Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Adv Biosyst ; 3(12): e1900184, 2019 12.
Article in English | MEDLINE | ID: mdl-32648681

ABSTRACT

The process of new bone formation following trauma requires the temporal recruitment of cells to the site, including mesenchymal stem cells, preosteoblasts, and osteoblasts, the latter of which deposit minerals. Hence, bone repair, a process that is assessed by the extent of mineralization within the defect, can take months before it is possible to determine if a treatment is successful. Here, a fluorescently tagged Osterix, an early key gene in the bone formation cascade, is used as a predictive measure of bone formation. Using a calvarial defect model in mice, the ability to noninvasively track the Osterix transcription factor in an Osterix-mCherry mouse model is evaluated as a measure for bone formation following treatment with recombinant human Bone-Morphogenetic-Protein 2 (rhBMP-2). Two distinct delivery materials are utilized, an injectable nanocomposite hydrogel and a collagen sponge, that afford distinct release kinetics and it is found that cherry-fluorescent protein can be detected as early as 2 weeks following treatment. Osterix intensity correlates with subsequent bone formation and hence can serve as a rapid screening tool for osteogenic drugs or for the evaluation and optimization of delivery platforms.


Subject(s)
Luminescent Proteins/metabolism , Osteogenesis/physiology , Skull/metabolism , Sp7 Transcription Factor/metabolism , Animals , Bone Morphogenetic Protein 2/pharmacology , Cells, Cultured , Luminescent Proteins/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/drug effects , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/pharmacology , Regeneration/drug effects , Sp7 Transcription Factor/genetics , Tissue Scaffolds/chemistry , Transforming Growth Factor beta/pharmacology , Red Fluorescent Protein
2.
ACS Nano ; 11(3): 2598-2610, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28221760

ABSTRACT

Hydrogels are an attractive class of biomaterials for minimally invasive local drug delivery given their injectability, tunability, high water content, and biocompatibility. Broad applicability though is challenged: relatively modest mechanical properties restrict use to soft tissues, while flow properties necessary for injectability limit implantation to dried, enclosed tissues to minimize material migration during gelation. To address these dual concerns, we designed an injectable nanocomposite hydrogel based on dextran aldehyde and a poly(amido amine) dendrimer doped with phyllosilicate nanoplatelet fillers. Balance of components allows for exfoliation of nanoplatelets, significantly changing macromer solution flow, facilitating injection and manipulation in a wide variety of implantation contexts while enhancing compressive modulus of hydrogels at low loading. Importantly, rheological and mechanical effects were dependent on aspect ratio, with high aspect ratio nanoplatelets having much stronger effects on mechanics and low aspect ratio nanoplatelets having stronger effects on rheology, enabling nearly independent control of rheological and mechanical properties. Nanoplatelets enhanced hydrogel properties at a filler loading substantially lower than that of comparably sized nanoparticles. We present a model to explain the role that aspect ratio plays in control of rheology and mechanics in nanoplatelet-containing hydrogels, with lessons for further nanocomposite hydrogel development. This low-cost biocompatible material may be useful as a drug delivery platform in challenging implantation environments.


Subject(s)
Biocompatible Materials/chemistry , Hydrogels/chemistry , Nanocomposites/chemistry , Rheology , Biocompatible Materials/chemical synthesis , Hydrogels/chemical synthesis , Particle Size , Surface Properties
3.
Clin Oral Implants Res ; 27(6): 676-85, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26183057

ABSTRACT

OBJECTIVE: To compare the ability of three non-glycosylated/rhBMP-2 (BMP-2) delivery systems to induce supracrestal bone. MATERIAL AND METHODS: Thirty-six custom SLActive dental implants were partially embedded in transverse orientation into the posterior mandibles of 18 adult rabbits with the head of the implant located 3 mm supracrestal. Delivery of BMP-2 (30 µg) from the following materials was studied: (1) Ti implant + BMP-2 with hydroxyapatite (HA)-coated collagen (Col/HA) scaffold, (2) Ti implant with Col/HA infused with PEG hydrogel + BMP-2, or (3) Ti implant with HA/ß-TCP/PEG hydrogel scaffold + BMP-2. Scaffolds were secured with a metal "umbrella." Non-BMP-2 contralateral controls were included. MicroCT imaging and histological analysis was performed after 10 weeks to assess new supracrestal bone formation. In vitro BMP-2 release studies were conducted. RESULTS: All treatment groups displayed new supracrestal bone formation. Ti + BMP-2 with Col/HA (3.0 ± 0.2 mm) and Ti with Col/HA/PEG hydrogel + BMP-2 (2.7 ± 0.4 mm) had significantly greater (P < 0.05) outcomes than without BMP-2. Maximum bone volume occurred in the Ti implant with HA/ß-TCP/PEG hydrogel scaffold + BMP-2 group. CONCLUSIONS: The use of an implant system composed of a partially inserted Ti implant, adjacent scaffold and scaffold stabilizer resulted in the formation of new supracrestal bone across all test groups with and without BMP-2. Delivery of BMP-2 directly from the Ti implant increased bone height, BIC and bone volume as compared to no BMP-2 when a Col/HA was used, but did not improve performance of the HA/ß-TCP/PEG scaffold.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Dental Implants , Osteogenesis/drug effects , Animals , Bone Morphogenetic Protein 2/administration & dosage , Calcium Phosphates/pharmacology , Collagen/pharmacology , Drug Delivery Systems , Durapatite/pharmacology , Hydrogels/pharmacology , Mandible/surgery , Polyethylene Glycols/pharmacology , Rabbits , Tissue Scaffolds , Titanium , X-Ray Microtomography
4.
Exp Gerontol ; 64: 62-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25681640

ABSTRACT

There is an age-associated reduction in the bone healing activity of bone morphogenetic protein-2 (BMP-2) that is currently addressed by administering higher doses of BMP-2 in elderly patients. The unwanted medical complications from high dose BMP-2 motivated this investigation to determine whether the addition of a low dose of fibroblast growth factor 2 (FGF-2) could enhance the ability of a lower dose of BMP-2 to heal calvarial bone defects in old mice (18-20 months old). FGF-2 (5 ng) and BMP-2 (2 µg) were administered by a controlled release two-phase biomaterial scaffold placed into the bone defect. FGF-2 released more rapidly and completely in vitro than BMP-2 (40% vs 2%). In vivo, both BMP-2 and FGF-2+BMP-2 groups formed more new bone in calvarial defects than scaffold alone (p < 0.001) or FGF-2 only groups (p < 0.01). The overall total volume of new bone was not statistically increased by the addition of FGF-2 to BMP-2 as measured by microCT, but the pattern of bone deposition was different. In old mice, but not young, there was enhanced bony fill in the central bone defect area when the BMP-2 was supplemented with FGF-2. Histological analysis of the center of the defect revealed an increased bone volume (%BV/TV (p = 0.004)) from the addition of FGF-2. These studies suggest that combining a low dose of FGF-2 with a low dose of BMP-2 has the potential to increase bone healing in old mice relative to BMP-2 alone.


Subject(s)
Aging , Bone Morphogenetic Protein 2/pharmacology , Bone Regeneration/drug effects , Fibroblast Growth Factor 2/pharmacology , Skull/pathology , Animals , Female , Mice , Mice, Inbred BALB C , Osteogenesis/drug effects , Skull/diagnostic imaging , Wound Healing/drug effects , X-Ray Microtomography
5.
Endocrinology ; 155(3): 965-74, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24424065

ABSTRACT

Repair of bone defects remains a significant clinical problem. Bone morphogenetic protein 2 (BMP2) is US Food and Drug Administration-approved for fracture healing but is expensive and has associated morbidity. Studies have shown that targeted overexpression of the 18-kDa low-molecular-weight fibroblast growth factor 2 isoform (LMW) by the osteoblastic lineage of transgenic mice increased bone mass. This study tested the hypotheses that overexpression of LMW would directly enhance healing of a critical size calvarial bone defect in mice and that this overexpression would have a synergistic effect with low-dose administration of BMP2 on critical size calvarial bone defect healing. Bilateral calvarial defects were created in LMW transgenic male mice and control/vector transgenic (Vector) male mice and scaffold with or without BMP2 was placed into the defects. New bone formation was assessed by VIVA-computed tomography of live animals over a 27-week period. Radiographic and computed tomography analysis revealed that at all time points, healing of the defect was enhanced in LMW mice compared with that in Vector mice. Although the very low concentration of BMP2 did not heal the defect in Vector mice, it resulted in complete healing of the defect in LMW mice. Histomorphometric and gene analysis revealed that targeted overexpression of LMW in osteoblast precursors resulted in enhanced calvarial defect healing due to increased osteoblast activity and increased canonical Wnt signaling.


Subject(s)
Bone Regeneration/drug effects , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation , Osteoblasts/metabolism , Skull/growth & development , Animals , Bone Morphogenetic Protein 2/metabolism , Cell Differentiation , Male , Mice , Mice, Transgenic , Microscopy, Fluorescence , Osteogenesis , Phosphorylation , Protein Isoforms/metabolism , Tomography, X-Ray Computed , Wnt Signaling Pathway , X-Ray Microtomography
6.
Tissue Eng Part A ; 20(1-2): 365-77, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23952622

ABSTRACT

The in vivo osteogenesis potential of mesenchymal-like cells derived from human embryonic stem cells (hESC-MCs) was evaluated in vivo by implantation on collagen/hydroxyapatite scaffolds into calvarial defects in immunodeficient mice. This study is novel because no osteogenic or chondrogenic differentiation protocols were applied to the cells prior to implantation. After 6 weeks, X-ray, microCT, and histological analysis showed that the hESC-MCs had consistently formed a highly vascularized new bone that bridged the bone defect and seamlessly integrated with host bone. The implanted hESC-MCs differentiated in situ to functional hypertrophic chondrocytes, osteoblasts, and osteocytes forming new bone tissue via an endochondral ossification pathway. Evidence for the direct participation of the human cells in bone morphogenesis was verified by two separate assays: with Alu and by human mitochondrial antigen positive staining in conjunction with co-localized expression of human bone sialoprotein in histologically verified regions of new bone. The large volume of new bone in a calvarial defect and the direct participation of the hESC-MCs far exceeds that of previous studies and that of the control adult hMSCs. This study represents a key step forward for bone tissue engineering because of the large volume, vascularity, and reproducibility of new bone formation and the discovery that it is advantageous to not over-commit these progenitor cells to a particular lineage prior to implantation. The hESC-MCs were able to recapitulate the mesenchymal developmental pathway and were able to repair the bone defect semi-autonomously without preimplantation differentiation to osteo- or chondroprogenitors.


Subject(s)
Bone Regeneration/physiology , Embryonic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Adult , Animals , Bone Regeneration/drug effects , Cell Differentiation/drug effects , Cell Shape/drug effects , Collagen/pharmacology , Durapatite/pharmacology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Morphogenesis/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Phenotype , Prosthesis Implantation , Regeneration/drug effects , Skull/drug effects , Skull/pathology , Stem Cell Transplantation , Tissue Scaffolds/chemistry
7.
J Gerontol A Biol Sci Med Sci ; 68(10): 1170-80, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23531867

ABSTRACT

Combined regimens of fibroblast growth factor-2 (FGF-2) and bone morphogenetic protein-2 (BMP-2) were investigated to stimulate osteogenic differentiation. In young mouse calvaria-derived cells, FGF-2 (0.16ng/mL) in combination with BMP-2 (50ng/mL) did not enhance mineralization, but in old mouse cells it resulted in more mineralization than BMP-2 alone. In young long bone mouse cultures, FGF-2 enhanced mineralization relative to BMP-2 alone, but in old cultures, lower dose of FGF-2 (0.016ng/mL) was necessary. In neonatal mouse calvarial cells, sequential delivery of low-dose FGF-2 and low-dose BMP-2 (5ng/mL) was more stimulatory than co-delivery. In young human cultures, 0.016ng/mL of FGF-2 did not enhance mineralization, in combination with 5ng/mL of BMP-2, but in older cultures, codelivery of FGF-2 and BMP-2 was superior to BMP-2 alone. In conclusion, BMP-2 treatment alone was sufficient for maximal mineralization in young osteoblast cultures. However, coadministration of FGF-2 and BMP-2 increases mineralization more than BMP-2 alone in cultures from old and young mouse long bones and old humans but not in young mouse calvarial cultures.


Subject(s)
Aging/pathology , Bone Morphogenetic Protein 2/administration & dosage , Fibroblast Growth Factor 2/administration & dosage , Osteogenesis/drug effects , Adult , Aged , Animals , Calcification, Physiologic/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred BALB C , Mice, Transgenic , Middle Aged , Osteoblasts/cytology , Osteoblasts/drug effects , Recombinant Proteins/administration & dosage , Young Adult
8.
J Mech Behav Biomed Mater ; 17: 269-77, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23127637

ABSTRACT

Self-reinforced composites (SRCs) are materials where both the matrix and fiber-reinforcing phase are made up of the same polymer. Improved bonding can be achieved with self-reinforced composites compared to traditional dual-polymer, fiber-reinforced composites owing to the identical chemistry of the components in SRCs. Bonding between the fiber and matrix phase is an important factor in applications where mechanical stability is required, such as in the field of bone repair. In this study, we prepared bioabsorbable poly(L-lactic acid)/hydroxyapatite (PLLA/HA) self-reinforced composites via a three-step process that includes surface etching of the fiber, the deposition of the HA coating onto the PLLA fibers through immersion in simulated body fluid (SBF), and hot compaction molding. Although coated with a layer of HA, self-reinforced composites were successfully generated by hot compaction. The effects of compaction time (15 and 30 min), compaction temperature (140, 150, 155, 160, 165, and 170 °C), and HA wt% (0, 5, 10, and 15 wt%) on flexural mechanical properties were studied. Mechanical test results indicated that in unfilled (no HA) PLLA SRCs, compaction time and temperature increased the flexural modulus of the composites tested. Based on the results obtained for unfilled composites, a single compaction time and temperature condition of 15 min and 170 °C were selected to study the effect of HA loading on the composite mechanical properties. HA was successfully loaded onto the fibers at 0, 5, 10, and 15 wt% before hot compaction and was found to significantly increase flexural modulus (P=0.0001). Modulus values ranged from 8.3 GPa±0.5 (0 wt% HA) to 9.7 GPa±0.6 (15 wt% HA). Microscopy results suggest that the HA in these composites forms a nodular-like structure along the fibers, which allows polymer-polymer contact yet prevents longitudinal shear. The procedure used successfully generated composites with flexural moduli near the lower range of bone that may have a possible clinical use for load-bearing bone-fixation devices.


Subject(s)
Biomimetics/methods , Coated Materials, Biocompatible/chemistry , Durapatite/chemistry , Lactic Acid/blood , Mechanical Phenomena , Materials Testing , Minerals/chemistry , Polyesters , Polymers
9.
Macromol Biosci ; 12(8): 1034-42, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22730306

ABSTRACT

These studies provide evidence for the ability of a commercially available, defined, hyaluronan-gelatin hydrogel, HyStem-C™, to maintain both mouse embryonic stem cells (mESCs) and human induced pluripotent stem cells (hiPSCs) in culture while retaining their growth and pluripotent characteristics. Growth curve and doubling time analysis show that mESCs and hiPSCs grow at similar rates on HyStem-C™ hydrogels and mouse embryonic fibroblasts and Matrigel™, respectively. Immunocytochemistry, flow cytometry, gene expression and karyotyping reveal that both human and murine pluripotent cells retain a high level of pluripotency on the hydrogels after multiple passages. The addition of fibronectin to HyStem-C™ enabled the attachment of hiPSCs in a xeno-free, fully defined medium.


Subject(s)
Biocompatible Materials/chemistry , Embryonic Stem Cells/cytology , Gelatin/chemistry , Hyaluronic Acid/chemistry , Induced Pluripotent Stem Cells/cytology , Animals , Biocompatible Materials/pharmacology , Biomarkers/metabolism , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Culture Media , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Fibronectins/chemistry , Gelatin/pharmacology , Humans , Hyaluronic Acid/pharmacology , Hydrogels , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Karyotyping , Lewis X Antigen/metabolism , Mice , Octamer Transcription Factor-3/metabolism , Tissue Engineering , Tissue Scaffolds
10.
J Gerontol A Biol Sci Med Sci ; 65(10): 1051-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20643704

ABSTRACT

The potential of fibroblast growth factor-2 (FGF-2) to stimulate osteoprogenitors in aging bone was investigated. Previous work showed a decrease in bone formation in cell cultures derived from bone of elderly female patients, but not in cells from age-matched male or younger female patients, with transforming growth factor ß increasing bone formation but not increasing osteoprogenitors. In the present study, FGF-2 was shown to significantly stimulate, in a dose-dependent manner, proliferation of mesenchyme-derived progenitor cells from bones of young and old mouse and humans. In proliferation assays, human cells were more responsive to lower concentrations (0.0016 ng/mL) of FGF-2 than mouse cells, but proliferation was less in cells from older bone. Immunofluorescence microscopy revealed that FGF-2 increased and prevented the decline in cells expressing activated leukocyte cell adhesion molecule, a novel marker for early lineage osteoblasts, but not α-smooth muscle actin. FGF-2 may have therapeutic potential for stimulating osteoblast progenitors in aging.


Subject(s)
Aging/physiology , Bone Marrow Cells/drug effects , Cell Proliferation/drug effects , Fibroblast Growth Factor 2/pharmacology , Mesenchymal Stem Cells/drug effects , Adult , Aged , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Female , Fluorescent Antibody Technique , Humans , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Middle Aged , Young Adult
11.
J Am Chem Soc ; 127(24): 8697-704, 2005 Jun 22.
Article in English | MEDLINE | ID: mdl-15954775

ABSTRACT

We have designed monolayers with weak intermolecular interactions for use as placeholders in intelligent self- and directed-assembly. We have shown that these 1-adamantanethiolate monolayers are labile with respect to displacement by exposing them to dilute solutions of alkanethiols. These self-assembled monolayers (SAMs) of 1-adamantanethiol on Au{111} were probed using ambient scanning tunneling microscopy (STM), and their assembled order was determined. Solution deposition of the molecules results in a highly ordered hexagonally close-packed molecular lattice with a measured nearest neighbor distance of 6.9 +/- 0.4 A. The SAMs exhibit several rotational domains, but lack the protruding domain boundaries typical of alkanethiolate SAMs, and are similarly stable at room temperature. Co-deposition of alkanethiol and 1-adamantanethiol from solution results in alkanethiolate SAMs, except when using extremely low alkanethiol to 1-adamantanethiol concentration ratios. Facile displacement of low interaction strength SAMs can be exploited to enhance patterning using soft nanolithography.

SELECTION OF CITATIONS
SEARCH DETAIL
...