Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters











Publication year range
1.
Biomacromolecules ; 25(6): 3312-3324, 2024 06 10.
Article in English | MEDLINE | ID: mdl-38728671

ABSTRACT

3D-printed hydrogel scaffolds biomimicking the extracellular matrix (ECM) are key in cartilage tissue engineering as they can enhance the chondrogenic differentiation of mesenchymal stem cells (MSCs) through the presence of active nanoparticles such as graphene oxide (GO). Here, biomimetic hydrogels were developed by cross-linking alginate, gelatin, and chondroitin sulfate biopolymers in the presence of GO as a bioactive filler, with excellent processability for developing bioactive 3D printed scaffolds and for the bioprinting process. A novel bioink based on our hydrogel with embedded human MSCs presented a cell survival rate near 100% after the 3D bioprinting process. The effects of processing and filler concentration on cell differentiation were further quantitatively evaluated. The nanocomposited hydrogels render high MSC proliferation and viability, exhibiting intrinsic chondroinductive capacity without any exogenous factor when used to print scaffolds or bioprint constructs. The bioactivity depended on the GO concentration, with the best performance at 0.1 mg mL-1. These results were explained by the rational combination of the three biopolymers, with GO nanoparticles having carboxylate and sulfate groups in their structures, therefore, biomimicking the highly negatively charged ECM of cartilage. The bioactivity of this biomaterial and its good processability for 3D printing scaffolds and 3D bioprinting techniques open up a new approach to developing novel biomimetic materials for cartilage repair.


Subject(s)
Alginates , Bioprinting , Cell Differentiation , Chondrogenesis , Chondroitin Sulfates , Gelatin , Hydrogels , Mesenchymal Stem Cells , Nanocomposites , Printing, Three-Dimensional , Tissue Scaffolds , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/pharmacology , Alginates/chemistry , Alginates/pharmacology , Gelatin/chemistry , Bioprinting/methods , Cell Differentiation/drug effects , Chondrogenesis/drug effects , Nanocomposites/chemistry , Tissue Scaffolds/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Tissue Engineering/methods , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Graphite/chemistry , Graphite/pharmacology , Cell Proliferation/drug effects , Cells, Cultured
2.
Domest Anim Endocrinol ; 74: 106572, 2021 01.
Article in English | MEDLINE | ID: mdl-33039930

ABSTRACT

The aim of this study was to differentiate canine adipose-derived mesenchymal stem cells (ADMSCs) into insulin-producing cells by using culture media with different compositions to determine the most efficient media. Stem cells isolated from the fat tissues close to the bitch uterus were distributed into 6 groups: (1) Dulbecco's modified Eagle medium (DMEM)-high glucose (HG), ß-mercaptoethanol, and nicotinamide; (2) DMEM-HG, ß-mercaptoethanol, nicotinamide, and exendin-4; (3) DMEM-HG, ß-mercaptoethanol, nicotinamide, exendin-4, B27, nonessential amino acids, and l-glutamine; (4) DMEM-HG, ß-mercaptoethanol, and nicotinamide (for the initial 8-d period), and DMEM-HG, ß-mercaptoethanol, nicotinamide, exendin-4, B27, nonessential amino acids, l-glutamine, and basic fibroblast growth factor (for the remaining 8-d period); (5) DMEM-HG and fetal bovine serum; and (6) DMEM-low glucose and fetal bovine serum (standard control group). Adipose-derived mesenchymal stem cells from groups 1 to 5 gradually became round in shape and gathered in clusters. These changes differed between the groups. In group 3, the cell clusters were apparently more in numbers and gathered as bigger aggregates. Dithizone staining showed that groups 3 and 4 were similar in terms of the mean area of each aggregate stained for insulin. However, only in group 4, the number of insulin aggregates and the total area of aggregates stained were significantly bigger than in the other groups. The mRNA expression of PDX1, BETA2, MafA, and Insulin were also confirmed in all the groups. We conclude that by manipulating the composition of the culture medium it is possible to induce canine ADMSCs into insulin-producing cells, and the 2-staged protocol that was used promoted the best differentiation.


Subject(s)
Cell Differentiation , Culture Media/pharmacology , Insulin/metabolism , Mesenchymal Stem Cells/physiology , Adipogenesis/drug effects , Adipogenesis/physiology , Animals , Carbazoles/chemistry , Carbazoles/pharmacology , Chondrogenesis/drug effects , Chondrogenesis/physiology , Culture Media/chemistry , Dogs , Immunophenotyping , Mercaptoethanol/pharmacology , Niacinamide/chemistry , Niacinamide/pharmacology , Osteogenesis/drug effects , Osteogenesis/physiology
3.
Biotechnol Lett ; 42(12): 2721-2734, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32785804

ABSTRACT

The development of new technologies to produce three-dimensional and biocompatible scaffolds associated with high-end cell culture techniques have shown to be promising for the regeneration of tissues and organs. Some biomedical devices, as meniscus prosthesis, require high flexibility and tenacity and such features are found in polyurethanes which represent a promising alternative. The Poly(PCL-TMC)urethane here presented, combines the mechanical properties of PCL with the elasticity attributed by TMC and presents great potential as a cellular carrier in cartilage repair. Scanning electron microscopy showed the presence of interconnected pores in the three-dimensional structure of the material. The scaffolds were submitted to proliferation and cell differentiation assays by culturing mesenchymal stem cells in bioreactor. The tests were performed in dynamic flow mode at the rate of 0.4 mL/min. Laser scanning confocal microscopy analysis showed that the flow rate promoted cell growth and cartilage ECM synthesis of aggrecan and type II collagen within the Poly(PCL-TMC)urethane scaffolds. This study demonstrated the applicability of the polymer as a cellular carrier in tissue engineering, as well as the ECM was incremented only when under oriented flow rate stimuli. Therefore, our results may also provide data on how oriented flow rate in dynamic bioreactors culture can influence cell activity towards cartilage ECM synthesis even when specific molecular stimuli are not present. This work addresses new perspectives for future clinical applications in cartilage tissue engineering when the molecular factors resources could be scarce for assorted reasons.


Subject(s)
Cartilage/chemistry , Chondrogenesis/drug effects , Extracellular Matrix/chemistry , Tissue Engineering , Bioreactors , Cartilage/drug effects , Cartilage/growth & development , Cartilage/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Methacrylates/chemistry , Methacrylates/pharmacology , Polyesters/chemistry , Polyesters/pharmacology , Polyurethanes/chemistry , Polyurethanes/pharmacology , Tissue Scaffolds/chemistry
4.
Arq. bras. med. vet. zootec. (Online) ; 71(2): 509-520, mar.-abr. 2019. graf, ilus
Article in English | VETINDEX, LILACS | ID: biblio-1011276

ABSTRACT

The aim of this study was to evaluate the effect of concentrations of caffeine on the viability, synthesis activity and gene expression in cultures of chondrocytes. Extracted articular cartilage from the femurs and tibias of 15 Wistar rats at three days old to isolate chondrocytes. Chondrocytes were cultured in chondrogenic medium (control) or supplemented with caffeine (0.5, 1.0, 2.0mM). Cell viability, alkaline phosphatase activity and collagen synthesis were assessed using colorimetric assays at 7, 14, 21 days. The chondrocyte cultures of all groups grown under coverslips were stained with hematoxylin-eosin to determine the percentage of cells/field and with PAS, safranin O, alcian blue to determine the percentage of matrix chondrogenic/field at 21 days. The expressions of gene transcripts for aggrecan, collagen-II, Sox-9, Runx-2 and alkaline phosphatase were also evaluated by RT-PCR at 21 days. The means were compared using Student-Newman-Keuls. Caffeine significantly reduced the conversion of MTT to formazan, percentage of cells/field, collagen synthesis, alkaline phosphatase activity, synthesis of PAS+, safranin O+ and alcian blue+ chondrogenic matrix, and the expression of aggrecan, Sox-9 and II collagen. It is concluded that caffeine at concentrations of 0.5, 1.0, 2.0mM has a direct inhibitory effect on chondrogenesis in cultures of chondrocytes from rats.(AU)


O objetivo deste estudo foi avaliar o efeito direto de concentrações de cafeína sobre a viabilidade, atividade de síntese e expressão gênica em culturas de condrócitos de ratos. As cartilagens dos fêmures e tíbias de 15 ratos Wistar com três dias foram extraídas para isolamento de condrócitos. Os condrócitos foram cultivados em meio condrogênico (controle) ou em meio acrescido de diferentes concentrações de cafeína (0,5, 1,0, 2,0mM). Foram avaliadas a viabilidade celular, a atividade da fosfatase alcalina e a síntese de colágeno por ensaios colorimétricos aos sete, 14 e 21 dias. Condrócitos cultivados sob lamínulas foram corados pela hematoxilina e eosina, para se determinar a porcentagem de células/campo, e pelo PAS, safranina O, alcian Blue, para se determinar a porcentagem de matriz condrogênica/campo aos 21 dias. Foi avaliada a expressão de transcriptos gênicos para Sox-9, Runx-2, agrecano, colágeno-II e fosfatase alcalina por qRT-PCR, aos 21 dias. As médias foram comparadas pelo Student-Newman-Keuls. A cafeína reduziu significativamente o MTT em cristais de formazan, a porcentagem de células/campo, a síntese de colágeno, a atividade da fosfatase alcalina e a síntese de matriz condrogênica PAS+, safranina O+, alcian blue+ e expressão de Sox-9 e colágeno-II. Conclui-se que a cafeína, nas concentrações de 0,5, 1,0, 2,0mM, apresenta efeito inibidor direto sobre a condrogênese em culturas de condrócitos de ratos.(AU)


Subject(s)
Animals , Female , Rats , Caffeine , Cartilage, Articular/drug effects , Chondrocytes/drug effects , Chondrogenesis/drug effects
5.
Arq. bras. med. vet. zootec. (Online) ; 71(2): 509-520, mar.-abr. 2019. graf, ilus
Article in English | VETINDEX | ID: vti-23535

ABSTRACT

The aim of this study was to evaluate the effect of concentrations of caffeine on the viability, synthesis activity and gene expression in cultures of chondrocytes. Extracted articular cartilage from the femurs and tibias of 15 Wistar rats at three days old to isolate chondrocytes. Chondrocytes were cultured in chondrogenic medium (control) or supplemented with caffeine (0.5, 1.0, 2.0mM). Cell viability, alkaline phosphatase activity and collagen synthesis were assessed using colorimetric assays at 7, 14, 21 days. The chondrocyte cultures of all groups grown under coverslips were stained with hematoxylin-eosin to determine the percentage of cells/field and with PAS, safranin O, alcian blue to determine the percentage of matrix chondrogenic/field at 21 days. The expressions of gene transcripts for aggrecan, collagen-II, Sox-9, Runx-2 and alkaline phosphatase were also evaluated by RT-PCR at 21 days. The means were compared using Student-Newman-Keuls. Caffeine significantly reduced the conversion of MTT to formazan, percentage of cells/field, collagen synthesis, alkaline phosphatase activity, synthesis of PAS+, safranin O+ and alcian blue+ chondrogenic matrix, and the expression of aggrecan, Sox-9 and II collagen. It is concluded that caffeine at concentrations of 0.5, 1.0, 2.0mM has a direct inhibitory effect on chondrogenesis in cultures of chondrocytes from rats.(AU)


O objetivo deste estudo foi avaliar o efeito direto de concentrações de cafeína sobre a viabilidade, atividade de síntese e expressão gênica em culturas de condrócitos de ratos. As cartilagens dos fêmures e tíbias de 15 ratos Wistar com três dias foram extraídas para isolamento de condrócitos. Os condrócitos foram cultivados em meio condrogênico (controle) ou em meio acrescido de diferentes concentrações de cafeína (0,5, 1,0, 2,0mM). Foram avaliadas a viabilidade celular, a atividade da fosfatase alcalina e a síntese de colágeno por ensaios colorimétricos aos sete, 14 e 21 dias. Condrócitos cultivados sob lamínulas foram corados pela hematoxilina e eosina, para se determinar a porcentagem de células/campo, e pelo PAS, safranina O, alcian Blue, para se determinar a porcentagem de matriz condrogênica/campo aos 21 dias. Foi avaliada a expressão de transcriptos gênicos para Sox-9, Runx-2, agrecano, colágeno-II e fosfatase alcalina por qRT-PCR, aos 21 dias. As médias foram comparadas pelo Student-Newman-Keuls. A cafeína reduziu significativamente o MTT em cristais de formazan, a porcentagem de células/campo, a síntese de colágeno, a atividade da fosfatase alcalina e a síntese de matriz condrogênica PAS+, safranina O+, alcian blue+ e expressão de Sox-9 e colágeno-II. Conclui-se que a cafeína, nas concentrações de 0,5, 1,0, 2,0mM, apresenta efeito inibidor direto sobre a condrogênese em culturas de condrócitos de ratos.(AU)


Subject(s)
Animals , Female , Rats , Caffeine , Cartilage, Articular/drug effects , Chondrocytes/drug effects , Chondrogenesis/drug effects , Models, Animal
6.
Cells ; 8(1)2019 01 16.
Article in English | MEDLINE | ID: mdl-30654437

ABSTRACT

Tissue engineering and cell-based therapy combine techniques that create biocompatible materials for cell survival, which can improve tendon repair. This study seeks to use a new fibrin sealant (FS) derived from the venom of Crotalus durissus terrificus, a biodegradable three-dimensional scaffolding produced from animal components only, associated with adipose-derived stem cells (ASC) for application in tendons injuries, considered a common and serious orthopedic problem. Lewis rats had tendons distributed in five groups: normal (N), transected (T), transected and FS (FS) or ASC (ASC) or with FS and ASC (FS + ASC). The in vivo imaging showed higher quantification of transplanted PKH26-labeled ASC in tendons of FS + ASC compared to ASC on the 14th day after transection. A small number of Iba1 labeled macrophages carrying PKH26 signal, probably due to phagocytosis of dead ASC, were observed in tendons of transected groups. ASC up-regulated the Tenomodulin gene expression in the transection region when compared to N, T and FS groups and the expression of TIMP-2 and Scleraxis genes in relation to the N group. FS group presented a greater organization of collagen fibers, followed by FS + ASC and ASC in comparison to N. Tendons from ASC group presented higher hydroxyproline concentration in relation to N and the transected tendons of T, FS and FS + ASC had a higher amount of collagen I and tenomodulin in comparison to N group. Although no marked differences were observed in the other biomechanical parameters, T group had higher value of maximum load compared to the groups ASC and FS + ASC. In conclusion, the FS kept constant the number of transplanted ASC in the transected region until the 14th day after injury. Our data suggest this FS to be a good scaffold for treatment during tendon repair because it was the most effective one regarding tendon organization recovering, followed by the FS treatment associated with ASC and finally by the transplanted ASC on the 21st day. Further investigations in long-term time points of the tendon repair are needed to analyze if the higher tissue organization found with the FS scaffold will improve the biomechanics of the tendons.


Subject(s)
Adipose Tissue/cytology , Fibrin Tissue Adhesive/therapeutic use , Stem Cell Transplantation , Stem Cells/cytology , Tendon Injuries/therapy , Adipogenesis/drug effects , Animals , Antigens, CD/metabolism , Biomarkers/metabolism , Biomechanical Phenomena , Birefringence , Calcium-Binding Proteins/metabolism , Cell Movement/drug effects , Chondrogenesis/drug effects , Collagen/metabolism , Fibrin Tissue Adhesive/pharmacology , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Expression Regulation/drug effects , Hydroxyproline/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Microfilament Proteins/metabolism , Osteogenesis/drug effects , Rats, Inbred Lew , Tendon Injuries/genetics , Tendon Injuries/pathology , Tendon Injuries/physiopathology
7.
Eur J Med Res ; 23(1): 52, 2018 Oct 24.
Article in English | MEDLINE | ID: mdl-30355362

ABSTRACT

BACKGROUND: A promising novel cell-free bioactive formulation for articular cartilage regeneration, called BIOF2, has recently been tested in pre-clinical trials. The aim of the present study was to evaluate the efficacy and safety of BIOF2 for intra-articular application in patients with severe osteoarthritis of the knee. METHODS: A prospective, randomized, 3-arm, parallel group clinical trial was conducted. It included 24 patients with severe osteoarthritis of the knee (WOMAC score 65.9 ± 17). Before they entered the study, all the patients were under osteoarthritis control through the standard treatment with nonsteroidal anti-inflammatory drugs (NSAIDs), prescribed by their family physician. Patients were distributed into three groups of 8 patients each (intra-articular BIOF2, total joint arthroplasty, or conservative treatment with NSAIDs alone). The WOMAC score, RAPID3 score, and Rasmussen clinical score were evaluated before treatment and at months 3, 6, and 12. BIOF2 was applied at months 0, 3, and 6. Complete blood count and blood chemistry parameters were determined in the BIOF2 group before treatment, at 72 h, and at months 1, 3, 6, and 12. In addition, articular cartilage volume was evaluated (according to MRI) at the beginning of the study and at month 12. RESULTS: The NSAID group showed no improvement at follow-up. Arthroplasty and BIOF2 treatments showed significant improvement in all the scoring scales starting at month 3. There were no statistically significant differences between the BIOF2 group and the arthroplasty group at month 6 (WOMAC score: 19.3 ± 18 vs 4.3 ± 5; P = 0.24) or month 12 (WOMAC score: 15.6 ± 15 vs 15.7 ± 17; P = 1.0). Arthroplasty and BIOF2 were successful at month 12 (according to a WOMAC score: ≤ 16) in 75% of the patients and the daily use of NSAIDs was reduced, compared with the group treated exclusively with NSAIDs (RR = 0.33, 95% CI 0.12-0.87, P = 0.02. This result was the same for BIOF2 vs NSAIDs and arthroplasty vs NSAIDs). BIOF2 significantly increased the articular cartilage by 22% (26.1 ± 10 vs 31.9 ± 10 cm2, P < 0.001) and produced a significant reduction in serum lipids. BIOF2 was well tolerated, causing slight-to-moderate pain only upon application. CONCLUSIONS: The intra-articular application of the new bioactive cell-free formulation (BIOF2) was well tolerated and showed no significative differences with arthroplasty for the treatment of severe osteoarthritis of the knee. BIOF2 can regenerate articular cartilage and is an easily implemented alternative therapy for the treatment of osteoarthritis. Trial registration Cuban Public Registry of Clinical Trials (RPCEC) Database RPCEC00000250. Registered 08/15/2017-Retrospectively registered, http://rpcec.sld.cu/en/trials/RPCEC00000250-En .


Subject(s)
Cartilage, Articular/drug effects , Mesenchymal Stem Cells/chemistry , Osteoarthritis, Knee/drug therapy , Steroids/administration & dosage , Adult , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arthroplasty, Replacement, Knee , Blood Cell Count , Cartilage, Articular/growth & development , Cell-Free System/chemistry , Cell-Free System/metabolism , Chondrocytes/drug effects , Chondrogenesis/drug effects , Female , Humans , Injections, Intra-Articular , Male , Mesenchymal Stem Cells/metabolism , Middle Aged , Osteoarthritis, Knee/blood , Osteoarthritis, Knee/physiopathology , Osteoarthritis, Knee/surgery , Regeneration/drug effects , Steroids/pharmacology , Treatment Outcome
8.
J Pharm Pharmacol ; 70(1): 89-100, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29105086

ABSTRACT

OBJECTIVES: Verify the in-vitro effect of triiodothyronine (T3) on the chondrogenic differentiation of female rat bone marrow mesenchymal stem cells (BMMSCs) over several time periods and at several doses. METHODS: CD54 + /CD73 + /CD90 +  BMMSCs from Wistar female rats were cultured in chondrogenic medium with or without T3 (0.01; 1; 100; 1000 nm). At seven, 14 and 21 days, the cell morphology, chondrogenic matrix formation and expression of Sox9 and collagen II were evaluated. KEY FINDINGS: The dose of 100 nm did not alter the parameters evaluated in any of the periods studied. However, the 0.01 nm T3 dose improved the chondrogenic potential by increasing the chondrogenic matrix formation and expression of Sox9 and collagen II in at least one of the evaluated periods; the 1 nm T3 dose also improved the chondrogenic potential by increasing the chondrogenic matrix formation and the expression of collagen II in at least one of the evaluated periods. The 1000 nm T3 dose improved the chondrogenic potential by increasing the chondrogenic matrix formation and Sox9 expression in at least one of the evaluated periods. CONCLUSIONS: T3 has a dose-dependent effect on the differentiation of BMMSCs from female rats.


Subject(s)
Cell Differentiation/drug effects , Chondrogenesis/drug effects , Mesenchymal Stem Cells/drug effects , Triiodothyronine/pharmacology , Animals , Cells, Cultured , Chondrocytes/cytology , Collagen Type II/genetics , Dose-Response Relationship, Drug , Female , Mesenchymal Stem Cells/cytology , Rats , Rats, Wistar , SOX9 Transcription Factor/genetics , Time Factors , Triiodothyronine/administration & dosage
9.
Biomed Res Int ; 2016: 5891925, 2016.
Article in English | MEDLINE | ID: mdl-27840829

ABSTRACT

Bisphosphonates such as alendronate are antiosteoporotic drugs that inhibit the activity of bone-resorbing osteoclasts and secondarily promote osteoblastic function. Diabetes increases bone-matrix-associated advanced glycation end products (AGEs) that impair bone marrow progenitor cell (BMPC) osteogenic potential and decrease bone quality. Here we investigated the in vitro effect of alendronate and/or AGEs on the osteoblastogenic, adipogenic, and chondrogenic potential of BMPC isolated from nondiabetic untreated rats. We also evaluated the in vivo effect of alendronate (administered orally to rats with insulin-deficient Diabetes) on long-bone microarchitecture and BMPC multilineage potential. In vitro, the osteogenesis (Runx2, alkaline phosphatase, type 1 collagen, and mineralization) and chondrogenesis (glycosaminoglycan production) of BMPC were both decreased by AGEs, while coincubation with alendronate prevented these effects. The adipogenesis of BMPC (PPARγ, intracellular triglycerides, and lipase) was increased by AGEs, and this was prevented by coincubation with alendronate. In vivo, experimental Diabetes (a) decreased femoral trabecular bone area, osteocyte density, and osteoclastic TRAP activity; (b) increased bone marrow adiposity; and (c) deregulated BMPC phenotypic potential (increasing adipogenesis and decreasing osteogenesis and chondrogenesis). Orally administered alendronate prevented all these Diabetes-induced effects on bone. Thus, alendronate could improve bone alterations in diabetic rats by preventing the antiosteogenic, antichondrogenic, and proadipocytic effects of AGEs on BMPC.


Subject(s)
Alendronate/administration & dosage , Bone Marrow Cells/cytology , Cell Differentiation/drug effects , Diabetes Mellitus, Experimental/drug therapy , Adipogenesis/drug effects , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Regeneration/drug effects , Chondrogenesis/drug effects , Diabetes Mellitus, Experimental/pathology , Glycation End Products, Advanced/administration & dosage , Glycation End Products, Advanced/metabolism , Humans , Osteogenesis/drug effects , Rats
10.
Genet Mol Res ; 15(3)2016 Jul 15.
Article in English | MEDLINE | ID: mdl-27525846

ABSTRACT

This study was designed to evaluate bone matrix gelatin (BMG)/fibrin glue and chitosan/gelatin composite scaffolds for cartilage tissue engineering. Chondrocytes were isolated from costal cartilage of Sprague-Dawley rats and seeded on BMG/fibrin glue or chitosan/gelatin composite scaffolds. After different in vitro culture durations, the scaffolds were subjected to hematoxylin and eosin, Masson's trichrome, and toluidine blue staining, anti-collagen II and anti-aggrecan immunohistochemistry, and scanning electronic microscopy (SEM) analysis. After 2 weeks of culture, chondrocytes were distributed evenly on the surfaces of both scaffolds. Cell numbers and the presence of extracellular matrix components were markedly increased after 8 weeks of culture, and to a greater extent on the chitosan/gelatin scaffold. The BMG/fibrin glue scaffold showed signs of degradation after 8 weeks. Immunofluorescence analysis confirmed higher levels of collagen II and aggrecan using the chitosan/gelatin scaffold. SEM revealed that the majority of cells on the surface of the BMG/fibrin glue scaffold demonstrated a round morphology, while those in the chitosan/gelatin group had a spindle-like shape, with pseudopodia. Chitosan/gelatin scaffolds appear to be superior to BMG/ fibrin glue constructs in supporting chondrocyte attachment, proliferation, and biosynthesis of cartilaginous matrix components.


Subject(s)
Chondrocytes/drug effects , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Adhesives/adverse effects , Aggrecans/genetics , Aggrecans/metabolism , Animals , Bone Matrix/chemistry , Cell Adhesion , Cells, Cultured , Chitosan/adverse effects , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrogenesis/drug effects , Collagen Type II/genetics , Collagen Type II/metabolism , Fibrin/adverse effects , Gelatin/adverse effects , Rats , Rats, Sprague-Dawley , Tissue Scaffolds/adverse effects
11.
Genet Mol Res ; 14(1): 339-48, 2015 Jan 23.
Article in English | MEDLINE | ID: mdl-25729966

ABSTRACT

The use of mesenchymal stem cells (MSCs) in experimental, clinical, and therapeutic trials has grown in recent years. However, the issue remains of whether these procedures are completely safe for transplant patients. Therefore, this study was designed and carried out with the aim of evaluating two different comet assay protocols for genomic damage pattern analysis in MSCs derived from adipose tissue. The analyzed and interpreted results suggest that genetic testing is needed to support clonal expansion safety in cell therapy procedures with MSCs. Furthermore, they also suggest that if the comet assay technique would be used as a genomic integrity screening assay, the protocol performed at pH = 12 (that yielded a frequency of damaged cells: tail intensity = 9.50 ± 0.60, tail moment = 0.0122 ± 0.0007; results are reported as means ± standard deviation) would be indicated as genomic damage, and that subsequent single-strand breaks occur at pH > 13 (frequency of damaged cells: tail intensity = 30.71 ± 4.23, tail moment = 0.0447 ± 0.0073). Our study demonstrates that, in the era of regenerative medicine, it is necessary to standardize and establish a battery of tests in order to identify genomic damage prior to MSC transplantation.


Subject(s)
Adipose Tissue/cytology , Comet Assay/methods , Genome , Mesenchymal Stem Cells/cytology , Adipogenesis/drug effects , Animals , Cell Count , Cell Shape/drug effects , Cell Survival/drug effects , Chondrogenesis/drug effects , DNA Damage , Hydrogen-Ion Concentration , Mesenchymal Stem Cells/drug effects , Mutagens/toxicity , Osteogenesis/drug effects , Rabbits
12.
J Appl Toxicol ; 35(11): 1390-7, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25619733

ABSTRACT

High levels of homocysteine (Hcy) are related to an increased risk of the occurrence of congenital anomalies, including limb defects. However, few evaluations about how toxic levels of Hcy affect limb development have been reported. We investigated whether Hcy can affect the cell cycle proteins and proteins involved in mesenchymal cell differentiation during limb development, in a chicken embryo model. Embryos were treated with 20 µmol d-l Hcy/50 µl saline at embryonic day 2 and analyzed at embryonic day 6. Untreated control embryos received exclusively 50 µl saline solution. To identify cells in proliferation and cell cycle proteins, as well as Pax1/9 and Sox9 proteins, we performed immunolocalization and flow cytometry analyses using the antibodies anti-phosphohistone H3, anti-p53, anti-p21, anti-proliferating cell nuclear antigen, anti-Pax1, anti-Pax9 and anti-Sox9. No significant differences in cell proliferation were observed between Hcy-treated and untreated embryos. We observed a decrease of the proliferating cell nuclear antigen and p21 proteins, both involved in the G1 phase of cell cycle progression. On the other hand, in mesenchymal cells of the limbs, Hcy induces an increase of p53 protein, which can be activated by DNA damage. In cell differentiation, Hcy induced an increase mainly of Pax9 and Sox9 proteins. Our data indicate that the treatment with Hcy changes the mesenchymal cell dynamics during limb development, but does not change the morphology of the cartilage molds. These findings provide information to understand better the cellular basis of the toxicity of Hcy on chondrogenesis during limb development.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chondrogenesis/drug effects , Homocysteine/pharmacology , Mesenchymal Stem Cells/drug effects , Organogenesis/drug effects , Animals , Chick Embryo , DNA Damage , Extremities/embryology , Mesenchymal Stem Cells/metabolism , PAX9 Transcription Factor/genetics , PAX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;47(8): 637-645, 08/2014. tab, graf
Article in English | LILACS | ID: lil-716279

ABSTRACT

Tissue engineering encapsulated cells such as chondrocytes in the carrier matrix have been widely used to repair cartilage defects. However, chondrocyte phenotype is easily lost when chondrocytes are expanded in vitro by a process defined as “dedifferentiation”. To ensure successful therapy, an effective pro-chondrogenic agent is necessary to overcome the obstacle of limited cell numbers in the restoration process, and dedifferentiation is a prerequisite. Gallic acid (GA) has been used in the treatment of arthritis, but its biocompatibility is inferior to that of other compounds. In this study, we modified GA by incorporating sulfamonomethoxine sodium and synthesized a sulfonamido-based gallate, JJYMD-C, and evaluated its effect on chondrocyte metabolism. Our results showed that JJYMD-C could effectively increase the levels of the collagen II, Sox9, and aggrecan genes, promote chondrocyte growth, and enhance secretion and synthesis of cartilage extracellular matrix. On the other hand, expression of the collagen I gene was effectively down-regulated, demonstrating inhibition of chondrocyte dedifferentiation by JJYMD-C. Hypertrophy, as a characteristic of chondrocyte ossification, was undetectable in the JJYMD-C groups. We used JJYMD-C at doses of 0.125, 0.25, and 0.5 µg/mL, and the strongest response was observed with 0.25 µg/mL. This study provides a basis for further studies on a novel agent in the treatment of articular cartilage defects.


Subject(s)
Animals , Rabbits , Benzamides/chemical synthesis , Cell Dedifferentiation/drug effects , Cell Proliferation/drug effects , Chondrocytes/drug effects , Phenotype , Pyrimidines/chemical synthesis , Aggrecans/genetics , Aggrecans/metabolism , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Benzamides/pharmacology , Cell Survival , Cell Dedifferentiation/immunology , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrogenesis/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type II/genetics , Collagen Type II/metabolism , Glycosaminoglycans/analysis , Immunohistochemistry , Laser Scanning Cytometry , Primary Cell Culture , Pyrimidines/pharmacology , Real-Time Polymerase Chain Reaction , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Tissue Engineering
14.
Braz J Med Biol Res ; 47(8): 637-45, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25003544

ABSTRACT

Tissue engineering encapsulated cells such as chondrocytes in the carrier matrix have been widely used to repair cartilage defects. However, chondrocyte phenotype is easily lost when chondrocytes are expanded in vitro by a process defined as "dedifferentiation". To ensure successful therapy, an effective pro-chondrogenic agent is necessary to overcome the obstacle of limited cell numbers in the restoration process, and dedifferentiation is a prerequisite. Gallic acid (GA) has been used in the treatment of arthritis, but its biocompatibility is inferior to that of other compounds. In this study, we modified GA by incorporating sulfamonomethoxine sodium and synthesized a sulfonamido-based gallate, JJYMD-C, and evaluated its effect on chondrocyte metabolism. Our results showed that JJYMD-C could effectively increase the levels of the collagen II, Sox9, and aggrecan genes, promote chondrocyte growth, and enhance secretion and synthesis of cartilage extracellular matrix. On the other hand, expression of the collagen I gene was effectively down-regulated, demonstrating inhibition of chondrocyte dedifferentiation by JJYMD-C. Hypertrophy, as a characteristic of chondrocyte ossification, was undetectable in the JJYMD-C groups. We used JJYMD-C at doses of 0.125, 0.25, and 0.5 µg/mL, and the strongest response was observed with 0.25 µg/mL. This study provides a basis for further studies on a novel agent in the treatment of articular cartilage defects.


Subject(s)
Benzamides/chemical synthesis , Cell Dedifferentiation/drug effects , Cell Proliferation/drug effects , Chondrocytes/drug effects , Phenotype , Pyrimidines/chemical synthesis , Aggrecans/genetics , Aggrecans/metabolism , Animals , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Benzamides/pharmacology , Cell Dedifferentiation/immunology , Cell Survival , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrogenesis/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type II/genetics , Collagen Type II/metabolism , Glycosaminoglycans/analysis , Immunohistochemistry , Laser Scanning Cytometry , Primary Cell Culture , Pyrimidines/pharmacology , Rabbits , Real-Time Polymerase Chain Reaction , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Tissue Engineering
15.
J Dent Res ; 93(4): 335-45, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24389809

ABSTRACT

Bone morphogenetic proteins (BMPs) are members of the TGF-ß superfamily, acting as potent regulators during embryogenesis and bone and cartilage formation and repair. Cell and molecular biology approaches have unveiled the great complexity of BMP action, later confirmed by transgenic animal studies. Genetic engineering allows for the production of large amounts of BMPs for clinical use, but they have systematically been associated with a delivery system, such as type I collagen and calcium phosphate ceramics, to ensure controlled release and to maximize their biological activity at the surgical site, avoiding systemic diffusion. Clinical orthopedic studies have shown the benefits of FDA-approved recombinant human BMPs (rhBMPs) 2 and 7, but side effects, such as swelling, seroma, and increased cancer risk, have been reported, probably due to high BMP dosage. Several studies have supported the use of BMPs in periodontal regeneration, sinus lift bone-grafting, and non-unions in oral surgery. However, the clinical use of BMPs is growing mainly in off-label applications, with robust evidence to ascertain rhBMPs' safety and efficacy through well-designed, randomized, and double-blind clinical trials. Here we review and discuss the critical data on BMP structure, mechanisms of action, and possible clinical applications.


Subject(s)
Bone Morphogenetic Proteins/physiology , Bone Morphogenetic Protein 2/therapeutic use , Bone Morphogenetic Protein 7/therapeutic use , Bone Morphogenetic Proteins/therapeutic use , Bone Regeneration/drug effects , Chondrogenesis/drug effects , Drug Delivery Systems , Humans , Osteogenesis/drug effects , Recombinant Proteins/therapeutic use , Signal Transduction/physiology , Structure-Activity Relationship , Transforming Growth Factor beta/therapeutic use
16.
Mol Cell Biomech ; 10(2): 85-105, 2013 Jun.
Article in English | MEDLINE | ID: mdl-24015478

ABSTRACT

The electrospinning technique is a method used to produce nano and microfibers using the influence of electrostatic forces. Porous three dimensional networks of continuous and interconnected fibers as scaffolds were obtained from a poly (lactic acid) solution. The concentration of the polymeric solution, 12.5% m/w, as well as the conditions of voltage (V = 11kV) and tip-metallic collector distance (H = 13cm) were established to develop these scaffolds through the electrospinning process. The characteristics of the scaffolds, such as fiber diameter, sintering and the biomimetics of the characteristics of a native extra cellular matrix were verified by Scanning Electron Microscopy (SEM). The orientation induced in the material as a consequence of the electrospinning forces was studied by Differential Scanning Calorimetry (DSC) and X-Ray Diffraction (XRD).The same techniques were used to study the hydrolytic degradation of samples in a ringer solution (pH = 7-7.4 at 37 degrees C) for 12 weeks and showed evidences of superficial degradation on the microfibers. The suitability of these scaffolds for tissue engineering was studied through the primary cell culture of chondrocytes, by observing adhesion and cellular proliferation developed during 14 days of assay.


Subject(s)
Biocompatible Materials/chemical synthesis , Chondrocytes/cytology , Chondrocytes/physiology , Lactic Acid/chemistry , Polymers/chemistry , Tissue Engineering/instrumentation , Tissue Scaffolds , Biocompatible Materials/pharmacology , Cells, Cultured , Chondrocytes/drug effects , Chondrogenesis/drug effects , Chondrogenesis/physiology , Electrochemistry/instrumentation , Electrochemistry/methods , Equipment Design , Equipment Failure Analysis , Humans , Lactic Acid/pharmacology , Materials Testing , Polyesters , Polymers/pharmacology , Rotation , Surface Properties
17.
Rheumatol Int ; 33(1): 121-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22238025

ABSTRACT

Umbilical cord blood contains undifferentiated mesenchymal stem cells (MSCs) with chondrogenic potential that may be used for the repair of joint damage. The role of growth factors during the process of chondrogenesis is still not entirely understood. The objective of this study was to evaluate the formation of chondrocytes, cartilaginous matrix and type II collagen from human umbilical cord blood stem cells exposed to two different growth factors, BMP-6 and BMP-2, while being cultured as a micromass or a monolayer. Umbilical cord blood was obtained from full-term deliveries, and then, mononuclear cells were separated and cultured for expansion. Afterward, these cells were evaluated by flow cytometry using antibodies specific for MSCs and induced to chondrogenic differentiation in micromass and monolayer cultures supplemented with BMP-2 and BMP-6. Cellular phenotype was evaluated after 7, 14 and 21 days by RT-PCR and Western blot analysis to identify the type II collagen and aggrecan. The expanded cells displayed surface antigens characteristic of mesenchymal progenitor cells and were negative for hematopoietic differentiation antigens. Type II collagen and aggrecan mRNAs were expressed from day 14 in cells stimulated with BMP-2 or BMP-6. Type II collagen was demonstrated by Western blotting in both groups, and the greatest expression was observed 21 days after the cells were stimulated with BMP-2 cultured in micromass. BMP-2 in micromass culture was more efficient to induce the chondrogenesis.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 6/pharmacology , Chondrocytes/drug effects , Chondrogenesis/drug effects , Mesenchymal Stem Cells/drug effects , Aggrecans/genetics , Aggrecans/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Chondrocytes/metabolism , Chondrogenesis/physiology , Collagen Type II/genetics , Collagen Type II/metabolism , Extracellular Matrix Proteins/metabolism , Fetal Blood/cytology , Gene Expression/drug effects , Humans , Mesenchymal Stem Cells/metabolism
18.
PLoS One ; 7(6): e39885, 2012.
Article in English | MEDLINE | ID: mdl-22768154

ABSTRACT

Dental pulp (DP) can be extracted from child's primary teeth (deciduous), whose loss occurs spontaneously by about 5 to 12 years. Thus, DP presents an easy accessible source of stem cells without ethical concerns. Substantial quantities of stem cells of an excellent quality and at early (2-5) passages are necessary for clinical use, which currently is a problem for use of adult stem cells. Herein, DPs were cultured generating stem cells at least during six months through multiple mechanical transfers into a new culture dish every 3-4 days. We compared stem cells isolated from the same DP before (early population, EP) and six months after several mechanical transfers (late population, LP). No changes, in both EP and LP, were observed in morphology, expression of stem cells markers (nestin, vimentin, fibronectin, SH2, SH3 and Oct3/4), chondrogenic and myogenic differentiation potential, even after cryopreservation. Six hours after DP extraction and in vitro plating, rare 5-bromo-2'-deoxyuridine (BrdU) positive cells were observed in pulp central part. After 72 hours, BrdU positive cells increased in number and were found in DP periphery, thus originating a multicellular population of stem cells of high purity. Multiple stem cell niches were identified in different zones of DP, because abundant expression of nestin, vimentin and Oct3/4 proteins was observed, while STRO-1 protein localization was restricted to perivascular niche. Our finding is of importance for the future of stem cell therapies, providing scaling-up of stem cells at early passages with minimum risk of losing their "stemness".


Subject(s)
Cell Separation/methods , Dental Pulp/cytology , Stem Cell Niche , Stem Cells/cytology , Biomarkers/metabolism , Bromodeoxyuridine/metabolism , Cell Count , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Child , Chondrogenesis/drug effects , Culture Media/pharmacology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Gene Expression Regulation/drug effects , Humans , Immunophenotyping , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Muscle Development/drug effects , Stem Cell Niche/drug effects , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/ultrastructure , Time Factors
19.
Clinics (Sao Paulo) ; 66(3): 487-92, 2011.
Article in English | MEDLINE | ID: mdl-21552678

ABSTRACT

INTRODUCTION: Numerous experimental efforts have been undertaken to induce the healing of lesions within articular cartilage by re-establishing competent repair tissue. Adult mesenchymal stem cells have attracted attention as a source of cells for cartilage tissue engineering. The purpose of this study was to investigate chondrogenesis employing periosteal mesenchymal cells. METHODS: Periosteum was harvested from patients who underwent orthopedic surgeries. Mesenchymal stem cells were characterized through flow cytometry using specific antibodies. The stem cells were divided into four groups. Two groups were stimulated with transforming growth factor ß3 (TGF-ß3), of which one group was cultivated in a monolayer culture and the other was cultured in a micromass culture. The remaining two groups were cultivated in monolayer or micromass cultures in the absence of TGF-ß3. Cell differentiation was verified through quantitative reverse transcription-polymerase chain reaction (RT-PCR) and using western blot analysis. RESULT: In the groups cultured without TGF-ß3, only the cells maintained in the micromass culture expressed type II collagen. Both the monolayer and the micromass groups that were stimulated with TGF-ß3 expressed type II collagen, which was observed in both quantitative RT-PCR and western blot analysis. The expression of type II collagen was significantly greater in the micromass system than in the monolayer system. CONCLUSION: The results of this study demonstrate that the interactions between the cells in the micromass culture system can regulate the proliferation and differentiation of periosteal mesenchymal cells during chondrogenesis and that this effect is enhanced by TGF-ß3.


Subject(s)
Chondrogenesis/drug effects , Mesenchymal Stem Cells/cytology , Periosteum/cytology , Transforming Growth Factor beta3/pharmacology , Adult , Analysis of Variance , Blotting, Western , Cell Differentiation , Cells, Cultured , Collagen Type II/biosynthesis , Gene Expression , Humans , Mesenchymal Stem Cells/drug effects , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction
20.
Clinics ; Clinics;66(3): 487-492, 2011. ilus, tab
Article in English | LILACS | ID: lil-585963

ABSTRACT

INTRODUCTION: Numerous experimental efforts have been undertaken to induce the healing of lesions within articular cartilage by re-establishing competent repair tissue. Adult mesenchymal stem cells have attracted attention as a source of cells for cartilage tissue engineering. The purpose of this study was to investigate chondrogenesis employing periosteal mesenchymal cells. METHODS: Periosteum was harvested from patients who underwent orthopedic surgeries. Mesenchymal stem cells were characterized through flow cytometry using specific antibodies. The stem cells were divided into four groups. Two groups were stimulated with transforming growth factor β3 (TGF-β3), of which one group was cultivated in a monolayer culture and the other was cultured in a micromass culture. The remaining two groups were cultivated in monolayer or micromass cultures in the absence of TGF-β3. Cell differentiation was verified through quantitative reverse transcription-polymerase chain reaction (RT-PCR) and using western blot analysis. RESULT: In the groups cultured without TGF-β3, only the cells maintained in the micromass culture expressed type II collagen. Both the monolayer and the micromass groups that were stimulated with TGF-β3 expressed type II collagen, which was observed in both quantitative RT-PCR and western blot analysis. The expression of type II collagen was significantly greater in the micromass system than in the monolayer system. CONCLUSION: The results of this study demonstrate that the interactions between the cells in the micromass culture system can regulate the proliferation and differentiation of periosteal mesenchymal cells during chondrogenesis and that this effect is enhanced by TGF-β3.


Subject(s)
Adult , Humans , Middle Aged , Chondrogenesis/drug effects , Mesenchymal Stem Cells , Periosteum/cytology , /pharmacology , Analysis of Variance , Blotting, Western , Cell Differentiation , Cells, Cultured , Collagen Type II/biosynthesis , Gene Expression , Mesenchymal Stem Cells , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL