Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.218
Filtrar
1.
J Cell Mol Med ; 28(11): e18443, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38837873

RESUMEN

The human auricle has a complex structure, and microtia is a congenital malformation characterized by decreased size and loss of elaborate structure in the affected ear with a high incidence. Our previous studies suggest that inadequate cell migration is the primary cytological basis for the pathogenesis of microtia, however, the underlying mechanism is unclear. Here, we further demonstrate that microtia chondrocytes show a decreased directional persistence during cell migration. Directional persistence can define a leading edge associated with oriented movement, and any mistakes would affect cell function and tissue morphology. By the screening of motility-related genes and subsequent confirmations, active Rac1 (Rac1-GTP) is identified to be critical for the impaired directional persistence of microtia chondrocytes migration. Moreover, Rho guanine nucleotide exchange factors (GEFs) and Rho GTPase-activating proteins (GAPs) are detected, and overexpression of Tiam1 significantly upregulates the level of Rac1-GTP and improves directional migration in microtia chondrocytes. Consistently, decreased expression patterns of Tiam1 and active Rac1 are found in microtia mouse models, Bmp5se/J and Prkralear-3J/GrsrJ. Collectively, our results provide new insights into microtia development and therapeutic strategies of tissue engineering for microtia patients.


Asunto(s)
Movimiento Celular , Condrocitos , Microtia Congénita , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Proteína de Unión al GTP rac1 , Animales , Femenino , Humanos , Masculino , Ratones , Condrocitos/metabolismo , Condrocitos/citología , Microtia Congénita/metabolismo , Microtia Congénita/genética , Microtia Congénita/patología , Modelos Animales de Enfermedad , Proteína de Unión al GTP rac1/metabolismo , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T/metabolismo , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T/genética
2.
Carbohydr Polym ; 339: 122251, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38823918

RESUMEN

In this study, the disulfide-linked hyaluronic acid (HA) hydrogels were optimised for potential application as a scaffold in tissue engineering through the Quality by Design (QbD) approach. For this purpose, HA was first modified by incorporating the cysteine moiety into the HA backbone, which promoted the formation of disulfide cross-linked HA hydrogel at physiological pH. Utilising a Design of Experiments (DoE) methodology, the critical factors to achieve stable biomaterials, i.e. the degree of HA substitution, HA molecular weight, and coupling agent ratio, were explored. To establish a design space, the DoE was performed with 65 kDa, 138 kDa and 200 kDa HA and variable concentrations of coupling agent to optimise conditions to obtain HA hydrogel with improved rheological properties. Thus, HA hydrogel with a 12 % degree of modification, storage modulus of ≈2321 Pa and loss modulus of ≈15 Pa, was achieved with the optimum ratio of coupling agent. Furthermore, biocompatibility assessments in C28/I2 chondrocyte cells demonstrated the non-toxic nature of the hydrogel, underscoring its potential for tissue regeneration. Our findings highlight the efficacy of the QbD approach in designing HA hydrogels with tailored properties for biomedical applications.


Asunto(s)
Materiales Biocompatibles , Condrocitos , Disulfuros , Ácido Hialurónico , Hidrogeles , Reología , Ingeniería de Tejidos , Ácido Hialurónico/química , Hidrogeles/química , Hidrogeles/síntesis química , Disulfuros/química , Condrocitos/efectos de los fármacos , Condrocitos/citología , Materiales Biocompatibles/química , Materiales Biocompatibles/síntesis química , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Humanos , Concentración de Iones de Hidrógeno
3.
Nat Commun ; 15(1): 4820, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38844479

RESUMEN

Chondrocyte differentiation controls skeleton development and stature. Here we provide a comprehensive map of chondrocyte-specific enhancers and show that they provide a mechanistic framework through which non-coding genetic variants can influence skeletal development and human stature. Working with fetal chondrocytes isolated from mice bearing a Col2a1 fluorescent regulatory sensor, we identify 780 genes and 2'704 putative enhancers specifically active in chondrocytes using a combination of RNA-seq, ATAC-seq and H3K27ac ChIP-seq. Most of these enhancers (74%) show pan-chondrogenic activity, with smaller populations being restricted to limb (18%) or trunk (8%) chondrocytes only. Notably, genetic variations overlapping these enhancers better explain height differences than those overlapping non-chondrogenic enhancers. Finally, targeted deletions of identified enhancers at the Fgfr3, Col2a1, Hhip and, Nkx3-2 loci confirm their role in regulating cognate genes. This enhancer map provides a framework for understanding how genes and non-coding variations influence bone development and diseases.


Asunto(s)
Condrocitos , Condrogénesis , Elementos de Facilitación Genéticos , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos , Animales , Elementos de Facilitación Genéticos/genética , Humanos , Condrocitos/metabolismo , Condrocitos/citología , Ratones , Condrogénesis/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Regulación del Desarrollo de la Expresión Génica , Desarrollo Óseo/genética , Extremidades/embriología , Masculino , Diferenciación Celular/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Femenino
4.
Int J Mol Sci ; 25(11)2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38891793

RESUMEN

Joint-resident chondrogenic precursor cells have become a significant therapeutic option due to the lack of regenerative capacity in articular cartilage. Progenitor cells are located in the superficial zone of the articular cartilage, producing lubricin/Prg4 to decrease friction of cartilage surfaces during joint movement. Prg4-positive progenitors are crucial in maintaining the joint's structure and functionality. The disappearance of progenitor cells leads to changes in articular hyaline cartilage over time, subchondral bone abnormalities, and the formation of ectopic ossification. Genetic labeling cell technology has been the main tool used to characterize Prg4-expressing progenitor cells of articular cartilage in vivo through drug injection at different time points. This technology allows for the determination of the origin of progenitor cells and the tracking of their progeny during joint development and cartilage damage. We endeavored to highlight the currently known information about the Prg4-producing cell population in the joint to underline the significance of the role of these cells in the development of articular cartilage and its homeostasis. This review focuses on superficial progenitors in the joint, how they contribute to postnatal articular cartilage formation, their capacity for regeneration, and the consequences of Prg4 deficiency in these cells. We have accumulated information about the Prg4+ cell population of articular cartilage obtained through various elegantly designed experiments using transgenic technologies to identify potential opportunities for further research.


Asunto(s)
Cartílago Articular , Proteoglicanos , Células Madre , Cartílago Articular/metabolismo , Cartílago Articular/citología , Animales , Humanos , Células Madre/metabolismo , Células Madre/citología , Proteoglicanos/metabolismo , Condrogénesis , Condrocitos/metabolismo , Condrocitos/citología , Diferenciación Celular , Regeneración
5.
Int J Mol Sci ; 25(11)2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38891860

RESUMEN

Osteoarthritis (OA) is a degenerative joint disease commonly found in elderly people and obese patients. Currently, OA treatments are determined based on their condition severity and a medical professional's advice. The aim of this study was to differentiate human Wharton's jelly-derived mesenchymal stem cells (hWJ-MSCs) into chondrocytes for transplantation in OA-suffering guinea pigs. hWJ-MSCs were isolated using the explant culture method, and then, their proliferation, phenotypes, and differentiation ability were evaluated. Subsequently, hWJ-MSCs-derived chondrocytes were induced and characterized based on immunofluorescent staining, qPCR, and immunoblotting techniques. Then, early-OA-suffering guinea pigs were injected with hyaluronic acid (HA) containing either MSCs or 14-day-old hWJ-MSCs-derived chondrocytes. Results showed that hWJ-MSCs-derived chondrocytes expressed specific markers of chondrocytes including Aggrecan, type II collagen, and type X collagen proteins and ß-catenin, Sox9, Runx2, Col2a1, Col10a1, and ACAN gene expression markers. Administration of HA plus hWJ-MSCs-derived chondrocytes (HA-CHON) produced a better recovery rate of degenerative cartilages than HA plus MSCs or only HA. Histological assessments demonstrated no significant difference in Mankin's scores of recovered cartilages between HA-CHON-treated guinea pigs and normal articular cartilage guinea pigs. Transplantation of hWJ-MSCs-derived chondrocytes was more effective than undifferentiated hWJ-MSCs or hyaluronic acid for OA treatment in guinea pigs. This study provides a promising treatment to be used in early OA patients to promote recovery and prevent disease progression to severe osteoarthritis.


Asunto(s)
Diferenciación Celular , Condrocitos , Modelos Animales de Enfermedad , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Osteoartritis , Cordón Umbilical , Gelatina de Wharton , Animales , Cobayas , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Condrocitos/metabolismo , Condrocitos/citología , Osteoartritis/terapia , Osteoartritis/patología , Osteoartritis/metabolismo , Humanos , Gelatina de Wharton/citología , Trasplante de Células Madre Mesenquimatosas/métodos , Cordón Umbilical/citología , Ácido Hialurónico/farmacología , Células Cultivadas
6.
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
7.
Cell Commun Signal ; 22(1): 342, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38907358

RESUMEN

BACKGROUND: Mechanical unloading of the knee articular cartilage results in cartilage matrix atrophy, signifying the osteoarthritic-inductive potential of mechanical unloading. In contrast, mechanical loading stimulates cartilage matrix production. However, little is known about the response of meniscal fibrocartilage, a major mechanical load-bearing tissue of the knee joint, and its functional matrix-forming fibrochondrocytes to mechanical unloading events. METHODS: In this study, primary meniscus fibrochondrocytes isolated from the inner avascular region of human menisci from both male and female donors were seeded into porous collagen scaffolds to generate 3D meniscus models. These models were subjected to both normal gravity and mechanical unloading via simulated microgravity (SMG) for 7 days, with samples collected at various time points during the culture. RESULTS: RNA sequencing unveiled significant transcriptome changes during the 7-day SMG culture, including the notable upregulation of key osteoarthritis markers such as COL10A1, MMP13, and SPP1, along with pathways related to inflammation and calcification. Crucially, sex-specific variations in transcriptional responses were observed. Meniscus models derived from female donors exhibited heightened cell proliferation activities, with the JUN protein involved in several potentially osteoarthritis-related signaling pathways. In contrast, meniscus models from male donors primarily regulated extracellular matrix components and matrix remodeling enzymes. CONCLUSION: These findings advance our understanding of sex disparities in knee osteoarthritis by developing a novel in vitro model using cell-seeded meniscus constructs and simulated microgravity, revealing significant sex-specific molecular mechanisms and therapeutic targets.


Asunto(s)
Menisco , Simulación de Ingravidez , Humanos , Menisco/citología , Masculino , Femenino , Células Cultivadas , Persona de Mediana Edad , Proliferación Celular , Condrocitos/metabolismo , Condrocitos/citología , Adulto , Transcriptoma/genética
8.
Connect Tissue Res ; 65(3): 237-252, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38739041

RESUMEN

PURPOSE/AIM OF STUDY: During the development of the vertebrate skeleton, the progressive differentiation and maturation of chondrocytes from mesenchymal progenitors is precisely coordinated by multiple secreted factors and signaling pathways. The WNT signaling pathway has been demonstrated to play a major role in chondrogenesis. However, the identification of secreted factors that fine-tune WNT activity has remained elusive. Here, in this study, we have identified PI15 (peptidase inhibitor 15, protease Inhibitor 15, SugarCrisp), a member of the CAP (cysteine rich secretory proteins, antigen 5, and pathogenesis related 1 proteins) protein superfamily, as a novel secreted WNT antagonist dynamically upregulated during chondrocyte differentiation. MATERIALS AND METHODS: ATDC5 cells, C3H10T1/2 micromass cultures and primary chondrocyte cells were used as in vitro models of chondrogenesis. PI15 levels were stably depleted or overexpressed by viral shRNA or expression vectors. Chondrogenesis was evaluated by qPCR gene expression analysis and Alcian blue staining. Protein interactions were determined by coimmunoprecipitation assays. RESULTS AND CONCLUSIONS: shRNA-mediated knockdown of PI15 in ATDC5 cells, C3H10T1/2 cells or primary chondrocytes inhibits chondrogenesis, whereas the overexpression of PI15 strongly enhances chondrogenic potential. Mechanistically, PI15 binds to the LRP6 WNT co-receptor and blocks WNT-induced LRP6 phosphorylation, thus repressing WNT-induced transcriptional activity and alleviating the inhibitory effect of WNT signaling on chondrogenesis. Altogether, our findings suggest that PI15 acts as a key regulator of chondrogenesis and unveils a mechanism through which chondrocyte-derived molecules can modulate WNT activity as differentiation proceeds, thereby creating a positive feedback loop that further drives differentiation.


Asunto(s)
Diferenciación Celular , Condrocitos , Condrogénesis , Vía de Señalización Wnt , Condrocitos/metabolismo , Condrocitos/efectos de los fármacos , Condrocitos/citología , Diferenciación Celular/efectos de los fármacos , Animales , Vía de Señalización Wnt/efectos de los fármacos , Ratones , Condrogénesis/efectos de los fármacos , Línea Celular , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo
9.
PLoS One ; 19(5): e0297947, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38768116

RESUMEN

In various biological systems, analyzing how cell behaviors are coordinated over time would enable a deeper understanding of tissue-scale response to physiologic or superphysiologic stimuli. Such data is necessary for establishing both normal tissue function and the sequence of events after injury that lead to chronic disease. However, collecting and analyzing these large datasets presents a challenge-such systems are time-consuming to process, and the overwhelming scale of data makes it difficult to parse overall behaviors. This problem calls for an analysis technique that can quickly provide an overview of the groups present in the entire system and also produce meaningful categorization of cell behaviors. Here, we demonstrate the application of an unsupervised method-the Variational Autoencoder (VAE)-to learn the features of cells in cartilage tissue after impact-induced injury and identify meaningful clusters of chondrocyte behavior. This technique quickly generated new insights into the spatial distribution of specific cell behavior phenotypes and connected specific peracute calcium signaling timeseries with long term cellular outcomes, demonstrating the value of the VAE technique.


Asunto(s)
Cartílago Articular , Condrocitos , Cartílago Articular/citología , Condrocitos/citología , Animales , Análisis por Conglomerados , Señalización del Calcio
10.
Tissue Eng Regen Med ; 21(5): 791-807, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38771465

RESUMEN

BACKGROUND: Tissue engineering is increasingly viewed as a promising avenue for functional cartilage reconstruction. However, chondrocyte dedifferentiation during in vitro culture remains an obstacle for clinical translation of tissue engineered cartilage. Re-differentiated induction have been employed to induce dedifferentiated chondrocytes back to their original phenotype. Regrettably, these strategies have been proven to be only moderately effective. METHODS: To explore underlying mechanism, RNA transcriptome sequencing was conducted on primary chondrocytes (P0), dedifferentiated chondrocytes (P5), and redifferentiated chondrocytes (redifferentiation-induction of P5, P5.R). Based on multiple bioinformatics analysis, LGR5 was identified as a target gene. Subsequently, stable cell lines with LGR5 knocking-down and overexpression were established using P0 chondrocytes. The phenotypic changes in P1 and P5 chondrocytes with either LGR5 knockdown or overexpression were assessed to ascertain the potential influence of LGR5 dysregulation on chondrocyte phenotypes. Regulatory mechanism was then investigated using bioinformatic analysis, protein-protein docking, immunofluorescence co-localization and immunoprecipitation. RESULTS: The current study found that dysregulation of LGR5 can significantly impact the dedifferentiated phenotypes of chondrocytes (P5). Upregulation of LGR5 appears to activate the PI3K/AKT signal via increasing the phosphorylation levels of AKT (p-AKT1). Moreover, the increase of p-AKT1 may stabilize ß-catenin and enhance the intensity of Wnt/ß-catenin signal, and help to restore the dedifferentated phenotype of chondrocytes. CONCLUSION: LGR5 can modulate the phenotypes of chondrocytes in P5 passage through PI3K/AKT signaling pathway.


Asunto(s)
Diferenciación Celular , Condrocitos , Fenotipo , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Receptores Acoplados a Proteínas G , Transducción de Señal , Condrocitos/metabolismo , Condrocitos/citología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Humanos , Desdiferenciación Celular , Células Cultivadas
11.
Sci Rep ; 14(1): 10182, 2024 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702382

RESUMEN

Progressive cartilage deterioration leads to chronic inflammation and loss of joint function, causing osteoarthritis (OA) and joint disease. Although symptoms vary among individuals, the disease can cause severe pain and permanent disability, and effective therapies are urgently needed. Human Adipose-Derived Stem Cells (ADSCs) may differentiate into chondrocytes and are promising for treating OA. Moreover, recent studies indicate that electromagnetic fields (EMFs) could positively affect the chondrogenic differentiation potential of ADSCs. In this work, we investigated the impact of EMFs with frequencies of 35 Hertz and 58 Hertz, referred to as extremely low frequency-EMFs (ELF-EMFs), on the chondrogenesis of ADSCs, cultured in both monolayer and 3D cell micromasses. ADSC cultures were daily stimulated for 36 min with ELF-EMFs or left unstimulated, and the progression of the differentiation process was evaluated by morphological analysis, extracellular matrix deposition, and gene expression profiling of chondrogenic markers. In both culturing conditions, stimulation with ELF-EMFs did not compromise cell viability but accelerated chondrogenesis by enhancing the secretion and deposition of extracellular matrix components at earlier time points in comparison to unstimulated cells. This study showed that, in an appropriate chondrogenic microenvironment, ELF-EMFs enhance chondrogenic differentiation and may be an important tool for supporting and accelerating the treatment of OA through autologous adipose stem cell therapy.


Asunto(s)
Tejido Adiposo , Diferenciación Celular , Condrogénesis , Campos Electromagnéticos , Células Madre Mesenquimatosas , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Tejido Adiposo/citología , Células Cultivadas , Condrocitos/citología , Condrocitos/metabolismo , Matriz Extracelular/metabolismo , Supervivencia Celular/efectos de la radiación
12.
Sci Rep ; 14(1): 11765, 2024 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-38782958

RESUMEN

In vitro use of articular cartilage on an organ-on-a-chip (OOAC) via microfluidics is challenging owing to the dense extracellular matrix (ECM) composed of numerous protein moieties and few chondrocytes, which has limited proliferation potential and microscale translation. Hence, this study proposes a novel approach for using a combination of biopolymers and decellularised ECM (dECM) as a bioink additive in the development of scalable OOAC using a microfluidic platform. The bioink was tested with native chondrocytes and mesenchymal stem cell-induced chondrocytes using biopolymers of alginate and chitosan composite hydrogels. Two-dimensional (2D) and three-dimensional (3D) biomimetic tissue construction approaches have been used to characterise the morphology and cellular marker expression (by histology and confocal laser scanning microscopy), viability (cell viability dye using flow cytometry), and genotypic expression of ECM-specific markers (by quantitative PCR). The results demonstrated that the bioink had a significant impact on the increase in phenotypic and genotypic expression, with a statistical significance level of p < 0.05 according to Student's t-test. The use of a cell-laden biopolymer as a bioink optimised the niche conditions for obtaining hyaline-type cartilage under culture conditions, paving the way for testing mechano-responsive properties and translating these findings to a cartilage-on-a-chip microfluidics system.


Asunto(s)
Alginatos , Cartílago Articular , Quitosano , Condrocitos , Matriz Extracelular , Ingeniería de Tejidos , Quitosano/química , Alginatos/química , Cartílago Articular/metabolismo , Cartílago Articular/citología , Animales , Matriz Extracelular/metabolismo , Condrocitos/metabolismo , Condrocitos/citología , Ingeniería de Tejidos/métodos , Biopolímeros/química , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Andamios del Tejido/química , Dispositivos Laboratorio en un Chip , Hidrogeles/química , Células Cultivadas , Supervivencia Celular , Sistemas Microfisiológicos
13.
Cells ; 13(9)2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38727280

RESUMEN

Regenerative medicine harnesses stem cells' capacity to restore damaged tissues and organs. In vitro methods employing specific bioactive molecules, such as growth factors, bio-inductive scaffolds, 3D cultures, co-cultures, and mechanical stimuli, steer stem cells toward the desired differentiation pathways, mimicking their natural development. Chondrogenesis presents a challenge for regenerative medicine. This intricate process involves precise modulation of chondro-related transcription factors and pathways, critical for generating cartilage. Cartilage damage disrupts this process, impeding proper tissue healing due to its unique mechanical and anatomical characteristics. Consequently, the resultant tissue often forms fibrocartilage, which lacks adequate mechanical properties, posing a significant hurdle for effective regeneration. This review comprehensively explores studies showcasing the potential of amniotic mesenchymal stem cells (AMSCs) and amniotic epithelial cells (AECs) in chondrogenic differentiation. These cells exhibit innate characteristics that position them as promising candidates for regenerative medicine. Their capacity to differentiate toward chondrocytes offers a pathway for developing effective regenerative protocols. Understanding and leveraging the innate properties of AMSCs and AECs hold promise in addressing the challenges associated with cartilage repair, potentially offering superior outcomes in tissue regeneration.


Asunto(s)
Amnios , Diferenciación Celular , Condrogénesis , Humanos , Amnios/citología , Animales , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Condrocitos/citología , Condrocitos/metabolismo , Medicina Regenerativa/métodos , Ingeniería de Tejidos/métodos
14.
J Biomed Mater Res B Appl Biomater ; 112(6): e35433, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38817048

RESUMEN

Ex vivo tissue engineering is an effective therapeutic approach for the treatment of severe cartilage diseases that require tissue replenishment or replacement. This strategy demands scaffolds that are durable enough for long-term cell culture to form artificial tissue. Additionally, such scaffolds must be biocompatible to prevent the transplanted matrix from taking a toll on the patient's body. From the viewpoint of structure and bio-absorbability, a ß-tricalcium phosphate (ß-TCP) fiber scaffold (ßTFS) is expected to serve as a good scaffold for tissue engineering. However, the fragility and high solubility of ß-TCP fibers make this matrix unsuitable for long-term cell culture. To solve this problem, we developed an alginate-coated ß-TCP fiber scaffold (ßTFS-Alg). To assess cell proliferation and differentiation in the presence of ßTFS-Alg, we characterized ATDC5 cells, a chondrocyte-like cell line, when grown in this matrix. We found that alginate coated the surface of ßTFS fiber and suppressed the elution of Ca2+ from ß-TCP fibers. Due to the decreased solubility of ßTFS-Alg compared with ß-TCP, the former provided an improved scaffold for long-term cell culture. Additionally, we observed superior cell proliferation and upregulation of chondrogenesis marker genes in ATDC5 cells cultured in ßTFS-Alg. These results suggest that ßTFS-Alg is suitable for application in tissue culture.


Asunto(s)
Alginatos , Fosfatos de Calcio , Andamios del Tejido , Fosfatos de Calcio/química , Fosfatos de Calcio/farmacología , Alginatos/química , Andamios del Tejido/química , Proliferación Celular , Ratones , Ácido Glucurónico/química , Animales , Ácidos Hexurónicos/química , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Línea Celular , Condrocitos/citología , Condrocitos/metabolismo , Ingeniería de Tejidos , Ensayo de Materiales , Diferenciación Celular , Humanos , Técnicas de Cultivo de Célula
15.
Colloids Surf B Biointerfaces ; 239: 113959, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38772085

RESUMEN

Cartilage repair remains a major challenge in clinical trials. These current cartilage repair materials can not effectively promote chondrocyte generation, limiting their practical application in cartilage repair. In this work, we develop an implantable scaffold of RADA-16 peptide hydrogel incorporated with TGF-ß1 to provide a microenvironment for stem cell-directed differentiation and chondrocyte adhesion growth. The longest release of growth factor TGF-ß1 release can reach up to 600 h under physiological conditions. TGF-ß1/RADA-16 hydrogel was demonstrated to be a lamellar porous structure. Based on the cell culture with hBMSCs, TGF-ß1/RADA-16 hydrogel showed excellent ability to promote cell proliferation, directed differentiation into chondrocytes, and functional protein secretion. Within 14 days, 80% of hBMSCs were observed to be directed to differentiate into vigorous chondrocytes in the co-culture of TGF-ß1/RADA-16 hydrogels with hBMSCs. Specifically, these newly generated chondrocytes can secrete and accumulate large amounts of collagen II within 28 days, which can effectively promote the formation of cartilage tissue. Finally, the exploration of RADA-16 hydrogel-based scaffolds incorporated with TGF-ß1 bioactive species would further greatly promote the practical clinical trials of cartilage remediation, which might have excellent potential to promote cartilage regeneration in areas of cartilage damage.


Asunto(s)
Cartílago , Diferenciación Celular , Condrocitos , Hidrogeles , Regeneración , Andamios del Tejido , Factor de Crecimiento Transformador beta1 , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Regeneración/efectos de los fármacos , Andamios del Tejido/química , Hidrogeles/química , Hidrogeles/farmacología , Humanos , Condrocitos/efectos de los fármacos , Condrocitos/citología , Condrocitos/metabolismo , Diferenciación Celular/efectos de los fármacos , Cartílago/efectos de los fármacos , Cartílago/fisiología , Cartílago/metabolismo , Proliferación Celular/efectos de los fármacos , Ingeniería de Tejidos/métodos , Células Cultivadas , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Animales , Condrogénesis/efectos de los fármacos , Péptidos
16.
Int J Biol Macromol ; 270(Pt 1): 132126, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38723805

RESUMEN

Articular cartilage is an avascular and almost acellular tissue with limited self-regenerating capabilities. Although injectable hydrogels have garnered a lot of attention as a promising treatment, a biocompatible hydrogel with adequate mechanical properties is yet to be created. In this study, an interpenetrating network hydrogel comprised of chitosan and silk fibroin was created through electrostatic and hydrophobic bonds, respectively. The polymeric network of the scaffold combined an effective microenvironment for cell activity with enhanced mechanical properties to address the current issues in cartilage scaffolds. Furthermore, microspheres (MS) were utilized for a controlled release of methylprednisolone acetate (MPA), around ~75 % after 35 days. The proposed scaffolds demonstrated great mechanical stability with ~0.047 MPa compressive moduli and ~145 kPa compressive strength. Moreover, the degradation rate of the samples (~45 % after 35 days) was optimized to match neo-cartilage formation. Furthermore, the use of natural biomaterials yielded good biocompatibility with ~76 % chondrocyte viability after 7 days. According to gross observation after 12 weeks the defect site of the treated groups was filled with minimally discernible boundary. These results were confirmed by histopathology assays were the treated groups showed higher chondrocyte count and collagen type II expression.


Asunto(s)
Cartílago Articular , Quitosano , Fibroínas , Hidrogeles , Microesferas , Regeneración , Quitosano/química , Fibroínas/química , Animales , Regeneración/efectos de los fármacos , Hidrogeles/química , Cartílago Articular/efectos de los fármacos , Andamios del Tejido/química , Condrocitos/efectos de los fármacos , Condrocitos/citología , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Ingeniería de Tejidos/métodos , Conejos , Supervivencia Celular/efectos de los fármacos , Inyecciones , Acetato de Metilprednisolona/química
17.
J Nanobiotechnology ; 22(1): 300, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38816719

RESUMEN

BACKGROUND: Extracellular vesicles (EVs) derived from human adipose-derived mesenchymal stem cells (hADSCs) have shown great therapeutic potential in plastic and reconstructive surgery. However, the limited production and functional molecule loading of EVs hinder their clinical translation. Traditional two-dimensional culture of hADSCs results in stemness loss and cellular senescence, which is unfavorable for the production and functional molecule loading of EVs. Recent advances in regenerative medicine advocate for the use of three-dimensional culture of hADSCs to produce EVs, as it more accurately simulates their physiological state. Moreover, the successful application of EVs in tissue engineering relies on the targeted delivery of EVs to cells within biomaterial scaffolds. METHODS AND RESULTS: The hADSCs spheroids and hADSCs gelatin methacrylate (GelMA) microspheres are utilized to produce three-dimensional cultured EVs, corresponding to hADSCs spheroids-EVs and hADSCs microspheres-EVs respectively. hADSCs spheroids-EVs demonstrate excellent production and functional molecule loading compared with hADSCs microspheres-EVs. The upregulation of eight miRNAs (i.e. hsa-miR-486-5p, hsa-miR-423-5p, hsa-miR-92a-3p, hsa-miR-122-5p, hsa-miR-223-3p, hsa-miR-320a, hsa-miR-126-3p, and hsa-miR-25-3p) and the downregulation of hsa-miR-146b-5p within hADSCs spheroids-EVs show the potential of improving the fate of remaining ear chondrocytes and promoting cartilage formation probably through integrated regulatory mechanisms. Additionally, a quick and innovative pipeline is developed for isolating chondrocyte homing peptide-modified EVs (CHP-EVs) from three-dimensional dynamic cultures of hADSCs spheroids. CHP-EVs are produced by genetically fusing a CHP at the N-terminus of the exosomal surface protein LAMP2B. The CHP + LAMP2B-transfected hADSCs spheroids were cultured with wave motion to promote the secretion of CHP-EVs. A harvesting method is used to enable the time-dependent collection of CHP-EVs. The pipeline is easy to set up and quick to use for the isolation of CHP-EVs. Compared with nontagged EVs, CHP-EVs penetrate the biomaterial scaffolds and specifically deliver the therapeutic miRNAs to the remaining ear chondrocytes. Functionally, CHP-EVs show a major effect on promoting cell proliferation, reducing cell apoptosis and enhancing cartilage formation in remaining ear chondrocytes in the M1 macrophage-infiltrated microenvironment. CONCLUSIONS: In summary, an innovative pipeline is developed to obtain CHP-EVs from three-dimensional dynamic culture of hADSCs spheroids. This pipeline can be customized to increase EVs production and functional molecule loading, which meets the requirements for regulating remaining ear chondrocyte fate in the M1 macrophage-infiltrated microenvironment.


Asunto(s)
Condrocitos , Vesículas Extracelulares , Células Madre Mesenquimatosas , Péptidos , Esferoides Celulares , Humanos , Condrocitos/metabolismo , Condrocitos/citología , Vesículas Extracelulares/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/citología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Péptidos/química , Péptidos/metabolismo , MicroARNs/metabolismo , MicroARNs/genética , Macrófagos/metabolismo , Macrófagos/citología , Células Cultivadas , Microesferas , Ingeniería de Tejidos/métodos , Técnicas de Cultivo Tridimensional de Células/métodos , Microambiente Celular , Cartílago Auricular/metabolismo , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Diferenciación Celular
18.
Stem Cells ; 42(6): 554-566, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38613477

RESUMEN

Microtia is a congenital auricle dysplasia with a high incidence and tissue engineering technology provides a promising strategy to reconstruct auricles. We previously described that the engineered cartilage constructed from microtia chondrocytes exhibited inferior levels of biochemical and biomechanical properties, which was proposed to be resulted of the decreased migration ability of microtia chondrocytes. In the current study, we found that Rho GTPase members were deficient in microtia chondrocytes. By overexpressing RhoA, Rac1, and CDC42, respectively, we further demonstrated that RhoA took great responsibility for the decreased migration ability of microtia chondrocytes. Moreover, we constructed PGA/PLA scaffold-based cartilages to verify the chondrogenic ability of RhoA overexpressed microtia chondrocytes, and the results showed that overexpressing RhoA was of limited help in improving the quality of microtia chondrocyte engineered cartilage. However, coculture of adipose-derived stem cells (ADSCs) significantly improved the biochemical and biomechanical properties of engineered cartilage. Especially, coculture of RhoA overexpressed microtia chondrocytes and ADSCs produced an excellent effect on the wet weight, cartilage-specific extracellular matrix, and biomechanical property of engineered cartilage. Furthermore, we presented that coculture of RhoA overexpressed microtia chondrocytes and ADSCs combined with human ear-shaped PGA/PLA scaffold and titanium alloy stent fabricated by CAD/CAM and 3D printing technology effectively constructed and maintained auricle structure in vivo. Collectively, our results provide evidence for the essential role of RhoA in microtia chondrocytes and a developed strategy for the construction of patient-specific tissue-engineered auricular cartilage.


Asunto(s)
Condrocitos , Técnicas de Cocultivo , Microtia Congénita , Ingeniería de Tejidos , Proteína de Unión al GTP rhoA , Condrocitos/metabolismo , Condrocitos/citología , Humanos , Ingeniería de Tejidos/métodos , Proteína de Unión al GTP rhoA/metabolismo , Proteína de Unión al GTP rhoA/genética , Microtia Congénita/metabolismo , Microtia Congénita/genética , Cartílago Auricular/citología , Cartílago Auricular/metabolismo , Células Madre/metabolismo , Células Madre/citología , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Condrogénesis/genética , Masculino , Andamios del Tejido/química , Femenino
19.
BMC Biotechnol ; 24(1): 25, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38689309

RESUMEN

The reconstruction of a stable, nipple-shaped cartilage graft that precisely matches the natural nipple in shape and size on the contralateral side is a clinical challenge. While 3D printing technology can efficiently and accurately manufacture customized complex structures, it faces limitations due to inadequate blood supply, which hampers the stability of nipple-shaped cartilage grafts produced using this technology. To address this issue, we employed a biodegradable biomaterial, Poly(lactic-co-glycolic acid) (PLGA), loaded with Cell-Free Fat Extract (Ceffe). Ceffe has demonstrated the ability to promote angiogenesis and cell proliferation, making it an ideal bio-ink for bioprinting precise nipple-shaped cartilage grafts. We utilized the Ceffe/PLGA scaffold to create a porous structure with a precise nipple shape. This scaffold exhibited favorable porosity and pore size, ensuring stable shape maintenance and satisfactory biomechanical properties. Importantly, it could release Ceffe in a sustained manner. Our in vitro results confirmed the scaffold's good biocompatibility and its ability to promote angiogenesis, as evidenced by supporting chondrocyte proliferation and endothelial cell migration and tube formation. Furthermore, after 8 weeks of in vivo culture, the Ceffe/PLGA scaffold seeded with chondrocytes regenerated into a cartilage support structure with a precise nipple shape. Compared to the pure PLGA group, the Ceffe/PLGA scaffold showed remarkable vascular formation, highlighting the beneficial effects of Ceffe. These findings suggest that our designed Ceffe/PLGA scaffold with a nipple shape represents a promising strategy for precise nipple-shaped cartilage regeneration, laying a foundation for subsequent nipple reconstruction.


Asunto(s)
Cartílago , Condrocitos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Impresión Tridimensional , Ingeniería de Tejidos , Andamios del Tejido , Andamios del Tejido/química , Animales , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Ingeniería de Tejidos/métodos , Condrocitos/citología , Cartílago/citología , Cartílago/crecimiento & desarrollo , Proliferación Celular/efectos de los fármacos , Materiales Biocompatibles/química , Conejos , Porosidad , Ácido Poliglicólico/química , Neovascularización Fisiológica/efectos de los fármacos
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...