Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 420
Filtrar
1.
Biomed Mater Eng ; 2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39240621

RESUMEN

BACKGROUND: Osteoarthritis is a prevalent joint disease affecting both humans and animals. It is characterized by articular cartilage degeneration and joint surface eburnation. Currently, no effective pharmacological treatment is available to restore the original function and structure of defective cartilage. OBJECTIVE: This study explores the potential of stem cell-based therapy in treating joint diseases involving cartilage degeneration, offering a promising avenue for future research and treatment. The primary aim was to compare the characteristics and, more importantly, the chondrogenic differentiation potential of human and rat adipose-derived mesenchymal stem cells (AD-MSCs). METHODS: Rat adipose tissue was collected from Sprague Dawley rats, while human adipose tissue was obtained in the form of lipoaspirate. The mesenchymal stem cells (MSCs) were then harvested using collagenase enzyme and subcultured. We meticulously evaluated and compared the cell morphology, percentage of cell viability, population doubling time, metabolic proliferation, and chondrogenic differentiation potential of MSCs harvested from both sources. Chondrogenic differentiation was induced at passage 3 using the 3D pellet culture method and assessed through histological and molecular analysis. RESULTS: The findings revealed that human and rat AD-MSCs were phenotypically identical, and an insignificant difference was found in cell morphology, percentage of cell viability, metabolic proliferation, and population doubling time. However, the chondrogenic differentiation potential of human AD-MSCs was evaluated as significantly higher than that of rat AD-MSCs. CONCLUSION: The current study suggests that research regarding chondrogenic differentiation of rat AD-MSCs can be effectively translated to humans. This discovery is a significant contribution to the field of regenerative medicine and has the potential to advance our understanding of stem cell-based therapy for joint diseases.

2.
Biology (Basel) ; 13(8)2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-39194498

RESUMEN

The limited self-repair capacity of articular cartilage is a challenge for healing injuries. While mesenchymal stem/stromal cells (MSCs) are a promising approach for tissue regeneration, the criteria for selecting a suitable cell source remain undefined. To propose a molecular criterion, dental pulp stem cells (DPSCs) with a Hox-negative expression pattern and bone marrow mesenchymal stromal cells (BMSCs), which actively express Hox genes, were differentiated towards chondrocytes in 3D pellets, employing a two-step protocol. The MSCs' response to preconditioning by cobalt chloride (CoCl2), a hypoxia-mimicking agent, was explored in an assessment of the chondrogenic differentiation's efficiency using morphological, histochemical, immunohistochemical, and biochemical experiments. The preconditioned DPSC pellets exhibited significantly elevated levels of collagen II and glycosaminoglycans (GAGs) and reduced levels of the hypertrophic marker collagen X. No significant effect on GAGs production was observed in the preconditioned BMSC pellets, but collagen II and collagen X levels were elevated. While preconditioning did not modify the ALP specific activity in either cell type, it was notably lower in the DPSCs differentiated pellets compared to their BMSCs counterparts. These results could be interpreted as demonstrating the higher plasticity of DPSCs compared to BMSCs, suggesting the contribution of their unique molecular characteristics, including their negative Hox expression pattern, to promote a chondrogenic differentiation potential. Consequently, DPSCs could be considered compelling candidates for future cartilage cell therapy.

3.
Biomed Pharmacother ; 179: 117349, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39191028

RESUMEN

OBJECT: Adipose-derived mesenchymal stem cells (ADSCs) have received significant attention in the field of cartilage tissue repair. Angelica sinensis polysaccharide (ASP) can enhance both the proliferation and differentiation of mesenchymal stem cells. Therefore, we intend to explore the effect of ASP on chondrogenic differentiation of ADSCs in vitro, and elucidate the underlying mechanisms. METHOD: ADSCs were treated with different concentrations of ASP to determine the optimal concentration. The chondrogenic differentiation of ADSCs was evaluated using Alcian blue staining, qRT-PCR, western blot, and IF staining. Transcriptome sequencing was performed to identify the expression profiles of ADSCs before and after ASP treatment, followed by bioinformatic analyses including differential expression analysis, enrichment analysis, and construction of PPI networks to identify differentially expressed genes (DEGs) associated with ASP and chondrogenic differentiation. RESULT: Surface markers of isolated rat-derived ADSCs were identified by CD44+CD90+CD45-CD106-, and exhibited the capacity for lipogenic, osteogenic, and chondrogenic differentiation. With increasing concentration of ASP treatment, there was an upregulation in the activity and acidic mucosubstance of ADSCs. The levels of Aggrecan, COL2A1, and Sox9 showed an increase in ADSCs after 28 days of 80 µg/ml ASP treatment. Transcriptome sequencing revealed that ASP-associated DEGs regulate extracellular matrix synthesis, immune response, inflammatory response, and cell cycle, and are involved in the NF-κB, AGE-RAGE, and calcium pathways. Moreover, Edn1, Frzb, Mdk, Nog, and Sulf1 are hub genes in DEGs. Notably, ASP upregulated MDK levels in ADSCs, while knockdown of MDK mitigated ASP-induced elevations in acidic mucosubstance, chondrogenic differentiation-related markers (Aggrecan, COL2A1, and Sox9), and the activity of the PI3K/AKT pathway. CONCLUSION: ASP enhances the proliferation and chondrogenic differentiation of ADSCs by activating the MDK-mediated PI3K/AKT pathway.


Asunto(s)
Angelica sinensis , Diferenciación Celular , Condrogénesis , Células Madre Mesenquimatosas , Polisacáridos , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Polisacáridos/farmacología , Condrogénesis/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Angelica sinensis/química , Transducción de Señal/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Sprague-Dawley , Ratas , Tejido Adiposo/citología , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Células Cultivadas , Masculino
4.
Small ; : e2405979, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39077937

RESUMEN

Biological effectors play critical roles in augmenting the repair of cartilage injuries, but it remains a challenge to control their release in a programmable, stepwise fashion. Herein, a hybrid system consisting of polydopamine (PDA) nanobottles embedded in a hydrogel matrix to manage the release of biological effectors for use in cartilage repair is reported. Specifically, a homing effector is load in the hydrogel matrix, together with the encapsulation of a cartilage effector in PDA nanobottles filled with phase-change material. In action, the homing effector is quickly released from the hydrogel in the initial step to recruit stem cells from the surroundings. Owing to the antioxidation effect of PDA, the recruited cells are shielded from reactive oxygen species. The cartilage effector is then slowly released from the nanobottles to promote chondrogenic differentiation, facilitating cartilage repair. Altogether, this strategy encompassing recruitment, protection, and differentiation of stem cells offers a viable route to tissue repair or regeneration through stem cell therapy.

5.
Gels ; 10(7)2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-39057446

RESUMEN

Functional articular cartilage regeneration remains an unmet medical challenge, increasing the interest for innovative biomaterial-based tissue engineering (TE) strategies. Hydrogels, 3D macromolecular networks with hydrophilic groups, present articular cartilage-like features such as high water content and load-bearing capacity. In this study, 3D porous polyethylene glycol diacrylate (PEGDA) hydrogels were fabricated combining the gas foaming technique and a UV-based crosslinking strategy. The 3D porous PEGDA hydrogels were characterized in terms of their physical, structural and mechanical properties. Our results showed that the size of the hydrogel pores can be modulated by varying the initiator concentration. In vitro cytotoxicity tests showed that 3D porous PEGDA hydrogels presented high biocompatibility both with human chondrocytes and osteoblast-like cells. Importantly, the 3D porous PEGDA hydrogels supported the viability and chondrogenic differentiation of human bone marrow-derived mesenchymal stem/stromal cell (hBM-MSC)-based spheroids as demonstrated by the positive staining of typical cartilage extracellular matrix (ECM) (glycosaminoglycans (GAGs)) and upregulation of chondrogenesis marker genes. Overall, the produced 3D porous PEGDA hydrogels presented cartilage-like mechanical properties and supported MSC spheroid chondrogenesis, highlighting their potential as suitable scaffolds for cartilage TE or disease modelling strategies.

6.
Int J Biol Macromol ; 276(Pt 1): 133868, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39009266

RESUMEN

Intervertebral disc degeneration is a clinical disease that reduces the quality of patient's life. The degeneration usually initiates in the nucleus pulposus (NP), hence the use of hydrogels represents a promising therapeutic approach. However, the viscoelastic nature of hydrogel and its ability to provide biomimetic architecture and biochemical cues influence the regeneration capability. This study focused on tuning the physical nature of a glycosaminoglycan hydrogel (κ-carrageenan) as well as the release kinetics of a chondrogenic factor (kartogenin - KGN) through physical cross-linking. For this, κ-carrageenan was cross linked with 2.5 % and 5 % potassium chloride (KCl) for 15 and 30 min and loaded with KGN molecule at 50 µM and 100 µM. The tight network structure with low water retention and degradation property was seen in hydrogel cross-linked with increased KCl concentration and time. However, optimal degradation along with NP mimicking viscoelastic nature was exhibited by 5 wt% KCl treated hydrogel (H3 hydrogel). All hydrogel groups exhibited burst KGN release at 24 h followed by a sustained release for 5 days. However, hydrogel cross-linked with 5 wt% KCl enhanced chondrogenic differentiation, mainly at lower KGN dose. In summary, this study shows the potential application of biomimetic KGN laden carrageenan hydrogel in NP regeneration.


Asunto(s)
Carragenina , Hidrogeles , Núcleo Pulposo , Ácidos Ftálicos , Regeneración , Carragenina/química , Carragenina/farmacología , Núcleo Pulposo/efectos de los fármacos , Hidrogeles/química , Regeneración/efectos de los fármacos , Ácidos Ftálicos/química , Ácidos Ftálicos/farmacología , Animales , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Condrogénesis/efectos de los fármacos , Humanos , Diferenciación Celular/efectos de los fármacos , Anilidas
7.
Sci Rep ; 14(1): 16396, 2024 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-39013921

RESUMEN

Most of the conditions involving cartilaginous tissues are irreversible and involve degenerative processes. The aim of the present study was to fabricate a biocompatible fibrous and film scaffolds using electrospinning and casting techniques to induce chondrogenic differentiation for possible application in cartilaginous tissue regeneration. Polycaprolactone (PCL) electrospun nanofibrous scaffolds and PCL film were fabricated and incorporated with multi-walled carbon nanotubes (MWCNTs). Thereafter, coating of chondroitin sulfate (CS) on the fibrous and film structures was applied to promote chondrogenic differentiation of human dental pulp stem cells (hDPSCs). First, the morphology, hydrophilicity and mechanical properties of the scaffolds were characterized by scanning electron microscopy (SEM), spectroscopic characterization, water contact angle measurements and tensile strength testing. Subsequently, the effects of the fabricated scaffolds on stimulating the proliferation of human dental pulp stem cells (hDPSCs) and inducing their chondrogenic differentiation were evaluated via electron microscopy, flow cytometry and RT‒PCR. The results of the study demonstrated that the different forms of the fabricated PCL-MWCNTs scaffolds analyzed demonstrated biocompatibility. The nanofilm structures demonstrated a higher rate of cellular proliferation, while the nanofibrous architecture of the scaffolds supported the cellular attachment and differentiation capacity of hDPSCs and was further enhanced with CS addition. In conclusion, the results of the present investigation highlighted the significance of this combination of parameters on the viability, proliferation and chondrogenic differentiation capacity of hDPSCs seeded on PCL-MWCNT scaffolds. This approach may be applied when designing PCL-based scaffolds for future cell-based therapeutic approaches developed for chondrogenic diseases.


Asunto(s)
Diferenciación Celular , Condrogénesis , Sulfatos de Condroitina , Pulpa Dental , Nanofibras , Nanotubos de Carbono , Poliésteres , Células Madre , Andamios del Tejido , Humanos , Pulpa Dental/citología , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacología , Poliésteres/química , Poliésteres/farmacología , Nanofibras/química , Diferenciación Celular/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Andamios del Tejido/química , Nanotubos de Carbono/química , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ingeniería de Tejidos/métodos
8.
Tissue Eng Regen Med ; 21(7): 1021-1036, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39037474

RESUMEN

BACKGROUND: Chondrogenic differentiation medium (CDM) is usually used to maintain chondrogenic activity during chondrocyte sheet production. However, tissue qualities remain to be determined as to what factors improve cell functions. Moreover, the relationship between CDM and cell migration proteins has not been reported. METHOD: In this study, the effect of CDM on the behavior of chondrocyte sheets was investigated. Structural analysis, mechanical testing and proteomics were performed to observe tissue qualities. The relationship between CDM and cell migration proteins were investigated using time-lapse observations and bioinformatic analysis. RESULTS: During 48 h, CDM affected the chondrocyte behaviors by reducing cell migration. Compared to the basal medium, CDM impacted the contraction of monolayered chondrocyte sheets. At day 7, the contracted sheets increased tissue thickness and improved tissue stiffness. Cartilage specific proteins were also upregulated. Remarkedly, the chondrocyte sheets in CDM displayed downregulated proteins related to cell migration. Bioinformatic analysis revealed that TGFß1 was shown to be associated with cartilage functions and cell migration. Pathway analysis of chondrocyte sheets in CDM also revealed the presence of a TGFß pathway without activating actin production, which might be involved in synthesizing cartilage-specific proteins. Cell migration pathway showed MAPK signaling in both cultures of the chondrocyte sheets. CONCLUSION: Reduced cell migration in the chondrocyte sheet affected the tissue quality. Using CDM, TGFß1 might trigger cartilage protein production through the TGFß pathway and be involved in cell migration via the MAPK signaling pathway. Understanding cell behaviors and their protein expression would be beneficial for developing high-quality tissue-engineered cartilage.


Asunto(s)
Movimiento Celular , Condrocitos , Condrocitos/metabolismo , Condrocitos/citología , Humanos , Condrogénesis , Factor de Crecimiento Transformador beta1/metabolismo , Cartílago/metabolismo , Diferenciación Celular , Células Cultivadas
9.
Bioengineering (Basel) ; 11(6)2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38927763

RESUMEN

Electrical stimulation (ES) is a widely discussed topic in the field of cartilage tissue engineering due to its ability to induce chondrogenic differentiation (CD) and proliferation. It shows promise as a potential therapy for osteoarthritis (OA). In this study, we stimulated mesenchymal stem cells (MSCs) incorporated into collagen hydrogel (CH) scaffolds, consisting of approximately 500,000 cells each, for 1 h per day using a 2.5 Vpp (119 mV/mm) 8 Hz sinusoidal signal. We compared the cell count, morphology, and CD on days 4, 7, and 10. The results indicate proliferation, with an increase ranging from 1.86 to 9.5-fold, particularly on day 7. Additionally, signs of CD were observed. The stimulated cells had a higher volume, while the stimulated scaffolds showed shrinkage. In the ES groups, up-regulation of collagen type 2 and aggrecan was found. In contrast, SOX9 was up-regulated in the control group, and MMP13 showed a strong up-regulation, indicating cell stress. In addition to lower stress levels, the control groups also showed a more spheroidic shape. Overall, scaffold-based ES has the potential to achieve multiple outcomes. However, finding the appropriate stimulation pattern is crucial for achieving successful chondrogenesis.

10.
Stem Cell Res Ther ; 15(1): 177, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38886785

RESUMEN

BACKGROUND: Cartilage is a kind of avascular tissue, and it is difficult to repair itself when it is damaged. In this study, we investigated the regulation of chondrogenic differentiation and vascular formation in human jaw bone marrow mesenchymal stem cells (h-JBMMSCs) by the long-chain noncoding RNA small nucleolar RNA host gene 1 (SNHG1) during cartilage tissue regeneration. METHODS: JBMMSCs were isolated from the jaws via the adherent method. The effects of lncRNA SNHG1 on the chondrogenic differentiation of JBMMSCs in vitro were detected by real-time fluorescence quantitative polymerase chain reaction (RT-qPCR), Pellet experiment, Alcian blue staining, Masson's trichrome staining, and modified Sirius red staining. RT-qPCR, matrix gel tube formation, and coculture experiments were used to determine the effect of lncRNA SNHG1 on the angiogenesis in JBMMSCs in vitro. A model of knee cartilage defects in New Zealand rabbits and a model of subcutaneous matrix rubber suppositories in nude mice were constructed for in vivo experiments. Changes in mitochondrial function were detected via RT-qPCR, dihydroethidium (DHE) staining, MitoSOX staining, tetramethyl rhodamine methyl ester (TMRM) staining, and adenosine triphosphate (ATP) detection. Western blotting was used to detect the phosphorylation level of signal transducer and activator of transcription 3 (STAT3). RESULTS: Alcian blue staining, Masson's trichrome staining, and modified Sirius Red staining showed that lncRNA SNHG1 promoted chondrogenic differentiation. The lncRNA SNHG1 promoted angiogenesis in vitro and the formation of microvessels in vivo. The lncRNA SNHG1 promoted the repair and regeneration of rabbit knee cartilage tissue. Western blot and alcian blue staining showed that the JAK inhibitor reduced the increase of STAT3 phosphorylation level and staining deepening caused by SNHG1. Mitochondrial correlation analysis revealed that the lncRNA SNHG1 led to a decrease in reactive oxygen species (ROS) levels, an increase in mitochondrial membrane potential and an increase in ATP levels. Alcian blue staining showed that the ROS inhibitor significantly alleviated the decrease in blue fluorescence caused by SNHG1 knockdown. CONCLUSIONS: The lncRNA SNHG1 promotes chondrogenic differentiation and angiogenesis of JBMMSCs. The lncRNA SNHG1 regulates the phosphorylation of STAT3, reduces the level of ROS, regulates mitochondrial energy metabolism, and ultimately promotes cartilage regeneration.


Asunto(s)
Diferenciación Celular , Condrogénesis , Células Madre Mesenquimatosas , Mitocondrias , ARN Largo no Codificante , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Humanos , Animales , Conejos , Mitocondrias/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Condrogénesis/genética , Ratones , Ratones Desnudos , Regeneración , Neovascularización Fisiológica , Cartílago/metabolismo , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Angiogénesis
11.
Int J Mol Sci ; 25(11)2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38891883

RESUMEN

Articular cartilage damage still remains a major problem in orthopedical surgery. The development of tissue engineering techniques such as autologous chondrocyte implantation is a promising way to improve clinical outcomes. On the other hand, the clinical application of autologous chondrocytes has considerable limitations. Mesenchymal stromal cells (MSCs) from various tissues have been shown to possess chondrogenic differentiation potential, although to different degrees. In the present study, we assessed the alterations in chondrogenesis-related gene transcription rates and extracellular matrix deposition levels before and after the chondrogenic differentiation of MSCs in a 3D spheroid culture. MSCs were obtained from three different tissues: umbilical cord Wharton's jelly (WJMSC-Wharton's jelly mesenchymal stromal cells), adipose tissue (ATMSC-adipose tissue mesenchymal stromal cells), and the dental pulp of deciduous teeth (SHEDs-stem cells from human exfoliated deciduous teeth). Monolayer MSC cultures served as baseline controls. Newly formed 3D spheroids composed of MSCs previously grown in 2D cultures were precultured for 2 days in growth medium, and then, chondrogenic differentiation was induced by maintaining them in the TGF-ß1-containing medium for 21 days. Among the MSC types studied, WJMSCs showed the most similarities with primary chondrocytes in terms of the upregulation of cartilage-specific gene expression. Interestingly, such upregulation occurred to some extent in all 3D spheroids, even prior to the addition of TGF-ß1. These results confirm that the potential of Wharton's jelly is on par with adipose tissue as a valuable cell source for cartilage engineering applications as well as for the treatment of osteoarthritis. The 3D spheroid environment on its own acts as a trigger for the chondrogenic differentiation of MSCs.


Asunto(s)
Diferenciación Celular , Condrocitos , Condrogénesis , Matriz Extracelular , Células Madre Mesenquimatosas , Esferoides Celulares , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Humanos , Condrogénesis/genética , Matriz Extracelular/metabolismo , Esferoides Celulares/citología , Esferoides Celulares/metabolismo , Condrocitos/citología , Condrocitos/metabolismo , Células Cultivadas , Gelatina de Wharton/citología , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Técnicas de Cultivo de Célula/métodos , Ingeniería de Tejidos/métodos , Cartílago/citología , Cartílago/metabolismo , Diente Primario/citología , Diente Primario/metabolismo , Pulpa Dental/citología , Pulpa Dental/metabolismo
12.
Cells ; 13(9)2024 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-38727293

RESUMEN

BACKGROUND: Since cytokine receptor-like factor 1 (CRLF1) has been implicated in tissue regeneration, we hypothesized that CRLF1 released by mesenchymal stem cells can promote the repair of osteochondral defects. METHODS: The degree of a femoral osteochondral defect repair in rabbits after intra-articular injections of bone marrow-derived mesenchymal stem cells (BMSCs) that were transduced with empty adeno-associated virus (AAV) or AAV containing CRLF1 was determined by morphological, histological, and micro computer tomography (CT) analyses. The effects of CRLF1 on chondrogenic differentiation of BMSCs or catabolic events of interleukin-1beta-treated chondrocyte cell line TC28a2 were determined by alcian blue staining, gene expression levels of cartilage and catabolic marker genes using real-time PCR analysis, and immunoblot analysis of Smad2/3 and STAT3 signaling. RESULTS: Intra-articular injections of BMSCs overexpressing CRLF1 markedly improved repair of a rabbit femoral osteochondral defect. Overexpression of CRLF1 in BMSCs resulted in the release of a homodimeric CRLF1 complex that stimulated chondrogenic differentiation of BMSCs via enhancing Smad2/3 signaling, whereas the suppression of CRLF1 expression inhibited chondrogenic differentiation. In addition, CRLF1 inhibited catabolic events in TC28a2 cells cultured in an inflammatory environment, while a heterodimeric complex of CRLF1 and cardiotrophin-like Cytokine (CLC) stimulated catabolic events via STAT3 activation. CONCLUSION: A homodimeric CRLF1 complex released by BMSCs enhanced the repair of osteochondral defects via the inhibition of catabolic events in chondrocytes and the stimulation of chondrogenic differentiation of precursor cells.


Asunto(s)
Diferenciación Celular , Condrocitos , Condrogénesis , Células Madre Mesenquimatosas , Animales , Conejos , Células Madre Mesenquimatosas/metabolismo , Condrogénesis/genética , Condrocitos/metabolismo , Receptores de Citocinas/metabolismo , Receptores de Citocinas/genética , Fémur/patología , Transducción de Señal , Línea Celular , Trasplante de Células Madre Mesenquimatosas
13.
J Oral Rehabil ; 51(8): 1507-1520, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38717032

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSCs) derived from the synovium, known as synovium mesenchymal stem cells (SMSCs), exhibit significant potential for articular cartilage regeneration owing to their capacity for chondrogenic differentiation. However, the microRNAs (miRNAs) governing this process and the associated mechanisms remain unclear. While mechanical stress positively influences chondrogenesis in MSCs, the miRNA-mediated response of SMSCs to mechanical stimuli is not well understood. OBJECTIVE: This study explores the miRNA-driven mechano-transduction in SMSCs chondrogenesis under mechanical stress. METHODS: The surface phenotype of SMSCs was analysed by flow cytometry. Chondrogenesis capacities of SMSCs were examined by Alcian blue staining. High throughput sequencing was used to screen mechano-sensitive miRNAs of SMSCs. The RNA expression level of COL2A1, ACAN, SOX9, BMPR2 and miR-143-3p of SMSCs were tested by quantitative real-time polymerase chain reaction (qRT-PCR). The interaction between miR-143-3p and TLR4 was confirmed by luciferase reporter assays. The protein expression levels of related genes were assessed by western blot. RESULTS: High-throughput sequencing revealed a notable reduction in miR-143-3p levels in mechanically stressed SMSCs. Gain- or loss-of-function strategies introduced by lentivirus demonstrated that miR-143-3p overexpression hindered chondrogenic differentiation, whereas its knockdown promoted this process. Bioinformatics scrutiny and luciferase reporter assays pinpointed a potential binding site for miR-143-3p within the 3'-UTR of bone morphogenetic protein receptor type 2 (BMPR2). MiR-143-3p overexpression decreased BMPR2 expression and phosphorylated Smad1, 5 and 8 levels, while its inhibition activated BMPR2-Smad pathway. CONCLUSION: This study elucidated that miR-143-3p negatively regulates SMSCs chondrogenic differentiation through the BMPR2-Smad pathway under mechanical tensile stress. The direct targeting of BMPR2 by miR-143-3p established a novel dimension to our understanding of mechano-transduction mechanism during SMSC chondrogenesis. This understanding is crucial for advancing strategies in articular cartilage regeneration.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II , Diferenciación Celular , Condrogénesis , Células Madre Mesenquimatosas , MicroARNs , Transducción de Señal , Estrés Mecánico , Membrana Sinovial , Humanos , Agrecanos/metabolismo , Agrecanos/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Diferenciación Celular/fisiología , Células Cultivadas , Condrogénesis/fisiología , Colágeno Tipo II/metabolismo , Colágeno Tipo II/genética , Células Madre Mesenquimatosas/metabolismo , MicroARNs/metabolismo , MicroARNs/genética , Transducción de Señal/fisiología , Proteínas Smad/metabolismo , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Membrana Sinovial/citología , Membrana Sinovial/metabolismo
14.
Tissue Cell ; 88: 102380, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38615643

RESUMEN

The use of mesenchymal stem cells (MSCs) in cartilage regeneration has gained significant attention in regenerative medicine. This paper reviews the molecular mechanisms underlying MSC-based cartilage regeneration and explores various therapeutic strategies to enhance the efficacy of MSCs in this context. MSCs exhibit multipotent capabilities and can differentiate into various cell lineages under specific microenvironmental cues. Chondrogenic differentiation, a complex process involving signaling pathways, transcription factors, and growth factors, plays a pivotal role in the successful regeneration of cartilage tissue. The chondrogenic differentiation of MSCs is tightly regulated by growth factors and signaling pathways such as TGF-ß, BMP, Wnt/ß-catenin, RhoA/ROCK, NOTCH, and IHH (Indian hedgehog). Understanding the intricate balance between these pathways is crucial for directing lineage-specific differentiation and preventing undesirable chondrocyte hypertrophy. Additionally, paracrine effects of MSCs, mediated by the secretion of bioactive factors, contribute significantly to immunomodulation, recruitment of endogenous stem cells, and maintenance of chondrocyte phenotype. Pre-treatment strategies utilized to potentiate MSCs, such as hypoxic conditions, low-intensity ultrasound, kartogenin treatment, and gene editing, are also discussed for their potential to enhance MSC survival, differentiation, and paracrine effects. In conclusion, this paper provides a comprehensive overview of the molecular mechanisms involved in MSC-based cartilage regeneration and outlines promising therapeutic strategies. The insights presented contribute to the ongoing efforts in optimizing MSC-based therapies for effective cartilage repair.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Regeneración , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Regeneración/fisiología , Animales , Condrogénesis/fisiología , Cartílago/metabolismo , Cartílago/fisiología , Diferenciación Celular , Condrocitos/metabolismo , Condrocitos/citología , Transducción de Señal
15.
Nanomedicine (Lond) ; 19(12): 1029-1034, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38573156

RESUMEN

Nano-sized piezoelectric materials allow for precise interaction with living systems to local deliver electrical cues. Recent innovations enhance their potential in tissue engineering and regenerative medicine.


Asunto(s)
Nanoestructuras , Humanos , Nanoestructuras/química , Nanoestructuras/uso terapéutico , Nanomedicina/métodos , Investigación Biomédica Traslacional , Animales , Nanotecnología/métodos , Sistemas de Liberación de Medicamentos/métodos
16.
Biomolecules ; 14(4)2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38672430

RESUMEN

Bovine serum albumin (BSA) plays a crucial role in cell culture media, influencing cellular processes such as proliferation and differentiation. Although it is commonly included in chondrogenic differentiation media, its specific function remains unclear. This study explores the effect of different BSA concentrations on the chondrogenic differentiation of human adipose-derived stromal/stem cells (hASCs). hASC pellets from six donors were cultured under chondrogenic conditions with three BSA concentrations. Surprisingly, a lower BSA concentration led to enhanced chondrogenesis. The degree of this effect was donor-dependent, classifying them into two groups: (1) high responders, forming at least 35% larger, differentiated pellets with low BSA in comparison to high BSA; (2) low responders, which benefitted only slightly from low BSA doses with a decrease in pellet size and marginal differentiation, indicative of low intrinsic differentiation potential. In all cases, increased chondrogenesis was accompanied by hypertrophy under low BSA concentrations. To the best of our knowledge, this is the first study showing improved chondrogenicity and the tendency for hypertrophy with low BSA concentration compared to standard levels. Once the tendency for hypertrophy is understood, the determination of BSA concentration might be used to tune hASC chondrogenic or osteogenic differentiation.


Asunto(s)
Diferenciación Celular , Condrogénesis , Células Madre Mesenquimatosas , Albúmina Sérica Bovina , Humanos , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Condrogénesis/efectos de los fármacos , Medios de Cultivo/química , Medios de Cultivo/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Albúmina Sérica Bovina/farmacología , Albúmina Sérica Bovina/química , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo
17.
Macromol Biosci ; 24(8): e2400003, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38597147

RESUMEN

Articular cartilage defects pose a significant challenge due to the limited self-healing ability of cartilage. However, traditional techniques face limitations including autologous chondrocyte expansion issues. This study aims to investigate the effects of the polylactic acid-glycolic acid (PLGA) and collagen-surface modified polylactic acid-glycolic acid (CPLGA) microspheres loaded with tetramethylpyrazine (TMP) on two cell types and the regeneration potential of articular cartilage. CPLGA microspheres are prepared by Steglich reaction and characterized. They evaluated the effect of TMP-loaded microspheres on HUVECs (Human Umbilical Vein Endothelial Cells) and examined the compatibility of blank microspheres with BMSCs (Bone marrow mesenchymal stromal cells) and their potential to promote cartilage differentiation. Subcutaneous implant immune tests and cartilage defect treatment are conducted to assess biocompatibility and cartilage repair potential. The results highlight the efficacy of CPLGA microspheres in promoting tissue regeneration, attributed to improved hydrophilicity and collagen-induced mitigation of degradation. Under hypoxic conditions, both CPLGA and PLGA TMP-loaded microspheres exhibit inhibitory effects on HUVEC proliferation, migration, and angiogenesis. Notably, CPLGA microspheres show enhanced compatibility with BMSCs, facilitating chondrogenic differentiation. Moreover, the CPLGA microsphere-composite hydrogel exhibits potential for cartilage repair by modulating angiogenesis and promoting BMSC differentiation.


Asunto(s)
Cartílago Articular , Colágeno , Células Endoteliales de la Vena Umbilical Humana , Hidrogeles , Microesferas , Pirazinas , Humanos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Cartílago Articular/efectos de los fármacos , Cartílago Articular/patología , Pirazinas/química , Pirazinas/farmacología , Animales , Colágeno/química , Colágeno/farmacología , Hidrogeles/química , Hidrogeles/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Diferenciación Celular/efectos de los fármacos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/farmacología , Condrogénesis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos
18.
In Vitro Cell Dev Biol Anim ; 60(4): 343-353, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38504085

RESUMEN

MicroRNAs (miRNAs) play an important role in articular cartilage damage in osteoarthritis (OA). However, the biological role of miRNAs in the chondrogenic differentiation of bone marrow mesenchymal stem cell (BMSC) remains largely unclear. Rabbit bone marrow mesenchymal stem cells (rBMSCs) were isolated, cultured, and identified. Afterwards, rBMSCs were induced to chondrogenic differentiation, examined by Alcian Blue staining. Differentially expressed miRNAs were identified in rBMSCs between induced and non-induced groups by miRNA sequencing analysis, part of which was validated via PCR assay. Cell viability and apoptosis were assessed by CCK-8 assay and Hoechst staining. Saffron O staining was utilized to assess chondrocyte hyperplasia. The expression of specific chondrogenic markers, including COL2A1, SOX9, Runx2, MMP-13, Aggrecan, and BMP-2, were measured at mRNA and protein levels. The association between beta-transducin repeat containing E3 ubiquitin protein ligase (BTRC) and miR-10a-5p in the miRNA family from rabbit (ocu-miR-10a-5p) was determined by luciferase reporter assay. A total of 76 differentially expressed miRNAs, including 52 downregulated and 24 upregulated miRNAs, were identified in rBMSCs from the induced group. Inhibition of ocu-miR-10a-5p suppressed rBMSC viability and chondrogenic differentiation, as well as downregulated the expression of ß-catenin, SOX9, COL2A1, MMP-13, and Runx2. BTRC was predicted and confirmed as a target of ocu-miR-10a-5p. Overexpression of BTRC rescued the promoting impacts of overexpressed ocu-miR-10a-5p on chondrogenic differentiation of rBMSCs and ß-catenin expression. Taken together, our data suggested that ocu-miR-10a-5p facilitated rabbit BMSC survival and chondrogenic differentiation by activating Wnt/ß-catenin signaling through BTRC.


Asunto(s)
Diferenciación Celular , Condrogénesis , Células Madre Mesenquimatosas , MicroARNs , Vía de Señalización Wnt , Animales , Conejos , MicroARNs/genética , MicroARNs/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Diferenciación Celular/genética , Condrogénesis/genética , Vía de Señalización Wnt/genética , Condrocitos/metabolismo , Condrocitos/citología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Apoptosis/genética , Supervivencia Celular , beta Catenina/metabolismo , beta Catenina/genética , Secuencia de Bases , Regulación de la Expresión Génica
19.
J Dent Sci ; 19(1): 86-91, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38303882

RESUMEN

Background/purpose: Aging severely impairs the beneficial effects of human dental pulp stem cells (hDPSCs) on cartilage regeneration. Lysine demethylase 3A (KDM3A) is involved in regulating mesenchymal stem cells (MSCs) senescence and bone aging. In this study, we investigated the role of KDM3A in hDPSCs aging and whether KDM3A could rejuvenate aged hDPSCs to enhance their chondrogenic differentiation capacity. Materials and methods: The cellular aging of hDPSCs was evaluated by senescence-associated ß-galactosidase (SA-ß-gal) staining. Protein levels were determined using Western blot analysis. KDM3A was overexpressed in aged hDPSCs by lentivirus infection. Quantitative reverse-transcription polymerase chain reaction (RT-qPCR) were used to determine the mRNA levels of stemness markers. Toluidine blue staining was used to evaluate the effect of KDM3A overexpression on the chondrogenic differentiation of aged hDPSCs. Results: hDPSCs at passage 12 or treated with etoposide exhibited augmented cellular senescence as evidenced by increased SA-ß-gal activity. KDM3A was significantly increased during senescence of hDPSCs. Overexpression of KDM3A did not affect the stemness properties but significantly promoted the chondrogenic differentiation of aged hDPSCs. Conclusion: Our findings indicate that KDM3A plays an important role in the maintenance of the chondrogenic differentiation capacity of aged hDPSCs and suggest that therapies targeting KDM3A may be a novel strategy to rejuvenate aged hDPSCs.

20.
Adv Healthc Mater ; 13(9): e2303255, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38253413

RESUMEN

Partial-thickness cartilage defect (PTCD) is a common and formidable clinical challenge without effective therapeutic approaches. The inherent anti-adhesive characteristics of the extracellular matrix within cartilage pose a significant impediment to the integration of cells or biomaterials with the native cartilage during cartilage repair. Here, an injectable photocrosslinked bioadhesive hydrogel, consisting of gelatin methacryloyl (GM), acryloyl-6-aminocaproic acid-g-N-hydroxysuccinimide (AN), and poly(lactic-co-glycolic acid) microspheres loaded with kartogenin (KGN) (abbreviated as GM/AN/KGN hydrogel), is designed to enhance interfacial integration and repair of PTCD. After injected in situ at the irregular defect, a stable and robust hydrogel network is rapidly formed by ultraviolet irradiation, and it can be quickly and tightly adhered to native cartilage through amide bonds. The hydrogel exhibits good adhesion strength up to 27.25 ± 1.22 kPa by lap shear strength experiments. The GM/AN/KGN hydrogel demonstrates good adhesion, low swelling, resistance to fatigue, biocompatibility, and chondrogenesis properties in vitro. A rat model with PTCD exhibits restoration of a smoother surface, stable seamless integration, and abundant aggrecan and type II collagen production. The injectable stable adhesive hydrogel with long-term chondrogenic differentiation capacity shows great potential to facilitate repair of PTCD.


Asunto(s)
Anilidas , Condrogénesis , Hidrogeles , Ácidos Ftálicos , Ratas , Animales , Hidrogeles/farmacología , Hidrogeles/química , Preparaciones de Acción Retardada/farmacología , Cartílago
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA