Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
Acta Biomater ; 94: 574-584, 2019 08.
Article in English | MEDLINE | ID: mdl-31141734

ABSTRACT

The use of bioactive glasses (e.g. silicates, phosphates, borates) has demonstrated to be an effective therapy for the restoration of bone fractures, wound healing and vascularization. Their partial dissolution towards the surrounding tissue has shown to trigger positive bioactive responses, without the necessity of using growth factors or cell therapy, which reduces money-costs, side effects and increases their translation to the clinics. However, bioactive glasses often need from stabilizers (e.g. SiO44-, Ti4+, Co2+, etc.) that are not highly abundant in the body and which metabolization is not fully understood. In this study, we were focused on synthesizing pure calcium phosphate glasses without the presence of such stabilizers. We combined a mixture of ethylphosphate and calcium 2-methoxyethoxide to synthesize nanoparticles with different compositions and degradability. Synthesis was followed by an in-depth nuclear magnetic resonance characterization, complemented with other techniques that helped us to correlate the chemical structure of the glasses with their physiochemical properties and reaction mechanism. After synthesis, the organically modified xerogel (i.e. calcium monoethylphosphate) was treated at 200 or 350 °C and its solubility was maintained and controlled due to the elimination of organics, increase of phosphate-calcium interactions and phosphate polycondensation. To the best of our knowledge, we are reporting the first sol-gel synthesis of binary (P2O5-CaO) calcium phosphate glass nanoparticles in terms of continuous polycondensated phosphate chains structure without the addition of extra ions. The main goal is to straightforward the synthesis, to get a safer metabolization and to modulate the bioactive ion release. Additionally, we shed light on the chemical structure, reaction mechanism and properties of calcium phosphate glasses with high calcium contents, which nowadays are poorly understood. STATEMENT OF SIGNIFICANCE: The use of bioactive inorganic materials (i.e. bioactive ceramics, glass-ceramics and glasses) for biomedical applications is attractive due to their good integration with the host tissue without the necessity of adding exogenous cells or growth factors. In particular, degradable calcium phosphate glasses are completely resorbable, avoiding the retention in the body of the highly stable silica network of silicate glasses, and inducing a more controllable degradability than bioactive ceramics. However, most calcium phosphate glasses include the presence of stabilizers (e.g. Ti4+, Na+, Co2+), which metabolization is not fully understood and complicates their synthesis. The development of binary calcium phosphate glasses with controlled degradability reduces these limitations, offering a simple and completely metabolizable material with higher transfer to the clinics.


Subject(s)
Calcium Compounds/chemistry , Glass/chemistry , Magnetic Resonance Spectroscopy , Nanoparticles/chemistry , Oxides/chemistry , Phosphates/chemistry
2.
Colloids Surf B Biointerfaces ; 177: 121-129, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30716697

ABSTRACT

Limbal epithelial stem cells (LESCs) are responsible for the renewal of corneal epithelium. Cultivated limbal epithelial transplantation is the current treatment of choice for restoring the loss or dysfunction of LESCs. To perform this procedure, a substratum is necessary for in vitro culturing of limbal epithelial cells and their subsequent transplantation onto the ocular surface. In this work, we evaluated poly-L/DL-lactic acid 70:30 (PLA) films functionalized with type IV collagen (col IV) as potential in vitro carrier substrata for LESCs. We first demonstrated that PLA-col IV films were biocompatible and suitable for the proliferation of human corneal epithelial cells. Subsequently, limbal epithelial cell suspensions, isolated from human limbal rings, were cultivated using culture medium that did not contain animal components. The cells adhered significantly faster to PLA-col IV films than to tissue culture plastic (TCP). The mRNA expression levels for the LESC specific markers, K15, P63α and ABCG2 were similar or greater (significantly in the case of K15) in limbal epithelial cells cultured on PLA-col IV films than limbal epithelial cells cultured on TCP. The percentage of cells expressing the corneal (K3, K12) and the LESC (P63α, ABCG2) specific markers was similar for both substrata. These results suggest that the PLA-col IV films promoted LESC attachment and helped to maintain their undifferentiated stem cell phenotype. Consequently, these substrata offer an alternative for the transplantation of limbal cells onto the ocular surface.


Subject(s)
Collagen Type IV/chemistry , Epithelial Cells/cytology , Epithelium, Corneal/cytology , Polyesters/chemistry , Stem Cells/cytology , Cell Proliferation , Cell Survival , Cells, Cultured , Humans
3.
Colloids Surf B Biointerfaces ; 158: 569-577, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28750339

ABSTRACT

Chitosan is a biodegradable natural polysaccharide that has been widely studied for regenerative purposes in the central nervous system. In this study we assessed the in vitro glial and neuronal cells response to chitosan either flat or patterned with grooves in the micrometric range. Chitosan demonstrated to be a good substrate for the attachment and growth of both neurons and glial cells. Chitosan micropatterns promoted glial cell maturation, suggesting astroglial activation. Nevertheless, those mature/reactive glial cells were permissive for axonal growth. Axons aligned and organized along the patterned grooves and the size of the linear topographic patterns is also affecting neurite and cell response. Patterns with 10µm width induced fasciculation of axons, which can be useful for CNS tissue engineering substrates when precise orientation of the axonal outgrowth is desired.


Subject(s)
Chitosan/chemistry , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism , Animals , Axons/metabolism , Brain/cytology , Brain/metabolism , Central Nervous System/cytology , Central Nervous System/metabolism , Humans
4.
Int J Nanomedicine ; 12: 4901-4919, 2017.
Article in English | MEDLINE | ID: mdl-28744124

ABSTRACT

The success of scaffold implantation in acellular tissue engineering approaches relies on the ability of the material to interact properly with the biological environment. This behavior mainly depends on the design of the graft surface and, more precisely, on its capacity to biodegrade in a well-defined manner (nature of ions released, surface-to-volume ratio, dissolution profile of this release, rate of material resorption, and preservation of mechanical properties). The assessment of the biological behavior of temporary templates is therefore very important in tissue engineering, especially for composites, which usually exhibit complicated degradation behavior. Here, blended polylactic acid (PLA) calcium phosphate ORMOGLASS (organically modified glass) nanofibrous mats have been incubated up to 4 weeks in physiological simulated conditions, and their morphological, topographical, and chemical changes have been investigated. The results showed that a significant loss of inorganic phase occurred at the beginning of the immersion and the ORMOGLASS maintained a stable composition afterward throughout the degradation period. As a whole, the nanostructured scaffolds underwent fast and heterogeneous degradation. This study reveals that an angiogenic calcium-rich environment can be achieved through fast-degrading ORMOGLASS/PLA blended fibers, which seems to be an excellent alternative for guided bone regeneration.


Subject(s)
Biocompatible Materials/chemistry , Nanostructures/chemistry , Tissue Scaffolds/chemistry , Calcium/chemistry , Calcium Phosphates/chemistry , Glass/chemistry , Lactic Acid/chemistry , Polyesters/chemistry
5.
Acta Biomater ; 54: 377-385, 2017 05.
Article in English | MEDLINE | ID: mdl-28242456

ABSTRACT

Insufficient angiogenesis remains a major hurdle in current bone tissue engineering strategies. An extensive body of work has focused on the use of angiogenic factors or endothelial progenitor cells. However, these approaches are inherently complex, in terms of regulatory and methodologic implementation, and present a high cost. We have recently demonstrate the potential of electrospun poly(lactic acid) (PLA) fiber-based membranes, containing calcium phosphate (CaP) ormoglass particles, to elicit angiogenesis in vivo, in a subcutaneous model in mice. Here we have devised an injectable composite, containing CaP glass-ceramic particles, dispersed within a (Hydroxypropyl)methyl cellulose (HPMC) matrix, with the capacity to release calcium in a more sustained fashion. We show that by tuning the release of calcium in vivo, in a rat bone defect model, we could improve both bone formation and increase angiogenesis. The bone regeneration kinetics was dependent on the Ca2+ release rate, with the faster Ca2+ release composite gel showing improved bone repair at 3weeks, in relation to control. In the same line, improved angiogenesis could be observed for the same gel formulation at 6weeks post implantation. This methodology allows to integrate two fundamental processes for bone tissue regeneration while using a simple, cost effective, and safe approach. STATEMENT OF SIGNIFICANCE: In current bone tissue engineering approaches the achievement of sufficient angiogenesis, during tissue regeneration, is a major limitation in order to attain full tissue functionality. Recently, we have shown that calcium ions, released by the degradation of calcium phosphate ormoglasses (CaP), are effective angiogenic promoters, in both in vitro and in a subcutaneous implantation model. Here, we devised an injectable composite, containing CaP glass-ceramic particles, dispersed within a HPMC matrix, enabling the release of calcium in a more sustained fashion. We show that by tuning the release of calcium in vivo, in a rat bone defect model, we could improve both bone formation and increase angiogenesis. This simple and cost effective approach holds great promise to translate to the clinics.


