Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 31
1.
Int J Biol Macromol ; 270(Pt 2): 132409, 2024 Jun.
Article En | MEDLINE | ID: mdl-38768918

Suture pull-through is a clinical problem in meniscus repair surgery due to the sharp leading edge of sutures. Several tissue adhesives have been developed as an alternative to traditional suturing; however, there is still no suitable tissue adhesive specific for meniscus repair treatment due to unsatisfactory biosafety, biodegradable, sterilizable, and tissue-bonding characteristics. In this study, we used a tissue adhesive composed of chitosan hydrochloride reacted with oxidative periodate-oxidized dextran (ChitHCl-DDA) combined with a chitosan-based hydrogel and oxidative dextran to attach to the meniscus. We conducted viscoelastic tests, viscosity tests, lap shear stress tests, Fourier transform infrared (FTIR) spectroscopy, swelling ratio tests, and degradation behavior tests to characterize these materials. An MTT assay, alcian blue staining, migration assay, cell behavior observations, and protein expression tests were used to understand cell viability and responses. Moreover, ex vivo and in vivo tests were used to analyze tissue regeneration and biocompatibility of the ChitHCl-DDA tissue adhesive. Our results revealed that the ChitHCl-DDA tissue adhesive provided excellent tissue adhesive strength, cell viability, and cell responses. This tissue adhesive has great potential for torn meniscus tissue repair and regeneration.


Biocompatible Materials , Chitosan , Regeneration , Tissue Adhesives , Tissue Adhesives/chemistry , Tissue Adhesives/pharmacology , Animals , Regeneration/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Chitosan/chemistry , Chitosan/pharmacology , Materials Testing , Meniscus/drug effects , Dextrans/chemistry , Cell Survival/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Rabbits , Tibial Meniscus Injuries/surgery , Humans , Injections
2.
Stem Cells Dev ; 30(10): 537-547, 2021 05 15.
Article En | MEDLINE | ID: mdl-33757298

Osteoarthritis (OA) is the most common type of arthritis, afflicting millions of people in the world. Elevation of inflammatory mediators and enzymatic matrix destruction is often associated with OA. Therefore, the objective of this study was to investigate the effects of conditioned medium from periodontal ligament-derived stem cells (PDLSCs) on inflammatory and catabolic gene expressions of chondrocytes, synoviocytes, and meniscus cells under in vitro inflammatory condition. Stem cells were isolated from human periodontal ligaments. Conditioned medium was collected and concentrated 20 × . Chondrocytes, synoviocytes, and meniscus cells were isolated from pig knees and divided into four experimental groups: serum-free media, serum-free media+interleukin-1ß (IL-1ß) (10 ng/mL), conditioned media (CM), and CM+IL-1ß. Protein content and extracellular vesicle (EV) miRNAs of CM were analyzed by liquid chromatography-tandem mass spectrometry and RNA sequencing, respectively. It was found that the IL-1ß treatment upregulated the expression of IL-1ß, tumor necrosis factor-α (TNF-α), MMP-13, and ADAMTS-4 genes in the three cell types, whereas PDLSC-conditioned medium prevented the upregulation of gene expression by IL-1ß in all three cell types. This study also found that there was consistency in anti-inflammatory effects of PDLSC CM across donors and cell subcultures, while PDLSCs released several anti-inflammatory factors and EV miRNAs at high levels. OA has been suggested as an inflammatory disease in which all intrasynovial tissues are involved. PDLSC-conditioned medium is a cocktail of trophic factors and EV miRNAs that could mediate different inflammatory processes in various tissues in the joint. Introducing PDLSC-conditioned medium to osteoarthritic joints could be a potential treatment to prevent OA progression by inhibiting inflammation.


Anti-Inflammatory Agents/pharmacology , Chondrocytes/drug effects , Culture Media, Conditioned/pharmacology , Meniscus/drug effects , Stem Cells/metabolism , Synoviocytes/drug effects , ADAMTS4 Protein/genetics , Animals , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Culture Media, Conditioned/metabolism , Culture Media, Serum-Free/pharmacology , Extracellular Vesicles/genetics , Gene Expression Regulation/drug effects , Humans , Interleukin-1beta/genetics , Interleukin-1beta/pharmacology , Matrix Metalloproteinase 13/genetics , Meniscus/cytology , Meniscus/metabolism , MicroRNAs/genetics , Periodontal Ligament/cytology , Stem Cells/cytology , Swine , Synoviocytes/cytology , Synoviocytes/metabolism , Tumor Necrosis Factor-alpha/genetics
3.
Int Immunopharmacol ; 91: 107191, 2021 Feb.
Article En | MEDLINE | ID: mdl-33359852

This study aims to investigate the effects and mechanisms of parathyroid hormone [1-34] (PTH1-34) on TNF-α-stimulated mice chondrocytes, as well as cartilage from a meniscus injury induced osteoarthritis (MIO) mice model. The C57BL/6J mice received medial meniscectomy, and then administrated with PTH1-34. The results showed that PTH1-34 administration decreased secondary allodynia and the pain-related transcripts. The IHC, ELISA, Micro-CT imaging and histopathology analysis revealed the significantly improved subchondral plate thickness and bone porosity, the reduced pro-inflammatory cytokines in serum and joint fluid. In vitro, mice chondrocyte was treated with TNF-α or co-cultured with synovial cells. The results showed that TNF-α markedly upregulated the MMP13 expression, and the ERK1/2, NF-κB or PI3K signaling pathway inhibitors could reverse the induction effect of TNF-α on expression of MMP13 in chondrocytes. PTH1-34 alone has no effect on the expression of MMP13 and NF-κB signaling pathways, but the PTH1-34 could reverse the induction effect of TNF-α on MMP13 expression and NF-κB signaling pathway activation in chondrocytes. In addition, PTH1-34 administration inhibited the expression of TNF-α and MMP13, and chondrocyte viability, while the PKA repressor reversed the effect of PTH1-34 in chondrocytes co-cultured with synovial cells. In conclusion, PTH1-34 has an obvious analgesic and anti-inflammatory effect, inhibits the matrix synthesis and alleviates the progression of osteoarthritis. In vitro, PTH1-34 inhibited TNF-α expression and antagonized TNF-α-induced MMP13 expression via the PKA pathway and the NF-κB signaling pathways, respectively.


Analgesics/pharmacology , Anti-Inflammatory Agents/pharmacology , Arthralgia/prevention & control , Chondrocytes/drug effects , Joints/drug effects , Matrix Metalloproteinase 13/metabolism , Meniscus/drug effects , Osteoarthritis/prevention & control , Teriparatide/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Animals , Arthralgia/enzymology , Arthralgia/etiology , Cells, Cultured , Chondrocytes/enzymology , Chondrocytes/pathology , Coculture Techniques , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Joints/enzymology , Joints/pathology , Meniscectomy , Meniscus/enzymology , Meniscus/pathology , Meniscus/surgery , Mice, Inbred C57BL , NF-kappa B/metabolism , Osteoarthritis/enzymology , Osteoarthritis/etiology , Osteoarthritis/pathology , Signal Transduction , Synovial Membrane/drug effects , Synovial Membrane/enzymology , Synovial Membrane/pathology
4.
Sci Rep ; 10(1): 13789, 2020 08 14.
Article En | MEDLINE | ID: mdl-32796866

Although intra-articular corticosteroid injections (IACI) are commonly used for the treatment of knee osteoarthritis (OA), there is controversy regarding possible deleterious effects on joint structure. In this line, this study investigates the effects of IACI on the evolution of knee OA structural changes and pain. Participants for this nested case-control study were from the Osteoarthritis Initiative. Knees of participants who had received an IACI and had magnetic resonance images (MRI) were named cases (n = 93), and each matched with one control (n = 93). Features assessed at the yearly visits and their changes within the follow-up period were from MRI (cartilage volume, meniscal thickness, bone marrow lesions, bone curvature, and synovial effusion size), X-ray (joint space width), and clinical (Western Ontario and McMaster Universities Osteoarthritis Index [WOMAC] pain score) data. Participants who received IACI experienced a transient and significantly greater rate of loss of the meniscal thickness (p = 0.006) and joint space width (p = 0.011) in the knee medial compartment in the year they received the injection, compared to controls. No significant effect of the IACI was found on the rate of cartilage loss nor on any other knee structural changes or WOMAC pain post-treatment. In conclusion, a single IACI in knee OA was shown to be safe with no negative impact on structural changes, but there was a transient meniscal thickness reduction, a phenomenon for which the clinical relevance is at present unknown.


Adrenal Cortex Hormones/therapeutic use , Cartilage, Articular/drug effects , Knee Joint/drug effects , Meniscus/drug effects , Osteoarthritis, Knee/prevention & control , Adrenal Cortex Hormones/administration & dosage , Aged , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Case-Control Studies , Disease Progression , Female , Humans , Injections, Intra-Articular , Knee Joint/diagnostic imaging , Knee Joint/physiopathology , Magnetic Resonance Imaging/methods , Male , Meniscus/metabolism , Meniscus/pathology , Middle Aged , Osteoarthritis, Knee/diagnostic imaging , Osteoarthritis, Knee/physiopathology , Pain/diagnosis , Pain/diagnostic imaging , Pain/prevention & control
5.
Acta Biomater ; 109: 73-81, 2020 06.
Article En | MEDLINE | ID: mdl-32344175

Knee meniscus injury is frequent, resulting in over 1 million surgeries annually in the United States and Europe. Because of the near-avascularity of this fibrocartilaginous tissue and its intrinsic lack of healing, tissue engineering has been proposed as a solution for meniscus repair and replacement. This study describes an approach employing bioactive stimuli to enhance both extracellular matrix content and organization of neomenisci toward augmenting their mechanical properties. Self-assembled fibrocartilages were treated with TGF-ß1, chondroitinase ABC, and lysyl oxidase-like 2 (collectively termed TCL) in addition to lysophosphatidic acid (LPA). TCL + LPA treatment synergistically improved circumferential tensile stiffness and strength, significantly enhanced collagen and pyridinoline crosslink content per dry weight, and achieved tensile anisotropy (circumferential/radial) values of neomenisci close to 4. This study utilizes a combination of bioactive stimuli for use in tissue engineering studies, providing a promising path toward deploying these neomenisci as functional repair and replacement tissues. STATEMENT OF SIGNIFICANCE: This study utilizes a scaffold-free approach, which strays from the tissue engineering paradigm of using scaffolds with cells and bioactive factors to engineer neotissue. While self-assembled neomenisci have attained compressive properties akin to native tissue, tensile properties still require improvement before being able to deploy engineered neomenisci as functional tissue repair or replacement options. In order to augment tensile properties, this study utilized bioactive factors known to augment matrix content in combination with a soluble factor that enhances matrix organization and anisotropy via cell traction forces. Using a bioactive factor to enhance matrix organization mitigates the need for bioreactors used to apply mechanical stimuli or scaffolds to induce proper fiber alignment.


Extracellular Matrix/metabolism , Fibrocartilage/metabolism , Meniscus/metabolism , Tissue Engineering/methods , Amino Acid Oxidoreductases/pharmacology , Animals , Cattle , Chondrocytes/metabolism , Chondroitin ABC Lyase/pharmacology , Elastic Modulus , Extracellular Matrix/drug effects , Fibrocartilage/drug effects , Humans , Lysophospholipids/pharmacology , Materials Testing , Meniscus/drug effects , Tensile Strength , Transforming Growth Factor beta1/pharmacology
6.
Theranostics ; 10(11): 5090-5106, 2020.
Article En | MEDLINE | ID: mdl-32308770

Meniscus deficiency, the most common and refractory disease in human knee joints, often progresses to osteoarthritis (OA) due to abnormal biomechanical distribution and articular cartilage abrasion. However, due to its anisotropic spatial architecture, complex biomechanical microenvironment, and limited vascularity, meniscus repair remains a challenge for clinicians and researchers worldwide. In this study, we developed a 3D printing-based biomimetic and composite tissue-engineered meniscus scaffold consisting of polycaprolactone (PCL)/silk fibroin (SF) with extraordinary biomechanical properties and biocompatibility. We hypothesized that the meticulously tailored composite scaffold could enhance meniscus regeneration and cartilage protection. Methods: The physical property of the scaffold was characterized by scanning electron microscopy (SEM) observation, degradation test, frictional force of interface assessment, biomechanical testing, and fourier transform infrared (FTIR) spectroscopy analysis. To verify the biocompatibility of the scaffold, the viability, morphology, proliferation, differentiation, and extracellular matrix (ECM) production of synovium-derived mesenchymal stem cell (SMSC) on the scaffolds were assessed by LIVE/DEAD staining, alamarBlue assay, ELISA analysis, and qRT-PCR. The recruitment ability of SMSC was tested by dual labeling with CD29 and CD90 by confocal microscope at 1 week after implantation. The functionalized hybrid scaffold was then implanted into the meniscus defects on rabbit knee joint for meniscus regeneration, comparing with the Blank group (no scaffold) and PS group. The regenerated meniscus tissue was evaluated by histological and immunohistochemistry staining, and biomechanical test. Macroscopic and histological scoring was performed to assess the outcome of meniscus regeneration and cartilage protection in vivo. Results: The combination of SF and PCL could greatly balance the biomechanical properties and degradation rate to match the native meniscus. SF sponge, characterized by fine elasticity and low interfacial shear force, enhanced energy absorption capacity of the meniscus and improved chondroprotection. The SMSC-specific affinity peptide (LTHPRWP; L7) was conjugated to the scaffold to further increase the recruitment and retention of endogenous SMSCs. This meticulously tailored scaffold displayed superior biomechanics, structure, and function, creating a favorable microenvironment for SMSC proliferation, differentiation, and extracellular matrix (ECM) production. After 24 weeks of implantation, the histological assessment, biochemical contents, and biomechanical properties demonstrated that the polycaprolactone/silk fibroin-L7 (PS-L7) group was close to the native meniscus group, showing significantly better cartilage protection than the PS group. Conclusion: This tissue engineering scaffold could greatly strengthen meniscus regeneration and chondroprotection. Compared with traditional cell-based therapies, the meniscus tissue engineering approach with advantages of one-step operation and reduced cost has a promising potential for future clinical and translational studies.


Cartilage, Articular/cytology , Fibroins/chemistry , Meniscus/cytology , Mesenchymal Stem Cells/cytology , Polyesters/chemistry , Printing, Three-Dimensional/instrumentation , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Biomechanical Phenomena , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cell Differentiation , Cells, Cultured , Meniscus/drug effects , Meniscus/metabolism , Mesenchymal Stem Cells/metabolism , Porosity , Rabbits
7.
Carbohydr Polym ; 234: 115902, 2020 Apr 15.
Article En | MEDLINE | ID: mdl-32070521

Injectable in situ gelling hydrogels are viable treatment options for meniscal injuries occurring in athletes. The present study aims to develop an injectable hydrogel via borax complexation of oxidized alginate, followed by a self-crosslinking reaction with gelatin through a Schiff's base reaction. Gelation kinetics and degree of crosslinking could be controlled by changing the concentration of components and the formation of Schiff ;'s base formation was confirmed by Raman spectroscopy. The injectable alginate dialdehyde-gelatin (15ADA20G) hydrogel showed 423 ±â€¯20 % water uptake, had an average pore size of 48 µm and compressive strength 295 ±â€¯32 kPa. Phase contrast images, scanning electron micrographs and actin staining depicted adhesion, profuse proliferation, and distribution of fibrochondrocytes on the hydrogel demonstrating its cytocompatibility. Application of hydrogel at the pig meniscal tear ex vivo showed good integration with the host meniscal tissue. Further, the histology of 15ADA20G hydrogel filled meniscus showed retention of hydrogel in the close proximity of meniscal tear even after 3days in culture. The self-crosslinking injectable hydrogel offers a niche for the growth of fibrochondrocytes.


Alginates/pharmacology , Biocompatible Materials/pharmacology , Cross-Linking Reagents/pharmacology , Gelatin/pharmacology , Meniscus/drug effects , Tissue Scaffolds/chemistry , Alginates/chemical synthesis , Alginates/chemistry , Animals , Biocompatible Materials/chemical synthesis , Biocompatible Materials/chemistry , Cell Movement , Cell Proliferation , Cross-Linking Reagents/chemical synthesis , Cross-Linking Reagents/chemistry , Gelatin/chemical synthesis , Gelatin/chemistry , Oxidation-Reduction , Particle Size , Surface Properties , Swine , Tissue Engineering
8.
Biomacromolecules ; 21(1): 240-249, 2020 01 13.
Article En | MEDLINE | ID: mdl-31596075

Rapid adhesion between tissue and synthetic materials is relevant to accelerate wound healing and to facilitate the integration of implantable medical devices. Most frequently, tissue adhesives are applied as a gel or a liquid formulation. This manuscript presents an alternative approach to mediate adhesion between synthetic surfaces and tissue. The strategy presented here is based on the modification of the surface of interest with a thin polymer film that can be transformed on-demand, using UV-light as a trigger, from a nonadhesive into a reactive and tissue adhesive state. As a first proof-of-concept, the feasibility of two photoreactive, thin polymer film platforms has been explored. Both of these films, colloquially referred to as polymer brushes, have been prepared using surface-initiated atom transfer radical polymerization (SI-ATRP) of 2-hydroxyethyl methacrylate (HEMA). In the first part of this study, it is shown that direct UV-light irradiation of PHEMA brushes generates tissue-reactive aldehyde groups and facilitates adhesion to meniscus tissue. While this strategy is very straightforward from an experimental point of view, a main drawback is that the generation of the tissue reactive aldehyde groups uses the 250 nm wavelength region of the UV spectrum, which simultaneously leads to extensive photodegradation of the polymer brush. The second part of this report outlines the synthesis of PHEMA brushes that are modified with 4-[3-(trifluoromethyl)-3H-diazirin-3-yl]benzoic acid (TFMDA) moieties. UV-irradiation of the TFMDA containing brushes transforms the diazirine moieties into reactive carbenes that can insert into C-H, N-H, and O-H bonds and mediate the formation of covalent bonds between the brush surface and meniscus tissue. The advantage of the TFMDA-modified polymer brushes is that these can be activated with 365 nm wavelength UV light, which does not cause photodegradation of the polymer films. While the work presented in this manuscript has used silicon wafers and fused silica substrates as a first proof-of-concept, the versatility of SI-ATRP should enable the application of this strategy to a broad range of biomedically relevant surfaces.


Methacrylates/chemistry , Tissue Adhesives/chemistry , Tissue Adhesives/radiation effects , Animals , Azirines , Benzoates , Cattle , Meniscus/drug effects , Meniscus/radiation effects , Methane/analogs & derivatives , Methane/chemistry , Photochemical Processes , Polymerization , Silicon Dioxide/chemistry , Surface Properties , Ultraviolet Rays
9.
Int J Mol Sci ; 21(1)2019 Dec 19.
Article En | MEDLINE | ID: mdl-31861690

Meniscal injuries, particularly in the avascular zone, have a low propensity for healing and are associated with the development of osteoarthritis. Current meniscal repair techniques are limited to specific tear types and have significant risk for failure. In previous work, we demonstrated the ability of meniscus-derived matrix (MDM) scaffolds to augment the integration and repair of an in vitro meniscus defect. The objective of this study was to determine the effects of percent composition and dehydrothermal (DHT) or genipin cross-linking of MDM bioscaffolds on primary meniscus cellular responses and integrative meniscus repair. In all scaffolds, the porous microenvironment allowed for exogenous cell infiltration and proliferation, as well as endogenous meniscus cell migration. The genipin cross-linked scaffolds promoted extracellular matrix (ECM) deposition and/or retention. The shear strength of integrative meniscus repair was improved with increasing percentages of MDM and genipin cross-linking. Overall, the 16% genipin cross-linked scaffolds were most effective at enhancing integrative meniscus repair. The ability of the genipin cross-linked scaffolds to attract endogenous meniscus cells, promote glycosaminoglycan and collagen deposition, and enhance integrative meniscus repair reveals that these MDM scaffolds are promising tools to augment meniscus healing.


Extracellular Matrix/metabolism , Iridoids/pharmacology , Meniscus/cytology , Tissue Engineering/methods , Animals , Cell Proliferation , Cells, Cultured , Female , Meniscus/drug effects , Meniscus/metabolism , Shear Strength , Swine , Tissue Scaffolds
10.
Theranostics ; 9(24): 7108-7121, 2019.
Article En | MEDLINE | ID: mdl-31695756

Rationale The small molecule Kartogenin (KGN) promotes cartilage regeneration in osteoarthritis (OA) by activating stem cells differentiation, but its pharmacological mode-of-action remains unclear. KGN can be cleaved into 4-aminobiphenyl (4-ABP) and phthalic acid (PA) following enzymolysis of an amide bond. Therefore, this study investigated whether 4-ABP or PA exerted the same action as KGN. Methods KGN, 4-ABP and PA were analyzed in cartilage of mice after oral, intravenous or intra-articular administration of KGN by liquid chromatography-mass spectrometry method. Their effect on proliferation and chondrogenic differentiation of mesenchymal stem cells (MSC) was evaluated in vitro. Furthermore, their effect on cartilage preservation was tested in mice OA model induced by destabilization of medial meniscus. OA severity was quantified using OARSI histological scoring. Transcriptional analysis was used to find the possible targets of the chemicals, which were further validated. Results We demonstrated that while oral or intra-articular KGN delivery effectively ameliorated OA phenotypes in mice, only 4-ABP was detectable in cartilage. 4-ABP could induce chondrogenic differentiation and proliferation of MSC in vitro and promote cartilage repair in OA mouse models mainly by increasing the number of CD44+/CD105+ stem-cell and prevention of matrix loss. These effect of 4-ABP was stronger than that of KGN. Transcriptional profiling of 4-ABP-stimulated MSC suggested that RPS6KA2 and the PI3K-Akt pathway were 4-ABP targets; 4-ABP could activate the PI3K-Akt pathway to promote MSC proliferation and repair OA injury, which was blocked in RPS6KA2-knockdown MSC or RPS6KA2-deficient mice.Conclusion 4-ABP bio-distribution in cartilage promotes proliferation and chondrogenic differentiation of MSC, and repairs osteoarthritic lesions via PI3K-Akt pathway activation.


Aminobiphenyl Compounds/metabolism , Anilides/metabolism , Cartilage/metabolism , Phthalic Acids/metabolism , Regeneration , Administration, Oral , Anilides/administration & dosage , Anilides/pharmacology , Animals , Antigens, CD/metabolism , Cartilage/drug effects , Cartilage/injuries , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chondrogenesis/drug effects , Humans , Hydrolysis , Male , Meniscus/drug effects , Meniscus/pathology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phthalic Acids/administration & dosage , Phthalic Acids/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Regeneration/drug effects , Signal Transduction/drug effects , Tissue Distribution/drug effects
11.
Acta Biomater ; 100: 173-183, 2019 12.
Article En | MEDLINE | ID: mdl-31546030

Fibrillar collagens are highly prevalent in the extracellular matrix of all connective tissues and therefore commonly used as a biomaterial in tissue engineering applications. In the native environment, collagen fibers are arranged in a complex hierarchical structure that is often difficult to recreate in a tissue engineered construct. Small leucine rich proteoglycans as well as hyaluronan binding proteoglycans, aggrecan and versican, have been implicated in regulating fiber formation. In this study, we modified proteoglycan production in vitro by altering culture medium glucose concentrations (4500, 1000, 500, 250, and 125 mg/L), and evaluated its effect on the formation of collagen fibers inside tissue engineered meniscal constructs. Reduction of extracellular glucose resulted in a dose dependent decrease in total sulfated glycosaminoglycan (GAG) production, but minimal decreases of decorin and biglycan. However, fibromodulin doubled in production between 125 and 4500 mg/L glucose concentration. A peak in fiber formation was observed at 500 mg/L glucose concentration and corresponded with reductions in total GAG production. Fiber formation reduction at 125 and 250 mg/L glucose concentrations are likely due to changes in metabolic activity associated with a limited supply of glucose. These results point to proteoglycan production as a means to manipulate fiber architecture in tissue engineered constructs. STATEMENT OF SIGNIFICANCE: Fibrillar collagens are highly prevalent in the extracellular matrix of all connective tissues; however achieving appropriate assembly and organization of collagen fibers in engineered connective tissues is a persistent challenge. Proteoglycans have been implicated in regulating collagen fiber organization both in vivo and in vitro, however little is known about methods to control proteoglycan production and the subsequent fiber organization in tissue engineered menisci. Here, we show that media glucose content can be optimized to control proteoglycan production and collagen fiber assembly, with optimal collagen fiber assembly occurring at sub-physiologic levels of glucose.


Fibrillar Collagens/metabolism , Glucose/pharmacology , Meniscus/physiology , Proteoglycans/biosynthesis , Tissue Engineering/methods , Animals , Cattle , Decorin/metabolism , Fibromodulin/metabolism , Meniscus/drug effects , Tissue Scaffolds/chemistry
12.
Stem Cell Res Ther ; 10(1): 201, 2019 07 08.
Article En | MEDLINE | ID: mdl-31287023

BACKGROUND: The meniscus tear is one of the most common knee injuries particularly seen in athletes and aging populations. Subchondral bone sclerosis, irreparable joint damage, and the early onset of osteoarthritis make the injured meniscus heal difficultly. METHODS: The study was performed by in vitro and in vivo experiments. The in vitro experiments were carried out using the bone marrow stem cells (BMSCs) isolated from the rabbits, and the stemness of the BMSCs was tested by immunostaining. The BMSCs positively expressed stem cell markers were cultured with various concentrations of kartogenin (KGN) for 2 weeks. The chondrogenesis of BMSCs induced by KGN was examined by histochemical staining and quantitative RT-PCR. The in vivo experiments were completed by a rabbit model. Three holes were created in each meniscus by a biopsy punch. The rabbits were treated with four different conditions in each group. Group 1 was treated with 20 µl of saline (saline); group 2 was treated with 5 µl of 100 µM KGN and 15 µl saline (KGN); group 3 was treated with 5 µl of 100 µM KGN, 5 µl of 10,000 U/ ml thrombin, and 10 µl of PRP (KGN+PRP); group 4 was treated with 10,000 BMSCs in 10 µl of PRP, 5 µl of saline solution, and 5 µl of 10,000 U/ml thrombin (PRP+BMSC); group 5 was treated with 10,000 BMSCs in 10 µl of PRP, 5 µl of 100 µM KGN, and 5 µl of 10,000 U/ml thrombin (KGN+PRP+BMSC). The menisci were collected at day 90 post-surgery for gross inspection and histochemical analysis. RESULTS: The histochemical staining showed that KGN induced chondrogenesis of BMSCs in a concentration-dependent manner. The RT-PCR results indicated that chondrocyte-related genes were also increased in the BMSCs cultured with KGN in a dose-dependent manner. The in vivo results showed that large unhealed wound areas were still found in the wounds treated with saline and KGN groups. The wounds treated with BMSCs-containing PRP gel healed much faster than the wounds treated without BMSCs. Furthermore, the wounds treated with BMSCs-containing KGN-PRP gel have healed completely and formed more cartilage-like tissues than the wounds treated with BMSCs-containing PRP gel. CONCLUSIONS: BMSCs could be differentiated into chondrocytes when they were cultured with KGN-PRP gel in vitro and formed more cartilage-like tissues in the wounded rabbit meniscus when the wounds were treated with BMSCs-containing KGN-PRP gel. The results indicated that the BMSCs-containing KGN-PRP gel is a good substitute for injured meniscus repair and regeneration.


Anilides/pharmacology , Meniscus/drug effects , Mesenchymal Stem Cell Transplantation , Osteoarthritis/therapy , Phthalic Acids/pharmacology , Anilides/chemistry , Animals , Cartilage/drug effects , Cartilage/growth & development , Cell Differentiation/drug effects , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrogenesis/drug effects , Disease Models, Animal , Humans , Meniscus/growth & development , Meniscus/injuries , Mesenchymal Stem Cells/cytology , Osteoarthritis/metabolism , Osteoarthritis/pathology , Phthalic Acids/chemistry , Platelet-Rich Plasma/chemistry , Rabbits , Wound Healing/drug effects , Wound Healing/genetics
13.
Nanomedicine ; 21: 102044, 2019 10.
Article En | MEDLINE | ID: mdl-31255792

In the present study, the polycaprolactone (PCL) nanofibers were investigated as a carrier to deliver phytochemicals for bone and cartilage tissue engineering. The PCL nanofibers was blended with phytochemicals hexadecanoic acid, octadecanoic acid and N,N-diisopropyl (2,2,3,3,3-pentafluoropropyl) amine isolated from a medicinal plant, Wattakaka volubilis. The scaffolds were characterized using scanning electron microscope (SEM) and Fourier transform infrared (FTIR) spectroscopy. The average diameter of control and phytochemical loaded nanofiber was 208 ±â€¯9.6 nm and 316 ±â€¯7.0 nm respectively. Biodegradation rate of nanofibers, impact of nanofiber on meniscus and osteoblast cell growth was analyzed using 3-(4,5-dimethyl thiazolyl-2)-2,5-diphenyl tetrazolium bromide (MTT) assay, DNA content and extra cellular matrix secretion. Hoechst stain and SEM images were used to visualize and monitor the cell growth on PCL scaffold. The phytochemicals incorporated PCL nanofibers enhanced the growth and proliferation of primary human meniscus and osteoblast like cells and hence may be suitable scaffold for bone and cartilage tissue engineering applications.


Apocynaceae/chemistry , Bone Regeneration/drug effects , Meniscus/drug effects , Phytochemicals/pharmacology , Tissue Engineering , Biodegradable Plastics/chemistry , Biodegradable Plastics/pharmacology , Cartilage/drug effects , Cartilage/growth & development , Cell Line , Humans , Meniscus/growth & development , Microscopy, Electron, Scanning , Nanofibers/chemistry , Osteoblasts/drug effects , Palmitic Acid/chemistry , Palmitic Acid/pharmacology , Phytochemicals/chemistry , Polyesters/chemistry , Polyesters/pharmacology , Stearic Acids/chemistry , Stearic Acids/pharmacology
14.
J Ultrasound Med ; 38(11): 2853-2859, 2019 Nov.
Article En | MEDLINE | ID: mdl-30887572

OBJECTIVES: Meniscal tears caused by acute trauma or degenerative fraying affect a wide array of individuals. An effective, long-lasting treatment has widely been sought after. Intra-articular corticosteroid injections have been among the methods of controlling pain for more than 60 years. However, such injections tend to produce short-lasting results, with profound effects lasting an average of up to 4 weeks. The purpose of this study was to determine the average duration and magnitude of pain relief after meniscal-targeted injections. METHODS: The electronic medical records of 135 patients were accessed for this retrospective chart review. Patients who had meniscal tears or degenerative fraying and were treated with meniscal-targeted injections were selected. Patients' visual analog scale (VAS) pain scores (before and after treatment) were recorded, along with the percentage of pain relief and duration of pain relief. RESULTS: Ultrasound-guided meniscus-targeted corticosteroid injections for meniscal tears or degenerative fraying produced 5.68 (SD, 5.28) weeks of pain relief on average, with a decrease in pain from initial to follow-up visits of 2.14 (P < .0001) as per the visual analog scale score, and an Integral of Pain Relief score of 3.98. CONCLUSIONS: Our findings indicate a substantial benefit from 20- or 40-mg meniscus-targeted triamcinolone injections, granted the limitations of chart review research and no control group comparison. Results highlight the need for future prospective research comparing meniscus-targeted injections with intra-articular injections to identify a better modality for treating patients with chronic knee pain caused by meniscal tears or degenerative fraying.


Adrenal Cortex Hormones/administration & dosage , Arthralgia/drug therapy , Chronic Pain/drug therapy , Knee Injuries/complications , Meniscus/drug effects , Tendon Injuries/complications , Adrenal Cortex Hormones/therapeutic use , Adult , Aged , Aged, 80 and over , Arthralgia/etiology , Chronic Pain/etiology , Female , Follow-Up Studies , Humans , Injections, Intra-Articular , Knee Injuries/diagnostic imaging , Male , Meniscus/diagnostic imaging , Middle Aged , Retrospective Studies , Tendon Injuries/diagnostic imaging , Treatment Outcome , Ultrasonography, Interventional/methods
15.
Int J Biol Macromol ; 122: 1152-1162, 2019 Feb 01.
Article En | MEDLINE | ID: mdl-30218727

In this study, porcine fibrochondrocyte-seeded agarose, methacrylated gelatin (GelMA), methacrylated hyaluronic acid (MeHA) and GelMA-MeHA blend hydrogels, and 3D printed PCL scaffolds were tested under dynamic compression for potential meniscal regeneration in vitro. Cell-carrying hydrogels produced higher levels of extracellular matrix (ECM) components after a 35-day incubation than the 3D printed PCL. Cells on GelMA exhibited strong cell adhesion (evidenced with intense paxillin staining) and dendritic cell morphology, and produced an order of magnitude higher level of collagen (p < 0.05) than other materials. On the other hand, cells in agarose exhibited low cell adhesion and round cell morphology, and produced higher levels of glycosaminoglycans (GAGs) (p < 0.05) than other materials. A low level of ECM production and a high level of cell proliferation were observed on the 3D printed PCL. Dynamic compression at 10% strain enhanced GAG production in agarose (p < 0.05), and collagen production in GelMA. These results show that hydrogels have a higher potential for meniscal regeneration than the 3D printed PCL, and depending on the material used, fibrochondrocytes could be directed to proliferate or produce cartilaginous or fibrocartilaginous ECM. Agarose and MeHA could be used for the regeneration of the inner region of meniscus, while GelMA for the outer region.


Gelatin/chemistry , Hyaluronic Acid/chemistry , Hydrogels/chemistry , Meniscus/drug effects , Regeneration/drug effects , Sepharose/chemistry , Sepharose/pharmacology , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Adhesion/drug effects , Cell Survival/drug effects , Female , Mechanical Phenomena , Meniscus/physiology , Polyesters/pharmacology , Printing, Three-Dimensional , Swine , Tissue Scaffolds
16.
ACS Appl Mater Interfaces ; 10(45): 38692-38699, 2018 Nov 14.
Article En | MEDLINE | ID: mdl-30335947

Despite the development of hydrogels with high mechanical properties, insufficient adhesion between these materials and biological surfaces significantly limits their use in the biomedical field. By controlling toughening processes, we designed a composite double-network hydrogel with ∼90% water content, which creates a dissipative interface and robustly adheres to soft tissues such as cartilage and meniscus. A double-network matrix composed of covalently cross-linked poly(ethylene glycol) dimethacrylate and ionically cross-linked alginate was reinforced with nanofibrillated cellulose. No tissue surface modification was needed to obtain high adhesion properties of the developed hydrogel. Instead, mechanistic principles were used to control interfacial crack propagation. Comparing to commercial tissue adhesives, the integration of the dissipative polymeric network on the soft tissue surfaces allowed a significant increase in the adhesion strength, such as ∼130 kPa for articular cartilage. Our findings highlight the significant role of controlling hydrogel structure and dissipation processes for toughening the interface. This research provides a promising path to the development of highly adhesive hydrogels for tissues repair.


Hydrogels/chemistry , Hydrogels/pharmacology , Tissue Adhesives/chemistry , Tissue Adhesives/pharmacology , Alginates/chemistry , Alginates/pharmacology , Animals , Bone and Bones/drug effects , Cartilage/drug effects , Cattle , Cellulose/chemistry , Cellulose/pharmacology , Meniscus/drug effects , Methacrylates/chemistry , Methacrylates/pharmacology , Nanostructures/chemistry , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology
17.
Biomaterials ; 182: 13-20, 2018 11.
Article En | MEDLINE | ID: mdl-30099277

Intra-articular injection of hyaluronic acid (HA) is used to treat osteoarthritis (OA) as a viscosupplement, yet it only provides short-term benefit because HA is cleaved by hyaluronidase and cleared out of the joint after several days. Therefore, we developed a new polymer biolubricant based on poly-oxanorbornane carboxylate to enhance joint lubrication for a prolonged time. Rheological and biotribological studies of the biolubricant reveal viscoelastic properties and coefficient of friction equivalent and superior to that of healthy synovial fluid, respectively. Furthermore, in an ex vivo bovine cartilage plug model, the biolubricant exhibits superior long-term reduction of friction and wear prevention compared to saline and healthy synovial fluid. ISO 10993 biocompatibility tests demonstrate that the biolubricant polymer is non-toxic. In an in vivo rat medial meniscal tear OA model, where the performance of the leading HA viscosupplement (Synvisc-one®) is comparable to the saline control, treatment with the biolubricant affords significant chondroprotection compared to the saline control.


Chondrocytes/drug effects , Furans/administration & dosage , Knee Joint/drug effects , Meniscus/drug effects , Polyenes/administration & dosage , Synovial Fluid/drug effects , Viscosupplements/administration & dosage , Animals , Biomechanical Phenomena , Cell Line , Chondrocytes/cytology , Chondrocytes/metabolism , Furans/pharmacology , Furans/therapeutic use , Humans , Injections, Intra-Articular , Knee Injuries/drug therapy , Knee Injuries/metabolism , Knee Joint/metabolism , Male , Meniscus/injuries , Meniscus/metabolism , Mice , NIH 3T3 Cells , Osteoarthritis/drug therapy , Polyenes/pharmacology , Polyenes/therapeutic use , Rats, Inbred Lew , Synovial Fluid/metabolism , Viscosupplements/pharmacology , Viscosupplements/therapeutic use
18.
J Mech Behav Biomed Mater ; 86: 314-324, 2018 10.
Article En | MEDLINE | ID: mdl-30006280

Meniscal injury is typically treated surgically via partial meniscectomy, which has been shown to cause cartilage degeneration in the long-term. Consequently, research has focused on meniscal prevention and replacement. However, none of the materials or implants developed for meniscal replacement have yet achieved widespread acceptance or demonstrated conclusive chondroprotective efficacy. A redesigned silk fibroin scaffold, which already displayed promising results regarding biocompatibility and cartilage protection in a previous study, was characterised in terms of its biomechanical, structural and biological functionality to serve as a potential material for permanent partial meniscal replacement. Therefore, different quasi-static but also dynamic compression tests were performed. However, the determined compressive stiffness (0.56 ±â€¯0.31 MPa and 0.30 ±â€¯0.12 MPa in relaxation and creep configuration, respectively) was higher in comparison to the native meniscal tissue, which could potentially disturb permanent integration into the host tissue. Nevertheless, µ-CT analysis met the postulated requirements for partial meniscal replacement materials in terms of the microstructural parameters, like mean pore size (215.6 ±â€¯10.9 µm) and total porosity (80.1 ±â€¯4.3%). Additionally, the biocompatibility was reconfirmed during cell culture experiments. The current study provides comprehensive mechanical and biological data for the characterisation of this potential replacement material. Although some further optimisation of the silk fibroin scaffold may be advantageous, the silk fibroin scaffold showed sufficient biomechanical competence to support loads already in the early postoperative phase.


Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Fibroins/chemistry , Fibroins/pharmacology , Mechanical Phenomena , Meniscus/drug effects , Meniscus/physiology , Animals , Biomechanical Phenomena , Materials Testing , Meniscus/diagnostic imaging , Mice , Tensile Strength , X-Ray Microtomography
19.
Cartilage ; 9(3): 293-303, 2018 07.
Article En | MEDLINE | ID: mdl-29986604

Objective Early intervention therapies targeting inflammation and cell death during the acute phase of cartilage injury have the potential to prevent posttraumatic osteoarthritis. The objective of this study was to investigate the effects of interleukin receptor antagonist protein (IRAP), hyaluronan (HA), dexamethasone (DEX), and mesenchymal stem cell (MSC) treatment on the expression of established genetic markers for matrix degradation, apoptosis, and inflammation in articular cartilage during the acute phase of injury. Design A custom impact device was used to create replicable injury ex vivo to intact porcine knee joint. One hour after impact, IRAP, HA, DEX, or MSCs was intra-articularly injected. At 8 hours postinjury, cartilage and meniscus samples were harvested for genetic expression analysis. Expression of miR-27b, miR-140, miR-125b, miR-16, miR-34a, miR-146a, miR-22, ADAMTS-4, ADAMTS-5, MMP-3, IL-1ß, and TNF-α was analyzed by real-time polymerase chain reaction. Results At 8 hours postinjury, expression of ADAMTS-4, ADAMTS-5, MMP-3, IL-1ß, and TNF-α in cartilage was significantly decreased in IRAP- and DEX-treated joints as compared to nontreated injured joints, whereas only IRAP upregulated expression of miR-140, miR-125b, miR-27b, miR-146a, and miR-22 in cartilage. HA and MSC treatments had no significant effects on catabolic and inflammatory gene expression in cartilage. However, HA treatment significantly upregulated expression of all miRNAs except miR-16. In addition, the treatments tested also exhibited significant influences on meniscus. Conclusions This study provides a valuable starting point for further research into potential targets for and efficacy of various early intervention strategies that may delay or prevent the progression of posttraumatic osteoarthritis after acute cartilage injury.


Anti-Inflammatory Agents/metabolism , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Osteoarthritis, Knee/genetics , ADAMTS4 Protein/drug effects , ADAMTS4 Protein/genetics , ADAMTS5 Protein/drug effects , ADAMTS5 Protein/genetics , Animals , Cartilage, Articular/drug effects , Cartilage, Articular/injuries , Cell Death/drug effects , Cells, Cultured/metabolism , Chondrocytes/drug effects , Dexamethasone/administration & dosage , Dexamethasone/therapeutic use , Gene Expression , Hyaluronic Acid/administration & dosage , Hyaluronic Acid/therapeutic use , Inflammation/metabolism , Injections, Intra-Articular/methods , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/genetics , Meniscus/drug effects , Meniscus/metabolism , Mesenchymal Stem Cell Transplantation/methods , MicroRNAs/genetics , Models, Animal , Osteoarthritis, Knee/prevention & control , Receptors, Interleukin/antagonists & inhibitors , Receptors, Interleukin/therapeutic use , Swine , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/genetics
20.
Biochem Biophys Res Commun ; 502(3): 370-374, 2018 07 20.
Article En | MEDLINE | ID: mdl-29852167

Obesity and associated metabolic factors are major risk factors for the development of osteoarthritis. Previously, we have shown that the free fatty acid palmitate induces endoplasmic reticulum (ER) stress and induces apoptosis in meniscus cells. However, the molecular mechanisms involved in these effects are not clearly understood. In our current study, we found that palmitate inhibits autophagy by modulating the protein levels of autophagy-related genes-5 (ATG5) that is associated with decreased lipidation of LC3 and increased activation of cleaved caspase 3. Pretreatment of meniscus cells with 4-phenyl butyric acid, a small molecule chemical chaperone that alleviates ER stress, or with MG-132, a proteasome inhibitor, restored normal levels of ATG5 and autophagosome formation, and decreased expression of cleaved caspase 3. Thus, our data suggest that palmitate downregulates autophagy in meniscus cells by degrading ATG5 protein via ER-associated protein degradation, and thus promotes apoptosis. This is the first study to demonstrate that palmitate-induced endoplasmic reticulum stress negatively regulates autophagy.


Autophagy-Related Protein 5/metabolism , Autophagy/physiology , Meniscus/cytology , Meniscus/metabolism , Palmitic Acid/metabolism , Animals , Autophagy/drug effects , Cells, Cultured , Down-Regulation/drug effects , Endoplasmic Reticulum Stress/drug effects , Humans , Leupeptins/pharmacology , Meniscus/drug effects , Osteoarthritis/etiology , Palmitic Acid/pharmacology , Phenylbutyrates/pharmacology , Proteasome Inhibitors/pharmacology , Proteolysis/drug effects , Swine
...