioactivity of the glasses. The CV analysis indicated that the Fe-doped MBGs could generate H2O2 in a cathodic potential higher than -0.2 V (vs. Ag/AgCl) in the O2-saturated Na2SO4 solution. Additionally, the data of the EIS test revealed that the Fe2O3-doped MBGs could increase the standard rate constant of Electro-Fenton's (EF) reaction up to 38.44 times as compared with the Fe-free glasses. In conclusion, Fe-doped 45S5-derived glasses may be useful in cancer therapy strategies due to their capability of activating Fenton's reaction and subsequent production of reactive oxygen species (ROS) such as •OH free radicals.

4.
Materials (Basel) ; 14(12)2021 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-34204198

RESUMEN

Exploring new therapies for managing skin wounds is under progress and, in this regard, mesoporous silica nanoparticles (MSNs) and mesoporous bioactive glasses (MBGs) offer great opportunities in treating acute, chronic, and malignant wounds. In general, therapeutic effectiveness of both MSNs and MBGs in different formulations (fine powder, fibers, composites etc.) has been proved over all the four stages of normal wound healing including hemostasis, inflammation, proliferation, and remodeling. The main merits of these porous substances can be summarized as their excellent biocompatibility and the ability of loading and delivering a wide range of both hydrophobic and hydrophilic bioactive molecules and chemicals. In addition, doping with inorganic elements (e.g., Cu, Ga, and Ta) into MSNs and MBGs structure is a feasible and practical approach to prepare customized materials for improved skin regeneration. Nowadays, MSNs and MBGs could be utilized in the concept of targeted therapy of skin malignancies (e.g., melanoma) by grafting of specific ligands. Since potential effects of various parameters including the chemical composition, particle size/morphology, textural properties, and surface chemistry should be comprehensively determined via cellular in vitro and in vivo assays, it seems still too early to draw a conclusion on ultimate efficacy of MSNs and MBGs in skin regeneration. In this regard, there are some concerns over the final fate of MSNs and MBGs in the wound site plus optimal dosages for achieving the best outcomes that deserve careful investigation in the future.

5.
Colloids Surf B Biointerfaces ; 206: 111932, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34175740

RESUMEN

The multifunctional biological properties of Ce ions including antioxidant, anti-inflammatory, antibacterial and anti-cancer effects are very encouraging for development of Ce-containing biomaterials with therapeutic properties. Herein, novel Ce3+/Ce4+ ions containing mesoporous bioactive glass ultrasmall nanoparticles (Ce-BGn) were prepared by a facile one-pot ultrasound-assisted sol-gel method. Interestingly, Ce2O3 incorporation exerted a significant influence on the particle size and textural properties of mesoporous BGn (SiO2 - CaO binary glass system). Ce-BGn exhibited ultrasmall nanoparticle size (< 30 nm), mesoporous texture (pore size up to 2.82 nm and pore volume up to 0.191 cm3/g) and large specific surface area ca. 132.9 m2/g. Notably, in situ formation of CeO2 nanospheres (3-6 nm) was detected at the surface and in the amorphous glass matrix of mesoporous Ce-BGn. Importantly, X-ray photoelectron spectroscopy (XPS) revealed the presence of 72.57 % Ce3+ and 27.43 % Ce4+ at the surface of mesoporous Ce-BGn with Ce3+/Ce4+ ratio = 2.66. Furthermore, mesoporous Ce-BGn exhibited high catalase-mimic activity and showed sustained release of Ce (2.5-32 ppm), Ca (85-327 ppm) and Si (54-200 ppm) ions within 4 weeks along with excellent bone-like hydroxyapatite formation. Finally, the in vitro biological behavior of mesoporous Ce-BGn in cell cultures of human skin fibroblasts (HSF) revealed that mesoporous Ce-BGn (with concentrations up to 300 µg/mL) possess good cyto-biocompatibility. Taken together, novel ultrasmall mesoporous Ce-BGn showed remarkable catalase-mimic activity via surface containing Ce3+/Ce4+ ions which can scavenge ROS (Ce3+↔ Ce4+) and decompose H2O2 molecules into H2O and O2. In addition to that, Ce-BGn demonstrated sustained release of bioactive ions (Ce, Ca and Si), excellent bone-like hydroxyapatite formation and good cyto-biocompatibility.


Asunto(s)
Cerio , Nanopartículas , Antioxidantes/farmacología , Catalasa , Vidrio , Humanos , Peróxido de Hidrógeno , Porosidad , Dióxido de Silicio
6.
Bioact Mater ; 6(1): 123-136, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32817919

RESUMEN

Therapeutic options are quite limited in clinics for the successful repair of infected/degenerated tissues. Although the prevalent treatment is the complete removal of the whole infected tissue, this leads to a loss of tissue function and serious complications. Herein the dental pulp infection, as one of the most common dental problems, was selected as a clinically relevant case to regenerate using a multifunctional nanotherapeutic approach. For this, a mesoporous bioactive glass nano-delivery system incorporating silicate, calcium, and copper as well as loading epidermal growth factor (EGF) was designed to provide antibacterial/pro-angiogenic and osteo/odontogenic multiple therapeutic effects. Amine-functionalized Cu-doped bioactive glass nanospheres (Cu-BGn) were prepared to be 50-60 nm in size, mesoporous, positive-charged and bone-bioactive. The Cu-BGn could release bioactive ions (copper, calcium and silicate ions) with therapeutically-effective doses. The Cu-BGn treatment to human umbilical vein endothelial cells (HUVEC) led to significant enhancement of the migration, tubule formation and expression of angiogenic gene (e.g. vascular endothelial growth factor, VEGF). Furthermore, the EGF-loaded Cu-BGn (EGF@Cu-BGn) showed pro-angiogenic effects with antibacterial activity against E. faecalis, a pathogen commonly involved in the pulp infection. Of note, under the co-culture condition of HUVEC with E. faecalis, the secretion of VEGF was up-regulated. In addition, the osteo/odontogenic stimulation of the EGF@Cu-BGn was evidenced with human dental pulp stem cells. The local administration of the EGF@Cu-BGn in a rat molar tooth defect infected with E. faecalis revealed significant in vivo regenerative capacity, highlighting the nanotherapeutic uses of the multifunctional nanoparticles for regenerating infected/damaged hard tissues.

7.
Mater Sci Eng C Mater Biol Appl ; 110: 110660, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32204088

RESUMEN

Bone-mimetic scaffolds are receiving much interest as such scaffolds exhibit excellent biocompatibility and very close mimic to bone structure and composition. Here, novel bone-mimetic nanohydroxyapatite (nHA)/collagen (Col) porous scaffolds (nHA/Col) were prepared from surface silanized mesoporous nanobioglass (NBG)/Col hybrid scaffold by biomimetic mineralization. Surface silanized mesoporous NBG was prepared by ultrasound-assisted sol-gel method and post treatment with 3-aminopropyltriethylsilane (APTS). The surface silanized mesoporous NBG was characterized by transmission electron microscopy (TEM), transmission electron microscopy-selected area electron diffraction (TEM-SAED) and X-ray photoelectron spectroscopy (XPS). The physicochemical/mechanical characterizations of the scaffolds included scanning electron microscopy (SEM) and TEM imaging of micro/nanostructure, energy dispersive X-ray (EDX) analysis of chemical composition, TEM-SAED and X-ray diffraction/Attenuated total Reflectance-Fourier Infrared spectroscopy (XRD/ATR-FTIR) analyses of amorphous-to-crystalline transformations, thermogravimetric/differential scanning calorimetric (TGA/DSC) analyses of thermal behaviour , porosity and dynamic mechanical analyses. The presence of NBG in collagen fibrillar network enabled progressive growth of HA nanocrystals and generation of a novel bone-mimetic hybrid structures while preserving the highly porous structure of collagen scaffold. The crystallinity, crystallite size and crystal morphology of the grown HA nanocrystals were controllable by regulation of the mineralization time. Furthermore, the osteogenic properties of the non-mineralized (NBG/Col) and mineralized (nHA/Col) hybrid porous scaffolds were examined in vivo using critical-sized calvarial bone defect model in rat. Histological and micro-computed tomography (Micro-CT) analyses after 6 weeks of implantation revealed that the mineralized scaffolds possess excellent in vivo osteogenic potential compared to the non-mineralized one. Collectively, by using surface silanized mesoporous NBG hybridization with collagen fibrillar network, we successfully introduced a new approach for developing novel bone-mimetic nanohydroxyapatite/collagen hybrid scaffolds that possess significant potential for bone tissue regeneration.


Asunto(s)
Materiales Biomiméticos/farmacología , Huesos/efectos de los fármacos , Cerámica/farmacología , Colágeno/farmacología , Durapatita/farmacología , Ensayo de Materiales , Silanos/farmacología , Andamios del Tejido/química , Animales , Rastreo Diferencial de Calorimetría , Colágeno/ultraestructura , Osteogénesis/efectos de los fármacos , Espectroscopía de Fotoelectrones , Porosidad , Ratas , Cráneo/diagnóstico por imagen , Cráneo/efectos de los fármacos , Espectroscopía Infrarroja por Transformada de Fourier , Difracción de Rayos X , Microtomografía por Rayos X
8.
J Tissue Eng ; 11: 2041731419900424, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32076499

RESUMEN

For skeletal muscle engineering, scaffolds that can stimulate myogenic differentiation of cells while possessing suitable mechanical properties (e.g. flexibility) are required. In particular, the elastic property of scaffolds is of importance which helps to resist and support the dynamic conditions of muscle tissue environment. Here, we developed highly flexible nanocomposite nanofibrous scaffolds made of polycarbonate diol and isosorbide-based polyurethane and hydrophilic nano-graphene oxide added at concentrations up to 8%. The nano-graphene oxide incorporation increased the hydrophilicity, elasticity, and stress relaxation capacity of the polyurethane-derived nanofibrous scaffolds. When cultured with C2C12 cells, the polyurethane-nano-graphene oxide nanofibers enhanced the initial adhesion and spreading of cells and further the proliferation. Furthermore, the polyurethane-nano-graphene oxide scaffolds significantly up-regulated the myogenic mRNA levels and myosin heavy chain expression. Of note, the cells on the flexible polyurethane-nano-graphene oxide nanofibrous scaffolds could be mechanically stretched to experience dynamic tensional force. Under the dynamic force condition, the cells expressed significantly higher myogenic differentiation markers at both gene and protein levels and exhibited more aligned myotubular formation. The currently developed polyurethane-nano-graphene oxide nanofibrous scaffolds, due to their nanofibrous morphology and high mechanical flexibility, along with the stimulating capacity for myogenic differentiation, are considered to be a potential matrix for future skeletal muscle engineering.

9.
Acta Biomater ; 99: 181-195, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31446049

RESUMEN

With donor organs not readily available, the need for a tissue-engineered oesophagus remains high, particularly for congenital childhood conditions such as atresia. Previous attempts have not been successful, and challenges remain. Small intestine submucosa (SIS) is an acellular matrix material with good biological properties; however, as is common with these types of materials, they demonstrate poor mechanical properties. In this work, electrospinning was performed to mechanically reinforce tubular SIS with polylactic-co-glycolic acid (PLGA) nanofibres. It was hypothesised that if attachment could be achieved between the two materials, then this would (i) improve the SIS mechanical properties, (ii) facilitate smooth muscle cell alignment to support directional growth of muscle cells and (iii) allow for the delivery of bioactive molecules (VEGF in this instance). Through a relatively simple multistage process, adhesion between the layers was achieved without chemically altering the SIS. It was also found that altering mandrel rotation speed affected the alignment of the PLGA nanofibres. SIS-PLGA scaffolds performed mechanically better than SIS alone; yield stress improvement was 200% and 400% along the longitudinal and circumferential directions, respectively. Smooth muscle cells cultured on the aligned fibres showed resultant unidirectional alignment. In vivo the SIS-PLGA scaffolds demonstrated limited foreign body reaction judged by the type and proportion of immune cells present and lack of fibrous encapsulation. The scaffolds remained intact at 4 weeks in vivo, and good cellular infiltration was observed. The incorporation of VEGF within SIS-PLGA scaffolds increased the blood vessel density of the surrounding tissues, highlighting the possible stimulation of endothelialisation by angiogenic factor delivery. Overall, the designed SIS-PLGA-VEGF hybrid scaffolds might be used as a potential matrix platform for oesophageal tissue engineering. In addition to this, achieving improved attachment between layers of acellular matrix materials and electrospun fibre layers offers the potential utility in other applications. STATEMENT OF SIGNIFICANCE: Because of its multi-layered nature and complex structure, the oesophagus tissue poses several challenges for successful clinical grafting. Therefore, it is promising to utilise tissue engineering strategies to mimic and form structural compartments for its recovery. In this context, we investigated the use of tubular small intestine submucosa (SIS) reinforced with polylactic-co-glycolic acid (PLGA) nanofibres by using electrospinning and also, amongst other parameters, the integrity of the bilayered structure created. This was carried out to facilitate smooth muscle cell alignment, support directional growth of muscle cells and allow the delivery of bioactive molecules (VEGF in this study). We evaluated this approach by using in vitro and in vivo models to determine the efficacy of this new system.


Asunto(s)
Esófago/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Intestino Delgado/efectos de los fármacos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Animales , Materiales Biocompatibles , Adhesión Celular/efectos de los fármacos , Supervivencia Celular , Sistemas de Liberación de Medicamentos , Electroquímica , Humanos , Microscopía Electrónica de Rastreo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Nanofibras/química , Neovascularización Fisiológica , Estrés Mecánico , Porcinos , Resistencia a la Tracción , Factor A de Crecimiento Endotelial Vascular/farmacología
10.
Biomaterials ; 207: 23-38, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30952042

RESUMEN

Inflammation prevailing conditions delay healing processes of damaged tissues, leading to a functional impairment. Although anti-inflammatory drugs are clinically available, they often cause unwanted side effects thus being considered suboptimal. Here we report drug-free synthetic nanoparticles that target and internalize pro-inflammatory cells and release ions, ultimately demonstrating profound anti-inflammatory functions. We introduce folate-functionalized bioactive glass nanoparticle BGN(F) that can bind to pro-inflammatory cells to endocytose and release ions. The folate-conjugation significantly enhanced the nanoparticle internalization to LPS-induced pro-inflammatory cells. The direct treatment of BGN(F) at proper doses (80-160 µg/mL) substantially down-regulated pro-inflammatory molecules, including TNF-α, IL-6, iNOS and COX-2, at both gene and protein levels. The phosphorylation of intracellular signaling molecules involved in the inflammatory events, such as p38 MAPK, ERK (1/2), SAPK/JANK, IκBα, and NF-κB, were significantly suppressed by the BGN(F) treatment. Furthermore, BGN(F) was potential to switch the macrophage polarization from M1 to M2. The released ions, not the physical interactions, of nanoparticles were observed to contribute in major part to the anti-inflammatory actions of BGN(F). The BGN(F), when locally administered to a Notexin-induced myoinjury tissue in mice, significantly down-regulated IL-6 and TNF-α, switched the macrophage phenotype from M1 to M2, and accelerated tissue healing. The current findings that demonstrate profound anti-inflammatory actions of BGN(F) in vitro and in vivo support their uses as novel drug-free nanotherapeutic platform for the treatment of inflamed tissues.


Asunto(s)
Antiinflamatorios/química , Antiinflamatorios/uso terapéutico , Ácido Fólico/química , Inflamación/tratamiento farmacológico , Nanopartículas/química , Animales , Supervivencia Celular/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Receptor 2 de Folato/metabolismo , Inmunohistoquímica , Inflamación/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica de Transmisión , FN-kappa B/metabolismo , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo , Espectroscopía Infrarroja por Transformada de Fourier , Factor de Necrosis Tumoral alfa/metabolismo
11.
ACS Biomater Sci Eng ; 5(11): 6117-6126, 2019 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-33405665

RESUMEN

Introducing new generations of injectable bioactive types of cement that fulfill excellent injectability, rapid self-setting, high bioactivity, proper biodegradability, and fast therapeutic ion-releasing capability is highly demanded for tooth and bone regeneration. Here, we announce therapeutic fast ion-releasing nanobiocements (NBCs) based on sol-gel-processed calcium silicate mesoporous nanobioactive glass with or without strontium (NBC, Sr-free and Sr-NBC, Sr-doped). The stimulating role of Sr ions in odontogenesis of stem cells derived from dental pulp (DPSCs) and in in vivo dentin formation has been investigated. The nanobiocement was formulated through the mixing of bioactive glass nanopowder with a phosphate-buffered saline (P/L = 0.5 g/mL) to form a soft cement paste that hardens within 5-10 min in the ambient environment. The self-setting originated from a setting reaction involving the deposition of hydroxyapatite as evidenced from X-ray diffraction. Both nanobiocements showed the rapid release of therapeutic ions with biologically effective doses, including strontium (Sr), calcium (Ca), or silicon (Si). In vitro cell cultures with DPSCs showed excellent biocompatibility and high odontogenic potential, especially from Sr-NBC. In an in vivo study, Sr-NBC showed more new dentin formation compared to that of NBC, revealed by two different animal models (odontogenesis in subcutaneous and natural tooth environment). Also, NBCs showed high loading capacities of simvastatin used as a model drug. Taken together, Sr-NBC could be considered as a multifunctional nanobiocement with high bioactivity, excellent biodegradability, fast therapeutic ion release, and high drug loading capability, which potentiates its application in dentin-pulp complex regeneration therapy.

12.
Sci Rep ; 8(1): 5663, 2018 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-29618810

RESUMEN

Restoration of hard tissue in conjunction with adhesive is a globally challenging issue in medicine and dentistry. Common clinical therapies involving application of adhesive and substitute material for functional or anatomical recovery are still suboptimal. Biomaterials with bioactivity and inhibitory effects of enzyme-mediated adhesive degradation can render a solution to this. Here, we designed a novel copper-doped bioactive glass nanoparticles (CuBGn) to offer multifunction: metalloproteinases (MMP) deactivation and remineralization and incorporated the CuBGn in resin-dentin adhesive systems, which showed most common failure of MMP mediated adhesive degradation among hard tissue adhesives, to evaluate proposed therapeutic effects. A sol-gel derived bioactive glass nanoparticles doping 10 wt% of Cu (Cu-BGn) for releasing Cu ions, which were well-known MMP deactivator, were successfully created and included in light-curing dental adhesive (DA), a filler-free co-monomer resin blend, at different concentrations (up to 2 wt%). These therapeutic adhesives (CuBGn-DA) showed enhanced (a)cellular bioactivity, cytocompatibility, microtensile bond strength and MMP deactivation-ability. In conclusion, the incorporation of Cu ions releasing nano-bioactive glass demonstrated multifunctional properties at the resin-dentin interface; MMP deactivation and remineralization, representing a suitable strategy to extend the longevity of adhesive-hard tissue (i.e. resin-dentin) interfaces.


Asunto(s)
Adhesivos , Cobre/farmacología , Retención de Dentadura , Vidrio/química , Metaloproteasas/antagonistas & inhibidores , Nanopartículas/química , Remineralización Dental/métodos , Humanos
13.
Acta Biomater ; 69: 218-233, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29410166

RESUMEN

Cell encapsulating hydrogels with tunable mechanical and biological properties are of special importance for cell delivery and tissue engineering. Silk fibroin and collagen, two typical important biological proteins, are considered potential as cell culture hydrogels. However, both have been used individually, with limited properties (e.g., collagen has poor mechanical properties and cell-mediated shrinkage, and silk fibroin from Bombyx mori (mulberry) lacks cell adhesion motifs). Therefore, the combination of them is considered to achieve improved mechanical and biological properties with respect to individual hydrogels. Here, we show that the cell-encapsulating hydrogels of mulberry silk fibroin / collagen are implementable over a wide range of compositions, enabled simply by combining the different gelation mechanisms. Not only the gelation reaction but also the structural characteristics, consequently, the mechanical properties and cellular behaviors are accelerated significantly by the silk fibroin / collagen hybrid hydrogel approach. Of note, the mechanical and biological properties are tunable to represent the combined merits of individual proteins. The shear storage modulus is tailored to range from 0.1 to 20 kPa along the iso-compositional line, which is considered to cover the matrix stiffness of soft-to-hard tissues. In particular, the silk fibroin / collagen hydrogels are highly elastic, exhibiting excellent resistance to permanent deformation under different modes of stress; without being collapsed or water-squeezed out (vs. not possible in individual proteins) - which results from the mechanical synergism of interpenetrating networks of both proteins. Furthermore, the role of collagen protein component in the hybrid hydrogels provides adhesive sites to cells, stimulating anchorage and spreading significantly with respect to mulberry silk fibroin gel, which lacks cell adhesion motifs. The silk fibroin / collagen hydrogels can encapsulate cells while preserving the viability and growth over a long 3D culture period. Our findings demonstrate that the silk / collagen hydrogels possess physical and biological properties tunable and significantly improved (vs. the individual protein gels), implying their potential uses for cell delivery and tissue engineering. STATEMENT OF SIGNIFICANCE: Development of cell encapsulating hydrogels with excellent physical and biological properties is important for the cell delivery and cell-based tissue engineering. Here we communicate for the first time the novel protein composite hydrogels comprised of 'Silk' and 'Collagen' and report their outstanding physical, mechanical and biological properties that are not readily achievable with individual protein hydrogels. The properties include i) gelation accelerated over a wide range of compositions, ii) stiffness levels covering 0.1 kPa to 20 kPa that mimic those of soft-to-hard tissues, iii) excellent elastic behaviors under various stress modes (bending, twisting, stretching, and compression), iv) high resistance to cell-mediated gel contraction, v) rapid anchorage and spreading of cells, and vi) cell encapsulation ability with a long-term survivability. These results come from the synergism of individual proteins of alpha-helix and beta-sheet structured networks. We consider the current elastic cell-encapsulating hydrogels of silk-collagen can be potentially useful for the cell delivery and tissue engineering in a wide spectrum of soft-to-hard tissues.


Asunto(s)
Células Inmovilizadas/metabolismo , Colágeno Tipo I/química , Hidrogeles/química , Células Madre Mesenquimatosas/metabolismo , Secuencias de Aminoácidos , Animales , Bombyx , Adhesión Celular , Células Inmovilizadas/citología , Fibroínas , Humanos , Células Madre Mesenquimatosas/citología , Ratas
14.
ACS Biomater Sci Eng ; 4(3): 1019-1028, 2018 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-33418785

RESUMEN

Healing and repair of damaged bones with various geometries are challenging issues in personalized regenerative medicine. Herein, we examine if the engineered electroblown scaffolds can be suitable to this purpose with the ability to shape and fill defects, populate stem cells, and stimulate the regeneration process of defected bone. The electroblowing method could generate bioactive nanocomposite scaffolds made of poly(caprolactone) and bioactive glass nanoparticles, of which the macrostructure is highly spaced and fibrous networked. The scaffolds were easily formable to different shapes with space filling ability, and were hydrophilic to soak water and blood rapidly. Multipotent stem cells from dental pulp effectively infiltrated the scaffold networks, anchored the fiber surface within few hours, proliferated actively over weeks, and were stimulated to differentiate into osteogenic cells. The cell/scaffold constructs, implanted to tooth-extracted, irregular-shaped alveolar bone defects, were shown to fit the defect and to stimulate early new bone formation. Taken together, the electroblown bioactive fibrous scaffolds and their constructs with multipotent dental stem cells may be useful as a potential 3D platform for future personalized bone tissue engineering.

15.
Dent Mater ; 33(10): e361-e372, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28778494

RESUMEN

OBJECTIVES: Even though polymethyl methacrylate (PMMA) resin is widely used as a dental material, it has poor microbial anti-adhesive properties, which accelerates oral infections. In this investigation, silver-sulfadiazine (AgSD)-loaded mesoporous silica nanoparticles (Ag-MSNs) were incorporated into PMMA to introduce long-term microbial anti-adhesive effects and to make PMMA a rechargeable resin. METHODS: After characterization of the Ag-MSNs in terms of their mesoporous characteristics and drug loading capacity, the 3 point flexural test and hardness were evaluated in PMMA incorporating Ag-MSNs (0.5, 1, 2.5 and 5%). Anti-adhesive effects were observed for Candida albicans and Streptococcus oralis with experimental specimens for up to 28days and after recharging with AgSD. RESULTS: A typical spherical morphology and high mesoporosity were observed for the MSNs used for loading AgSD. Incorporation of Ag-MSNs into PMMA (0.5, 1, 2.5 and 5%) sustained its flexural strength but increased its surface hardness. Anti-adhesive effects were observed after 1h of exposure to both microbial species, and the effects accelerated with increasing Ag-MSN incorporation into PMMA. Long-term microbial anti-adhesive effects were observed for up to 14 days, and further long-term (7 days) anti-adhesive effects were observed after reloading the Ag-MSN-incorporated PMMA (aged for 28 days) with AgSD; these effects were largely caused by released silver ions and partially by changes in surface hydrophilicity. No cytotoxicity to keratinocytes was observed. CONCLUSIONS: The improved mechanical properties and the prolonged microbial anti-adhesive effects, which lasted after reloading of the drug, suggest the potential usefulness of Ag-MSN-incorporated PMMA as a microbial anti-adhesive dental material. SIGNIFICANCE: Ag-MSN-incorporated PMMA can be used as a microbial anti-adhesive dental material for dentures, orthodontic devices and provisional restorative materials.


Asunto(s)
Cementos Dentales , Polimetil Metacrilato , Sulfadiazina de Plata , Ensayo de Materiales , Dióxido de Silicio
16.
Acta Biomater ; 60: 38-49, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28754647

RESUMEN

Here we communicate the generation of biomimetically grown apatite spheres from aggregated bioglass nanoparticles and the potential properties applicable for drug delivery and cell/tissue engineering. Ion releasing nanoparticulates of bioglass (85%SiO2-15%CaO) in a mineralizing medium show an intriguing dynamic phenomenon - aggregation, mineralization to apatite, integration and growth into micron-sized (1.5-3µm) spheres. During the progressive ionic dissolution/precipitation reactions, nano-to-micro-morphology, glass-to-crystal composition, and the physico-chemical properties (porosity, surface area, and charge) change dynamically. With increasing reaction period, the apatite becomes more crystallized with increased crystallinity and crystal size, and gets a composition closer to the stoichiometry. The developed microspheres exhibit hierarchical surface nanostructure, negative charge (ς-potential of -20mV), and ultrahigh mesoporosity (mesopore size of 6.1nm, and the resultant surface area of 63.7m2/g and pore volume of 0.153cm3/g) at 14days of mineralization, which are even higher than those of its precursor bioglass nanoparticles. Thanks to these properties, the biomimetic mineral microspheres take up biological molecules effectively, i.e., loading capacity of positive-charged protein is over 10%. Of note, the release is highly sustainable at a constant rate, i.e., profiling almost 'zero-order' kinetics for 4weeks, suggesting the potential usefulness as protein delivery systems. The biomimetic mineral microspheres hold some remnant Si in the core region, and release calcium, phosphate, and silicate ions over the test period, implying the long-term ionic-related therapeutic functions. The mesenchymal stem cells favour the biomimetic spheres with an excellent viability. Due to the merit of sizes (a few micrometers), the spheres can be intercalated into cells, mediating cellular interactions in 3D cell-spheroid engineering, and also can stimulate osteogenic differentiation of cells when incorporated into cell-laden gels. The intriguing properties observed in this study, including biomimetic composition, high mesoporosity, release of therapeutic ions, effective loading and long-term release of proteins, and diverse yet favorable 3D cellular interactions, suggest great potential of the newly developed biomimetic microspheres in biomedical applications, such as drug delivery and cell/tissue engineering. STATEMENT OF SIGNIFICANCE: This work reports the generation of apatite spheres with a few micrometers in size biomimetically grown from bioactive glass nanoparticles, through a series of intriguing yet unprecedented phenomenon involving aggregation of nanoparticles, mineralization and sphere growth. The mineral microspheres possess some unique physico-chemical properties including mesoporosity, ultrahigh surface area, and therapeutic ionic release. Furthermore, the spheres show excellent loading and delivery capacity of protein molecules, and mediate favorable cellular interactions in 2D and 3D culture conditions, demonstrating a future multifunctional microcarrier platform for the therapeutics delivery and cell/tissue engineering.


Asunto(s)
Apatitas , Materiales Biomiméticos , Sistemas de Liberación de Medicamentos/métodos , Vidrio/química , Células Madre Mesenquimatosas/metabolismo , Nanopartículas/química , Adulto , Animales , Apatitas/química , Apatitas/farmacología , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Células Cultivadas , Femenino , Humanos , Masculino , Ensayo de Materiales/métodos , Células Madre Mesenquimatosas/citología , Porosidad , Ratas , Ratas Sprague-Dawley
17.
Acta Biomater ; 60: 93-108, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28713017

RESUMEN

Inducing differentiation and maturation of resident multipotent stem cells (MSCs) is an important strategy to regenerate hard tissues in mal-calcification conditions. Here we explore a co-delivery approach of therapeutic molecules comprised of ion and drug through a mesoporous bioglass nanoparticle (MBN) for this purpose. Recently, MBN has offered unique potential as a nanocarrier for hard tissues, in terms of high mesoporosity, bone bioactivity (and possibly degradability), tunable delivery of biomolecules, and ionic modification. Herein Sr ion is structurally doped to MBN while drug Phenamil is externally loaded as a small molecule activator of BMP signaling, for the stimulation of osteo/odontogenesis and mineralization of human MSCs derived from dental pulp. The Sr-doped MBN (85Si:10Ca:5Sr) sol-gel processed presents a high mesoporosity with a pore size of ∼6nm. In particular, Sr ion is released slowly at a daily rate of ∼3ppm per mg nanoparticles for up to 7days, a level therapeutically effective for cellular stimulation. The Sr-MBN is internalized to most MSCs via an ATP dependent macropinocytosis within hours, increasing the intracellular levels of Sr, Ca and Si ions. Phenamil is loaded maximally ∼30% into Sr-MBN and then released slowly for up to 7days. The co-delivered molecules (Sr ion and Phenamil drug) have profound effects on the differentiation and maturation of cells, i.e., significantly enhancing expression of osteo/odontogenic genes, alkaline phosphatase activity, and mineralization of cells. Of note, the stimulation is a result of a synergism of Sr and Phenamil, through a Trb3-dependent BMP signaling pathway. This biological synergism is further evidenced in vivo in a mal-calcification condition involving an extracted tooth implantation in dorsal subcutaneous tissues of rats. Six weeks post operation evidences the osseous-dentinal hard tissue formation, which is significantly stimulated by the Sr/Phenamil delivery, based on histomorphometric and micro-computed tomographic analyses. The bioactive nanoparticles releasing both Sr ion and Phenamil drug are considered to be a promising therapeutic nanocarrier platform for hard tissue regeneration. Furthermore, this novel ion/drug co-delivery concept through nanoparticles can be extensively used for other tissues that require different therapeutic treatment. STATEMENT OF SIGNIFICANCE: This study reports a novel design concept in inorganic nanoparticle delivery system for hard tissues - the co-delivery of therapeutic molecules comprised of ion (Sr) and drug (Phenamil) through a unique nanoparticle of mesoporous bioactive glass (MBN). The physico-chemical and biological properties of MBN enabled an effective loading of both therapeutic molecules and a subsequently sustained/controlled release. The co-delivered Sr and Phenamil demonstrated significant stimulation of adult stem cell differentiation in vitro and osseous/dentinal regeneration in vivo, through BMP signaling pathways. We consider the current combination of Sr ion with Phenamil is suited for the osteo/odontogenesis of stem cells for hard tissue regeneration, and further, this ion/drug co-delivery concept can extend the applications to other areas that require specific cellular and tissue functions.


Asunto(s)
Proteínas Morfogenéticas Óseas , Calcificación Fisiológica/efectos de los fármacos , Portadores de Fármacos , Células Madre Mesenquimatosas/metabolismo , Nanopartículas , Transducción de Señal/efectos de los fármacos , Estroncio , Amilorida/análogos & derivados , Amilorida/química , Amilorida/farmacocinética , Amilorida/farmacología , Técnicas de Cultivo de Célula , Células Cultivadas , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/farmacología , Humanos , Células Madre Mesenquimatosas/citología , Nanopartículas/química , Nanopartículas/uso terapéutico , Odontogénesis/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Porosidad , Estroncio/química , Estroncio/farmacocinética , Estroncio/farmacología
18.
Biomaterials ; 142: 62-76, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28727999

RESUMEN

Regeneration of infected tissues is a globally challenging issue in medicine and dentistry. Common clinical therapies involving a complete removal of infected areas together with a treatment of antimicrobial drugs are often suboptimal. Biomaterials with anti-bacterial and pro-regenerative potential can offer a solution to this. Here we design a novel nanocarrier based on a mesoporous silicate-calcium glass by doping with Ag ions and simultaneously loading antimicrobial drugs onto mesopores. The nanocarriers could controllably release multiple ions (silver, calcium, and silicate) and drugs (tetracycline or chlorohexidine) to levels therapeutically relevant, and effectively internalize to human dental stem cells (∼90%) with excellent viability, ultimately stimulating odontogenic differentiation. The release of Ag ions had profound effects on most oral bacteria species through a membrane rupture, and the antibiotic delivery complemented the antibacterial functions by inhibiting protein synthesis. Of note, the nanocarriers easily anchored to bacteria membrane helping the delivery of molecules to an intra-bacterial space. When administered to an infected dentin-pulp defect in rats, the therapeutic nanocarriers effectively regenerated tissues following a complete bacterial killing. This novel concept of multiple-delivering ions and drug can be extensively applied to other infectious tissues that require relayed biological functions (anti-bacterial then pro-regenerative) for successful healing.


Asunto(s)
Pulpa Dental/efectos de los fármacos , Pulpa Dental/microbiología , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Iones/farmacología , Nanopartículas/química , Regeneración/efectos de los fármacos , Células Madre Adultas/citología , Animales , Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Cerámica/química , Pulpa Dental/patología , Humanos , Masculino , Pruebas de Sensibilidad Microbiana , Nanopartículas/ultraestructura , Nanosferas/química , Neovascularización Fisiológica/efectos de los fármacos , Odontogénesis/efectos de los fármacos , Porosidad , Ratas Sprague-Dawley , Plata/farmacología , Tetraciclina/farmacología
19.
J Tissue Eng ; 8: 2041731417707339, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28560015

RESUMEN

Angiogenesis is considered an important issue in the development of biomaterials for the successful regeneration of tissues including bone. While growth factors are commonly used with biomaterials to promote angiogenesis, some ions released from biomaterials can also contribute to angiogenic events. Many silica-based biomaterials have been widely used for the repair and regeneration of tissues, mainly hard tissues such as bone and tooth structure. They have shown excellent performance in bone formation by stimulating angiogenesis. The release of silicate and others (Co and Cu ions) has therefore been implicated to play critical roles in the angiogenesis process. In this short review, we highlight the in vitro and in vivo findings of angiogenesis (and the related bone formation) stimulated by the various types of silicon-containing biomaterials where silicate ions released might play essential roles. We discuss further the possible molecular mechanisms underlying in the ion-induced angiogenic events.

20.
ACS Appl Mater Interfaces ; 9(3): 2059-2073, 2017 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-28029246

RESUMEN

Providing a nanotopological physical cue in concert with a bioactive chemical signal within 3D scaffolds, while it being considered a promising approach for bone regeneration, has yet to be explored. Here, we develop 3D porous scaffolds that are networked to be a nanofibrous structure and incorporated with bioactive glass nanoparticles (BGn) to tackle this issue. The presence of BGn and nanofibrous structure (BGn + nanofibrous) substantially increased the surface area, hydro-affinity and protein loading capacity of scaffolds. In particular, the BGn released Si and Ca ions to the levels known to be biologically effective, offering the bone scaffold an ability to deliver therapeutic ions. The mesenchymal stem cells (MSCs) from rats exhibited significantly accelerated adhesion events including cell anchorage, cytoskeletal extensions, and the expression of adhesion signaling molecules on the BGn/nanofibrous scaffolds. The cells gained a more rapid proliferation and migration (penetration) ability over 2 weeks within the BGn + nanofibrous scaffolds than within either nanofibrous or BGn scaffolds. The osteogenesis of MSCs, as confirmed by the expressions of bone-associated genes and proteins, as well as the cellular mineralization was significantly stimulated by the BGn and nanofibrous topology in a synergistic manner. The behaviors of endothelial cells (HUVECs) including cell migration and tubule networking were also enhanced when influenced by the BGn and nanofibrous scaffolds (but more by BGn than by nanofiber). A subcutaneous tissue implantation of the scaffolds further evidenced the in vivo stimulation of neo-blood vessel formation by the BGn + nanofibrous cues, suggesting the possible promising role in bone regeneration. Taken together, the therapeutic ions and nanofibrous topology implemented within 3D scaffolds are considered to play synergistic actions in osteogenesis and angiogenesis, implying the potential usefulness of the BGn + nanofibrous scaffolds for bone tissue engineering.


Asunto(s)
Nanofibras , Nanopartículas , Animales , Regeneración Ósea , Diferenciación Celular , Células Cultivadas , Señales (Psicología) , Células Madre Mesenquimatosas , Osteogénesis , Ratas , Ingeniería de Tejidos , Andamios del Tejido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...