Subject(s)
Drug Evaluation, Preclinical , Endothelial Progenitor Cells , Neovascularization, Physiologic/drug effects , Osteogenesis/drug effects , Animals , Calcium/chemistry , Calcium/pharmacology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacology , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/transplantation , Heterografts , Humans , Mice , Polyesters/chemistry , Polyesters/pharmacology , Rats , Rats, Wistar
6.
BMC Anesthesiol ; 16: 78, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27619387

ABSTRACT

BACKGROUND: The use of elastomeric devices for ambulatory intravenous pain treatment in Major Ambulatory Surgery (MAS) has been described to improve postoperative pain management. The objective of the study was to describe the first 3 years experience of the use of elastomeric devices for ambulatory intravenous pain treatment in MAS implemented at our site since 2010. METHODS: Data were retrieved from the medical records for all patients who, between January 2010 and March 2014, underwent surgical procedures at the ambulatory surgical centre at our hospital and were prescribed a home-based continuous intravenous analgesia. RESULTS: Data were retrieved from the medical records of 1128 patients. The most frequent surgical interventions included orthopedic and proctology surgeries. 80 % of patients were discharged home without pain; during the first 48 h after discharge roughly 40 % of subjects were completely free of pain, 50 % reported mild pain (VAS 1 to 3) and 9 % reported higher pain scores (4 and above). Peripheral nerve block was associated to better pain control in the immediate postoperative period. Vomiting in the first 24 h was 4.6 % before introducing haloperidol into the drug schemes, and 2.6 % thereafter. Complications related with the intravenous route required treatment withdrawal in 1.1 % cases. Only 3.5 % of patients returned to the hospital in the first 72 h, mainly for non-pain related reasons. Overall, 99.5 % of patients were satisfied with the treatment received at home. CONCLUSION: Our initial experience suggest that outpatient multimodal intravenous analgesia in patients undergoing day-case surgery is a feasible alternative in our setting, that allows an effective management of postoperative pain with a small rate of adverse events and complications requiring readmission.


Subject(s)
Ambulatory Surgical Procedures/methods , Analgesics/administration & dosage , Analgesics/therapeutic use , Pain, Postoperative/drug therapy , Administration, Intravenous , Adolescent , Adult , Aged , Aged, 80 and over , Analgesics/adverse effects , Elastomers/administration & dosage , Female , Humans , Infusion Pumps , Male , Middle Aged , Outpatients , Pain Management/methods , Patient Satisfaction , Retrospective Studies , Young Adult
7.
Acta Biomater ; 45: 349-356, 2016 11.
Article in English | MEDLINE | ID: mdl-27639311

ABSTRACT

The current study reports on the manufacturing by rapid prototyping technique of three-dimensional (3D) scaffolds based on silicon substituted hydroxyapatite with Elastin-like Recombinamers (ELRs) functionalized surfaces. Silicon doped hydroxyapatite (Si-HA), with Ca10(PO4)5.7(SiO4)0.3(OH)1.7h0.3 nominal formula, was surface functionalized with two different types of polymers designed by genetic engineering: ELR-RGD that contain cell attachment specific sequences and ELR-SNA15/RGD with both hydroxyapatite and cells domains that interact with the inorganic phase and with the cells, respectively. These hybrid materials were subjected to in vitro assays in order to clarify if the ELRs coating improved the well-known biocompatible and bone regeneration properties of calcium phosphates materials. The in vitro tests showed that there was a total and homogeneous colonization of the 3D scaffolds by Bone marrow Mesenchymal Stromal Cells (BMSCs). In addition, the BMSCs were viable and able to proliferate and differentiate into osteoblasts. STATEMENT OF SIGNIFICANCE: Bone tissue engineering is an area of increasing interest because its main applications are directly related to the rising life expectancy of the population, which promotes higher rates of several bone pathologies, so innovative strategies are needed for bone tissue regeneration therapies. Here we use the rapid prototyping technology to allow moulding ceramic 3D scaffolds and we use different bio-polymers for the functionalization of their surfaces in order to enhance the biological response. Combining the ceramic material (silicon doped hydroxyapatite, Si-HA) and the Elastin like Recombinamers (ELRs) polymers with the presence of the integrin-mediate adhesion domain alone or in combination with SNA15 peptide that possess high affinity for hydroxyapatite, provided an improved Bone marrow Mesenchymal Stromal Cells (BMSCs) differentiation into osteoblastic linkage.


Subject(s)
Bone and Bones/physiology , Durapatite/pharmacology , Elastin/pharmacology , Regenerative Medicine/methods , Silicon/pharmacology , Tissue Scaffolds/chemistry , Alkaline Phosphatase/metabolism , Amino Acid Sequence , Animals , Bone and Bones/drug effects , Cell Differentiation , Cell Proliferation/drug effects , Cell Survival/drug effects , Elastin/chemistry , Electrophoresis, Polyacrylamide Gel , Green Fluorescent Proteins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Osteocalcin/metabolism , Rats , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transfection
8.
Acta Biomater ; 29: 435-445, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26441126

ABSTRACT

In current bone tissue engineering strategies the achievement of sufficient angiogenesis during tissue regeneration is still a major limitation in order to attain full functionality. Several strategies have been described to tackle this problem, mainly by the use of angiogenic factors or endothelial progenitor cells. However, when facing a clinical scenario these approaches are inherently complex and present a high cost. As such, more cost effective alternatives are awaited. Here, we demonstrate the potential of electrospun poly(lactic acid) (PLA) fiber-based membranes, containing calcium phosphate ormoglass (CaP) particles, to elicit angiogenesis in vivo, in a subcutaneous model in mice. We show that the current approach elicited the local expression of angiogenic factors, associated to a chemotactic effect on macrophages, and sustained angiogenesis into the biomaterial. As both PLA and CaP are currently accepted for clinical application these off-the-shelf novel membranes have great potential for guided bone regeneration applications. STATEMENT OF SIGNIFICANCE: In current bone tissue engineering approaches the achievement of sufficient angiogenesis, during tissue regeneration, is a major limitation in order to attain full tissue functionality. Recently, our group has found that calcium ions released by the degradation of calcium phosphate ormoglasses (CaP) are effective angiogenic promoters. Based on this, in this work we successfully produced hybrid fibrous mats with different contents of CaP nanoparticles and thus with different calcium ion release rates, using an ormoglass - poly(lactic acid) blend approach. We show that these matrices, upon implantation in a subcutaneous site, could elicit the local expression of angiogenic factors, associated to a chemotactic effect on macrophages, and sustained angiogenesis into the biomaterial, in a CaP dose dependent manner. This off-the-shelf cost effective approach presents great potential to translate to the clinics.


Subject(s)
Calcium Phosphates , Calcium , Lactic Acid , Membranes, Artificial , Neovascularization, Physiologic/drug effects , Polymers , Adult , Animals , Calcium/chemistry , Calcium/pharmacokinetics , Calcium/pharmacology , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacokinetics , Calcium Phosphates/pharmacology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Drug Evaluation, Preclinical , Female , Humans , Lactic Acid/chemistry , Lactic Acid/pharmacokinetics , Lactic Acid/pharmacology , Male , Mice , Polyesters , Polymers/chemistry , Polymers/pharmacokinetics , Polymers/pharmacology
9.
Biomaterials ; 68: 42-53, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26264645

ABSTRACT

Bone tissue engineering demands alternatives overcoming the limitations of traditional approaches in the context of a constantly aging global population. In the present study, elastin-like recombinamers hydrogels were produced by means of carbodiimide-catalyzed crosslinking with citric acid, a molecule suggested to be essential for bone nanostructure. By systematically studying the effect of the relative abundance of reactive species on gelation and hydrogel properties such as functional groups content, degradation and structure, we were able to understand and to control the crosslinking reaction to achieve hydrogels mimicking the fibrillary nature of the extracellular matrix. By studying the effect of polymer concentration on scaffold mechanical properties, we were able to produce hydrogels with a stiffness value of 36.13 ± 10.72 kPa, previously suggested to be osteoinductive. Microstructured and mechanically-tailored hydrogels supported the growth of human mesenchymal stem cells and led to higher osteopontin expression in comparison to their non-tailored counterparts. Additionally, tailored hydrogels were able to rapidly self-mineralize in biomimetic conditions, evidencing that citric acid was successfully used both as a crosslinker and a bioactive molecule providing polymers with calcium phosphate nucleation capacity.


Subject(s)
Bone Regeneration/physiology , Citric Acid/pharmacokinetics , Hydrogels/chemical synthesis , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Tissue Scaffolds , Animals , Biomimetic Materials/chemical synthesis , Bone Substitutes/chemical synthesis , Calcification, Physiologic/physiology , Cell Differentiation/physiology , Cell Proliferation/physiology , Cells, Cultured , Cross-Linking Reagents/chemistry , Extracellular Matrix/chemistry , Humans , Materials Testing , Mesenchymal Stem Cells/physiology , Nanoparticles , Osteoblasts/physiology , Osteogenesis/physiology , Rats
10.
Acta Biomater ; 18: 59-67, 2015 May.
Article in English | MEDLINE | ID: mdl-25702533

ABSTRACT

Many cell therapies rely on the ability of mesenchymal stromal cells (MSCs) to diffuse and localize throughout the target tissue - such as tumoral and ischemic tissues-, in response to specific cytokine signals, rather than being concentrated at the site of implantation. Therefore, it is fundamental to engineer biomaterial carriers as reservoirs, from which cells can migrate, possibly in a controlled manner. In this work, microcarriers (µCs) made of polylactic acid are characterized as MSC delivery vehicles capable of modulating key chemotactic pathways. The effect of different functionalization strategies on MSC migratory behavior from the µCs is studied in vitro in relation to SDF-1α/CXCR4 axis, - a major actor in MSC recruitment, chemotaxis and homing. Collagen and arginine-glycine-aspartic acid (RGD) peptides were either covalently grafted or physisorbed on µC surface. While stable covalent modifications promoted better cell adhesion and higher proliferation compared to physisorption, the functionalization method of the µCs also affected the cells migratory behavior in response to SDF-1α (CXCL12) stimulation. Less stable coatings (physisorbed) showed sensibly higher number of migrating cells than covalent collagen/RGD coatings. The combination of physic-chemical cues provided by protein/peptide functionalization and stimuli induced by 3D culture on µCs improved MSC expression of CXCR4, and exerted a control over cell migration, a condition suitable to promote cell homing after transplantation in vivo. These are key findings to highlight the impact of surface modification approaches on chemokine-triggered cell release, and allow designing biomaterials for efficient and controlled cell delivery to damaged tissues.


Subject(s)
Cell Movement/drug effects , Coated Materials, Biocompatible/pharmacology , Extracellular Matrix/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Microspheres , Peptides/pharmacology , Animals , Cell Adhesion/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Chemokine CXCL12/pharmacology , Extracellular Matrix/drug effects , Flow Cytometry , Fluorescent Antibody Technique , Rats, Inbred Lew , Receptors, CXCR4/metabolism
11.
ACS Biomater Sci Eng ; 1(10): 927-934, 2015 Oct 12.
Article in English | MEDLINE | ID: mdl-33429524

ABSTRACT

Considering that vitronectin (VN) can promote both cell adhesion and matrix degradation, it is likely to play a dual role at the cell-biomaterial interface. In this paper we therefore describe details of the dynamic interplay between matrix adhesion and pericellular proteolysis in endothelial cells adhered to glass model substratum. Initially we show that coating concentration determines protein organization at the surface. When the protein coating density approached saturation (63 ng cm-2), VN spontaneously organized itself in multimeric aggregates at the surface (30-50 nm in diameter). At subsaturation protein density (17 ng cm-2) VN molecules were present predominantly as single entities, indicating that a minimum coating density was required for VN multimerization. By fluorescent visualization of surface-associated VN in different ways, we provide the first evidence of significant proteolytic remodelling of VN by endothelial cells (HUVECs) at the sites of αv integrin clusters. The degree of proteolysis was estimated using a novel approach relying on dequenching of FITC-labeled VN upon proteolytic activity, showing that about one-third of the surface-associated VN was proteolytically altered by adhering HUVECs. In addition, we demonstrate that HUVECs can internalize surface-associated VN and deposit it in a linear pattern along longitudinal actin filaments. Deposited VN was partly colocalized with urokinase receptors. Taken altogether, we elucidate the complex and dynamic behavior of VN during initial cell-biomaterials interactions, the equilibrium if which could have a significant impact on the biocompatibility of any blood contacting implants.

12.
Acta Biomater ; 12: 242-249, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25448348

ABSTRACT

Major limitations of calcium phosphate cements (CPCs) are their relatively slow degradation rate and the lack of macropores allowing the ingrowth of bone tissue. The development of self-setting cement foams has been proposed as a suitable strategy to overcome these limitations. In previous work we developed a gelatine-based hydroxyapatite foam (G-foam), which exhibited good injectability and cohesion, interconnected porosity and good biocompatibility in vitro. In the present study we evaluated the in vivo performance of the G-foam. Furthermore, we investigated whether enrichment of the foam with soybean extract (SG-foam) increased its bioactivity. G-foam, SG-foam and non-foamed CPC were implanted in a critical-size bone defect in the distal femoral condyle of New Zealand white rabbits. Bone formation and degradation of the materials were investigated after 4, 12 and 20weeks using histological and biomechanical methods. The foams maintained their macroporosity after injection and setting in vivo. Compared to non-foamed CPC, cellular degradation of the foams was considerably increased and accompanied by new bone formation. The additional functionalization with soybean extract in the SG-foam slightly reduced the degradation rate and positively influenced bone formation in the defect. Furthermore, both foams exhibited excellent biocompatibility, implying that these novel materials may be promising for clinical application in non-loaded bone defects.


Subject(s)
Biocompatible Materials , Durapatite/chemistry , Gelatin/chemistry , Glycine max/chemistry , Animals , Biomechanical Phenomena , Female , Rabbits , X-Ray Diffraction
13.
Biotechnol Lett ; 37(4): 935-42, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25502922

ABSTRACT

Bioactive nanocomposite scaffolds with cell-adhesive surface have excellent bone regeneration capacities. Fibronectin (FN)-immobilized nanobioactive glass (nBG)/polycaprolactone (PCL) (FN-nBG/PCL) scaffolds with an open pore architecture were generated by a robotic-dispensing technique. The surface immobilization level of FN was significantly higher on the nBG/PCL scaffolds than on the PCL scaffolds, mainly due to the incorporated nBG that provided hydrophilic chemical-linking sites. FN-nBG/PCL scaffolds significantly improved cell responses, including initial anchorage and subsequent cell proliferation. Although further in-depth studies on cell differentiation and the in vivo animal responses are required, bioactive nanocomposite scaffolds with cell-favoring surface are considered to provide promising three-dimensional substrate for bone regeneration.


Subject(s)
Cell Adhesion , Fibronectins/metabolism , Osteocytes/physiology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Immobilized Proteins/metabolism , Protein Binding , Rats, Sprague-Dawley
14.
J Biomed Mater Res B Appl Biomater ; 103(6): 1287-93, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25355602

ABSTRACT

Electrospinning is a method that can be used to efficiently produce scaffolds that mimic the fibrous structure of natural tissue, such as muscle structures or the extracellular matrix of bone. The technique is often used as a way of depositing composites (organic/inorganic materials) to obtain bioactive nanofibers which have the requisite mechanical properties for use in tissue engineering. However, many factors can influence the formation and collection of fibers, including experimental variables such as the parameters of the solution of the electrospun slurry. In this study, we assessed the influence of the polymer concentration, glass content and glass hydrolysis level on the morphology and thickness of fibers produced by electrospinning for a PCL-(Si-Ca-P2 ) bioactive ormoglass-organically modified glass-blend. Based on previous assays, this combination of materials shows good angiogenic and osteogenic properties, which gives it great potential for use in tissue engineering. The results of our study showed that blend preparation directly affected the features of the resulting fibers, and when the parameters of the blend are precisely controlled, fibers with a regular diameter could be produced fairly easily when 2,2,2-trifluoroethanol was used as a solvent instead of tetrahydrofuran. The diameter of the homogeneous fibers ranged from 360 to 620 nm depending on the experimental conditions used. This demonstrates that experimental optimization of the electrospinning process is crucial in order to obtain a deposit of hybrid nanofibers with a regular shape.


Subject(s)
Nanofibers/chemistry , Polyesters/chemistry , Silicon Compounds/chemistry
15.
Mater Sci Eng C Mater Biol Appl ; 44: 183-90, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25280695

ABSTRACT

In this study gelatin (Gel) modified with calcium phosphate nanoparticles (SG5) and polycaprolactone (PCL) were used to prepare a 3D bi-layer scaffold by collecting electrospun PCL and gelatin/SG5 fibers separately in the same collector. The objective of this study was to combine the desired properties of PCL and Gel/SG5 in the same scaffold in order to enhance mineralization, thus improving the ability of the scaffold to bond to the bone tissue. The scanning electron microscopy (SEM), Fourier transform infrared spectroscopy (FTIR) and the wide angle X-ray diffraction (WAXD) measurements confirmed that SG5 nanoparticles were successfully incorporated into the fibrous gelatin matrix. The composite Gel/SG5/PCL scaffold exhibited more enhanced mechanical properties than individual Gel and Gel/SG5 scaffolds. The presence of SG5 nanoparticles accelerated the nucleation and growth of apatite crystals on the surface of the composite Gel/SG5/PCL scaffold in simulated body fluid (SBF). The osteoblast response in vitro to developed electrospun scaffolds (PCL and Gel/SG5/PCL) was investigated by using normal human primary NHOst cell lines. NHOst cell culture studies showed that higher alkaline phosphatase (ALP) activity and better mineralization were obtained in the case of composite materials than in pure PCL scaffolds. The mechanically strong PCL scaffold served as a skeleton, while the Gel/SG5 fibers facilitated cell spreading and mineralization of the scaffold.


Subject(s)
Calcium Phosphates/chemistry , Gelatin/chemistry , Polyesters/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Bone and Bones/drug effects , Bone and Bones/metabolism , Humans , Microscopy, Electron, Scanning , Osteoblasts/cytology , Osteoblasts/drug effects , Spectroscopy, Fourier Transform Infrared , X-Ray Diffraction
16.
Biofabrication ; 6(3): 035020, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25048797

ABSTRACT

Bioprinting allows the fabrication of living constructs with custom-made architectures by spatially controlled deposition of multiple bioinks. This is important for the generation of tissue, such as osteochondral tissue, which displays a zonal composition in the cartilage domain supported by the underlying subchondral bone. Challenges in fabricating functional grafts of clinically relevant size include the incorporation of cues to guide specific cell differentiation and the generation of sufficient cells, which is hard to obtain with conventional cell culture techniques. A novel strategy to address these demands is to combine bioprinting with microcarrier technology. This technology allows for the extensive expansion of cells, while they form multi-cellular aggregates, and their phenotype can be controlled. In this work, living constructs were fabricated via bioprinting of cell-laden microcarriers. Mesenchymal stromal cell (MSC)-laden polylactic acid microcarriers, obtained via static culture or spinner flask expansion, were encapsulated in gelatin methacrylamide-gellan gum bioinks, and the printability of the composite material was studied. This bioprinting approach allowed for the fabrication of constructs with high cell concentration and viability. Microcarrier encapsulation improved the compressive modulus of the hydrogel constructs, facilitated cell adhesion, and supported osteogenic differentiation and bone matrix deposition by MSCs. Bilayered osteochondral models were fabricated using microcarrier-laden bioink for the bone compartment. These findings underscore the potential of this new microcarrier-based biofabrication approach for bone and osteochondral constructs.


Subject(s)
Bioprinting/methods , Lactic Acid/chemistry , Mesenchymal Stem Cells/cytology , Polymers/chemistry , Tissue Engineering/instrumentation , Tissue Scaffolds/chemistry , Animals , Cell Survival , Lactic Acid/chemical synthesis , Osteogenesis , Polyesters , Polymers/chemical synthesis , Rats , Rats, Inbred Lew
17.
ACS Appl Mater Interfaces ; 6(10): 7512-22, 2014 May 28.
Article in English | MEDLINE | ID: mdl-24754868

ABSTRACT

In bone regeneration, silicon-based calcium phosphate glasses (Bioglasses) have been widely used since the 1970s. However, they dissolve very slowly because of their high amount of Si (SiO2 > 45%). Recently, our group has found that calcium ions released by the degradation of glasses in which the job of silicon is done by just 5% of TiO2 are effective angiogenic promoters, because of their stimulation of a cell-membrane calcium sensing receptor (CaSR). Based on this, other focused tests on angiogenesis have found that Bioglasses also have the potential to be angiogenic promoters even with high contents of silicon (80%); however, their slow degradation is still a problem, as the levels of silicon cannot be decreased any lower than 45%. In this work, we propose a new generation of hybrid organically modified glasses, ormoglasses, that enable the levels of silicon to be reduced, therefore speeding up the degradation process. Using electrospinning as a faithful way to mimic the extracellular matrix (ECM), we successfully produced hybrid fibrous mats with three different contents of Si (40, 52, and 70%), and thus three different calcium ion release rates, using an ormoglass-polycaprolactone blend approach. These mats offered a good platform to evaluate different calcium release rates as osteogenic promoters in an in vivo subcutaneous environment. Complementary data were collected to complement Ca(2+) release analysis, such as stiffness evaluation by AFM, ζ-potential, morphology evaluation by FESEM, proliferation and differentiation analysis, as well as in vivo subcutaneous implantations. Material and biological characterization suggested that compositions of organic/inorganic hybrid materials with a Si content equivalent to 40%, which were also those that released more calcium, were osteogenic. They also showed a greater ability to form blood vessels. These results suggest that Si-based ormoglasses can be considered an efficient tool for calcium release modulation, which could play a key role in the angiogenic promoting process.


Subject(s)
Biocompatible Materials/chemistry , Calcium/metabolism , Glass/chemistry , Polyesters/chemistry , Silicon/chemistry , Animals , Biocompatible Materials/pharmacology , Bone Regeneration/drug effects , Calcium/chemistry , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Hydrogen-Ion Concentration , Ions/chemistry , Mice , Microscopy, Atomic Force , Neovascularization, Physiologic/drug effects , Prostheses and Implants , Rats , Receptors, Calcium-Sensing/metabolism , Skin/pathology , Surface Properties , Thermogravimetry
18.
Biomaterials ; 35(17): 4769-81, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24636215

ABSTRACT

Regenerative medicine strategies to promote recovery following traumatic brain injuries are currently focused on the use of biomaterials as delivery systems for cells or bioactive molecules. This study shows that cell-free biomimetic scaffolds consisting of radially aligned electrospun poly-l/dl lactic acid (PLA70/30) nanofibers release L-lactate and reproduce the 3D organization and supportive function of radial glia embryonic neural stem cells. The topology of PLA nanofibers supports neuronal migration while L-lactate released during PLA degradation acts as an alternative fuel for neurons and is required for progenitor maintenance. Radial scaffolds implanted into cavities made in the postnatal mouse brain fostered complete implant vascularization, sustained neurogenesis, and allowed the long-term survival and integration of the newly generated neurons. Our results suggest that the endogenous central nervous system is capable of regeneration through the in vivo dedifferentiation induced by biophysical and metabolic cues, with no need for exogenous cells, growth factors, or genetic manipulation.


Subject(s)
Brain/blood supply , Brain/physiology , Lactic Acid/administration & dosage , Nanofibers/chemistry , Neural Stem Cells/transplantation , Neurogenesis , Tissue Scaffolds/chemistry , Animals , Biomimetic Materials/chemistry , Brain/pathology , Cells, Cultured , Drug Delivery Systems , Lactic Acid/chemistry , Mice , Nanofibers/ultrastructure , Neovascularization, Physiologic , Neural Stem Cells/cytology , Polyesters , Polymers/chemistry , Regeneration
19.
Mater Sci Eng C Mater Biol Appl ; 38: 55-62, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24656352

ABSTRACT

Achieving high quality 3D-printed structures requires establishing the right printing conditions. Finding processing conditions that satisfy both the fabrication process and the final required scaffold properties is crucial. This work stresses the importance of studying the outcome of the plasticizing effect of PEG on PLA-based blends used for the fabrication of 3D-direct-printed scaffolds for tissue engineering applications. For this, PLA/PEG blends with 5, 10 and 20% (w/w) of PEG and PLA/PEG/bioactive CaP glass composites were processed in the form of 3D rapid prototyping scaffolds. Surface analysis and differential scanning calorimetry revealed a rearrangement of polymer chains and a topography, wettability and elastic modulus increase of the studied surfaces as PEG was incorporated. Moreover, addition of 10 and 20% PEG led to non-uniform 3D structures with lower mechanical properties. In vitro degradation studies showed that the inclusion of PEG significantly accelerated the degradation rate of the material. Results indicated that the presence of PEG not only improves PLA processing but also leads to relevant surface, geometrical and structural changes including modulation of the degradation rate of PLA-based 3D printed scaffolds.


Subject(s)
Lactic Acid/chemistry , Polyethylene Glycols/chemistry , Polymers/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Interferometry , Mechanical Phenomena , Microscopy, Atomic Force , Microscopy, Electron, Scanning , Polyesters , Porosity , Temperature
20.
J Mater Sci Mater Med ; 25(7): 1781-7, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24668270

ABSTRACT

Surface biofunctionalisation of many biodegradable polymers is one of the used strategies to improve the biological activity of such materials. In this work, the introduction of collagen type I over the surface of a biodegradable polymer (poly lactic acid) processed in the forms of films and fibers leads to an enhancing of the cellular adhesion of human dermal fibroblast when compared to unmodified polymer and biomolecule-physisorbed polymer surface. The change of topography of the material does not affect the cellular adhesion but results in a higher proliferation of the fibroblast cultured over the fibers. Moreover, the difference of topography governs the cellular morphology, i.e. cells adopt a more stretched conformation where cultured over the films while a more elongated with lower area morphology are obtained for the cells grown over the fibers. This study is relevant for designing and modifying different biodegradable polymers for their use as scaffolds for different applications in the field of Tissue Engineering and Regenerative Medicine.


Subject(s)
Biocompatible Materials/chemistry , Collagen Type I/chemistry , Fibroblasts/cytology , Animals , Cattle , Cell Adhesion , Cell Proliferation , Collagen/chemistry , Fibroblasts/metabolism , Humans , Lactic Acid/chemistry , Microscopy, Fluorescence , Polyesters , Polymers/chemistry , Recombinant Proteins/chemistry , Skin/metabolism , Surface Properties , Tissue Engineering/methods , Viscosity
SELECTION OF CITATIONS
SEARCH DETAIL
...