Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 52
1.
Biomater Sci ; 12(2): 479-494, 2024 Jan 16.
Article En | MEDLINE | ID: mdl-38090986

Cartilage defects can be difficult to heal, potentially leading to complications such as osteoarthritis. Recently, a tissue engineering approach that uses scaffolds and growth factors has been proposed to regenerate new cartilage tissues. Herein, we investigated the application of hyaluronic acid (HA) gel loaded with transforming growth factor-beta 3 (TGF-ß3) for enhanced cartilage regeneration. We assessed the clinical conditions required to efficiently enhance the ability of the modified HA gel to repair defective cartilage. Based on our findings, the prepared HA gel exhibited good physicochemical and mechanical properties and was non-toxic and non-inflammatory. Moreover, HA gel-loaded TGF-ß3 (HAT) had improved biocompatibility and promoted the synthesis of cartilage-specific matrix and collagen, further improving its ability to repair defects. The application of HAT resulted in an initial burst release of HA, which degraded slowly in vivo. Finally, HAT combined with microfracture-inducing bone marrow stem cells could significantly improve the cartilage microenvironment and regeneration of cartilage defects. Our results indicate that HA is a suitable material for developing growth factor carriers, whereas HAT is a promising candidate for cartilage regeneration. Furthermore, this differentiated strategy provides a rapid and effective clinical approach for next-generation cartilage regeneration.


Hyaluronic Acid , Mesenchymal Stem Cells , Hyaluronic Acid/chemistry , Transforming Growth Factor beta3/chemistry , Hydrogels/chemistry , Cartilage/metabolism , Transforming Growth Factors/metabolism , Transforming Growth Factors/pharmacology
2.
Sci Rep ; 11(1): 22365, 2021 11 16.
Article En | MEDLINE | ID: mdl-34785671

Antibody function is typically entirely dictated by the Complementarity Determining Regions (CDRs) that directly bind to the antigen, while the framework region acts as a scaffold for the CDRs and maintains overall structure of the variable domain. We recently reported that the rabbit monoclonal antibody 4A11 (rbt4A11) disrupts signaling through both TGFß2 and TGFß3 (Sun et al. in Sci Transl Med, 2021. https://doi.org/10.1126/scitranslmed.abe0407 ). Here, we report a dramatic, unexpected discovery during the humanization of rbt4A11 where, two variants of humanized 4A11 (h4A11), v2 and v7 had identical CDRs, maintained high affinity binding to TGFß2/3, yet exhibited distinct differences in activity. While h4A11.v7 completely inhibited TGFß2/3 signaling like rbt4A11, h4A11.v2 did not. We solved crystal structures of TGFß2 complexed with Fab fragments of h4A11.v2 or h4A11.v7 and identified a novel interaction between the two heavy chain molecules in the 2:2 TGFb2:h4A11.v2-Fab complex. Further characterization revealed that framework residue variations at either position 19, 79 or 81 (Kabat numbering) of the heavy chain strikingly converts h4A11.v2 into an inhibitory antibody. Our work suggests that in addition to CDRs, framework residues and interactions between Fabs in an antibody could be engineered to further modulate activity of antibodies.


Amino Acid Substitution , Antibodies, Monoclonal, Humanized/chemistry , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Variable Region/chemistry , Transforming Growth Factor beta2/chemistry , Transforming Growth Factor beta3/chemistry , Animals , Antibodies, Monoclonal, Humanized/genetics , Crystallography, X-Ray , Humans , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Variable Region/genetics , Protein Structure, Quaternary , Rabbits , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta3/genetics
3.
Int J Biol Macromol ; 172: 381-393, 2021 Mar 01.
Article En | MEDLINE | ID: mdl-33476613

Current implantable materials are limited in terms of function as native tissue, and there is still no effective clinical treatment to restore articular impairments. Hereby, a functionalized polyacrylamide (PAAm)-alginate (Alg) Double Network (DN) hydrogel acting as an articular-like tissue is developed. These hydrogels sustain their mechanical stability under different temperature (+4 °C, 25 °C, 40 °C) and humidity conditions (60% and 75%) over 3 months. As for the functionalization, transforming growth factor beta-3 (TGF-ß3) encapsulated (NPTGF-ß3) and empty poly(lactide-co-glycolide) (PLGA) nanoparticles (PLGA NPs) are synthesized by using microfluidic platform, wherein the mean particle sizes are determined as 81.44 ± 9.2 nm and 126 ± 4.52 nm with very low polydispersity indexes (PDI) of 0.194 and 0.137, respectively. Functionalization process of PAAm-Alg hydrogels with ester-end PLGA NPs is confirmed by FTIR analysis, and higher viscoelasticity is obtained for functionalized hydrogels. Moreover, cartilage regeneration capability of these hydrogels is evaluated with in vitro and in vivo experiments. Compared with the PAAm-Alg hydrogels, functionalized formulations exhibit a better cell viability. Histological staining, and score distribution confirmed that proposed hydrogels significantly enhance regeneration of cartilage in rats due to stable hydrogel matrix and controlled release of TGF-ß3. These findings demonstrated that PAAm-Alg hydrogels showed potential for cartilage repair and clinical application.


Acrylic Resins/chemistry , Alginates/chemistry , Biocompatible Materials/chemistry , Cartilage, Articular/drug effects , Hydrogels/chemistry , Nanoparticles/chemistry , Transforming Growth Factor beta3/pharmacokinetics , Absorbable Implants , Animals , Biocompatible Materials/pharmacology , Cartilage, Articular/growth & development , Cartilage, Articular/injuries , Cell Survival/drug effects , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/physiology , Drug Compounding/methods , Hindlimb/drug effects , Male , Nanoparticles/ultrastructure , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta3/chemistry , Transforming Growth Factor beta3/metabolism , Treatment Outcome
4.
Int J Biol Macromol ; 167: 787-795, 2021 Jan 15.
Article En | MEDLINE | ID: mdl-33278443

Transforming growth factor beta 3 (TGFß3) exhibits a complex native structure featuring the presence of multiple disulfide bonds forming the active dimer. Consequently, its heterologous expression in microbial system invariably leads to inclusion body (IB) formation. In this study, we observed an interesting phenomenon of switching a significant fraction of misfolded TGFß3 to folded form by modulating the cellular protein folding machinery. We carried out co-expression experiments with chaperones and demonstrated the requirement of a coordinated action of DnaK-DnaJ-GrpE and GroESL, to achieve the native soluble conformation of TGFß3, during over-expression in E. coli. The novelty of this study lies in the fact that orchestration of a group of chaperones to work in concert for efficient folding and assembly of TGFß3-like cytokines has not been widely explored. Additionally, we have also demonstrated that presence of osmolytes (sorbitol or trehalose) in the growth media have an appreciable impact on the solubility of TGFß3. We have further shown a synergism between the effects of molecular chaperone and osmolytes on the solubility of TGFß3. We have confirmed the functionality of soluble TGFß3 by performing binding interactions with its cognate receptor TßRII. Our study delineates the fact that an effective combination of chaperones or optimum concentration of compatible osmolyte, can efficiently abrogate competing aggregation pathways and help attain the native conformation of a cysteine rich cytokine in a facile manner.


Escherichia coli/genetics , Gene Expression , Protein Engineering , Transforming Growth Factor beta3/chemistry , Transforming Growth Factor beta3/genetics , Disulfides/chemistry , Escherichia coli/metabolism , Models, Molecular , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Protein Conformation , Protein Folding , Recombinant Proteins , Solubility , Solvents , Structure-Activity Relationship , Transforming Growth Factor beta3/biosynthesis , Transforming Growth Factor beta3/isolation & purification
5.
Regen Med ; 15(6): 1735-1747, 2020 06.
Article En | MEDLINE | ID: mdl-32811280

Aim: We aimed to evaluate the capacity of the bilayer polylactic-co-glycolic acid (PLGA)/TGF-ß3/adipose-derived mesenchymal stem cell (ADSC) construct used to repair cartilage defects and the role of ADSCs in the repair process in vivo. Materials & methods: Defects were created surgically on the femoropatellar groove of knee joints in 64 rabbits. All the rabbits were randomly divided into four groups: defect group, PLGA group, PLGA/TGF-ß3 group and PLGA/TGF-ß3/ADSC group. In vivo MRI and Prussian blue staining were applied. Quantitative real-time PCR and western blot methods were used to analyze the gene and protein expression. Results & conclusion: The result showed that TGF-ß3 could effectively stimulate the expressions of aggrecan, collagen type II and SRY-related HMG box 9 (SOX9). The bilayer PLGA/TGF-ß3/ADSC construct showed a promising repair effect.


Cartilage Diseases/therapy , Cartilage, Articular/physiology , Magnetic Iron Oxide Nanoparticles/chemistry , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Transforming Growth Factor beta3/metabolism , Animals , Cartilage, Articular/injuries , Cell Differentiation , Cells, Cultured , Lipid Bilayers/chemistry , Rabbits , Tissue Engineering , Tissue Scaffolds/chemistry , Transforming Growth Factor beta3/chemistry
6.
Colloids Surf B Biointerfaces ; 189: 110843, 2020 May.
Article En | MEDLINE | ID: mdl-32044676

Medical treatment of certain diseases and biomedical implants are tending to use delivery systems on the nanoscale basis for biologically active factors including drugs (e. g. antibiotics) or growth factors. Nanoparticles are a useful tool to deliver bioactive substances of different chemical nature directly to the site where it is required in the patient. Here we developed three innovative delivery systems based on different polysaccharides in order to induce a sustained release of TGF-ß3 to mediate chondrogenesis of human mesenchymal stromal cells. We were able to encapsulate the protein into nanoparticles and subsequently release TGF-ß3 from these particles. The protein was still active and was able to induce chondrogenic differentiation of human mesenchymal stromal cells.


Alginates/chemistry , Chitosan/chemistry , Chondrogenesis/drug effects , Nanoparticles/chemistry , Polyphosphates/chemistry , Transforming Growth Factor beta3/pharmacology , Cell Differentiation/drug effects , Drug Delivery Systems , Humans , Mesenchymal Stem Cells/drug effects , Particle Size , Surface Properties , Transforming Growth Factor beta3/chemistry
7.
Carbohydr Polym ; 229: 115551, 2020 Feb 01.
Article En | MEDLINE | ID: mdl-31826469

The field of cartilage tissue engineering has been evolved in the last decade and a myriad of scaffolding biomaterials and bioactive agents have been proposed. Controlled release of growth factors encapsulated in the polymeric nanomaterials has been of interest notably for the repair of damaged articular cartilage. Here, we proposed an on-chip hydrodynamic flow focusing microfluidic approach for synthesis of alginate nanogels loaded with the transforming growth factor beta 3 (TGF-ß3) through an ionic gelation method in order to achieve precise release profile of these bioactive agents during chondrogenic differentiation of mesenchymal stem cells (MSCs). Alginate nanogels with adjustable sizes were synthesized by fine-tuning the flow rate ratio (FRR) in the microfluidic device consisting of cross-junction microchannels. The result of present study showed that the proposed approach can be a promising tool to synthesize bioactive -loaded polymeric nanogels for applications in drug delivery and tissue engineering.


Alginates/chemistry , Microfluidics , Nanogels/chemistry , Transforming Growth Factor beta3/chemistry , Adult , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrogenesis/drug effects , Humans , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Nanogels/toxicity , Particle Size , Transforming Growth Factor beta3/metabolism , Transforming Growth Factor beta3/pharmacology
8.
Int J Mol Sci ; 20(20)2019 Oct 09.
Article En | MEDLINE | ID: mdl-31600954

Periodontal disease is the main reason for tooth loss in adults. Tissue engineering and regenerative medicine are advanced technologies used to manage soft and hard tissue defects caused by periodontal disease. We developed a transforming growth factor-ß3/chitosan sponge (TGF-ß3/CS) to repair periodontal soft and hard tissue defects. We investigated the proliferation and osteogenic differentiation behaviors of primary human periodontal ligament stem cells (hPDLSCs) to determine the bioactivity and potential application of TGF-ß3 in periodontal disease. We employed calcein-AM/propidium iodide (PI) double labeling or cell membranes (CM)-Dil labeling coupled with fluorescence microscopy to trace the survival and function of cells after implantation in vitro and in vivo. The mineralization of osteogenically differentiated hPDLSCs was confirmed by measuring alkaline phosphatase (ALP) activity and calcium content. The levels of COL I, ALP, TGF-ßRI, TGF-ßRII, and Pp38/t-p38 were assessed by western blotting to explore the mechanism of bone repair prompted by TGF-ß3. When hPDLSCs were implanted with various concentrations of TGF-ß3/CS (62.5-500 ng/mL), ALP activity was the highest in the TGF-ß3 (250 ng/mL) group after 7 d (p < 0.05 vs. control). The calcium content in each group was increased significantly after 21 and 28 d (p < 0.001 vs. control). The optimal result was achieved by the TGF-ß3 (500 ng/mL) group. These results showed that TGF-ß3/CS promotes osteogenic differentiation of hPDLSCs, which may involve the p38 mitogen-activated protein kinase (MAPK) signaling pathway. TGF-ß3/CS has the potential for application in the repair of incomplete alveolar bone defects.


Cell Differentiation/drug effects , Chitosan , Osteogenesis/drug effects , Periodontal Ligament/cytology , Stem Cells/drug effects , Transforming Growth Factor beta3/pharmacology , Biomarkers , Cell Proliferation/drug effects , Cells, Cultured , Chitosan/chemistry , Humans , Immunohistochemistry , MAP Kinase Signaling System , Stem Cells/cytology , Transforming Growth Factor beta3/chemistry
9.
Acta Biomater ; 90: 179-191, 2019 05.
Article En | MEDLINE | ID: mdl-30936036

Although there are numerous medical applications to recover damaged skin tissue, scarless wound healing is being extensively investigated to provide a better therapeutic outcome. The exogenous delivery of therapeutic growth factors (GFs) is one of the engineering strategies for skin regeneration. This study presents an exogenous GF delivery platform developed using coacervates (Coa), a tertiary complex of poly(ethylene argininyl aspartate diglyceride) (PEAD) polycation, heparin, and cargo GFs (i.e., transforming growth factor beta 3 (TGF-ß3) and interleukin 10 (IL-10)). Coa encompasses the advantage of high biocompatibility, facile preparation, protection of cargo GFs, and sustained GF release. We therefore speculated that coacervate-mediated dual delivery of TGF-ß3/IL-10 would exhibit synergistic effects for the reduction of scar formation during physiological wound healing. Our results indicate that the exogenous administration of dual GF via Coa enhances the proliferation and migration of skin-related cells. Gene expression profiles using RT-PCR revealed up-regulation of ECM formation at early stage of wound healing and down-regulation of scar-related genes at later stages. Furthermore, direct injection of the dual GF Coa into the edges of damaged skin in a rat skin wound defect model demonstrated accelerated wound closure and skin regeneration after 3 weeks. Histological evaluation and immunohistochemical staining also revealed enhanced formation of the epidermal layer along with facilitated angiogenesis following dual GF Coa delivery. Based on these results, we conclude that polycation-mediated Coa fabrication and exogenous dual GF delivery via the Coa platform effectively augments both the quantity and quality of regenerated skin tissues without scar formation. STATEMENT OF SIGNIFICANCE: This study was conducted to develop a simple administration platform for scarless skin regeneration using polycation-based coacervates with dual GFs. Both in vitro and in vivo studies were performed to confirm the therapeutic efficacy of this platform toward scarless wound healing. Our results demonstrate that the platform developed by us enhances the proliferation and migration of skin-related cells. Sequential modulation in various gene expression profiles suggests a balanced collagen-remodeling process by dual GFs. Furthermore, in vivo histological evaluation demonstrates that our technique enhances clear epidermis formation with less scab and thicker woven structure of collagen bundle, similar to that of a normal tissue. We propose that simple administration of dual GFs with Coa has the potential to be applied as a clinical approach for fundamental scarless skin regeneration.


Cicatrix/prevention & control , Dermis/metabolism , Drug Delivery Systems , Fibroblasts/metabolism , Interleukin-10 , Transforming Growth Factor beta3 , Wound Healing/drug effects , Cicatrix/metabolism , Cicatrix/pathology , Dermis/pathology , Fibroblasts/pathology , Humans , Interleukin-10/chemistry , Interleukin-10/pharmacokinetics , Interleukin-10/pharmacology , Transforming Growth Factor beta3/chemistry , Transforming Growth Factor beta3/pharmacokinetics , Transforming Growth Factor beta3/pharmacology
10.
Acta Biomater ; 93: 111-122, 2019 07 15.
Article En | MEDLINE | ID: mdl-30862549

Fibrocartilage is typically found in regions subject to complex, multi-axial loads and plays a critical role in musculoskeletal function. Mesenchymal stem cell (MSC)-mediated fibrocartilage regeneration may be guided by administration of appropriate chemical and/or physical cues, such as by culturing cells on polymer nanofibers in the presence of the chondrogenic growth factor TGF-ß3. However, targeted delivery and maintenance of effective local factor concentrations remain challenges for implementation of growth factor-based regeneration strategies in clinical settings. Thus, the objective of this study was to develop and optimize the bioactivity of a biomimetic nanofiber scaffold system that enables localized delivery of TGF-ß3. To this end, we fabricated TGF-ß3-releasing nanofiber meshes that provide sustained growth factor delivery and demonstrated their potential for guiding synovium-derived stem cell (SDSC)-mediated fibrocartilage regeneration. TGF-ß3 delivery enhanced cell proliferation and synthesis of relevant fibrocartilaginous matrix in a dose-dependent manner. By designing a scaffold that eliminates the need for exogenous or systemic growth factor administration and demonstrating that fibrochondrogenesis requires a lower growth factor dose compared to previously reported, this study represents a critical step towards developing a clinical solution for regeneration of fibrocartilaginous tissues. STATEMENT OF SIGNIFICANCE: Fibrocartilage is a tissue that plays a critical role throughout the musculoskeletal system. However, due to its limited self-healing capacity, there is a significant unmet clinical need for more effective approaches for fibrocartilage regeneration. We have developed a nanofiber-based scaffold that provides both the biomimetic physical cues, as well as localized delivery of the chemical factors needed to guide stem cell-mediated fibrocartilage formation. Specifically, methods for fabricating TGF-ß3-releasing nanofibers were optimized, and scaffold-mediated TGF-ß3 delivery enhanced cell proliferation and synthesis of fibrocartilaginous matrix, demonstrating for the first time, the potential for nanofiber-based TGF-ß3 delivery to guide stem cell-mediated fibrocartilage regeneration. This nanoscale delivery platform represents an exciting new strategy for fibrocartilage regeneration.


Drug Carriers/chemistry , Fibrocartilage/drug effects , Nanofibers/chemistry , Tissue Scaffolds/chemistry , Transforming Growth Factor beta3/chemistry , Animals , Cattle , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chondrogenesis , Collagen/chemistry , Drug Liberation , Extracellular Matrix/metabolism , Humans , Knee Joint/cytology , Mesenchymal Stem Cells/drug effects , Polyesters/chemistry , Polyglactin 910/chemistry , Proteoglycans/chemistry , Regeneration , Surface Properties , Tissue Engineering , Transforming Growth Factor beta3/pharmacokinetics
11.
J Biomater Sci Polym Ed ; 30(5): 415-436, 2019 04.
Article En | MEDLINE | ID: mdl-30688157

In the present study a combination of Transforming Growth Factor Beta 3 (TGF-ß3) and Bone Morphogenetic Protein-2 (BMP-2) loaded gelatin films sandwiched between poly (L-lactide) (PLLA)/poly (ε-caprolactone) (PCL) matrices were produced to enhance bone formation in alveolar bone defects. Osteogenic properties of tissue constructs were tested in alveolar bone defect model in rats. Bone healing was assessed by osteogenic gene expression levels of bone sialoprotein (BSP), alkaline phosphatase (ALP), osteonectin (ON, SPARC), osteocalcin (OC), runt-related transcription factor 2 (RUNX2), bone specific alkaline phosphatase (BALP) activity, histomorphometry and microtomography. Increase in osteogenic gene expression levels and BALP activity results showed that new bone formation was significantly accelerated in TGF-ß3 + BMP-2 loaded scaffold group compared to growth factor free and only BMP-2 loaded groups. The micro-computed tomography (µ-CT) data from the 4th months revealed that (TGF-ß3+ BMP-2) loaded scaffolds displayed increased bone formation and was able to fulfill 84% of the defect area (p < 0.05). Accelerated bone formation in the S-GF-B-T group compared to that of the S-GF group at the end of the 4th month was further verified via histomorphometric analysis (p = 0.008). Gene expression, BALP activity, microtomography and histomorphometry analysis indicated that (TGF-ß3 + BMP-2) loaded PLLA/PCL scaffolds increased the new bone formation. BMP-2 loaded scaffolds were less effective than combination of TGF-ß3 and BMP-2 loaded scaffolds. These findings demonstrated that focusing on the PLLA/PCL hybrid scaffolds combined with (TGF-ß3 + BMP-2) may lay the groundwork for future therapy-oriented efforts to enhance bone formation in alveolar defects.


Alveolar Process/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Bone Morphogenetic Protein 2/chemistry , Drug Carriers/chemistry , Drug Liberation , Transforming Growth Factor beta3/chemistry , Alkaline Phosphatase/metabolism , Alveolar Process/diagnostic imaging , Alveolar Process/metabolism , Animals , Gelatin/chemistry , Humans , Male , Polyesters/chemistry , Rats , Rats, Wistar , X-Ray Microtomography
12.
Acta Biomater ; 83: 167-176, 2019 01 01.
Article En | MEDLINE | ID: mdl-30458242

Cell-loaded hydrogels are frequently applied in cartilage tissue engineering for their biocompatibility, ease of application, and ability to conform to various defect sites. As a bioactive adjunct to the biomaterial, transforming growth factor beta (TGF-ß) has been shown to be essential for cell differentiation into a chondrocyte phenotype and maintenance thereof, but the low amounts of endogenous TGF-ß in the in vivo joint microenvironment necessitate a mechanism for controlled delivery and release of this growth factor. In this study, TGF-ß3 was directly loaded with human bone marrow-derived mesenchymal stem cells (MSCs) into poly-d,l-lactic acid/polyethylene glycol/poly-d,l-lactic acid (PDLLA-PEG) hydrogel, or PDLLA-PEG with the addition of hyaluronic acid (PDLLA/HA), and cultured in vitro. We hypothesize that the inclusion of HA within PDLLA-PEG would result in a controlled release of the loaded TGF-ß3 and lead to a robust cartilage formation without the use of TGF-ß3 in the culture medium. ELISA analysis showed that TGF-ß3 release was effectively slowed by HA incorporation, and retention of TGF-ß3 in the PDLLA/HA scaffold was detected by immunohistochemistry for up to 3 weeks. By means of both in vitro culture and in vivo implantation, we found that sulfated glycosaminoglycan production was higher in PDLLA/HA groups with homogenous distribution throughout the scaffold than PDLLA groups. Finally, with an optimal loading of TGF-ß3 at 10 µg/mL, as determined by RT-PCR and glycosaminoglycan production, an almost twofold increase in Young's modulus of the construct was seen over a 4-week period compared to TGF-ß3 delivery in the culture medium. Taken together, our results indicate that the direct loading of TGF-ß3 and stem cells in PDLLA/HA has the potential to be a one-step point-of-care treatment for cartilage injury. STATEMENT OF SIGNIFICANCE: Stem cell-seeded hydrogels are commonly used in cell-based cartilage tissue engineering, but they generally fail to possess physiologically relevant mechanical properties suitable for loading. Moreover, degradation of the hydrogel in vivo with time further decreases mechanical suitability of the hydrogel due in part to the lack of TGF-ß3 signaling. In this study, we demonstrated that incorporation of hyaluronic acid (HA) into a physiologically stiff PDLLA-PEG hydrogel allowed for slow release of one-time preloaded TGF-ß3, and when loaded with adult mesenchymal stem cells and cultured in vitro, it resulted in higher chondrogenic gene expression and constructs of significantly higher mechanical strength than constructs cultured in conventional TGF-ß3-supplemented medium. Similar effects were also observed in constructs implanted in vivo. Our results indicate that direct loading of TGF-ß3 combined with HA in the physiologically stiff PDLLA-PEG hydrogel has the potential to be used for one-step point-of-care treatment of cartilage injury.


Cell Differentiation/drug effects , Chondrogenesis/drug effects , Hyaluronic Acid , Hydrogels , Mesenchymal Stem Cells/metabolism , Transforming Growth Factor beta3 , Cell Culture Techniques , Cells, Cultured , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacology , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Mesenchymal Stem Cells/classification , Time Factors , Transforming Growth Factor beta3/chemistry , Transforming Growth Factor beta3/pharmacology
13.
Eur J Pharm Sci ; 127: 225-232, 2019 Jan 15.
Article En | MEDLINE | ID: mdl-30423434

TGF-ß3, a subtype of transforming growth factor-ß (TGF-ß), is essential to various biological processes, including endoderm development, organogenesis, epithelial hyperplasia, synthesis of extracellular matrix, and immune response. Essentially, TGF-ß3 engages the TGF-ß1/Smad signaling pathway to stimulate mesenchymal lineage cells, inhibit epithelial or neuroectodermal lineage cells, and regulate repair, remodeling, and potential scarring after cutaneous wounding. We have now expressed recombinant human TGF-ß3 in Escherichia coli Origami B (DE3), with yield 300 ±â€¯17 mg/L monomeric protein at pilot scale. Identity was confirmed by western blot and HPLC-based peptide mapping. After purification and refolding, dimeric proteins were found to induce chondro-related genes in adipose-derived stem cells, and to suppress scarring in injured rabbit ears. Thus, the recombinant protein has excellent potential for medical applications.


Transforming Growth Factor beta3 , Wound Healing/drug effects , Adipose Tissue/cytology , Animals , Cells, Cultured , Escherichia coli/genetics , Female , Male , Protein Folding , Protein Multimerization , Rabbits , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Stem Cells/drug effects , Stem Cells/metabolism , Transforming Growth Factor beta3/chemistry , Transforming Growth Factor beta3/genetics , Transforming Growth Factor beta3/metabolism , Transforming Growth Factor beta3/pharmacology
14.
Biomater Sci ; 7(1): 233-246, 2018 Dec 18.
Article En | MEDLINE | ID: mdl-30511062

Nanogels were prepared by ionotropic gelation of chitosan (CS) with tripolyphosphate (TPP). The use of such nanogels to prepare coatings by layer-by-layer deposition (LbL) was studied. The nanogels were characterized in terms of particle size, zeta-potential and stability. Nanogel suspensions were used to build polyelectrolyte multilayers on silicon wafers and on PCL fiber mats by LbL-deposition. Three different polysaccharides were used as polyanions, namely chondroitin sulfate, alginate and hyaluronic acid. The ellipsometric thickness was demonstrated to depend significantly on the type of polyanion. XPS analysis with depth profiling further substantiated the differences in the chemical composition of the films with the different polyanions. Furthermore, XPS data clearly indicated a strong penetration of the polyanions into the CS-TPP layer, resulting in a complete exchange and release of the TPP ions. The LbL-deposition also was studied with PCL fiber mats, which were modified with a chitosan-PCL-graft polymer and alginate. The possibility to create graded coatings on the fiber mats was shown employing fluorescently labelled CS-TPP nanoparticles. The potential of the coatings as drug delivery system for therapeutic proteins was exemplified with the release of Transforming Growth Factor ß3 (TGF-ß3). The CS-TPP nanogels were shown to encapsulate and release therapeutic proteins. In combination with the layer-by-layer deposition they will allow the creation of PCL fiber mat implants having with drug gradients for applications at tissue transitions.


Chitosan/analogs & derivatives , Coated Materials, Biocompatible/chemistry , Drug Carriers/chemistry , Nanofibers/chemistry , Nanoparticles/chemistry , Polyesters/chemistry , Transforming Growth Factor beta3/administration & dosage , Alginates/chemistry , Chitosan/chemistry , Chondroitin Sulfates/chemistry , Drug Delivery Systems , Drug Liberation , Humans , Hyaluronic Acid/chemistry , Nanofibers/ultrastructure , Nanoparticles/ultrastructure , Transforming Growth Factor beta3/chemistry
15.
Artif Cells Nanomed Biotechnol ; 46(sup1): 985-995, 2018.
Article En | MEDLINE | ID: mdl-29448837

Long segment tracheal stenosis often has a poor prognosis due to the limited availability of materials for tracheal reconstruction. Tissue engineered tracheal patches based on electrospun scaffolds and stem cells present ideal solutions to this medical challenge. However, the established engineering process is inefficient and time-consuming. In our research, to optimize the engineering process, core-shell nanofilms encapsulating TGF-ß3 were fabricated as scaffolds for tracheal patches. The morphological and mechanical characteristics, degradation and biocompatibility of poly(l-lactic acid-co-ε-caprolactone)/collagen (PLCL/collagen) scaffolds with different compositions (PLCL:collagen 75:25, 50:50 and 25:75, respectively) were comparatively evaluated to determine the preferable compositional ratio. Then the chondrogenesis-inducing potential is investigated, and tracheal patches based on electrospun scaffolds and bone marrow mesenchymal stem cells (BMSCs) were constructed to restore tracheal defects in rabbit models. The results indicated that core-shell scaffolds with a PLCL/collagen proportion of 75:25 were eligible for tracheal patches. The stable and sustained release of TGF-ß3 from scaffolds could efficiently promote the chondrogenic differentiation of BMSCs and shorten the incubation time. Tracheal integrity was well maintained for 2 months after restoration; meanwhile, re-epithelialization also achieved. In conclusion, TGF-ß3-encapsulating core-shell electrospun scaffolds with a PLCL/collagen proportion of 75:25 could be used to optimize engineering process of tracheal patches.


Collagen/chemistry , Electricity , Polyesters/chemistry , Tissue Engineering , Tissue Scaffolds/chemistry , Trachea/drug effects , Transforming Growth Factor beta3/chemistry , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Capsules , Cell Differentiation/drug effects , Chondrogenesis/drug effects , Drug Liberation , Mechanical Phenomena , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Rabbits , Trachea/cytology
16.
Acta Biomater ; 64: 148-160, 2017 12.
Article En | MEDLINE | ID: mdl-29017973

The ideal tissue engineering (TE) strategy for ligament regeneration should recapitulate the bone - calcified cartilage - fibrocartilage - soft tissue interface. Aligned electrospun-fibers have been shown to guide the deposition of a highly organized extracellular matrix (ECM) necessary for ligament TE. However, recapitulating the different tissues observed in the bone-ligament interface using such constructs remains a challenge. This study aimed to explore how fiber alignment and growth factor stimulation interact to regulate the chondrogenic and ligamentous differentiation of mesenchymal stem cells (MSCs). To this end aligned and randomly-aligned electrospun microfibrillar scaffolds were seeded with bone marrow derived MSCs and stimulated with transforming growth factor ß3 (TGFß3) or connective tissue growth factor (CTGF), either individually or sequentially. Without growth factor stimulation, MSCs on aligned-microfibers showed higher levels of tenomodulin (TNMD) and aggrecan gene expression compared to MSCs on randomly-oriented fibers. MSCs on aligned-microfibers stimulated with TGFß3 formed cellular aggregates and underwent robust chondrogenesis, evidenced by increased type II collagen expression and sulphated glycosaminoglycans (sGAG) synthesis compared to MSCs on randomly-oriented scaffolds. Bone morphogenetic protein 2 (BMP2) and type I collagen gene expression were higher on randomly-oriented scaffolds stimulated with TGFß3, suggesting this substrate was more supportive of an endochondral phenotype. In the presence of CTGF, MSCs underwent ligamentous differentiation, with increased TNMD expression on aligned compared to randomly aligned scaffolds. Upon sequential growth factor stimulation, MSCs expressed types I and II collagen and deposited higher overall levels of collagen compared to scaffolds stimulated with either growth factor in isolation. These findings demonstrate that modulating the alignment of microfibrillar scaffolds can be used to promote either an endochondral, chondrogenic, fibrochondrogenic or ligamentous MSC phenotype upon presentation of appropriate biochemical cues. STATEMENT OF SIGNIFICANCE: Polymeric electrospun fibers can be tuned to match the fibrillar size and anisotropy of collagen fibers in ligaments, and can be mechanically competent. Therefore, their use is attractive when attempting to tissue engineer the bone-ligament interface. A central challenge in this field is recapitulating the cellular phenotypes observed across the bone-ligament interface. Here we demonstrated that it is possible to direct MSCs seeded onto aligned electrospun fibres towards either a ligamentogenic, chondrogenic or fibrochondrogenic phenotype upon presentation of appropriate biochemical cues. This opens the possibility of using aligned microfibrillar scaffolds that are spatially functionalized with specific growth factors to direct MSC differentiation for engineering the bone-ligament interface.


Cell Differentiation/drug effects , Connective Tissue Growth Factor , Extracellular Matrix/chemistry , Ligaments/metabolism , Mesenchymal Stem Cells/metabolism , Tissue Engineering/methods , Transforming Growth Factor beta3 , Animals , Antigens, Differentiation/biosynthesis , Cell Culture Techniques/methods , Connective Tissue Growth Factor/chemistry , Connective Tissue Growth Factor/pharmacology , Ligaments/cytology , Mesenchymal Stem Cells/cytology , Swine , Transforming Growth Factor beta3/chemistry , Transforming Growth Factor beta3/pharmacology
17.
J Biol Chem ; 292(44): 18091-18097, 2017 11 03.
Article En | MEDLINE | ID: mdl-28912269

Glycoprotein A repetitions predominant (GARP) (encoded by the Lrrc32 gene) plays important roles in cell-surface docking and activation of TGFß. However, GARP's role in organ development in mammalian systems is unclear. To determine the function of GARP in vivo, we generated a GARP KO mouse model. Unexpectedly, the GARP KO mice died within 24 h after birth and exhibited defective palatogenesis without apparent abnormalities in other major organs. Furthermore, we observed decreased apoptosis and SMAD2 phosphorylation in the medial edge epithelial cells of the palatal shelf of GARP KO embryos at embryonic day 14.5 (E14.5), indicating a defect in the TGFß signaling pathway in the GARP-null developing palates. Of note, the failure to develop the secondary palate and concurrent reduction of SMAD phosphorylation without other defects in GARP KO mice phenocopied TGFß3 KO mice, although GARP has not been suggested previously to interact with TGFß3. We found that GARP and TGFß3 co-localize in medial edge epithelial cells at E14.5. In vitro studies confirmed that GARP and TGFß3 directly interact and that GARP is indispensable for the surface expression of membrane-associated latent TGFß3. Our findings indicate that GARP is essential for normal morphogenesis of the palate and demonstrate that GARP plays a crucial role in regulating TGFß3 signaling during embryogenesis. In conclusion, we have uncovered a novel function of GARP in positively regulating TGFß3 activation and function.


Gene Expression Regulation, Developmental , Membrane Proteins/metabolism , Organogenesis , Palate/metabolism , Protein Processing, Post-Translational , Smad2 Protein/metabolism , Transforming Growth Factor beta3/agonists , Animals , Animals, Newborn , Apoptosis , Cleft Palate/embryology , Cleft Palate/metabolism , Cleft Palate/pathology , Embryo, Mammalian/abnormalities , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Female , Gene Knock-In Techniques , HEK293 Cells , Heterozygote , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice, Knockout , Palate/abnormalities , Palate/embryology , Palate/pathology , Phosphorylation , Pregnancy , Protein Multimerization , Protein Transport , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Signal Transduction , Transforming Growth Factor beta3/chemistry , Transforming Growth Factor beta3/genetics , Transforming Growth Factor beta3/metabolism
18.
J Phys Chem B ; 121(22): 5483-5498, 2017 06 08.
Article En | MEDLINE | ID: mdl-28497965

Transforming growth factors (TGF-ßs) are proteins that regulate cell growth by binding to their receptors. In contrast to transforming growth factor (TGF) ß1, TGF-ß3 homodimer is believed to exist also in an open conformation, in which both of its monomers are loosely packed against each other. At the origin of this difference is the H3-helix. Its sequence and degree of structuration seem to govern the outcome of TGF dimerization. We docked two monomers of TGF-ß3 with intact and altered H3 α-helix against each other using HADDOCK. TGF-ß3 monomer with an intact H3-helix exclusively forms closed conformations of homodimer, whereas the open conformation may coexist with the closed one when a part of the H3 α-helix is destabilized. We quantify the difference in its conformational preference for the open versus the closed structure by calculating the binding energy between monomers using the MMPBSA approach. We compare the wild type (wt) TGFß3/TGFß1 homodimers in the Protein Data Bank to a swapped mutant where all residues of the H3-helix were mutated to the respective TGFß1/TGFß3 sequence. Swapping stabilizes the closed conformation and destabilizes the open conformation of TGFß3. Further detailed insight is derived from molecular dynamics simulation studies suggesting that Val 61 of the H3-helix may act as an anchor residue for the closed conformation of TGFß3. Computational alanine scanning mutagenesis confirms that several residues of the H3-helix are the hot residues for the closed conformation of TGFß3. These observations may bear relevance to general conformational transitions in proteins and specifically in the TGFß superfamily.


Molecular Dynamics Simulation , Transforming Growth Factor beta3/chemistry , Protein Conformation, alpha-Helical , Transforming Growth Factor beta3/genetics
19.
Tissue Eng Part A ; 23(17-18): 1011-1021, 2017 09.
Article En | MEDLINE | ID: mdl-28285569

Articular cartilage has a limited capacity to heal after damage from injury or degenerative disease. Tissue engineering constructs that more closely mimic the native cartilage microenvironment can be utilized to promote repair. Glycosaminoglycans (GAGs), a major component of the cartilage extracellular matrix, have the ability to sequester growth factors due to their level and spatial distribution of sulfate groups. This study evaluated the use of a GAG mimetic, cellulose sulfate, as a scaffolding material for cartilage tissue engineering. Cellulose sulfate can be synthesized to have a similar level and spatial distribution of sulfates as chondroitin sulfate C (CSC), the naturally occurring GAG. This partially sulfated cellulose (pSC) was incorporated into a fibrous gelatin construct by the electrospinning process. Scaffolds were characterized for fiber morphology and overall stability over time in an aqueous environment, growth factor interaction, and for supporting mesenchymal stem cell (MSC) chondrogenesis in vitro. All scaffold groups had micron-sized fibers and maintained overall stability in aqueous environments. Increasing concentrations of the transforming growth factor-beta 3 (TGF-ß3) were detected on scaffolds with increasing pSC. MSC chondrogenesis was enhanced on the scaffold with the highest pSC concentration as seen with the highest collagen type II production, collagen type II immunostaining, expression of cartilage-specific genes, and ratio of collagen type II to collagen type I production. These studies demonstrated the potential of pSC sulfate as a scaffolding material for cartilage tissue engineering.


Cellulose/chemistry , Chondrogenesis , Gelatin/chemistry , Mesenchymal Stem Cells/metabolism , Tissue Scaffolds/chemistry , Adolescent , Adult , Female , Humans , Male , Mesenchymal Stem Cells/cytology , Transforming Growth Factor beta3/chemistry , Transforming Growth Factor beta3/pharmacology
20.
J Mater Sci Mater Med ; 28(1): 22, 2017 Jan.
Article En | MEDLINE | ID: mdl-28025802

The aim of the current in vitro study was to investigate if tissue surface modification with collagenase and addition of the TGF-ß3 can increase the number of cells present in meniscus tears repaired with the use of newly developed tissue adhesives based on isocyanate-terminated block copolymers. Cylindrical explants were harvested from the inner part of bovine menisci. To simulate a full-thickness tear, the central core of the explants was removed and glued back into the defect, with or without incubation in collagenase solution prior to gluing. The repair constructs were then cultured with or without addition of TGF-ß3, and assessed for their histological appearance. The histological staining of the constructs confirmed that both developed adhesives were not cytotoxic. After 28 days, meniscus cells were present in direct contact with the glues. The addition of TGF-ß3 to the culture medium resulted in the presence of cells that formed a sheath inside the simulated tear and in increased cell numbers at the edges of annulus of the explants. In the group in which the tissue was incubated in collagenase and cultured in medium containing TGF-ß3, thicker layers of cells were observed. These results suggest that repairing the torn meniscus with tissue adhesives after pre-treatment of the tissue with collagenase and stimulation with TGF-ß3 is a very promising treatment method, especially when treating the inner avascular part of the meniscus. Nevertheless, longer-term in vitro and in vivo studies are needed to confirm the beneficial effects of this combination therapy.


Collagenases/chemistry , Tibial Meniscus Injuries/therapy , Tissue Adhesives/chemistry , Transforming Growth Factor beta3/chemistry , Animals , Biocompatible Materials/chemistry , Cattle , Cell Movement , Culture Media , Isocyanates/chemistry , Menisci, Tibial/cytology , Rupture/pathology , Spectroscopy, Fourier Transform Infrared , Surface Properties , Tissue Engineering/methods , Wound Healing/drug effects
...