Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Tissue Eng Regen Med ; 10(5): E275-85, 2016 05.
Article in English | MEDLINE | ID: mdl-23723146

ABSTRACT

Controlled delivery of growth factors from biodegradable biomatrices could accelerate and improve impaired wound healing. The study aim was to determine whether platelet-derived growth factor AB (PDGF.AB) with a transglutaminase (TG) crosslinking substrate site released from a fibrin biomatrix improves wound healing in severe thermal injury. The binding and release kinetics of TG-PDGF.AB were determined in vitro. Third-degree contact burns (dorsum of Yorkshire pigs) underwent epifascial necrosectomy 24 h post-burn. Wound sites were covered with autologous meshed (3:1) split-thickness skin autografts and either secured with staples or attached with sprayed fibrin sealant (FS; n = 8/group). TG-PDGF.AB binds to the fibrin biomatrix using the TG activity of factor XIIIa, and is subsequently released through enzymatic cleavage. Three doses of TG-PDGF.AB in FS (100 ng, 1 µg and 11 µg/ml FS) were tested. TG-PDGF.AB was bound to the fibrin biomatrix as evidenced by western blot analysis and subsequently released by enzymatic cleavage. A significantly accelerated and improved wound healing was achieved using sprayed FS containing TG-PDGF.AB compared to staples alone. Low concentrations (100 ng-1 µg TG-PDGF.AB/ml final FS clot) demonstrated to be sufficient to attain a nearly complete closure of mesh interstices 14 days after grafting. TG-PDGF.AB incorporated in FS via a specific binding technology was shown to be effective in grafted third-degree burn wounds. The adhesive properties of the fibrin matrix in conjunction with the prolonged growth factor stimulus enabled by this binding technology could be favourable in many pathological situations associated with wound-healing disturbances. Copyright © 2013 John Wiley & Sons, Ltd.


Subject(s)
Burns/drug therapy , Extracellular Matrix/chemistry , Fibrin , Platelet-Derived Growth Factor , Proto-Oncogene Proteins c-sis , Wound Healing/drug effects , Animals , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Fibrin/chemistry , Fibrin/pharmacokinetics , Fibrin/pharmacology , Platelet-Derived Growth Factor/chemistry , Platelet-Derived Growth Factor/pharmacokinetics , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins c-sis/chemistry , Proto-Oncogene Proteins c-sis/pharmacokinetics , Proto-Oncogene Proteins c-sis/pharmacology , Swine
2.
J Biomed Mater Res A ; 103(10): 3117-27, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25720595

ABSTRACT

Electrospun polymer/ceramic composites have gained interest for use as scaffolds for bone tissue engineering applications. In this study, we investigated methods to incorporate Platelet Derived Growth Factor-BB (PDGF-BB) in electrospun polycaprolactone (PCL) or PCL prepared with polyethylene oxide (PEO), where both contained varying levels (up to 30 wt %) of ceramic composed of biphasic calcium phosphates, hydroxyapatite (HA)/ß-tricalcium phosphate (TCP). Using a model protein, lysozyme, we compared two methods of protein incorporation, adsorption and emulsion electrospinning. Adsorption of lysozyme on scaffolds with ceramic resulted in minimal release of lysozyme over time. Using emulsion electrospinning, lysozyme released from scaffolds containing a high concentration of ceramic where the majority of the release occurred at later time points. We investigated the effect of reducing the electrostatic interaction between the protein and the ceramic on protein release with the addition of the cationic surfactant, cetyl trimethylammonium bromide (CTAB). In vitro release studies demonstrated that electrospun scaffolds prepared with CTAB released more lysozyme or PDGF-BB compared with scaffolds without the cationic surfactant. Human mesenchymal stem cells (MSCs) on composite scaffolds containing PDGF-BB incorporated through emulsion electrospinning expressed higher levels of osteogenic markers compared to scaffolds without PDGF-BB, indicating that the bioactivity of the growth factor was maintained. This study revealed methods for incorporating growth factors in polymer/ceramic scaffolds to promote osteoinduction and thereby facilitate bone regeneration.


Subject(s)
Mesenchymal Stem Cells/metabolism , Muramidase , Proto-Oncogene Proteins c-sis , Tissue Engineering , Tissue Scaffolds/chemistry , Becaplermin , Calcium Phosphates/chemistry , Cells, Cultured , Ceramics/chemistry , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Durapatite/chemistry , Humans , Hydroxyapatites/chemistry , Mesenchymal Stem Cells/cytology , Muramidase/chemistry , Muramidase/pharmacology , Polyesters/chemistry , Polyethylene Glycols/chemistry , Proto-Oncogene Proteins c-sis/chemistry , Proto-Oncogene Proteins c-sis/pharmacokinetics
3.
J Biomed Mater Res A ; 103(1): 282-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24610890

ABSTRACT

This study aimed to design a growth factor loaded copolyester of 3-hydroxybutyrate and 3-hydroxyhexanoate (PHBHHx) nanoparticles containing 3D collagen matrix to achieve growth factor sustained release for long-term stimulation of human mesenchymal stem cells (hMSCs) proliferation/differentiation for tissue engineer application. Platelet-derived growth factor-BB (PDGF-BB), which is known to enhance hMSCs proliferation in human serum, was selected as a model growth factor, and biodegradable copolyester of PHBHHx was chosen to be the sustained release vehicle. PDGF-BB phospholipid complex encapsulated PHBHHx nanoparticles were fabricated, and their effect on hMSCs proliferation was investigated via assays of CCK-8 and live-dead staining to cells inoculated in 2D tissue culture plates and 3D collagen gel scaffolds, respectively. The resulting spherical PHBHHx nanoparticles were stable in terms of their mean particle size, polydispersity index and zeta potential before and after lyophilization. In vitro study revealed a sustained release of PDGF-BB with a low burst release. Furthermore, sustained released PDGF-BB was revealed to significantly promote hMSCs proliferation in both cell monolayer and cell seeded 3D collagen scaffolds inoculated in serum-free media. Therefore, the 3D collagen matrices with locally sustained release growth factor nanoparticles hold promise to be used for stem cell tissue engineering.


Subject(s)
Hydrogels , Mesenchymal Stem Cells/metabolism , Models, Biological , Nanoparticles , Proto-Oncogene Proteins c-sis/metabolism , Becaplermin , Cells, Cultured , Freeze Drying , Humans , Microscopy, Electron, Scanning , Prohibitins , Proto-Oncogene Proteins c-sis/pharmacokinetics
4.
Biomaterials ; 33(28): 6709-20, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22770570

ABSTRACT

Heparin-conjugated electrospun poly(ε-caprolactone) (PCL)/gelatin scaffolds were developed to provide controlled release of platelet-derived growth factor-BB (PDGF-BB) and allow prolonged bioactivity of this molecule. A mixture of PCL and gelatin was electrospun into three different morphologies. Next, heparin molecules were conjugated to the reactive surface of the scaffolds. This heparin-conjugated scaffold allowed the immobilization of PDGF-BB via electrostatic interaction. In vitro PDGF-BB release profiles indicated that passive physical adsorption of PDGF-BB to non-heparinized scaffolds resulted in an initial burst release of PDGF-BB within 5 days, which then leveled off. However, electrostatic interaction between PDGF-BB and the heparin-conjugated scaffolds gave rise to a sustained release of PDGF-BB over the course of 20 days without an initial burst. Moreover, PDGF-BB that was strongly bound to the heparin-conjugated scaffolds enhanced smooth muscle cell (SMC) proliferation. In addition, scaffolds composed of 3.0 µm diameter fibers that were immobilized with PDGF-BB accelerated SMC infiltration into the scaffold when compared to scaffolds composed of smaller diameter fibers or scaffolds that did not release PDGF-BB. We concluded that the combination of the large pore structure in the scaffolds and the heparin-mediated delivery of PDGF-BB provided the most effective cellular interactions through synergistic physical and chemical cues.


Subject(s)
Myocytes, Smooth Muscle/metabolism , Proto-Oncogene Proteins c-sis/administration & dosage , Proto-Oncogene Proteins c-sis/pharmacokinetics , Tissue Scaffolds/chemistry , Analysis of Variance , Becaplermin , Cell Proliferation/drug effects , Delayed-Action Preparations , Gelatin/chemistry , Heparin/chemistry , Humans , Microscopy, Electron, Scanning , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Polyesters/chemistry , Proto-Oncogene Proteins c-sis/chemistry , Tissue Engineering/methods
5.
J Biomed Nanotechnol ; 7(6): 823-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22416582

ABSTRACT

Electrospinning is a versatile technique used to fabricate potential tissue engineering scaffolds with a structure similar to the native extracellular matrix (ECM). In this study, Platelet-Derived Growth Factor (PDGF)-BB with BSA as a carrier protein was incorporated into an electrospun PLGA/PEG-PLA composite scaffold for induction of cell migration, an early process necessary for tissue regeneration and wound healing. Incorporating PDGF-BB into the fibers did not change the overall morphology of the scaffold, with the exception of a slight increase (approximately 12%) in the number of fibers with diameters ranging from 1-100 nm. Following a strong burst of release during the initial 24 hours, approximately 20% of the total incorporated PDGF-BB was released from the scaffold over 5 days, as determined by ELISA. The presence of the released PDGF-BB was also confirmed via SDS-PAGE. Using an in vitro agarose-cell migration assay with MC3T3 pre-osteoblastic cells, the preserved bioactivity of the released PDGF-BB was demonstrated via its ability to stimulate robust cell migration, equivalent to that of pure unincorporated (control) PDGF-BB. Overall, this study demonstrates that it is feasible to incorporate and deliver bioactive PDGF-BB via an electrospun scaffold for potential tissue repair applications, especially as a potent inducer of cell migration.


Subject(s)
Cell Movement/drug effects , Lactic Acid/pharmacology , Nanofibers/chemistry , Polyethylene Glycols/pharmacology , Polyglycolic Acid/pharmacology , Proto-Oncogene Proteins c-sis/pharmacology , Tissue Scaffolds/chemistry , Analysis of Variance , Animals , Becaplermin , Cell Culture Techniques/methods , Cell Line , Electrochemical Techniques , Humans , Lactic Acid/chemistry , Mice , Polyethylene Glycols/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Proto-Oncogene Proteins c-sis/chemistry , Proto-Oncogene Proteins c-sis/pharmacokinetics
6.
Mol Pharm ; 7(5): 1724-33, 2010 Oct 04.
Article in English | MEDLINE | ID: mdl-20681555

ABSTRACT

New blood vessel formation is a critical requirement for treating many vascular and ischemia related diseases, as well as for many tissue engineering applications. Angiogenesis and vasculogenesis, in fact, represent crucial processes for the functional regeneration of complex tissues through tissue engineering strategies. Several growth factors (GFs) and signaling molecules involved in blood vessels formation have been identified, but their application to the clinical setting is still strongly limited by their extremely short half-life in the body. To overcome these limitations, we have developed a new injectable controlled release device based on polymeric nanoparticles for the delivery of two natural proangiogenic GFs: platelet derived growth factor (PDGF-BB) and fibroblast growth factor (FGF-2). The nanoparticle system was prepared by a modified solvent diffusion technique, encapsulating the GF both in presence and in the absence of two stabilizing agents: bovine serum albumin (BSA) and heparin sodium salt (Hp). The developed nanocarriers were characterized for morphology, size, encapsulation efficiency, release kinetics in vitro and GF activity in cell cultures. The results have indicated that the coencapsulation of stabilizing agents can preserve the GF active structure and, in addition, increase their encapsulation efficiency into nanoparticles. Through this optimization process, we were able to raise the encapsulation efficiency of FGF-2 to 63%, and that of PDGF-BB to 87%. These PLGA:poloxamer blend nanoparticles loaded with GFs were able to release PDGF-BB and FGF-2 in a sustained fashion for more than a month. This work also confirms other positive features of PLGA:poloxamer nanoparticles. Namely, they are able to maintain their stability in simulated biological medium, and they are also nontoxic to cell culture models. Incubation of nanoparticles loaded with FGF-2 or PDGF-BB with endothelial cell culture models has confirmed that GFs are released in a bioactive form. Altogether, these results underline the interest of PLGA:poloxamer nanoparticles for the controlled delivery of GFs and substantiate their potential for the treatment of ischemic diseases and for tissue engineering applications.


Subject(s)
Angiogenesis Inducing Agents/administration & dosage , Fibroblast Growth Factor 2/administration & dosage , Proto-Oncogene Proteins c-sis/administration & dosage , Animals , Becaplermin , Cattle , Drug Delivery Systems , Drug Stability , Fibroblast Growth Factor 2/pharmacokinetics , Freeze Drying , Hep G2 Cells , Humans , Lactic Acid/chemistry , Lactic Acid/toxicity , Nanocapsules/administration & dosage , Nanocapsules/chemistry , Nanocapsules/toxicity , Nanocapsules/ultrastructure , Poloxamer/chemistry , Poloxamer/toxicity , Polyglycolic Acid/chemistry , Polyglycolic Acid/toxicity , Polylactic Acid-Polyglycolic Acid Copolymer , Proto-Oncogene Proteins c-sis/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...