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











Publication year range
1.
Front Immunol ; 12: 653088, 2021.
Article in English | MEDLINE | ID: mdl-34122410

ABSTRACT

Allogeneic islet transplantation is a promising cell-based therapy for Type 1 Diabetes (T1D). The long-term efficacy of this approach, however, is impaired by allorejection. Current clinical practice relies on long-term systemic immunosuppression, leading to severe adverse events. To avoid these detrimental effects, poly(lactic-co-glycolic acid) (PLGA) microparticles (MPs) were engineered for the localized and controlled release of immunomodulatory TGF-ß1. The in vitro co-incubation of TGF-ß1 releasing PLGA MPs with naïve CD4+ T cells resulted in the efficient generation of both polyclonal and antigen-specific induced regulatory T cells (iTregs) with robust immunosuppressive function. The co-transplantation of TGF-ß1 releasing PLGA MPs and Balb/c mouse islets within the extrahepatic epididymal fat pad (EFP) of diabetic C57BL/6J mice resulted in the prompt engraftment of the allogenic implants, supporting the compatibility of PLGA MPs and local TGF-ß1 release. The presence of the TGF-ß1-PLGA MPs, however, did not confer significant graft protection when compared to untreated controls, despite measurement of preserved insulin expression, reduced intra-islet CD3+ cells invasion, and elevated CD3+Foxp3+ T cells at the peri-transplantation site in long-term functioning grafts. Examination of the broader impacts of TGF-ß1/PLGA MPs on the host immune system implicated a localized nature of the immunomodulation with no observed systemic impacts. In summary, this approach establishes the feasibility of a local and modular microparticle delivery system for the immunomodulation of an extrahepatic implant site. This approach can be easily adapted to deliver larger doses or other agents, as well as multi-drug approaches, within the local graft microenvironment to prevent transplant rejection.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Drug Carriers/chemistry , Graft Rejection/prevention & control , Immunologic Factors/administration & dosage , Islets of Langerhans Transplantation/adverse effects , Transforming Growth Factor beta1/administration & dosage , Animals , Blood Glucose/analysis , Coculture Techniques , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/diagnosis , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/diagnosis , Drug Liberation , Feasibility Studies , Glucose Tolerance Test , Graft Rejection/immunology , Humans , Immunomodulation , Islets of Langerhans Transplantation/methods , Male , Mice , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Primary Cell Culture , Rats , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Streptozocin/administration & dosage , Streptozocin/toxicity , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta1/pharmacokinetics , Transplantation, Homologous/adverse effects
2.
J Biomed Mater Res B Appl Biomater ; 108(5): 2041-2062, 2020 07.
Article in English | MEDLINE | ID: mdl-31872975

ABSTRACT

Degeneration of articular cartilage due to damages, diseases, or age-related factors can significantly decrease the mobility of the patients. Various tissue engineering approaches which take advantage of stem cells and growth factors in a three-dimensional constructs have been used for reconstructing articular tissue. Proliferative impact of basic fibroblast growth factor (bFGF) and chondrogenic differentiation effect of transforming growth factor-beta 1 (TGF-ß1) over mesenchymal stem cells have previously been verified. In this study, silk fibroin (SF) and of poly(ethylene glycol) dimethacrylate (PEGDMA) were used to provide a versatile platform for preparing hydrogels with tunable mechanical, swelling and degradation properties through physical and chemical crosslinking as a microenvironment for chondrogenic differentiation in the presence of bFGF and TGF-ß1 releasing nanoparticles (NPs) for the first time. Scaffolds with compressive moduli ranging from 95.70 ± 17.82 to 338.05 ± 38.24 kPa were obtained by changing both concentration PEGDMA and volume ratio of PEGDMA with 8% SF. Highest cell viability was observed in PEGDMA 10%-SF 8% (1:1) [PEG10-SF8(1:1)] hydrogel group. Release of bFGF and TGF-ß1 within PEG10-SF8(1:1) hydrogels resulted in higher DNA and glycosaminoglycans amounts indicating synergistic effect of dual release over proliferation and chondrogenic differentiation of dental pulp stem cells in hydrogels, respectively. Our results suggested that simultaneous delivery of bFGF and TGF-ß1 through utilization of PLGA NPs within PEG10-SF8(1:1) hydrogel provided a novel and versatile means for articular cartilage regeneration as they allow for dosage- and site-specific multiple growth factor delivery.


Subject(s)
Antimicrobial Cationic Peptides/chemistry , Cartilage, Articular/metabolism , Fibroins/chemistry , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Nanocapsules/chemistry , Tissue Scaffolds/chemistry , Transforming Growth Factor beta1/chemistry , Biocompatible Materials/chemistry , Cell Differentiation , Cell Survival , Chondrogenesis , Collagen Type II/chemistry , Drug Liberation , Glycosaminoglycans/chemistry , Humans , Mechanical Tests , Methacrylates/chemistry , Polyethylene Glycols/chemistry , Prosthesis Implantation , Tissue Engineering , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacokinetics
3.
Regen Med ; 14(8): 753-768, 2019 08.
Article in English | MEDLINE | ID: mdl-31474179

ABSTRACT

Aim: To investigate whether platelet-rich gel (PRG) incorporation could promote meniscal regeneration of the silk scaffold. Materials & methods: A PRG-incorporated silk sponge was fabricated for reconstruction of the meniscus in a rabbit meniscectomy model. Subsequently, characterization of the scaffold, as well as the in vitro cytocompatibility and in vivo function was evaluated. Results: Our results showed that the PRG-incorporated silk scaffold provided a sustained release of TGF-ß1 over 1 week. The PRG enhanced the cytocompatibility in vitro and cell infiltration in vivo of the silk sponge. Meanwhile, the implantation of the composite in situ ameliorated the cartilage degeneration in knee at 3 months. Conclusion: These findings indicated that PRG-incorporated silk scaffold could promote functional regeneration of the meniscus and effectively prevented subsequent osteoarthritis after meniscectomy.


Subject(s)
Blood Platelets , Meniscus/physiology , Regeneration , Silk , Tissue Scaffolds/chemistry , Transforming Growth Factor beta1 , Animals , Drug Implants/chemistry , Drug Implants/pharmacokinetics , Drug Implants/pharmacology , Gels/chemistry , Gels/pharmacology , Humans , Meniscectomy , Meniscus/surgery , Rabbits , Silk/chemistry , Silk/pharmacology , Transforming Growth Factor beta1/chemistry , Transforming Growth Factor beta1/pharmacokinetics , Transforming Growth Factor beta1/pharmacology
4.
Biomed Res Int ; 2019: 4923767, 2019.
Article in English | MEDLINE | ID: mdl-31223618

ABSTRACT

The aim of the present study was to investigate growth factors release kinetics for the combination of fresh platelet-rich fibrin (F-PRF) and lyophilized PRF (L-PRF) with different ratios to promote bone tissue regeneration. First, we quantified the level of transforming growth factor-ß1 (TGF-ß1), vascular endothelial growth factor (VEGF), and platelet-derived growth factor-AB (PDGF-AB) in vitro and analyzed their release kinetics from F-PRF, L-PRF, and the fresh/lyophilized PRF in different weight ratios (F:L=1:1, 1:3, 1:5). The second experimental phase was to investigate the proliferation and differentiation of bone mesenchymal stem cells (BMSCs) as a functional response to the factors released. To further test the osteogenic potential in vivo, different scaffolds (F-PRF, or L-PRF, or F:L=1:1) were implanted in rabbit cranial bone defects. There was a statistically significant increase in proliferation and differentiation of BMSCs when the culture medium contained different PRF exudates collected at day 14 compared with the negative control group. The results showed that the new bone formation in the fresh/lyophilized PRF (1:1) was much more than that of other groups in defects at both 6 and 12 weeks. Our data suggested growth factor concentration and release kinetics as a consequence of fresh and lyophilized PRF combination, which is an effective way for promoting bone regeneration.


Subject(s)
Bone Marrow Cells/metabolism , Bone Regeneration/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Mesenchymal Stem Cells/metabolism , Platelet-Rich Fibrin/chemistry , Transforming Growth Factor beta1 , Animals , Bone Marrow Cells/pathology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Humans , Male , Mesenchymal Stem Cells/pathology , Rabbits , Transforming Growth Factor beta1/chemistry , Transforming Growth Factor beta1/pharmacokinetics , Transforming Growth Factor beta1/pharmacology
5.
ACS Appl Mater Interfaces ; 11(16): 14608-14618, 2019 Apr 24.
Article in English | MEDLINE | ID: mdl-30938503

ABSTRACT

Continuous delivery of growth factors to the injury site is crucial to creating a favorable microenvironment for cartilage injury repair. In the present study, we fabricated a novel sustained-release scaffold, stromal-derived factor-1α (SDF-1α)/transforming growth factor-ß1 (TGF-ß1)-loaded silk fibroin-porous gelatin scaffold (GSTS). GSTS persistently releases SDF-1α and TGF-ß1, which enhance cartilage repair by facilitating cell homing and chondrogenic differentiation. Scanning electron microscopy showed that GSTS is a porous microstructure and the protein release assay demonstrated the sustainable release of SDF-1α and TGF-ß1 from GSTS. Bone marrow-derived mesenchymal stem cells (MSCs) maintain high in vitro cell activity and excellent cell distribution and phenotype after seeding into GSTS. Furthermore, MSCs acquired enhanced chondrogenic differentiation capability in the TGF-ß1-loaded scaffolds (GSTS or GST: loading TGF-ß1 only) and the conditioned medium from SDF-1α-loaded scaffolds (GSTS or GSS: loading SDF-1α only) effectively promoted MSCs migration. GSTS was transplanted into the osteochondral defects in the knee joint of rats, and it could promote cartilage regeneration and repair the cartilage defects at 12 weeks after transplantation. Our study shows that GSTS can facilitate in vitro MSCs homing, migration, chondrogenic differentiation and SDF-1α and TGF-ß1 have a synergistic effect on the promotion of in vivo cartilage forming. This SDF-1α and TGF-ß1 releasing GSTS have promising therapeutic potential in cartilage repair.


Subject(s)
Cartilage , Chemokine CXCL12 , Chondrogenesis/drug effects , Fibroins , Gelatin , Transforming Growth Factor beta1 , Animals , Cartilage/injuries , Cartilage/metabolism , Cartilage/pathology , Cell Differentiation/drug effects , Cell Movement/drug effects , Chemokine CXCL12/chemistry , Chemokine CXCL12/pharmacokinetics , Chemokine CXCL12/pharmacology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Fibroins/chemistry , Fibroins/pharmacokinetics , Fibroins/pharmacology , Gelatin/chemistry , Gelatin/pharmacokinetics , Gelatin/pharmacology , Male , Porosity , Rats , Transforming Growth Factor beta1/chemistry , Transforming Growth Factor beta1/pharmacokinetics , Transforming Growth Factor beta1/pharmacology
6.
Mar Drugs ; 17(1)2019 Jan 19.
Article in English | MEDLINE | ID: mdl-30669426

ABSTRACT

Articular cartilage is an avascular, non-innervated connective tissue with limited ability to regenerate. Articular degenerative processes arising from trauma, inflammation or due to aging are thus irreversible and may induce the loss of the joint function. To repair cartilaginous defects, tissue engineering approaches are under intense development. Association of cells and signalling proteins, such as growth factors, with biocompatible hydrogel matrix may lead to the regeneration of the healthy tissue. One current strategy to enhance both growth factor bioactivity and bioavailability is based on the delivery of these signalling proteins in microcarriers. In this context, the aim of the present study was to develop microcarriers by encapsulating Transforming Growth Factor-ß1 (TGF-ß1) into microparticles based on marine exopolysaccharide (EPS), namely GY785 EPS, for further applications in cartilage engineering. Using a capillary microfluidic approach, two microcarriers were prepared. The growth factor was either encapsulated directly within the microparticles based on slightly sulphated derivative or complexed firstly with the highly sulphated derivative before being incorporated within the microparticles. TGF-ß1 release, studied under in vitro model conditions, revealed that the majority of the growth factor was retained inside the microparticles. Bioactivity of released TGF-ß1 was particularly enhanced in the presence of highly sulphated derivative. It comes out from this study that GY785 EPS based microcarriers may constitute TGF-ß1 reservoirs spatially retaining the growth factor for a variety of tissue engineering applications and in particular cartilage regeneration, where the growth factor needs to remain in the target location long enough to induce robust regenerative responses.


Subject(s)
Alteromonas/chemistry , Drug Carriers/chemistry , Polysaccharides/chemistry , Transforming Growth Factor beta1/administration & dosage , Biological Availability , Cartilage, Articular/drug effects , Cartilage, Articular/physiology , Cell Line , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Drug Carriers/isolation & purification , Drug Compounding/methods , Drug Implants , Drug Liberation , Humans , Hydrothermal Vents/microbiology , Microfluidics , Polysaccharides/isolation & purification , Regeneration/drug effects , Tissue Scaffolds/chemistry , Transforming Growth Factor beta1/pharmacokinetics
7.
J Biomed Mater Res A ; 107(2): 403-413, 2019 02.
Article in English | MEDLINE | ID: mdl-30485631

ABSTRACT

Providing affinity sites on alginate (ALG) matrix enables specific binding of growth factors to the polymer backbone and allows their release in a controlled fashion. In this study, we used a blend of alginate sulfate (ALG-S) and polyvinyl alcohol (PVA) to fabricate electrospun scaffolds capable of delivering a heparin-like growth factor, transforming growth factor-beta1 (TGF-ß1). The alginate was sulfated with different degrees of sulfation (DS, from 0.8, 3.4 to 12.4%) by a simple process. The success of sulfation was determined by Fourier-transform infrared spectroscopy (FTIR), nuclear magnetic resonance spectroscopy (NMR), elemental analysis, ultraviolet (UV) spectroscopy and staining with dimethylmethylene blue. The physical-mechanical properties of nanofibrous mats were characterized by scanning electron microscopy (SEM), FTIR, energy-dispersive X-ray spectroscopy (EDX), tensile strength and mass loss analysis. Additionally, the release kinetics of transforming growth factor-ß1 (TGF-ß1) from PVA/ALG-S and PVA/ALG scaffolds were compared. The results showed that the binding and entrapment of TGF-ß1 to the nanofibrous scaffolds are improved by the addition of sulfate group to alginate. In conclusion, our results support that nanofibrous scaffold based on PVA/ALG-S can deliver growth factors in tissue engineering application. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 403-413, 2019.


Subject(s)
Alginates/chemistry , Drug Carriers/chemistry , Nanofibers/chemistry , Polyvinyl Alcohol/chemistry , Transforming Growth Factor beta1/administration & dosage , Cell Line , Drug Delivery Systems , Drug Liberation , Humans , Mesenchymal Stem Cells/cytology , Nanofibers/ultrastructure , Sulfates/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Transforming Growth Factor beta1/pharmacokinetics
8.
Int J Nanomedicine ; 13: 3177-3188, 2018.
Article in English | MEDLINE | ID: mdl-29922054

ABSTRACT

INTRODUCTION: Stimulating the proliferation and differentiation of chondrocytes for the regeneration of articular cartilage is a promising strategy, but it is currently ineffective. Although both physical stimulation and growth factors play important roles in cartilage repair, their interplay remains unclear and requires further investigation. In this study, we aimed to clarify their contribution using a magnetic drug carrier that not only can deliver growth factors but also provide an external stimulation to cells in the two-dimensional environment. MATERIALS AND METHODS: We developed a nanocapsule (transforming growth factor-ß1 [TGF-ß1]-loaded magnetic amphiphilic gelatin nanocapsules [MAGNCs]; TGF-ß1@MAGNCs) composed of hexanoic-anhydride-grafted gelatin and iron oxide nanoparticles to provide a combination treatment of TGF-ß1 and magnetically induced physical stimuli. With the expression of Arg-Gly-Asp peptide in the gelatin, the TGF-ß1@MAGNCs have an inherent affinity for chondrogenic ATDC5 cells. RESULTS: In the absence of TGF-ß1, ATDC5 cells treated with a magnetic field show significantly upregulated Col2a1 expression. Moreover, TGF-ß1 slowly released from biodegradable TGF-ß1@ MAGNCs further improves the differentiation with increased expression of Col2a1 and Aggrecan. CONCLUSION: Our study shows the time-dependent interplay of physical stimuli and growth factors on chondrogenic regeneration, and demonstrates the promising use of TGF-ß1@MAGNCs for articular cartilage repair.


Subject(s)
Chondrocytes/cytology , Chondrogenesis/drug effects , Nanocapsules/chemistry , Transforming Growth Factor beta1/pharmacokinetics , Aggrecans/metabolism , Animals , Cell Differentiation/drug effects , Cell Line , Chondrocytes/drug effects , Chondrogenesis/physiology , Collagen Type II/metabolism , Gelatin/chemistry , Humans , Magnetic Fields , Mice , Oligopeptides/chemistry , Tissue Engineering/methods , Transforming Growth Factor beta1/genetics
9.
Int J Mol Sci ; 18(10)2017 Sep 30.
Article in English | MEDLINE | ID: mdl-28973964

ABSTRACT

Dysregulation of the transforming growth factor-ß1 (TGF-ß1)/selected small mother against decapentaplegic (SMAD) pathway can be implicated in development of age-related macular degeneration (AMD), and the delivery of TGF-ß1 could be beneficial for AMD. We developed a new ophthalmic formulation of TGF-ß1 assessing the ocular pharmacokinetic profile of TGF-ß1 in the rabbit eye. Small unilamellar vesicles (SUV) loaded with TGF-ß1 were complemented with Annexin V and Ca2+, and the vitreous bioavailability of TGF-ß1 was assessed after topical ocular administration by a commercial ELISA kit. We detected high levels of TGF-ß1 (Cmax 114.7 ± 12.40 pg/mL) in the vitreous after 60 min (Tmax) from the topical application of the liposomal suspension. Ocular tolerability was also assessed by a modified Draize's test. The new formulation was well tolerated. In conclusion, we demonstrated that the novel formulation was able to deliver remarkable levels of TGF-ß1 into the back of the eye after topical administration. Indeed, this TGF-ß1 delivery system may be useful in clinical practice to manage ophthalmic conditions such as age-related macular degeneration, skipping invasive intraocular injections.


Subject(s)
Eye/metabolism , Transforming Growth Factor beta1/administration & dosage , Transforming Growth Factor beta1/pharmacokinetics , Administration, Ophthalmic , Animals , Humans , Liposomes , Macular Degeneration/drug therapy , Models, Molecular , Ophthalmic Solutions/administration & dosage , Ophthalmic Solutions/pharmacokinetics , Rabbits , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics
10.
Sci Rep ; 7(1): 10553, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28874815

ABSTRACT

Chondral defects pose a great challenge for clinicians to manage owing to the limited capacity for self-healing. Various traditional approaches have been adopted for the repair of these defects with unsatisfactory results. Cartilage tissue engineering techniques have emerged as promising strategies to enhance regeneration and overcome these traditional shortcomings. The cell-homing based technique is considered the most promising owing to its unique advantages. Thermosensitive hydrogels have been applied as scaffolds for biomedical applications with smart sol-gel response for altering environmental temperature. Transforming growth factor (TGF)-ß1 is considered to be capable of promoting chondrogenesis. In this study, a novel TGF-ß1-loaded poly(ε-caprolactone)-poly(ethylene glycol)-poly(ε-caprolactone) (PCEC) hydrogel was fabricated using simple procedures. Hydrogel characterization, rheological testing, component analysis, and assessment of sol-gel transition, in vitro degradation, and TGF-ß1 release confirmed that this material possesses a porous microstructure with favorable injectability and sustained drug release. Full-thickness cartilage defects were induced on rat knees for in vivo cartilage repair for eight weeks. Micro-CT and histological evaluation provided further evidence of the optimal capacity of this novel hydrogel for cartilage regeneration with respect to that of other methods. Moreover, our results demonstrated that the cell-free hydrogel is thermosensitive, injectable, biodegradable, and capable of in vivo cartilage repair and possesses high potential and benefits for acellular cartilage tissue engineering and clinical application in the future.


Subject(s)
Cartilage/physiology , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Regeneration , Transforming Growth Factor beta1/pharmacology , Animals , Cartilage/drug effects , Drug Liberation , Polyesters/chemistry , Polyethylene Glycols/chemistry , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta1/administration & dosage , Transforming Growth Factor beta1/pharmacokinetics
11.
Drug Des Devel Ther ; 10: 3043-3051, 2016.
Article in English | MEDLINE | ID: mdl-27703332

ABSTRACT

Bioactive mediators, cytokines, and chemokines have an important role in regulating and optimizing the synergistic action of materials, cells, and cellular microenvironments for tissue engineering. RADA self-assembling peptide hydrogels have been proved to have an excellent ability to promote cell proliferation, wound healing, tissue repair, and drug delivery. Here, we report that D-RADA16 and L-RADA16-RGD self-assembling peptides can form stable second structure and hydrogel scaffolds, affording the slow release of growth factor (transforming growth factor cytokine-beta 1 [TGF-beta 1]). In vitro tests demonstrated that the plateau release amount can be obtained till 72 hours. Moreover, L-RADA16, D-RADA16, and L-RADA16-RGD self-assembling peptide hydrogels containing TGF-beta 1 were used for 3D cell culture of bone mesenchymal stem cells of rats for 2 weeks. The results revealed that these three RADA16 peptide hydrogels had a significantly favorable influence on proliferation of bone mesenchymal stem cells and hold some promise in slow and sustained release of growth factor.


Subject(s)
Cytokines/chemistry , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Peptides/chemistry , Transforming Growth Factor beta1/chemistry , Transforming Growth Factor beta1/pharmacokinetics , Animals , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Delayed-Action Preparations , Peptides/metabolism , Rats
12.
J Tissue Eng Regen Med ; 10(12): 1041-1056, 2016 12.
Article in English | MEDLINE | ID: mdl-24737693

ABSTRACT

The degradation of elastic matrix in the infrarenal aortic wall is a critical parameter underlying the formation and progression of abdominal aortic aneurysms. It is mediated by the chronic overexpression of matrix metalloprotease (MMP)-2 and MMP-9, leading to a progressive loss of elasticity and weakening of the aortic wall. Delivery of therapeutic agents to inhibit MMPs, while concurrently coaxing cell-based regenerative repair of the elastic matrix represents a potential strategy for slowing or arresting abdominal aortic aneurysm growth. Previous studies have demonstrated elastogenic induction of healthy and aneurysmal aortic smooth muscle cells and inhibition of MMPs, following exogenous delivery of elastogenic factors such as transforming growth factor (TGF)-ß1, as well as MMP-inhibitors such as doxycycline (DOX) in two-dimensional culture. Based on these findings, and others that demonstrated elastogenic benefits of nanoparticulate delivery of these agents in two-dimensional culture, poly(lactide-co-glycolide) nanoparticles were developed for localized, controlled and sustained delivery of DOX and TGF-ß1 to human aortic smooth muscle cells within a three-dimensional gels of type I collagen, which closely simulate the arterial tissue microenvironment. DOX and TGF-ß1 released from these nanoparticles influenced elastogenic outcomes positively within the collagen constructs over 21 days of culture, which were comparable to that induced by exogenous supplementation of DOX and TGF-ß1 within the culture medium. However, this was accomplished at doses ~20-fold lower than the exogenous dosages of the agents, illustrating that their localized, controlled and sustained delivery from nanoparticles embedded within a three-dimensional scaffold is an efficient strategy for directed elastogenesis. Copyright © 2014 John Wiley & Sons, Ltd.


Subject(s)
Aorta/metabolism , Doxycycline , Drug Delivery Systems/methods , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Polyglactin 910/chemistry , Transforming Growth Factor beta1 , Aorta/cytology , Doxycycline/chemistry , Doxycycline/pharmacokinetics , Doxycycline/pharmacology , Humans , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Transforming Growth Factor beta1/chemistry , Transforming Growth Factor beta1/pharmacokinetics , Transforming Growth Factor beta1/pharmacology
13.
IET Nanobiotechnol ; 9(2): 76-84, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25829173

ABSTRACT

The study was aimed at evaluating the effect of electrospun scaffold containing TGF-ß1 on promoting human mesenchymal stem cells (MSCs) differentiation towards a nucleus pulposus-like phenotype under hypoxia. Two kinds of nanofibrous scaffolds containing TGF-ß1 were fabricated using uniaxial electrospinning (Group I) and coaxial electrospinning (Group II). Human MSCs were seeded on both kinds of scaffolds and cultured in a hypoxia chamber (2% O2), and then the scaffolds were characterised. Cell proliferation and differentiation were also evaluated after 3 weeks of cell culture. Results showed that both kinds of scaffolds shared similar diameter distributions and protein release. However, Group I scaffolds were more hydrophilic than that of Group II. Both kinds of scaffolds induced the MSCs to differentiate towards the nucleus pulposus-type phenotype in vitro. In addition, the expression of nucleus pulposus-associated genes (aggrecan, type II collagen, HIF-1α and Sox-9) in Group I increased more than that of Group II. These results indicate that electrospinning nanofibrous scaffolds containing TGF-ß1 supports the differentiation of MSCs towards the pulposus-like phenotype in a hypoxia chamber, which would be a more appropriate choice for nucleus pulposus regeneration.


Subject(s)
Cell Hypoxia/physiology , Cell Physiological Phenomena/physiology , Mesenchymal Stem Cells/cytology , Tissue Scaffolds/chemistry , Transforming Growth Factor beta1/chemistry , Cells, Cultured , Electrochemical Techniques , Humans , Nanofibers/chemistry , Nanotechnology , Phenotype , Transforming Growth Factor beta1/pharmacokinetics
14.
Asian Pac J Trop Med ; 7(3): 241-3, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24507648

ABSTRACT

OBJECTIVE: To observe the influence of the the transforming growth factor ß1 (TGF-ß1) eye drops on rabbit aqueous humor TGF-ß1 concentration, and to analyze the best drug concentration. METHODS: A total of 30 New Zealand white rabbits were randomly divided into 5 groups with 6 in each. Rabits in control group had PBS eye drops, group A, B, C, D adopted TGF-ß1 eye drops at 0.5, 1.0, 2.0, 4.0 mg/L, respectively, 4 times a day. Aqueous humor of right eye was extracted 1 week after administration to detect concentration changes of TGF-ß1 by ELISA; rabbits in fpur hroups adopted 2.0 mg/L eye drops to left eyes 4 times a day, 0.2 mL aqueous humor was extracted left eye at the scheduled time point 0, 30 min, 2 h, 4 h, 24 h for testing, the slit lamp was used to observe the cornea, chamber and lens. RESULTS: No obvious pathological changes in conjunctiva, cornea, rabbit conjunctival, anterior chamber, and the lens was found. Concentration of TGF-ß1 in rabbit aqueous humor in C, D group was significantly higher than the control group (P<0.05). CONCLUSIONS: TGF-ß1 eye drops at 2.0 mg/L, 4.0 mg/L can significantly increase concentration of TGF-ß1 in rabbit aqueous humor, withe good ocular surfac permeability.


Subject(s)
Aqueous Humor/chemistry , Transforming Growth Factor beta1/analysis , Animals , Aqueous Humor/metabolism , Cornea/chemistry , Cornea/metabolism , Diagnostic Techniques, Ophthalmological , Ophthalmic Solutions/administration & dosage , Rabbits , Random Allocation , Transforming Growth Factor beta1/administration & dosage , Transforming Growth Factor beta1/pharmacokinetics
15.
Ultrasound Med Biol ; 40(4): 765-74, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24433746

ABSTRACT

Unlike lipid-shelled microbubbles (MBs), albumin-shelled microbubbles (MBs) have not been reported to be actively targeted to cells without the assistance of antibodies. Recent studies indicate that the albumin molecule is similar to transforming growth factor ß (TGF-ß) both structurally and functionally. The TGF-ß superfamily is important during early tumor outgrowth, with an elevated TGF-ß being tumor suppressive; at later stages, this switches to malignant conversion and progression, including breast cancer. TGF-ß receptors I and II play crucial roles in both the binding and endocytosis of albumin. However, until now, no specific albumin receptor has been found. On the basis of the above-mentioned information, we hypothesized that non-antibody-conjugated albumin-shelled MBs can be used to deliver drugs to breast cancer cells. We also studied the possible roles of TGF-ß1 and radiation force in the behavior of cells and albumin-shelled MBs. The results indicate that albumin-shelled MBs loaded with paclitaxel (PTX) induce breast cancer cell apoptosis without the specific targeting produced by an antibody. Applying either an acoustic radiation force or cavitation alone to cells with PTX-loaded albumin MBs increased the apoptosis rate to 23.2% and 26.3% (p < 0.05), respectively. We also found that albumin-shelled MBs can enter MDA-MB-231 breast cancer cells and remain there for at least 24 h, even in the presence of PTX loading. Confocal micrographs revealed that 70.5% of the breast cancer cells took up albumin-shelled MBs spontaneously after 1 d of incubation. Applying an acoustic radiation force further increased the percentage to 91.9% in our experiments. However, this process could be blocked by TGF-ß1, even with subsequent exposure to the radiation force. From these results, we conclude that TGF-ß1 receptors are involved in the endocytotic process by which albumin-shelled MBs enter breast cancer cells. The acoustic radiation force increases the contact rate between albumin-shelled MBs and tumor cells. Combining a radiation force and cavitation yields an apoptosis rate of 31.3%. This in vitro study found that non-antibody-conjugated albumin-shelled MBs provide a useful method of drug delivery. Further in vivo studies of the roles of albumin MBs and TGF-ß in different stages of cancer are necessary.


Subject(s)
Albumins/pharmacokinetics , Breast Neoplasms/drug therapy , Capsules/pharmacokinetics , Capsules/radiation effects , Paclitaxel/administration & dosage , Sonication/methods , Transforming Growth Factor beta1/pharmacokinetics , Albumins/radiation effects , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Capsules/therapeutic use , Cell Line, Tumor , High-Energy Shock Waves , Humans , Transforming Growth Factor beta1/radiation effects , Treatment Outcome
16.
J Mater Sci Mater Med ; 24(2): 503-13, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23160914

ABSTRACT

A three-dimensional (3D) scaffolding system for chondrocytes culture has been produced by agglomeration of cells and gelatin microparticles with a mild centrifuging process. The diameter of the microparticles, around 10 µ, was selected to be in the order of magnitude of the chondrocytes. No gel was used to stabilize the construct that maintained consistency just because of cell and extracellular matrix (ECM) adhesion to the substrate. In one series of samples the microparticles were charged with transforming growth factor, TGF-ß1. The kinetics of growth factor delivery was assessed. The initial delivery was approximately 48 % of the total amount delivered up to day 14. Chondrocytes that had been previously expanded in monolayer culture, and thus dedifferentiated, adopted in this 3D environment a round morphology, both with presence or absence of growth factor delivery, with production of ECM that intermingles with gelatin particles. The pellet was stable from the first day of culture. Cell viability was assessed by MTS assay, showing higher absorption values in the cell/unloaded gelatin microparticle pellets than in cell pellets up to day 7. Nevertheless the absorption drops in the following culture times. On the contrary the cell viability of cell/TGF-ß1 loaded gelatin microparticle pellets was constant during the 21 days of culture. The formation of actin stress fibres in the cytoskeleton and type I collagen expression was significantly reduced in both cell/gelatin microparticle pellets (with and without TGF-ß1) with respect to cell pellet controls. Total type II collagen and sulphated glycosaminoglycans quantification show an enhancement of the production of ECM when TGF-ß1 is delivered, as expected because this growth factor stimulate the chondrocyte proliferation and improve the functionality of the tissue.


Subject(s)
Cartilage , Gelatin/chemistry , Tissue Engineering , Tissue Scaffolds/chemistry , Capsules/chemistry , Capsules/pharmacology , Cartilage/cytology , Cartilage/drug effects , Cartilage/physiology , Cell Culture Techniques , Cell Survival/drug effects , Cells, Cultured , Chemical Precipitation , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/physiology , Gelatin/pharmacology , Humans , Microspheres , Particle Size , Tissue Engineering/instrumentation , Tissue Engineering/methods , Transforming Growth Factor beta1/administration & dosage , Transforming Growth Factor beta1/pharmacokinetics , Water/metabolism
17.
Rev. esp. patol. torac ; 24(3): 257-261, jul.-sept. 2012. ilus, tab
Article in Spanish | IBECS | ID: ibc-106178

ABSTRACT

Objetivo: Establecer cuál es la dosis de TGF-β1 más adecuada para la estimulación de cultivos de fibroblastos pulmonares humanos y el tiempo necesario de incubación de los mismos para obtener la máxima respuesta. Material y métodos: Diseñamos un estudio de dosis respuesta con TGF-β1 sobre una línea celular de fibroblastos pulmonares humanos. Analizamos la producción de factor básico de crecimiento de fibroblastos (b-FGF) como marcador de estímulo fibrogénico. Para ello se cultivaron fibroblastos humanos procedentes de una línea celular (MRC5) obtenida de la ECCC (European Collection of Cell Culture, UK). Las células se cultivaron en placas de 33cm2, cuando estuvieron confluentes se les estimuló con diferentes dosis de TGF-β1 (Peprotech, USA): 5, 10 ng/ml. Las células se incubaron durante 12, 24, 48 y 72 horas. Se usaron como control células no estimuladas con TGF-β1. Los niveles de factor básico de crecimiento de fibroblastos (b-FGF) se midieron por ELISA (R&D System, Minneapolis, MN). Se analizó la viabilidad celular mediante Azul Trypan a las 24, 48 y 72 horas de la estimulación con TGF-β1. Resultados: La mayor producción de b-FGF se produjo a las 24 horas tras la estimulación con la dosis de 10 ng/ml de TGF-β1, siendo la producción de b-FGF igual a 501 pg/ml. La viabilidad celular tiene su mayor valor a las 48 horas, disminuyendo en horas sucesivas, alcanzando los niveles más bajos a las 72 horas. Conclusiones: Existe un efecto estimulador del TGF-β1 sobre los fibroblastos pulmonares humanos in vitro. Esta acción del TGF-β1 es dosis dependiente y alcanza el nivel máximo de proliferación con la dosis de 10 ng/ml a las 24 horas de su tratamiento (AU)


Objective: To establish the most appropriate dose of TGF-β1 to stimulate human lung fibroblasts cultures and their necessary incubation time to obtain the maximum response. Material and methods: A dose response study was designed with TGF - β1 based on human lung fibroblast cells. The production of basic fibroblast growth factor (b-FGF) was analyzed as a marker for fibrogenic stimulus. Human fibroblasts from a cell line (MRC5) were obtained from the ECCC (European Collection of Cell Culture, UK) and cultivated. Cells were cultured on 33 cm2 plates; once confluent, they were stimulated with various doses of TGF - β1 (Peprotech, USA): 5, 10 ng/ml. The cells were incubated for 12, 24, 48 and 72 hours. Cells not stimulated with TGF - β1 were used as a control. The levels of basic fibroblast growth factor (b-FGF) were measured using ELISA (R&D System, Minneapolis, MN). Cell viability was analyzed using Trypan Blue at 24, 48 and 72 hours following the stimulation with TGF - β1. Results: The greatest production of b-FGF took place 24 hours after the stimulation with the dose of 10ng/ml of TGF - β1, with the production of b-FGF being equal to 501 pg/ml. The cell viability reached its greatest value at the 48 hours, decreasing in the hours thereafter, to reach the lowest levels at 72 hours. Conclusions: TGF-β1 has a stimulating effect on human lugn fibroblasts in vitro. This action of the TGF - β1 is dose dependent and reaches maximum proliferation levels with a dose of 10ng/ml 24 hours following treatment (AU)


Subject(s)
Humans , Fibroblast Growth Factor 2 , Transforming Growth Factor beta1/pharmacokinetics , Idiopathic Pulmonary Fibrosis/drug therapy , Dose-Response Relationship, Drug
18.
Injury ; 43(3): 334-42, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22035848

ABSTRACT

Bone regeneration is a complex process that involves multiple cell types, growth factors (GFs) and cytokines. A synergistic contribution of various GFs and a crosstalk between their signalling pathways was suggested as determinative for the overall osteogenic outcome. The purpose of this work was to develop a brushite-PLGA system, which controls the release rate of the integrated growth factors (GFs) to enhance bone formation. The brushite cement implants were prepared by mixing a phosphate solid phase with an acid liquid phase. PDGF (250 ng) and TGF-ß1 (100 ng) were incorporated into the liquid phase. PLGA microsphere-encapsulated VEGF (350 ng) was pre-blended with the solid phase. VEGF, PDGF and TGF-ß1 release kinetics and tissue distributions were determined using iodinated ((125)I) GFs. In vivo results showed that PDGF and TGF-ß1 were delivered more rapidly from these systems implanted in an intramedullary defect in rabbit femurs than VEGF. The three GFs released from the brushite-PLGA system remained located around the implantation site (5 cm) with negligible systemic exposure. Bone peak concentrations of approximately 4 ng/g and 1.5 ng/g of PDGF and TGF-ß1, respectively were achieved on day 3. Thereafter, PDGF and TGF-ß1 concentrations stayed above 1 ng/g during the first week. The scaffolds also provided a VEGF peak concentration of nearly 6 ng/g on day 7 and a local concentration of approximately 1.5 ng/g during at least 4 weeks. Four weeks post implantation bone formation was considerably enhanced with the brushite-PLGA system loaded with each of the three GFs separately as well as with the combination of PDGF and VEGF. The addition of TGF-ß1 did not further improve the outcome. In conclusion, the herein presented brushite-PLGA system effectively controlled the release kinetics and localisation of the three GFs within the defect site resulting in markedly enhanced bone regeneration.


Subject(s)
Bone Regeneration/drug effects , Calcium Phosphates/pharmacology , Femoral Fractures/drug therapy , Lactic Acid/pharmacology , Platelet-Derived Growth Factor/pharmacokinetics , Polyglycolic Acid/pharmacology , Transforming Growth Factor beta1/pharmacokinetics , Vascular Endothelial Growth Factor A/pharmacokinetics , Animals , Bone Cements/pharmacology , Delayed-Action Preparations/pharmacokinetics , Male , Microspheres , Polylactic Acid-Polyglycolic Acid Copolymer , Rabbits , Tissue Scaffolds
19.
J Biomed Nanotechnol ; 6(2): 106-16, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20738064

ABSTRACT

The purpose of this work is to produce microspheres loaded with transforming growth factor beta1 TGFbeta1 and basic fibroblast growth factor FGF-2; to ensure the protein protection from degradation during the encapsulation and storage steps, to evaluate the release rate and the microspheres toxicity. The water in oil in water double emulsion technique was adapted to avoid the protein degradation during the encapsulation. The obtained microspheres were deeply characterized to evaluate their size, morphology, toxicity, the way of degradation, the protein stability and release rate. The microspheres were found to be biocompatible and the encapsulation efficiency was about 35%. It was observed that the obtained microspheres increase the shelf life of the growth factors. The diffusion coefficient was quantified using Fick's law of diffusion that was combined to an empirical equation representing the decrease in the protein stability. Such modelling helped to give indirect information about the microspheres morphology and drug distribution within the microspheres. The main conclusion consists of the formation of a higher compact polymer matrix when smaller particles are produced, which has different distinct effects: the encapsulation efficiency and the stability of the encapsulated growth factor are enhanced while both the growth factor diffusion and the polymer degradation rates decrease.


Subject(s)
Drug Delivery Systems/methods , Fibroblast Growth Factor 2/pharmacokinetics , Microspheres , Transforming Growth Factor beta1/pharmacokinetics , Fibroblast Growth Factor 2/chemistry , Fluorescein-5-isothiocyanate/analogs & derivatives , Humans , Lactic Acid , Methylene Chloride , Microscopy, Confocal , Microscopy, Electron, Scanning , Particle Size , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Protein Stability , Serum Albumin, Bovine , Surface Properties , Transforming Growth Factor beta1/chemistry
20.
Biomaterials ; 29(10): 1518-25, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18166223

ABSTRACT

Growth factors have become an important component for tissue engineering and regenerative medicine. Insulin-like growth factor-I (IGF-I) and transforming growth factor-beta1 (TGF-beta 1) in particular have great significance in cartilage tissue engineering. Here, we describe sequential release of IGF-I and TGF-beta 1 from modular designed poly(l,d-lactic-co-glycolic acid) (PLGA) scaffolds. Growth factors were encapsulated in PLGA microspheres using spontaneous emulsion, and in vitro release kinetics was characterized by ELISA. Incorporating BSA in the IGF-I formulations decreased the initial burst from 80% to 20%, while using uncapped PLGA rather than capped decreased the initial burst of TGF-beta 1 from 60% to 0% upon hydration. The bioactivity of released IGF-I and TGF-beta 1 was determined using MCF-7 proliferation assay and HT-2 inhibition assay, respectively. Both growth factors were released for up to 70 days in bioactive form. Scaffolds were fabricated by fusing bioactive IGF-I and TGF-beta 1 microspheres with dichloromethane vapor. Three scaffolds with tailored release kinetics were fabricated: IGF-I and TGF-beta 1 released continuously, TGF-beta 1 with IGF-I released sequentially after 10 days, and IGF-I with TGF-beta 1 released sequentially after 7 days. Scaffold swelling and degradation were characterized, indicating a peak swelling ratio of 4 after 7 days of incubation and showing 50% mass loss after 28 days, both consistent with scaffold release kinetics. The ability of these scaffolds to release IGF-I and TGF-beta 1 sequentially makes them very useful for cartilage tissue engineering applications.


Subject(s)
Insulin-Like Growth Factor I/chemistry , Lactic Acid/chemistry , Microspheres , Polyglycolic Acid/chemistry , Transforming Growth Factor beta1/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatography, Gel , Drug Carriers , Enzyme-Linked Immunosorbent Assay , Humans , Insulin-Like Growth Factor I/pharmacokinetics , Insulin-Like Growth Factor I/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer , Transforming Growth Factor beta1/pharmacokinetics , Transforming Growth Factor beta1/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL