Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Biomed Mater Res A ; 109(12): 2545-2555, 2021 12.
Article in English | MEDLINE | ID: mdl-34173706

ABSTRACT

Bone repair in elderly mice has been shown to be improved or negatively impacted by supplementing the highly osteogenic bone morphogenetic protein-2 (BMP-2) with fibroblast growth factor-2 (FGF-2). To better predict the outcome of FGF-2 supplementation, we investigated whether endogenous levels of FGF-2 play a role in optimal dosing of FGF-2 for augmenting BMP-2 activity in elderly mice. In vivo calvarial bone defect studies in Fgf2 knockout mice with wildtype controls were conducted with the growth factors delivered in a highly localized manner from a biomimetic calcium phosphate/polyelectrolyte multilayer coating applied to a bone graft substitute. Endogenous FGF-2 levels were measured in old mice versus young and found to decrease with age. Optimal dosing for improving bone defect repair correlated with levels of endogenous FGF-2, with a larger dose of FGF-2 required to have a positive effect on bone healing in the Fgf2 knockout mice. The same dose in wildtype old mice, with higher levels of FGF-2, promoted chondrogenesis and increased osteoclast activity. The results suggest a personalized medicine approach, based on a knowledge of endogenous levels of FGF-2, should guide FGF-2 supplementation in order to avoid provoking excessive bone resorption and cartilage formation, both of which inhibited calvarial bone repair.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Bone and Bones/abnormalities , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/pharmacology , Skull/drug effects , Skull/growth & development , Aging/pathology , Animals , Biomimetics , Bone Resorption , Bone Transplantation , Calcium Phosphates , Cartilage/growth & development , Coated Materials, Biocompatible , Drug Delivery Systems , Female , Fracture Healing , Mice , Mice, Knockout
2.
Biomaterials ; 196: 90-99, 2019 03.
Article in English | MEDLINE | ID: mdl-30075952

ABSTRACT

Older adults suffer from weakened and delayed bone healing due to age-related alterations in bone cells and in the immune system. Given the interaction between the immune system and skeletal cells, therapies that address deficiencies in both the skeletal and the immune system are required to effectively treat bone injuries of older patients. The sequence of macrophage activation observed in healthy tissue repair involves a transition from a pro-inflammatory state followed by a pro-reparative state. In older patients, inflammation is slower to resolve and impedes healing. The goal of this study was to design a novel drug delivery system for temporal guidance of the polarization of macrophages using bone grafting materials. A biomimetic calcium phosphate coating (bCaP) physically and temporally separated the pro-inflammatory stimulus interferon-gamma (IFNγ) from the pro-reparative stimulus simvastatin (SIMV). Effective doses were identified using a human monocyte line (THP-1) and testing culminated with bone marrow macrophages obtained from old mice. Sequential M1-to-M2 activation was achieved with both cell types. These results suggest that this novel immunomodulatory drug delivery system holds potential for controlling macrophage activation in bones of older patients.


Subject(s)
Calcium Phosphates/pharmacology , Cellular Senescence , Coated Materials, Biocompatible/pharmacology , Macrophages/cytology , Animals , Cellular Senescence/drug effects , Dose-Response Relationship, Drug , Drug Delivery Systems , Gene Expression Regulation/drug effects , Humans , Interferon-gamma/pharmacology , Kinetics , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , Simvastatin/pharmacology , THP-1 Cells , Time Factors
3.
Tissue Eng Part A ; 23(23-24): 1490-1501, 2017 12.
Article in English | MEDLINE | ID: mdl-28946792

ABSTRACT

A drug delivery coating for synthetic bone grafts has been developed to provide sequential delivery of multiple osteoinductive factors to better mimic aspects of the natural regenerative process. The coating is composed of a biomimetic calcium phosphate (bCaP) layer that is applied to a synthetic bone graft and then covered with a poly-l-Lysine/poly-l-Glutamic acid polyelectrolyte multilayer (PEM) film. Bone morphogenetic protein-2 (BMP-2) was applied before the coating process directly on the synthetic bone graft and then, bCaP-PEM was deposited followed by adsorption of fibroblast growth factor-2 (FGF-2) into the PEM layer. Cells access the FGF-2 immediately, while the bCaP-PEM temporally delays the cell access to BMP-2. In vitro studies with cells derived from mouse calvarial bones demonstrated that Sca-1 and CD-166 positive osteoblast progenitor cells proliferated in response to media dosing with FGF-2. Coated scaffolds with BMP-2 and FGF-2 were implanted in mouse calvarial bone defects and harvested at 1 and 3 weeks. After 1 week in vivo, proliferation of cells, including Sca-1+ progenitors, was observed with low dose FGF-2 and BMP-2 compared to BMP-2 alone, indicating that in vivo delivery of FGF-2 activated a similar population of cells as shown by in vitro testing. At 3 weeks, FGF-2 and BMP-2 delivery increased bone formation more than BMP-2 alone, particularly in the center of the defect, confirming that the proliferation of the Sca-1 positive osteoprogenitors by FGF-2 was associated with increased bone healing. Areas of bone mineralization were positive for double fluorochrome labeling of calcium and alkaline phosphatase staining of osteoblasts, along with increased TRAP+ osteoclasts, demonstrating active bone formation distinct from the bone-like collagen/hydroxyapatite scaffold. In conclusion, the addition of a bCaP layer to PEM delayed access to BMP-2 and allowed the FGF-2 stimulated progenitors to populate the scaffold before differentiating in response to BMP-2, leading to improved bone defect healing.


Subject(s)
Biomimetic Materials , Bone Morphogenetic Protein 2 , Calcium Phosphates , Coated Materials, Biocompatible , Fibroblast Growth Factor 2 , Skull , Animals , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Bone Morphogenetic Protein 2/chemistry , Bone Morphogenetic Protein 2/pharmacology , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Female , Fibroblast Growth Factor 2/chemistry , Fibroblast Growth Factor 2/pharmacology , Mice , Mice, Transgenic , NIH 3T3 Cells , Skull/injuries , Skull/metabolism , Skull/pathology
4.
Regen Eng Transl Med ; 3(2): 94-105, 2017 Jun.
Article in English | MEDLINE | ID: mdl-29457125

ABSTRACT

Dental caries (tooth decay) is the most common chronic disease. Dental tissue engineering is a promising alternative approach to alleviate the shortcomings of the currently available restorative materials. Mimicking the natural extracellular matrix (ECM) could enhance the performance of tissue engineering scaffolds. In this study, we developed microtubular (~20 µm diameter) polymethyl methacrylate (PMMA) scaffolds resembling the tubular (~2.5 µm diameter) structure of dentin, the collagen-based mineralized tissue that forms the major portion of teeth, to study the effect of scaffold architecture on differentiation of mouse dental pulp cells in vitro. Flat (control), plasma-treated solid and microtubular PMMA scaffolds with densities of 240±15, 459±51 and 480±116 tubules/mm2 were first characterized using scanning electron microscopy and contact angle measurements. Dental pulp cells were cultured on the surface of the scaffolds for up to 21 days and examined using various assays. Cell proliferation and mineralization were examined using Alamar Blue and Xylenol Orange (XO) staining assays, respectively. The differentiation of pulp cells into odontoblasts was examined by immunostaining for Nestin and by quantitative PCR analysis for dentin matrix protein 1 (Dmp1), dentin sialophosphoprotein (Dspp) and osteocalcin (Ocn). Our results showed that the highest tubular density scaffolds significantly (p<0.05) enhanced differentiation of pulp cells into odontoblasts as compared to control flat scaffolds, as evidenced by increased expression of Nestin (5.4x). However, mineralization was suppressed on all surfaces, possibly due to low cell density. These results suggest that the microtubular architecture may be a desirable feature of scaffolds developed for clinical applications. LAY SUMMARY: Regenerative engineering of diseased or traumatized tooth structure could avoid the deficiencies of traditional dental restorative (filling) materials. Cells in the dental pulp have the potential to differentiate to dentin-producing odontoblast cells. Furthermore, cell-supporting scaffolds that mimic a natural extracellular matrix (ECM) are known to influence behavior of progenitor cells. Accordingly, we hypothesized that a dentin-like microtubular scaffold would enhance differentiation of dental pulp cells. The hypothesis was proven true and differentiation to odontoblasts increased with increasing density of the microtubules. However, mineralization was suppressed, possibly due to a low density of cells. The results demonstrate the potential benefits of a microtubular scaffold design to promote odontoblast cells for regeneration of dentin.

5.
Int J Pharm ; 517(1-2): 58-66, 2017 Jan 30.
Article in English | MEDLINE | ID: mdl-27923697

ABSTRACT

Nanoparticles as drug delivery carriers have been investigated over the last few decades, particularly for cancer treatment. The rationale in developing such nanoparticles is to maximize drug efficacy while minimizing toxic side effects. This can be most effectively achieved through target specific drug delivery. A novel biocompatible nanoparticle platform prepared using the core-shell self-assembly technique is reported. The core consists of calcium phosphate which is biocompatible and pH-sensitive, and the shell is composed of biocompatible polymers (hyaluronic acid, CD44 targeting moiety; and chitosan, physical cross-linker). Cisplatin was selected as a model drug and incorporated between the core and the shell. The nanoparticle composition was optimized for high serum stability and low protein binding. These nanoparticles demonstrated target specific delivery in human lung cancer cells (which overexpress CD44 receptors). The targeting ability of the nanoparticles was confirmed with an 8-fold increase of drug efficacy (IC50) compared to cisplatin. Furthermore, the pH-sensitive core of the nanoparticle platform led to controlled drug release through destabilization in acidic conditions. This platform technology provides a simple approach for the design of targeted biocompatible nanoparticles for cancer therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Biocompatible Materials/chemistry , Cisplatin/administration & dosage , Drug Carriers/chemistry , Nanoparticles/chemistry , A549 Cells , Calcium Phosphates/chemistry , Cell Survival/drug effects , Chitosan/chemistry , Drug Compounding , Drug Delivery Systems , Drug Liberation , Drug Stability , Humans , Hyaluronan Receptors/chemistry , Hyaluronic Acid/chemistry , Hydrogen-Ion Concentration , Particle Size
6.
J Integr Med ; 14(5): 389-99, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27641610

ABSTRACT

OBJECTIVE: To assess the effect of human biofield therapy, an integrative medicine modality, on the development of tumors and metastasis, and immune function in a mouse breast cancer model. METHODS: Mice were injected with 66cl4 mammary carcinoma cells. In study one, mice received biofield therapy after cell injection. In study two, mice were treated by the biofield practitioner only prior to cell injection. Both studies had two control groups of mock biofield treatments and phosphate-buffered saline injection. Mice were weighed and tumor volume was determined. Blood samples were collected and 32 serum cytokine/chemokine markers were measured. Spleens/popliteal lymph nodes were isolated and dissociated for fluorescent-activated cell sorting (FACS) analysis of immune cells or metastasis assays in cell culture. RESULTS: No significant differences were found in weight, tumor size or metastasis. Significant effects were found in the immune responses in study one but no additional effects were found in study two. In study one, human biofield treatment significantly reduced percentage of CD4(+)CD44loCD25(+) and percentage of CD8(+) cells, elevated by cancer in the lymph nodes, to control levels determined by FACS analysis. In the spleen, only CD11b(+) macrophages were increased with cancer, and human biofield therapy significantly reduced them. Of 11 cytokines elevated by cancer, only interferon-γ, interleukin-1, monokine induced by interfer-γ, interleukin-2 and macrophage inflammatory protein-2 were significantly reduced to control levels with human biofield therapy. CONCLUSION: Human biofield therapy had no significant effect on tumor size or metastasis but produced significant effects on immune responses apparent in the down-regulation of specific lymphocytes and serum cytokines in a mouse breast cancer model.


Subject(s)
Breast Neoplasms/therapy , Integrative Medicine , Animals , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cytokines/blood , Female , Flow Cytometry , Humans , Lymphocytes/immunology , Mice , Mice, Inbred BALB C , Research Design , Tumor Burden , Xenograft Model Antitumor Assays
7.
Dent Mater ; 32(1): 43-53, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26423007

ABSTRACT

OBJECTIVES: Our goal is to review design strategies for the fabrication of calcium phosphate ceramic scaffolds (CPS), in light of their transient role in bone tissue engineering and associated requirements for effective bone regeneration. METHODS: We examine the various design options available to meet mechanical and biological requirements of CPS and later focus on the importance of proper characterization of CPS in terms of architecture, mechanical properties and time-sensitive properties such as biodegradability. Finally, relationships between in vitro versus in vivo testing are addressed, with an attempt to highlight reliable performance predictors. RESULTS: A combinatory design strategy should be used with CPS, taking into consideration 3D architecture, adequate surface chemistry and topography, all of which are needed to promote bone formation. CPS represent the media of choice for delivery of osteogenic factors and anti-infectives. Non-osteoblast mediated mineral deposition can confound in vitro osteogenesis testing of CPS and therefore the expression of a variety of proteins or genes including collagen type I, bone sialoprotein and osteocalcin should be confirmed in addition to increased mineral content. CONCLUSIONS: CPS are a superior scaffold material for bone regeneration because they actively promote osteogenesis. Biodegradability of CPS via calcium and phosphate release represents a unique asset. Structural control of CPS at the macro, micro and nanoscale and their combination with cells and polymeric materials is likely to lead to significant developments in bone tissue engineering.


Subject(s)
Bone Regeneration , Bone Substitutes/chemistry , Calcium Phosphates/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Ceramics/chemistry , Materials Testing , Microscopy, Electron, Scanning , Osseointegration , Osteogenesis , Surface Properties
8.
J Cell Biochem ; 117(3): 721-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26332075

ABSTRACT

FGF-2 stimulates preosteoblast replication, and knockout of the FGF-2 gene in mice resulted in osteopenia with age, associated with decreased Wnt-ß-Catenin signaling. In addition, targeted expression of FGF-2 in osteoblast progenitors increased bone mass in mice via Wnt-ß-Catenin signaling. We posited that diminution of the intrinsic proliferative capacity of human mesenchyme-derived progenitor cells (HMDPCs) with age is due in part to reduction in FGF-2. To test this hypothesis HMDPCs from young (27-38), middle aged (47-56), and old (65-76) female human subjects were isolated from bone discarded after orthopedic procedures. HMDPCs cultures were mostly homogeneous with greater than 90% mesenchymal progenitor cells, determined by fluorescence-activated cell sorting. There was a progressive decrease in FGF-2 and FGFR1 mRNA and protein in HMDPCs with age. Since FGF-2 activates ß-catenin, which can enhance bone formation, we also assessed its age-related expression in HMDPCs. An age-related decrease in total-ß-Catenin mRNA and protein expression was observed. However there were increased levels of p-ß-Catenin and decreased levels of activated-ß-Catenin in old HMDSCs. FGF-2 treatment increased FGFR1 and ß-Catenin protein, reduced the level of p-ß-Catenin and increased activated-ß-Catenin in aged HMDPCs. In conclusion, reduction in FGF-2 expression could contribute to age-related impaired function of HMDPCs via modulation of Wnt-ß-catenin signaling.


Subject(s)
Aging , Fibroblast Growth Factor 2/physiology , Mesenchymal Stem Cells/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , beta Catenin/metabolism , Adult , Aged , Cells, Cultured , Female , Gene Expression , Humans , Middle Aged , Receptor, Fibroblast Growth Factor, Type 1/genetics , Wnt Signaling Pathway , Young Adult , beta Catenin/genetics
9.
Int J Cell Biol ; 2015: 249573, 2015.
Article in English | MEDLINE | ID: mdl-26448751

ABSTRACT

Carboxymethyl hyaluronic acid (CMHA) is a semisynthetic derivative of HA that is recognized by HA binding proteins but contains an additional carboxylic acid on some of the 6-hydroxyl groups of the N-acetyl glucosamine sugar units. These studies tested the ability of CMHA to stabilize the formation of calcium phosphate nanoparticles and evaluated their potential to target therapy resistant, CD44(+)/CD24(-/low) human breast cancer cells (BT-474EMT). CMHA stabilized particles (nCaP(CMHA)) were loaded with the chemotherapy drug cis-diamminedichloroplatinum(II) (CDDP) to form nCaP(CMHA)CDDP. nCaP(CMHA)CDDP was determined to be poorly crystalline hydroxyapatite, 200 nm in diameter with a -43 mV zeta potential. nCaP(CMHA)CDDP exhibited a two-day burst release of CDDP that tapered resulting in 86% release by 7 days. Surface plasmon resonance showed that nCaP(CMHA)CDDP binds to CD44, but less effectively than CMHA or hyaluronan. nCaP(CMHA-AF488) was taken up by CD44(+)/CD24(-) BT-474EMT breast cancer cells within 18 hours. nCaP(CMHA)CDDP was as cytotoxic as free CDDP against the BT-474EMT cells. Subcutaneous BT-474EMT tumors were more reproducibly inhibited by a near tumor dose of 2.8 mg/kg CDDP than a 7 mg/kg dose nCaP(CMHA)CDDP. This was likely due to a lack of distribution of nCaP(CMHA)CDDP throughout the dense tumor tissue that limited drug diffusion.

10.
Article in English | MEDLINE | ID: mdl-26113869

ABSTRACT

Evidence-based integrative medicine therapies have been introduced to promote wellness and offset side-effects from cancer treatment. Energy medicine is an integrative medicine technique using the human biofield to promote well-being. The biofield therapy chosen for study was Therapeutic Touch (TT). Breast cancer tumors were initiated in mice by injection of metastatic 66cl4 mammary carcinoma cells. The control group received only vehicle. TT or mock treatments were performed twice a week for 10 minutes. Two experienced TT practitioners alternated treatments. At 26 days, metastasis to popliteal lymph nodes was determined by clonogenic assay. Changes in immune function were measured by analysis of serum cytokines and by fluorescent activated cells sorting (FACS) of immune cells from the spleen and lymph nodes. No significant differences were found in body weight gain or tumor size. Metastasis was significantly reduced in the TT-treated mice compared to mock-treated mice. Cancer significantly elevated eleven cytokines. TT significantly reduced IL-1-a, MIG, IL-1b, and MIP-2 to control/vehicle levels. FACS demonstrated that TT significantly reduced specific splenic lymphocyte subsets and macrophages were significantly elevated with cancer. Human biofield therapy had no significant effect on primary tumor but produced significant effects on metastasis and immune responses in a mouse breast cancer model.

11.
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
12.
J Biomed Mater Res A ; 103(2): 500-10, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24733780

ABSTRACT

End-capping by covalently binding functional groups to the ends of polymer chains offers potential advantages for tissue engineering scaffolds, but the ability of such polymers to influence cell behavior has not been studied. As a demonstration, polylactide (PLA) was end-capped with lithium carboxylate ionic groups (hPLA13kLi) and evaluated. Thin films of the hPLA13kLi and PLA homopolymer were prepared with and without surface texturing. Murine osteoblast progenitor cells from collagen 1α1 transgenic reporter mice were used to assess cell attachment, proliferation, differentiation, and mineralization. Measurement of green fluorescent protein expressed by these cells and xylenol orange staining for mineral allowed quantitative analysis. The hPLA13kLi was biologically active, increasing initial cell attachment and enhancing differentiation, while reducing proliferation and strongly suppressing mineralization, relative to PLA. These effects of bound lithium ions (Li(+) ) had not been previously reported, and were generally consistent with the literature on soluble additions of lithium. The surface texturing generated here did not influence cell behavior. These results demonstrate that end-capping could be a useful approach in scaffold design, where a wide range of biologically active groups could be employed, while likely retaining the desirable characteristics associated with the unaltered homopolymer backbone.


Subject(s)
Calcification, Physiologic , Cell Differentiation , Lithium/chemistry , Membranes, Artificial , Osteoblasts/metabolism , Polyesters/chemistry , Stem Cells/metabolism , Animals , Mice , Mice, Transgenic , Osteoblasts/cytology , Stem Cells/cytology
15.
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
16.
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
17.
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
18.
PLoS One ; 7(3): e33225, 2012.
Article in English | MEDLINE | ID: mdl-22457746

ABSTRACT

Controlled differentiation of human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) into cells that resemble adult mesenchymal stem cells (MSCs) is an attractive approach to obtain a readily available source of progenitor cells for tissue engineering. The present study reports a new method to rapidly derive MSC-like cells from hESCs and hiPSCs, in one step, based on culturing the cells on thin, fibrillar, type I collagen coatings that mimic the structure of physiological collagen. Human H9 ESCs and HDFa-YK26 iPSCs were singly dissociated in the presence of ROCK inhibitor Y-27632, plated onto fibrillar collagen coated plates and cultured in alpha minimum essential medium (alpha-MEM) supplemented with 10% fetal bovine serum, 50 uM magnesium L-ascorbic acid phosphate and 100 nM dexamethasone. While fewer cells attached on the collagen surface initially than standard tissue culture plastic, after culturing for 10 days, resilient colonies of homogenous spindle-shaped cells were obtained. Flow cytometric analysis showed that a high percentage of the derived cells expressed typical MSC surface markers including CD73, CD90, CD105, CD146 and CD166 and were negative as expected for hematopoietic markers CD34 and CD45. The MSC-like cells derived from pluripotent cells were successfully differentiated in vitro into three different lineages: osteogenic, chondrogenic, and adipogenic. Both H9 hES and YK26 iPS cells displayed similar morphological changes during the derivation process and yielded MSC-like cells with similar properties. In conclusion, this study demonstrates that bioimimetic, fibrillar, type I collagen coatings applied to cell culture plates can be used to guide a rapid, efficient derivation of MSC-like cells from both human ES and iPS cells.


Subject(s)
Collagen , Mesenchymal Stem Cells/cytology , Pluripotent Stem Cells/cytology , Adipose Tissue/cytology , Antigens, CD/analysis , Bone and Bones/cytology , Cartilage/cytology , Cell Differentiation , Flow Cytometry , Humans , Immunophenotyping , Mesenchymal Stem Cells/immunology , Pluripotent Stem Cells/immunology
19.
J Biomed Opt ; 16(6): 066009, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21721810

ABSTRACT

We have developed a novel nitroimidazole indocyanine dye conjugate for tumor-targeted hypoxia fluorescence tomography. The hypoxia probe has been evaluated in vitro using tumor cell lines and in vivo with tumor targeting in mice. The in vitro cell studies were performed to assess fluorescence labeling differences between hypoxia and normoxia conditions. When treated with the hypoxia probe, a fluorescence emission ratio of 2.5-fold was found between the cells incubated under hypoxia compared to the cells in normoxia condition. Hypoxia specificity was also confirmed by comparing the cells treated with indocyanine dye alone. In vivo tumor targeting in mice showed that the fluorescence signals measured at the tumor site were twice those at the normal site after 150 min post-injection of the hypoxia probe. On the other hand, the fluorescence signals measured after injection of indocyanine dye were the same at tumor and normal sites. In vivo fluorescence tomography images of mice injected with the hypoxia probe showed that the probe remained for more than 5 to 7 h in the tumors, however, the images of mice injected with indocyanine only dye confirmed that the unbound dye washed out in less than 3 h. These findings are supported with fluorescence images of histological sections of tumor samples using a Li-COR scanner and immunohistochemistry technique for tumor hypoxia.


Subject(s)
Cell Hypoxia/physiology , Molecular Imaging/methods , Neoplasms/metabolism , Spectrometry, Fluorescence/methods , Spectroscopy, Near-Infrared/methods , Tomography/methods , Animals , Female , Immunohistochemistry , Indocyanine Green/chemistry , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Neoplasms/pathology , Nitroimidazoles/chemistry
20.
J Biomater Sci Polym Ed ; 21(10): 1371-87, 2010.
Article in English | MEDLINE | ID: mdl-20534191

ABSTRACT

A simple method is reported for fabricating polystyrene disk inserts coated with biomimetic carbonated hydroxyapatite (cHA) to be used for culturing osteoprogenitor cells or other stem cells. Roughened disks cut from tissue-culture polystyrene (TCPS) were coated in simulated body fluid with 5 x normal physiologic ionic concentrations (SBFx5) by a 2-step, 2-day method. The coatings were rigorously characterized by various methods and assessed in cell culture. An adherent, nearly 10 mm thick, relatively uniform layer of single-phase cHA was formed in two days. MC3T3-E1 and mouse calvaria-derived osteoprogenitor cells (pCOBs) were cultured on the cHA for various time points. Despite less initial attachment of both cell types to the cHA, proliferation rates on cHA were similar to that on TCPS. Two-fold greater cell attachment (P < 0.05) of the MC3T3-E1 cells was observed relative to the pCOBs, on both the TCPS and the cHA. Importantly, the coatings were relatively smooth, without the extensive agglomerates observed in other studies and remained adherent and morphologically unchanged after 21 days of culture. This technique can be used to rapidly produce high-quality cHA-coated TCPS disks for cell-culture studies.


Subject(s)
Coated Materials, Biocompatible/chemistry , Durapatite/chemistry , Osteoblasts/cytology , Polystyrenes/chemistry , Stem Cells/cytology , Animals , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , Coated Materials, Biocompatible/adverse effects , Mice , Microscopy, Electron, Scanning , Osteoblasts/drug effects , Osteoblasts/ultrastructure , Spectrometry, X-Ray Emission , Spectroscopy, Fourier Transform Infrared , Stem Cells/drug effects , Stem Cells/ultrastructure , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL
...