Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
1.
Biomacromolecules ; 25(6): 3312-3324, 2024 Jun 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.
Stem Cell Res Ther ; 14(1): 70, 2023 04 07.
Article in English | MEDLINE | ID: mdl-37024989

ABSTRACT

BACKGROUND: Adipose tissue-derived stromal vascular fraction (SVF) harbors multipotent cells with potential therapeutic relevance. We developed a method to form adipose spheroids (AS) from the SVF with complex organoid structure and enhanced leptin secretion upon insulin stimulation. METHODS: SVF was generated from the interscapular brown adipose tissue of newborn mice. Immunophenotype and stemness of cultured SVF were determined by flow cytometry and in vitro differentiation, respectively. Spheroids were generated in hanging drops and non-adherent plates and compared by morphometric methods. The adipogenic potential was compared between preadipocyte monolayers and spheroids. Extracellular leptin was quantified by immunoassay. Lipolysis was stimulated with isoprenaline and quantified by colorimetric methods. AS viability and ultrastructure were determined by confocal and transmission electron microscopy analyses. RESULTS: Cultured SVF contained Sca1 + CD29 + CD44 + CD11b- CD45- CD90- cells with adipogenic and chondrogenic but no osteogenic potential. Culture on non-adherent plates yielded the highest quantity and biggest size of spheroids. Differentiation of AS for 15 days in a culture medium supplemented with insulin and rosiglitazone resulted in greater Pparg, Plin1, and Lep expression compared to differentiated adipocytes monolayers. AS were viable and maintained leptin secretion even in the absence of adipogenic stimulation. Glycerol release after isoprenaline stimulation was higher in AS compared to adipocytes in monolayers. AS were composed of outer layers of unilocular mature adipocytes and an inner structure composed of preadipocytes, immature adipocytes and an abundant loose extracellular matrix. CONCLUSION: Newborn mice adipose SVF can be efficiently differentiated into leptin-secreting AS. Prolonged stimulation with insulin and rosiglitazone allows the formation of structurally complex adipose organoids able to respond to adrenergic lipolytic stimulation.


Subject(s)
Adipocytes , Adipose Tissue, Brown , Cell Differentiation , Leptin , Leptin/metabolism , Organoids , Insulin/pharmacology , Animals , Mice , Adipose Tissue, Brown/cytology , Rosiglitazone/pharmacology , Cells, Cultured , Animals, Newborn , Immunophenotyping , Osteogenesis , Chondrogenesis , Adipocytes/ultrastructure , Lipolysis , Primary Cell Culture
3.
Cells ; 11(16)2022 08 17.
Article in English | MEDLINE | ID: mdl-36010629

ABSTRACT

The preservation of the chondrogenic phenotype and hypoxia-related physiological microenvironment are major challenges in the 2D culture of primary human chondrocytes. To address this problem, we develop a 3D culture system generating scaffold-free spheroids from human chondrocytes. Our results highlight the chondrogenic potential of cultured human articular chondrocytes in a 3D system combined with hypoxia independently of the cartilage source. After 14 days of culture, we developed spheroids with homogenous diameter and shape from hyaline cartilage donors. Spheroids generated in hypoxia showed a significantly increased glycosaminoglycans synthesis and up-regulated the expression of SOX9, ACAN, COL2A1, COMP, and SNAI1 compared to those obtained under normoxic conditions. Therefore, we conclude that spheroids developed under hypoxic conditions modulate the expression of chondrogenesis-related genes and native tissue features better than 2D cultures. Thus, this scaffold-free 3D culture system represents a novel in vitro model that can be used for cartilage biology research.


Subject(s)
Cartilage, Articular , Chondrocytes , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Chondrogenesis , Humans , Hypoxia/metabolism
4.
Bone ; 154: 116186, 2022 01.
Article in English | MEDLINE | ID: mdl-34520899

ABSTRACT

OBJECTIVE: The process of longitudinal bone growth occurs at the growth plate where the chondrocytes undergo apparent structural and molecular changes to promote growth. Recent reports suggest that radial shockwave treatment (rSWT) stimulates bone length in cultured fetal rat metatarsals. Therefore, we investigated if rSWT has similar growth promoting effects on cultured human growth plate fragments and addressed the same in a preclinical in vivo rabbit model by subjecting their growth plates to rSWT. METHODS: Short-term effects of high-energy rSWT were evaluated in a unique model of cultured human growth plate cartilage (n = 5) wherein samples exposed to rSWT were assessed for chondrogenic markers at 24 h in comparison to unexposed samples obtained from the same limb. Local in vivo effects were studied in six-week-old rabbits who had their distal femurs exposed to four weekly sessions of rSWT at low- and high-energy levels (n = 4 each). At sacrifice, histomorphometric and immunohistochemistry analyses were performed. For effect on longitudinal growth, proximal tibiae of 22-week-old rabbits (n = 12) were asymmetrically exposed to rSWT; the contralateral side served as untreated controls. At sacrifice, the final bone length was measured. RESULTS: In the ex vivo model of cultured human growth plate cartilage, rSWT exposure upregulated SOX9 and COL2A1 compared to control. In the immature rabbit model, an increased number of proliferative chondrocytes and column density was seen for both the energy levels. In the adolescent rabbits, an increase in tibial length was observed after the fourth session of high-energy rSWT and until six-weeks after rSWT compared to the untreated limb. CONCLUSIONS: Our preliminary experimental results suggest that rSWT may serve as a non-invasive treatment and possibly a safe strategy to stimulate longitudinal bone growth. However, further studies are needed to assess the in vivo effects of rSWT in models of disturbed bone growth.


Subject(s)
Chondrogenesis , Growth Plate , Animals , Bone Development , Cartilage , Chondrocytes , Humans , Rabbits , Rats
5.
Ciênc. rural (Online) ; 52(12): e20210509, 2022. graf, ilus
Article in English | VETINDEX | ID: biblio-1375155

ABSTRACT

The objective of this study was to investigate the in vitro action of triiodothyronine (T3) on the chondrogenic differentiation of adipose tissue-derived stem cells (ASCs) of female rats, with different time periods and doses. ASCs were extracted from female Wistar rats and were cultured in chondrogenic medium with and without the presence of T3. Five groups were established: 1) ASCs without T3; and 2,3,4,5) ASCs with 0.01, 1, 100 and 1,000 nM T3, respectively). After 7, 14 and 21 days, cell morphology, chondrogenic matrix formation, and expression of Sox9, aggrecan, collagen II, and collagen X were evaluated. The Student-Newman-Keuls test was used. ASCs showed CD54, CD73, and CD90 before chondrogenic differentiation. The hormone treatment did not alter chondrogenic matrix formation, Sox9 expression at 14 or 21 days, or expression of collagen II or collagen X at any time. However, the 0.01, 1, and 1000 nM T3 doses decreased Sox9 expression at 7 days. In conclusion, chondrogenic differentiation of ASCs of female rats is not influenced by T3.


O objetivo do presente trabalho foi verificar o efeito in vitro da triiodotironina (T3) na diferenciação condrogênica de células tronco mesenquimais do tecido adiposo (CTM-TA) de ratas, durante vários períodos e em várias doses. CTM-TA foram coletadas de ratas Wistar e cultivadas em meio condrogênico com ou sem a presença de T3. Constitui-se cinco grupos: 1) CTM-TA sem T3; e 2,3,4,5) CTM-TA com T3 (0,01; 1; 100 e 1000 nM, respectivamente). Após sete, 14 e 21 dias, foram avaliados morfologia celular, formação de matriz condrogênica e expressão de Sox9, agrecano, colágeno II e colágeno X. Para as análises foi utilizado o teste de Student Newman Keuls. CTM-TA expressaram CD54, CD73 e CD90 antes da diferenciação condrogênica. O tratamento hormonal não alterou a formação de matriz condrogênica e a expressão de Sox9 aos 14 e 21 dias e expressão dos colágenos II e X em nenhum dos períodos avaliados. No entanto, as doses de 0,01; 1 e 1000 nM T3 diminuíram a expressão de Sox9 aos 7 dias. Conclui-se que a diferenciação condrogênica de CTM-TA de ratas não é influenciada pela T3.


Subject(s)
Animals , Female , Rats , Stem Cells/physiology , Triiodothyronine/analysis , Adipose Tissue/physiology , Chondrogenesis/physiology
6.
Cells ; 10(9)2021 09 05.
Article in English | MEDLINE | ID: mdl-34571972

ABSTRACT

Cell therapy strategies using mesenchymal stem cells (MSCs) carried in fibrin glue have shown promising results in regenerative medicine. MSCs are crucial for tissue healing because they have angiogenic, anti-apoptotic and immunomodulatory properties, in addition to the ability to differentiate into several specialized cell lines. Fibrin sealant or fibrin glue is a natural polymer involved in the coagulation process. Fibrin glue provides a temporary structure that favors angiogenesis, extracellular matrix deposition and cell-matrix interactions. Additionally, fibrin glue maintains the local and paracrine functions of MSCs, providing tissue regeneration through less invasive clinical procedures. Thus, the objective of this systematic review was to assess the potential of fibrin glue combined with MSCs in bone or cartilage regeneration. The bibliographic search was performed in the PubMed/MEDLINE, LILACS and Embase databases, using the descriptors ("fibrin sealant" OR "fibrin glue") AND "stem cells" AND "bone regeneration", considering articles published until 2021. In this case, 12 preclinical and five clinical studies were selected to compose this review, according to the eligibility criteria. In preclinical studies, fibrin glue loaded with MSCs, alone or associated with bone substitute, significantly favored bone defects regeneration compared to scaffold without cells. Similarly, fibrin glue loaded with MSCs presented considerable potential to regenerate joint cartilage injuries and multiple bone fractures, with significant improvement in clinical parameters and absence of postoperative complications. Therefore, there is clear evidence in the literature that fibrin glue loaded with MSCs, alone or combined with bone substitute, is a promising strategy for treating lesions in bone or cartilaginous tissue.


Subject(s)
Bone Regeneration , Chondrogenesis , Fibrin Tissue Adhesive/therapeutic use , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Osteogenesis , Regenerative Medicine , Tissue Scaffolds , Animals , Fibrin Tissue Adhesive/adverse effects , Humans , Mesenchymal Stem Cell Transplantation/adverse effects , Models, Animal , Rabbits , Rats , Treatment Outcome , Wound Healing
7.
Rev. Bras. Ortop. (Online) ; 56(3): 333-339, May-June 2021. tab, graf
Article in English | LILACS | ID: biblio-1288682

ABSTRACT

Abstract Objective To evaluate clinically and radiologically the results of the treatment of chondral lesions using collagen membrane - autologous matrix-induced chondrogenesis (AMIC). Methods This is a series of observational cases, in which 15 patients undergoing AMIC were analyzed. The clinical evaluation was made by comparing the Lysholm and International Knee Document Commitee (IKDC) scores in the pre- and postoperative period of 12 months, and radiological evaluation using the Magnetic Resonance Observation of Cartilage Repair Tissue (MOCART) score in the same postoperative period. Results The mean age of the patients was 39.2 years old, and the mean size of the chondral lesions was 1.55cm2. There was a significant improvement in clinical scores, with a mean increase of 24.6 points on Lysholm and of 24.3 on IKDC after 12 months. In the radiological evaluation, MOCART had a mean of 65 points. It was observed that the larger the size of the lesion, the greater the improvement in scores. Conclusion Evaluating subjective clinical scores, the treatment of chondral lesions with the collagen membrane showed good results, as well as the evaluation of MOCART, with greater benefit in larger lesions.


Resumo Objetivo Avaliar clínica e radiologicamente os resultados do tratamento das lesões condrais com a membrana de colágeno - condrogênese autóloga induzida por matriz. Métodos Trata-se de uma série de casos observacional, na qual foram analisados 15 pacientes submetidos a condrogênese autóloga induzida por matriz. A avaliação clínica foi feita comparando os escores de Lysholm e International Knee Document Commitee (IKDC, na sigla em inglês) no pré- e pós-operatório de 12 meses, e avaliação radiológica através do escore de Magnetic Resonance Observation of Cartilage Repair Tissue (MOCART, na sigla em inglês) no mesmo período de pós-operatório. Resultados A média de idade dos pacientes foi 39,2 anos, e a média do tamanho das lesões condrais foi de 1,55cm2. Houve uma melhora significativa nos escores clínicos, com média de aumento de 24,6 pontos no Lysholm e de 24,3 no IKDC, após 12 meses. Na avaliação radiológica, o MOCART teve média de 65 pontos. Observou-se que quanto maior o tamanho da lesão, maior foi a melhora nos escores. Conclusão Avaliando escores clínicos subjetivos, o tratamento das lesões condrais com a membrana de colágeno mostrou bons resultados, assim como a avaliação de MOCART, com maior benefício em lesões maiores.


Subject(s)
Humans , Male , Female , Adolescent , Adult , Middle Aged , Postoperative Period , Magnetic Resonance Spectroscopy , Cartilage, Articular , Collagen , Chondrogenesis , Knee Injuries
8.
Sci Rep ; 11(1): 3063, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33542256

ABSTRACT

Articular chondral lesions, caused either by trauma or chronic cartilage diseases such as osteoarthritis, present very low ability to self-regenerate. Thus, their current management is basically symptomatic, progressing very often to invasive procedures or even arthroplasties. The use of amniotic fluid stem cells (AFSCs), due to their multipotentiality and plasticity, associated with scaffolds, is a promising alternative for the reconstruction of articular cartilage. Therefore, this study aimed to investigate the chondrogenic potential of AFSCs in a micromass system (high-density cell culture) under insulin-like growth factor 1 (IGF-1) stimuli, as well as to look at their potential to differentiate directly when cultured in a porous chitosan-xanthan (CX) scaffold. The experiments were performed with a CD117 positive cell population, with expression of markers (CD117, SSEA-4, Oct-4 and NANOG), selected from AFSCs, after immunomagnetic separation. The cells were cultured in both a micromass system and directly in the scaffold, in the presence of IGF-1. Differentiation to chondrocytes was confirmed by histology and by using immunohistochemistry. The construct cell-scaffold was also analyzed by scanning electron microscopy (SEM). The results demonstrated the chondrogenic potential of AFSCs cultivated directly in CX scaffolds and also in the micromass system. Such findings support and stimulate future studies using these constructs in osteoarthritic animal models.


Subject(s)
Adult Stem Cells/cytology , Cartilage, Articular/drug effects , Chondrogenesis/genetics , Osteoarthritis/genetics , Tissue Scaffolds/chemistry , Adult Stem Cells/transplantation , Amniotic Fluid/cytology , Cartilage, Articular/growth & development , Cartilage, Articular/ultrastructure , Cell Culture Techniques , Cell Differentiation/drug effects , Chitosan/pharmacology , Gene Expression Regulation, Developmental/drug effects , Humans , Insulin-Like Growth Factor I/pharmacology , Microscopy, Electron, Scanning , Osteoarthritis/pathology , Osteoarthritis/therapy , Polysaccharides, Bacterial/pharmacology , Proto-Oncogene Proteins c-kit/genetics , Tissue Engineering/methods
9.
Dev Biol ; 469: 86-95, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33058830

ABSTRACT

During digit development, the correct balance of chondrogenic signals ensures the recruitment of undifferentiated cells into the cartilage lineage or the maintenance of cells at the undifferentiated stage. WNT/ß catenin maintains the pool of progenitor cells, whereas TGFß signalling promotes cartilage differentiation by inducing Sox9 expression. Moreover, WNT5A promotes the degradation of ß catenin during mouse limb development. Although these mechanisms are well established, it is still unknown whether the signalling pathway downstream WNT5A is also involved in early chondrogenesis during digit formation. Thus, the aim of this study was to determine the role of WNT5A during the recruitment of progenitor cells during digit development. Our results showed that WNT5A activated calcium (Ca2+) release in the undifferentiated region during digit development. Further, the blockade of Ca2+ release or calcineurin (CaN) or nuclear factor of activated T-cells (NFAT) functions resulted in an inhibition of cartilage differentiation. Together, our results demonstrate that non canonical WNT5A-Ca2+-CaN-NFAT signalling plays a key role during embryonic digit development in vivo promoting the competence for chondrogenic signals and also acts as a permissive factor for chondrogenesis independently of cell death mechanisms.


Subject(s)
Calcium Signaling , Chondrogenesis , NFATC Transcription Factors/metabolism , Toes/embryology , Wnt-5a Protein/physiology , Animals , Calcineurin/metabolism , Calcium/metabolism , Chick Embryo , Extremities/embryology , SOX9 Transcription Factor/metabolism
10.
Knee Surg Sports Traumatol Arthrosc ; 29(6): 1990-1999, 2021 Jun.
Article in English | MEDLINE | ID: mdl-32852566

ABSTRACT

PURPOSE: To examine healing adaptations over 17 weeks post Achilles tendon (AT) rupture in the injured region (IR) compared to an uninjured region (UIR) of the AT. METHODS: Twenty-four rats were subjected to a complete right-sided AT rupture, while the left side served as a control. ATs were harvested at 1, 2, 8 and 17 weeks post-rupture and stained with antibodies specific to Collagen type I (Col I) and II (Col II) as well as Alcian Blue and Picrosirius Red staining techniques. Histopathological changes, proteoglycan content, collagen alignment and immunoexpression were assessed. RESULTS: Both regions examined, IR and UIR, exhibited over weeks 1-17 similar healing adaptations of increasing collagen alignment, decreasing Col I immunoexpression, as well as increasing proteoglycan content and Col II occurrence. Increased proteoglycan content was found already at week 2 in the UIR, while it first increased at week 8 in the IR. The area positive to Col II was increased compared to controls at week 8 in the UIR, whereas it first raised at week 17 in the IR. Collagen disorganization successively declined to reach control levels at week 17 in the UIR, but was still higher in the IR. CONCLUSION: This study demonstrated that uninjured areas of the AT remote from the rupture site also undergo pronounced remodeling, although with time-span differences relative to injured AT portions. These changes including the pathologic heterotopic mineralization and chondrogenic differentiation observed in both regions may have implications in the choice of rehabilitation regimes in order to prevent secondary rupture.


Subject(s)
Achilles Tendon/injuries , Achilles Tendon/physiopathology , Wound Healing/physiology , Achilles Tendon/pathology , Animals , Chondrogenesis , Collagen Type I/metabolism , Collagen Type II/metabolism , Female , Models, Animal , Proteoglycans/metabolism , Rats, Sprague-Dawley , Rupture/pathology , Rupture/physiopathology
11.
J Biomater Sci Polym Ed ; 32(4): 504-523, 2021 03.
Article in English | MEDLINE | ID: mdl-33176594

ABSTRACT

Cartilage tissue engineering (CTE) has the general objective of restoring and improving damaged cartilage. A very interesting strategy of CTE is to combine different polymers to obtain a viscoelastic material. In this study, we have evaluated the applicability of poly(2-hydroxyethyl methacrylate) networks semi-interpenetrated with sodium alginate for CTE. Alginate-containing hydrogels show an increase in scaffold porosity and swelling capacity, when compared with nonporous poly(2-hydroxyethyl methacrylate) scaffolds. Primary chondrocytes from young rats were cultured on the hydrogels, and an increase in chondrocyte proliferation and chondrocytic markers was observed in alginate-containing hydrogels. Chondrocytic phenotype was preserved on hydrogels containing the lowest amount of crosslinker and initiator (SEMI 3 and SEMI 4). In addition, nitric oxide production by RAW264.7 macrophages grown on hydrogels was tested, and none of the hydrogels showed high levels of this inflammatory marker after 2 days. These results indicate that our alginate-containing hydrogels could be useful for CTE.


Subject(s)
Alginates , Hydrogels , Animals , Chondrocytes , Chondrogenesis , Methacrylates , Rats , Tissue Engineering
12.
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
13.
Tissue Eng Part A ; 27(5-6): 311-327, 2021 03.
Article in English | MEDLINE | ID: mdl-30734654

ABSTRACT

A stabilized cartilage construct without signs of hypertrophy in chondrocytes is still a challenge. Suspensions of adipose stem/stromal cells (ASCs) and cartilage progenitor cells (CPCs) were seeded into micromolded nonadhesive hydrogel to produce spheroids (scaffold- and serum-free method) characterized by size, immunohistochemistry, fusion, and biomechanical properties. After cell dissociation, they were characterized for mesenchymal cell surface markers, cell viability, and quantitative real-time polymerase chain reaction. Both targeted and nontargeted (shotgun mass spectrometry) analyses were conducted on the culture supernatants. Induced ASC spheroids (ø = 350 µm) showed high cell viability and CD73 downregulation contrasting to CD90. The transforming growth factor (TGF)-ß3/TGF-ß1 ratio and SOX9 increased (p < 0.05), whereas interleukin (IL)-6, IL-8, RUNX2, and ALPL decreased. Induced ASC spheroids were able to completely fuse and showed a higher force required to compression at day 14 (p < 0.0001). Strong collagen type II in situ was associated with gradual decrease of collagen type X and a lower COLXA1 gene expression at day 14 compared with day 7 (p = 0.0352). The comparison of the secretome content of induced and non-induced ASCs and CPCs identified 138 proteins directly relevant to chondrogenesis of 704 proteins in total. Although collagen X was absent, thrombospondin-1 (TSP-1), described as antiangiogenic and antihypertrophic, and cartilage oligomeric matrix protein (COMP), a biomarker of chondrogenesis, were upregulated in induced ASC spheroids. Our scaffold- and serum-free method mimics stable cartilage acting as a tool for biomarker discovery and for regenerative medicine protocols. Impact Statement Promising adult stem cell sources for cartilage regeneration include adipose stem/stromal cells (ASCs) from subcutaneous adipose tissue. Our main objective was the development of a reproducible and easy-to-handle scaffold- and serum-free method to obtain stable cartilage from induced ASC spheroids. In addition to targeted protein profiling and biomechanical analysis, we provide the first characterization of the secretome composition for ASC spheroids, providing a useful tool to monitor in vitro chondrogenesis and a noninvasive quality control of tissue-engineered constructs. Furthermore, our secretome analysis revealed a potential novel biomarker-thrombospondin-1 (TSP-1), known by its antiangiogenic properties and recently described as an antihypertrophic protein.


Subject(s)
Cartilage , Mesenchymal Stem Cells , Adipose Tissue , Cell Differentiation , Cells, Cultured , Chondrocytes , Chondrogenesis , Humans , Thrombospondin 1 , Tissue Engineering
14.
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
15.
In Vitro Cell Dev Biol Anim ; 56(5): 399-411, 2020 May.
Article in English | MEDLINE | ID: mdl-32535758

ABSTRACT

Murine 3T3 cell lines constitute a standard model system for in vitro study of mammalian adipogenesis although they do not faithfully reflect the biology of the human adipose cells. Several human adipose cell lines and strains have been used to recapitulate human adipogenesis in vitro, but to date there is no generally accepted in vitro model for human adipogenesis. We obtained a clonal strain of human subcutaneous adipose stromal cells, IPI-SA3-C4, and characterized its utility as an in vitro model for human subcutaneous adipogenesis. IPI-SA3-C4 cells showed a high proliferative potential for at least 30 serial passages, reached 70 cumulative population doublings and exhibited a population doubling time of 47 h and colony forming efficiency of 12% at the 57th cumulative population doublings. IPI-SA3-C4 cells remained diploid (46XY) even at the 56th cumulative population doublings and expressed the pluripotency markers POU5F1, NANOG, KLF4, and MYC even at 50th cumulative population doublings. Under specific culture conditions, IPI-SA3-C4 cells displayed cellular hallmarks and molecular markers of adipogenic, osteogenic, and chondrogenic lineages and showed adipogenic capacity even at the 66th cumulative population doublings. These characteristics show IPI-SA3-C4 cells as a promising potential model for human subcutaneous adipogenesis in vitro.


Subject(s)
Adipocytes/cytology , Adipogenesis , Models, Biological , Multipotent Stem Cells/cytology , Animals , Biomarkers/metabolism , Carcinogenesis/pathology , Cell Line , Cell Lineage , Cell Proliferation , Cellular Senescence , Chondrogenesis , Diploidy , Humans , Infant , Karyotype , Kruppel-Like Factor 4 , Male , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , beta-Galactosidase/metabolism
16.
Biomedica ; 40(1): 72-88, 2020 03 01.
Article in English, Spanish | MEDLINE | ID: mdl-32220165

ABSTRACT

INTRODUCTION: The olfactory neuro-epithelium has an intrinsic capability of renewal during lifetime provided by the existence of globose and horizontal olfactory precursor cells. Additionally, mesenchymal stromal olfactory cells also support the homeostasis of the olfactory mucosa cell population. Under in vitro culture conditions with Dulbecco modified eagle/F12 medium supplemented with 10% fetal bovine serum, tissue biopsies from upper turbinate have generated an adherent population of cells expressing mainly mesenchymal stromal phenotypic markers. A closer examination of these cells has also found co-expression of olfactory precursors and ensheathing cell phenotypic markers. These results were suggestive of a unique property of olfactory mesenchymal stromal cells as potentially olfactory progenitor cells. OBJECTIVE: To study whether the expression of these proteins in mesenchymal stromal cells is modulated upon neuronal differentiation. MATERIALS AND METHODS: We observed the phenotype of olfactory stromal cells under DMEM/F12 plus 10% fetal bovine serum in comparison to cells from spheres induced by serum-free medium plus growth factors inducers of neural progenitors. RESULTS: The expression of mesenchymal stromal (CD29+, CD73+, CD90+, CD45-), horizontal basal (ICAM-1/CD54+, p63+, p75NGFr+), and ensheathing progenitor cell (nestin+, GFAP+) proteins was determined in the cultured population by flow cytometry. The determination of Oct 3/4, Sox-2, and Mash-1 transcription factors, as well as the neurotrophins BDNF, NT3, and NT4 by RT-PCR in cells, was indicative of functional heterogeneity of the olfactory mucosa tissue sample. CONCLUSIONS: Mesenchymal and olfactory precursor proteins were downregulated by serum-free medium and promoted differentiation of mesenchymal stromal cells into neurons and astroglial cells.


Introducción. El recambio celular del neuroepitelio olfatorio ocurre durante la vida del individuo gracias a precursores olfatorios. Además, las células mesenquimales del estroma también contribuyen a la homeostasis de la mucosa. Cuando un explante de una biopsia de mucosa se cultiva en un medio esencial mínimo, se genera una población predominante de células adherentes que expresan proteínas típicas de las células mesenquimales del estroma. La coexpresión de marcadores fenotípicos de precursores olfatorios y de células del recubrimiento del nervio olfatorio constituiría una propiedad única de las células mesenquimales del estroma. Objetivo. Determinar si la diferenciación celular de las células mesenquimales hacia fenotipos neurales modula la expresión de los marcadores mesenquimales característicos. Materiales y métodos. Se compararon las células aisladas de la mucosa olfatoria en un medio de cultivo con suplemento de 10 % de suero fetal bovino con esferas generadas en un medio sin suero más factores de crecimiento. Resultados. Se determinó la expresión de proteínas de las células mesenquimales del estroma (CD29+, CD73+, CD90+, CD45-), de las basales horizontales (ICAM-1/CD54+, p63+, p75NGFr+), y de las del recubrimiento del nervio olfatorio (nestin+, GFAP+) en la misma población cultivada. La determinación de Oct 3/4, Sox-2 y Mash-1, así como de las neurotrofinas BDNF, NT3 y NT4, sugirió que las células del estroma son funcionales. La expresión de las proteínas de las células mesenquimales y los precursores olfatorios, disminuyó en las células de las mesenesferas inducidas por ausencia de suero en el medio de cultivo. Conclusión. Las células mesenquimales del estroma de la mucosa olfatoria presentan una tendencia dominante hacia la diferenciación neural.


Subject(s)
Mesenchymal Stem Cells/metabolism , Nasal Mucosa/cytology , Olfactory Mucosa/cytology , Protein Biosynthesis , Adipogenesis , Antigens, Differentiation/analysis , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Chondrogenesis , Culture Media/pharmacology , Culture Media, Serum-Free/pharmacology , Glial Fibrillary Acidic Protein/biosynthesis , Glial Fibrillary Acidic Protein/genetics , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Mesenchymal Stem Cells/drug effects , Nasal Mucosa/metabolism , Nerve Growth Factors/biosynthesis , Nerve Growth Factors/genetics , Nestin/biosynthesis , Nestin/genetics , Neuroglia/metabolism , Neurons/metabolism , Olfactory Mucosa/metabolism , Osteogenesis , Recombinant Proteins/pharmacology , Spheroids, Cellular , Transcription Factors/biosynthesis , Transcription Factors/genetics , Turbinates
17.
Artif Organs ; 44(7): E288-E299, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31950507

ABSTRACT

Human adipose stem/stromal cell (ASC) spheroids were used as a serum-free in vitro model to recapitulate the molecular events and extracellular matrix organization that orchestrate a hypertrophic cartilage phenotype. Induced-ASC spheroids (ø = 450 µm) showed high cell viability throughout the period of culture. The expression of collagen type X alpha 1 chain (COLXA1) and matrix metallopeptidase 13 (MMP-13) was upregulated at week 2 in induced-ASC spheroids compared with week 5 (P < .001) evaluated by quantitative real-time PCR. In accordance, secreted levels of IL-6 (P < .0001), IL-8 (P < .0001), IL-10 (P < .0001), bFGF (P < .001), VEGF (P < .0001), and RANTES (P < .0001) were the highest at week 2. Strong in situ staining for collagen type X and low staining for TSP-1 was associated with the increase of hypertrophic genes expression at week 2 in induced-ASC spheroids. Collagen type I, osteocalcin, biglycan, and tenascin C were detected at week 5 by in situ staining, in accordance with the highest expression of alkaline phosphatase (ALPL) gene and the presence of calcium deposits as evaluated by Alizarin Red O staining. Induced-ASC spheroids showed a higher force required to compression at week 2 (P < .0001). The human ASC spheroids under serum-free inducer medium and normoxic culture conditions were induced to a hypertrophic cartilage phenotype, opening a new perspective to recapitulate endochondral ossification in vivo.


Subject(s)
Cartilage/growth & development , Chondrogenesis/physiology , Mesenchymal Stem Cells/physiology , Primary Cell Culture/methods , Tissue Engineering/methods , Adipose Tissue/cytology , Cartilage/cytology , Cartilage/ultrastructure , Cell Differentiation/physiology , Cells, Cultured , Collagen Type X/metabolism , Culture Media, Serum-Free , Extracellular Matrix/metabolism , Humans , Hypertrophy , Matrix Metalloproteinase 13/metabolism , Microscopy, Electron, Transmission , Spheroids, Cellular/physiology , Spheroids, Cellular/ultrastructure , Stromal Cells/physiology
18.
ACS Appl Mater Interfaces ; 12(4): 4343-4357, 2020 Jan 29.
Article in English | MEDLINE | ID: mdl-31909967

ABSTRACT

Scaffolds based on bioconjugated hydrogels are attractive for tissue engineering because they can partly mimic human tissue characteristics. For example, they can further increase their bioactivity with cells. However, most of the hydrogels present problems related to their processability, consequently limiting their use in 3D printing to produce tailor-made scaffolds. The goal of this work is to develop bioconjugated hydrogel nanocomposite inks for 3D printed scaffold fabrication through a micro-extrusion process having improved both biocompatibility and processability. The hydrogel is based on a photocrosslinkable alginate bioconjugated with both gelatin and chondroitin sulfate in order to mimic the cartilage extracellular matrix, while the nanofiller is based on graphene oxide to enhance the printability and cell proliferation. Our results show that the incorporation of graphene oxide into the hydrogel inks considerably improved the shape fidelity and resolution of 3D printed scaffolds because of a faster viscosity recovery post extrusion of the ink. Moreover, the nanocomposite inks produce anisotropic threads after the 3D printing process because of the templating of the graphene oxide liquid crystal. The in vitro proliferation assay of human adipose tissue-derived mesenchymal stem cells (hADMSCs) shows that bioconjugated scaffolds present higher cell proliferation than pure alginate, with the nanocomposites presenting the highest values at long times. Live/Dead assay otherwise displays full viability of hADMSCs adhered on the different scaffolds at day 7. Notably, the scaffolds produced with nanocomposite hydrogel inks were able to guide the cell proliferation following the direction of the 3D printed threads. In addition, the bioconjugated alginate hydrogel matrix induced chondrogenic differentiation without exogenous pro-chondrogenesis factors as concluded from immunostaining after 28 days of culture. This high cytocompatibility and chondroinductive effect toward hADMSCs, together with the improved printability and anisotropic structures, makes these nanocomposite hydrogel inks a promising candidate for cartilage tissue engineering based on 3D printing.


Subject(s)
Alginates/chemistry , Bioprinting/instrumentation , Graphite/chemistry , Hydrogels/chemistry , Mesenchymal Stem Cells/cytology , Tissue Scaffolds/chemistry , Cell Adhesion , Cell Proliferation , Chondrogenesis , Humans , Printing, Three-Dimensional/instrumentation , Tissue Engineering/instrumentation
19.
Tissue Eng Part B Rev ; 26(1): 1-12, 2020 02.
Article in English | MEDLINE | ID: mdl-31744404

ABSTRACT

Background: Symptomatic cartilage lesions and early osteoarthritis produce significant clinical and economic burdens. Cartilage repair can improve the symptoms and delay arthroplasty. The complete healing of damaged cartilage with the consistent reproduction of normal hyaline cartilage has not yet been achieved. The choice of harvesting site might influence the cells' abilities to modulate immunologic and inflammatory responses. Recently, dental pulp has been shown to contain a stem cell niche consisting of dental pulp stem cells (DPSCs) that maintain their self-renewal capacity due to the active environment in the dental pulp of deciduous teeth. Objective: The aim of this study was to critically review the current literature on the potential and limitations of the use of dental pulp-derived mesenchymal stem cells in cell-based therapies for cartilage regeneration. Methods: An electronic, customized search of scientific articles was conducted using the PubMed/MEDLINE and EMBASE databases from their inception to December 2018. The inclusion criteria were applied, and the articles that described the use of DPSC in cartilage treatment were selected for complete evaluation. The articles were classified according to the scaffold used, experimental model, chondrogenic differentiation features, defect location, cartilage evaluation, and results. After the application of the eligibility criteria, a total of nine studies were selected and fully analyzed. Results: A variety of animal models were used, including mice, rats, rabbits, and miniature pigs, to evaluate the quality and safety of human DPSCs in the repair of cartilage defects. Among the articles, two studies focused on preclinical models of cartilage tissue engineering. Five studies implanted DPSCs in other animal sites. Conclusion: The use of DPSCs is a potential new stem cell therapy for articular cartilage repair. The preclinical evidence discussed in this article provides a solid foundation for future clinical trials. Impact statement Osteoarthritis presents an ever-increasing clinical and socioeconomic burden. While cartilage repair has the potential to improve symptoms and delay joint replacement, complete regeneration of hyaline cartilage has been an elusive goal. Dental pulp has been shown to contain a niche that protects dental pulp stem cells (DPSCs) from the cumulative effects of genetic and environmental factors and maintains their self-renewal capacity due to the active environment. Transplantation and preclinical trials have demonstrated the strong potential of regenerative tissue-engineering protocols using DPSCs.


Subject(s)
Cartilage Diseases/therapy , Cartilage, Articular/cytology , Chondrogenesis , Dental Pulp/cytology , Regeneration , Stem Cells/cytology , Tissue Engineering/methods , Humans , Stem Cell Transplantation
20.
J Tissue Eng Regen Med ; 13(8): 1418-1429, 2019 08.
Article in English | MEDLINE | ID: mdl-31066519

ABSTRACT

There is no therapy currently available for fully repairing articular cartilage lesions. Our laboratory has recently developed a visible light-activatable methacrylated gelatin (mGL) hydrogel, with the potential for cartilage regeneration. In this study, we further optimized mGL scaffolds by supplementing methacrylated hyaluronic acid (mHA), which has been shown to stimulate chondrogenesis via activation of critical cellular signalling pathways. We hypothesized that the introduction of an optimal ratio of mHA would enhance the biological properties of mGL scaffolds and augment chondrogenesis of human bone marrow-derived mesenchymal stem cells (hBMSCs). To test this hypothesis, hybrid scaffolds consisting of mGL and mHA at different weight ratios were fabricated with hBMSCs encapsulated at 20 × 106  cells/ml and maintained in a chondrogenesis-promoting medium. The chondrogenenic differentiation of hBMSCs, within different scaffolds, was estimated after 8 weeks of culture. Our results showed that mGL/mHA at a 9:1 (%, w/v) ratio resulted in the lowest hBMSC hypertrophy and highest glycosaminoglycan production, with a slightly increased volume of the entire construct. The applicability of this optimally designed mGL/mHA hybrid scaffold for cartilage repair was then examined in vivo. A full-thickness cylindrical osteochondral defect was surgically created in the rabbit femoral condyle, and a three-dimensional cell-biomaterial construct was fabricated by in situ photocrosslinking to fully fill the lesion site. The results showed that implantation of the mGL/mHA (9:1) construct resulted in both cartilage and subchondral bone regeneration after 12 weeks, supporting its use as a promising scaffold for repair and resurfacing of articular cartilage defects, in the clinical setting.


Subject(s)
Cartilage, Articular/pathology , Cross-Linking Reagents/chemistry , Gelatin/chemistry , Hyaluronic Acid/chemistry , Light , Tissue Scaffolds/chemistry , Wound Healing , Animals , Cell Count , Cell Survival , Chondrogenesis , Gene Expression Regulation , Glycosaminoglycans/metabolism , Humans , Hydrogels/chemistry , Hypertrophy , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Methacrylates/chemistry , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL