Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.981
Filtrar
1.
Sci Rep ; 14(1): 20161, 2024 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-39215168

RESUMEN

Insulin-like growth factor (IGF) signaling is required for proper growth and skeletal development in vertebrates. Consequently, its dysregulation may lead to abnormalities of growth or skeletal structures. IGF is involved in the regulation of cell proliferation and differentiation of chondrocytes. However, the availability of bioactive IGF may be controlled by antagonizing IGF binding proteins (IGFBPs) in the circulation and tissues. As the metalloproteinase PAPP-A specifically cleaves members of the IGFBP family, we hypothesized that PAPP-A activity liberates bioactive IGF in cartilage. In PAPP-A knockout mice, the femur length was reduced and the mice showed a disorganized columnar organization of growth plate chondrocytes. Similarly, zebrafish lacking pappaa showed reduced length of Meckel's cartilage and disorganized chondrocytes, reminiscent of the mouse knockout phenotype. Expression of chondrocyte differentiation markers (sox9a, ihha, and col10a1) was markedly affected in Meckel's cartilage of pappaa knockout zebrafish, indicating that differentiation of chondrocytes was compromised. Additionally, the zebrafish pappaa knockout phenotype was mimicked by pharmacological inhibition of IGF signaling, and it could be rescued by treatment with exogenous recombinant IGF-I. In conclusion, our data suggests that IGF activity in the growing cartilage, and hence IGF signaling in chondrocytes, requires the presence of PAPP-A. The absence of PAPP-A causes aberrant chondrocyte organization and compromised growth in both mice and zebrafish.


Asunto(s)
Diferenciación Celular , Condrocitos , Proteína Plasmática A Asociada al Embarazo , Pez Cebra , Animales , Ratones , Cartílago/metabolismo , Cartílago/citología , Condrocitos/metabolismo , Condrocitos/citología , Condrogénesis , Placa de Crecimiento/metabolismo , Placa de Crecimiento/citología , Ratones Noqueados , Proteína Plasmática A Asociada al Embarazo/metabolismo , Proteína Plasmática A Asociada al Embarazo/genética , Transducción de Señal , Somatomedinas/metabolismo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
2.
Proc Inst Mech Eng H ; 238(7): 741-754, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39109566

RESUMEN

The objectives of this study were to compare the chondrogenic potential of cells derived from different layers of Mandibular condyle cartilage and to gain further understanding of the impact of chondrogenic cues when embedded into a novel hydrogel scaffold (PGH, a polymer blend of poly (ethylene glycol), gelatin, and heparin) compared to a gelatin hydrogel scaffold (GEL). Cartilage layer cells (CLCs) and fibroblastic superficial layer cells (SLCs) were harvested from the mandibular condyle of boer goats obtained from a local abattoir. After expansion, cells were seeded into PGH and GEL hydrogels and cultured in chondrogenic media for 3 weeks. Scaffolds were harvested at 0, 1, and 3 week(s) and processed for gross appearance, histochemical, biochemical, and mechanical assays. In terms of chondrogenesis, major differences were observed between scaffold materials, but not cell types. Glycosaminoglycan (GAG) staining showed GEL scaffolds deposited GAG during the 3 week period, which was also confirmed with the biochemical testing. Moreover, GEL scaffolds had significantly higher compressive modulus and peak stress than PGH scaffolds at all time points with the largest difference seen in week 3. It can be concluded that GEL outperformed PGH in chondrogenesis. It can also be concluded that materials play a more important role in the process of chondrogenesis than the tested cell populations. Fibroblastic SLCs were shown to have similar chondrogenic potential as CLCs cells, suggesting a rich pool of progenitor cells in the superficial fibroblastic layer capable of undergoing chondrogenesis given appropriate physical and chemical cues.


Asunto(s)
Cartílago , Condrogénesis , Gelatina , Cabras , Hidrogeles , Articulación Temporomandibular , Hidrogeles/química , Gelatina/química , Animales , Cartílago/citología , Articulación Temporomandibular/citología , Andamios del Tejido/química , Polimerizacion , Cóndilo Mandibular/citología , Cóndilo Mandibular/fisiología , Procesos Fotoquímicos , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo
3.
Matrix Biol ; 133: 33-42, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39097037

RESUMEN

Serine proteinase inhibitors (serpins) are a family of structurally similar proteins which regulate many diverse biological processes from blood coagulation to extracellular matrix (ECM) remodelling. Chondrogenesis involves the condensation and differentiation of mesenchymal stem cells (MSCs) into chondrocytes which occurs during early development. Here, and for the first time, we demonstrate that one serpin, SERPINA3 (gene name SERPINA3, protein also known as alpha-1 antichymotrypsin), plays a critical role in chondrogenic differentiation. We observed that SERPINA3 expression was markedly induced at early time points during in vitro chondrogenesis. We examined the expression of SERPINA3 in human cartilage development, identifying significant enrichment of SERPINA3 in developing cartilage compared to total limb, which correlated with well-described markers of cartilage differentiation. When SERPINA3 was silenced using siRNA, cartilage pellets were smaller and contained lower proteoglycan as determined by dimethyl methylene blue assay (DMMB) and safranin-O staining. Consistent with this, RNA sequencing revealed significant downregulation of genes associated with cartilage ECM formation perturbing chondrogenesis. Conversely, SERPINA3 silencing had a negligible effect on the gene expression profile during osteogenesis suggesting the role of SERPINA3 is specific to chondrocyte differentiation. The global effect on cartilage formation led us to investigate the effect of SERPINA3 silencing on the master transcriptional regulator of chondrogenesis, SOX9. Indeed, we observed that SOX9 protein levels were markedly reduced at early time points suggesting a role for SERPINA3 in regulating SOX9 expression and activity. In summary, our data support a non-redundant role for SERPINA3 in enabling chondrogenesis via regulation of SOX9 levels.


Asunto(s)
Diferenciación Celular , Condrocitos , Condrogénesis , Matriz Extracelular , Células Madre Mesenquimatosas , Serpinas , Condrogénesis/genética , Humanos , Condrocitos/metabolismo , Condrocitos/citología , Matriz Extracelular/metabolismo , Matriz Extracelular/genética , Serpinas/genética , Serpinas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Cartílago/metabolismo , Cartílago/crecimiento & desarrollo , Cartílago/citología , Regulación del Desarrollo de la Expresión Génica , Biomarcadores/metabolismo , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Células Cultivadas
4.
Biofabrication ; 16(4)2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39136309

RESUMEN

Multicellular spheroids such as microtissues and organoids have demonstrated great potential for tissue engineering applications in recent years as these 3D cellular units enable improved cell-cell and cell-matrix interactions. Current bioprinting processes that use multicellular spheroids as building blocks have demonstrated limited control on post printing distribution of cell spheroids or moderate throughput and printing efficiency. In this work, we presented a laser-assisted bioprinting approach able to transfer multicellular spheroids as building blocks for larger tissue structures. Cartilaginous multicellular spheroids formed by human periosteum derived cells (hPDCs) were successfully bioprinted possessing high viability and the capacity to undergo chondrogenic differentiation post printing. Smaller hPDC spheroids with diameters ranging from ∼100 to 150µm were successfully bioprinted through the use of laser-induced forward transfer method (LIFT) however larger spheroids constituted a challenge. For this reason a novel alternative approach was developed termed as laser induced propulsion of mesoscopic objects (LIPMO) whereby we were able to bioprint spheroids of up to 300µm. Moreover, we combined the bioprinting process with computer aided image analysis demonstrating the capacity to 'target and shoot', through automated selection, multiple large spheroids in a single sequence. By taking advantage of target and shoot system, multilayered constructs containing high density cell spheroids were fabricated.


Asunto(s)
Bioimpresión , Cartílago , Rayos Láser , Esferoides Celulares , Ingeniería de Tejidos , Bioimpresión/métodos , Humanos , Esferoides Celulares/citología , Ingeniería de Tejidos/métodos , Cartílago/citología , Cartílago/fisiología , Periostio/citología , Impresión Tridimensional , Condrogénesis , Diferenciación Celular , Células Cultivadas , Supervivencia Celular
5.
Biofabrication ; 16(4)2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39142325

RESUMEN

Hyalocytes, which are considered to originate from the monocyte/macrophage lineage, play active roles in vitreous collagen and hyaluronic acid synthesis. Obtaining a hyalocyte-compatible bioink during the 3D bioprinting of eye models is challenging. In this study, we investigated the suitability of a cartilage-decellularized extracellular matrix (dECM)-based bioink for printing a vitreous body model. Given that achieving a 3D structure and environment identical to those of the vitreous body necessitates good printability and biocompatibility, we examined the mechanical and biological properties of the developed dECM-based bioink. Furthermore, we proposed a 3D bioprinting strategy for volumetric vitreous body fabrication that supports cell viability, transparency, and self-sustainability. The construction of a 3D structure composed of bioink microfibers resulted in improved transparency and hyalocyte-like macrophage activity in volumetric vitreous mimetics, mimicking real vitreous bodies. The results indicate that our 3D structure could serve as a platform for drug testing in disease models and demonstrate that the proposed printing technology, utilizing a dECM-based bioink and volumetric vitreous body, has the potential to facilitate the development of advanced eye models for future studies on floater formation and visual disorders.


Asunto(s)
Bioimpresión , Matriz Extracelular , Tinta , Impresión Tridimensional , Cuerpo Vítreo , Cuerpo Vítreo/metabolismo , Cuerpo Vítreo/citología , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Animales , Bioimpresión/métodos , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Humanos , Cartílago/citología , Cartílago/química , Cartílago/metabolismo , Supervivencia Celular , Macrófagos/metabolismo , Macrófagos/citología
6.
Tissue Cell ; 90: 102475, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39059134

RESUMEN

Decellularized cortical bone powder derived from adult animals has been shown to induce bone remodeling. Furthermore, it is increasingly evident that the extracellular matrix (ECM) within decellularized tissues differs depending on the source tissue and the age of the animal, leading to distinct effects on cells. In this study, we prepared powders from decellularized fetal and adult porcine bone tissues and conducted biological analyses to determine if the decellularized tissue could induce adipose-derived stem cell differentiation. Decellularized fetal tissues and adult cortical bone were converted into powder by cryomilling, but decellularized adult bone marrow and cartilage were not powdered through this process. In vitro assessments revealed that decellularized fetal tissues, decellularized adult cartilage extract, and decellularized fetal cartilage powder can induce osteoblast differentiation. This study suggests that decellularized fetal bone tissues and adult cartilage contain ECM components that can induce osteoblast differentiation. Additionally, it highlights the utility of decellularized fetal cartilage powder for bone reconstruction.


Asunto(s)
Cartílago , Diferenciación Celular , Matriz Extracelular , Feto , Osteogénesis , Animales , Cartílago/citología , Cartílago/metabolismo , Matriz Extracelular/metabolismo , Porcinos , Feto/citología , Huesos/citología , Osteoblastos/citología , Osteoblastos/metabolismo , Matriz Extracelular Descelularizada/farmacología
7.
Biointerphases ; 19(4)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-39051723

RESUMEN

Once damaged, cartilage has poor intrinsic capacity to repair itself. Current cartilage repair strategies cannot restore the damaged tissue sufficiently. It is hypothesized that biomimetic scaffolds, which can recapitulate important properties of the cartilage extracellular matrix, play a beneficial role in supporting cell behaviors such as growth, cartilage differentiation, and integration with native cartilage, ultimately facilitating tissue recovery. Adipose-derived stem cells regenerated cartilage upon the sequential release of transforming growth factor ß1(TGFß1) and fibroblast growth factor 2(FGF2) using a nanofibrous scaffold, in order to get the recovery of functional cartilage. Experiments in vitro have demonstrated that the release sequence of growth factors FGF2 to TGFß1 is the most essential to promote adipose-derived stem cells into chondrocytes that then synthesize collagen II. Mouse subcutaneous implantation indicated that the treatment sequence of FGF2 to TGFß1 was able to significantly induce multiple increase in cartilage regeneration in vivo. This result demonstrates that the group treated with FGF2 to TGFß1 released from a nanofibrous scaffold provides a good strategy for cartilage regeneration by making a favorable microenvironment for cell growth and cartilage regeneration.


Asunto(s)
Diferenciación Celular , Factor 2 de Crecimiento de Fibroblastos , Nanofibras , Células Madre , Andamios del Tejido , Factor de Crecimiento Transformador beta1 , Animales , Factor 2 de Crecimiento de Fibroblastos/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Ratones , Nanofibras/química , Diferenciación Celular/efectos de los fármacos , Andamios del Tejido/química , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/fisiología , Condrogénesis/efectos de los fármacos , Cartílago/efectos de los fármacos , Cartílago/citología , Cartílago/fisiología , Tejido Adiposo/citología , Condrocitos/citología , Condrocitos/efectos de los fármacos , Condrocitos/fisiología , Células Cultivadas , Ingeniería de Tejidos/métodos
8.
J Mech Behav Biomed Mater ; 157: 106658, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39018919

RESUMEN

Although the sacroiliac (SI) joint can be a source of lower back and buttock pain, no comprehensive characterization studies on SI cartilage have been conducted. Using the minipig as a large animal model, this study conducted the first biomechanical, biochemical, and histological characterization of SI joint cartilage. Because previous literature has reported that sacral cartilage and iliac cartilage within the SI joint are histologically distinct, concomitantly it was expected that functional properties of the sacral cartilage would differ from those of the iliac cartilage. Creep indentation, uniaxial tension, biochemical, and histological analyses were conducted on the sacral and iliac cartilage of skeletally mature female Yucatan minipigs (n = 6-8 for all quantitative tests). Concurring with prior literature, the iliac cartilage appeared to be more fibrous than the sacral cartilage. Glycosaminoglycan content was 2.2 times higher in the sacral cartilage. The aggregate modulus of the sacral cartilage was 133 ± 62 kPa, significantly higher than iliac cartilage, which only had an aggregate modulus of 51 ± 61 kPa. Tensile testing was conducted in both cranial-caudal and ventral-dorsal axes, and Young's modulus values ranged from 2.5 ± 1.5 MPa to 13.6 ± 1.5 MPa, depending on anatomical structure (i.e., sacral vs. iliac) and orientation of the tensile test. The Young's modulus of sacral cartilage was 5.5 times higher in the cranial-caudal axis and 2.0 times higher in the ventral-dorsal axis than the iliac cartilage. The results indicate that the sacral and iliac cartilages are functionally distinct from each other. Understanding the distinct differences between sacral and iliac cartilage provides insight into the structure and function of the SI joint, which may inform future research aimed at repairing SI joint cartilage.


Asunto(s)
Fenómenos Mecánicos , Articulación Sacroiliaca , Porcinos Enanos , Animales , Porcinos , Fenómenos Biomecánicos , Femenino , Cartílago/fisiología , Cartílago/citología , Ensayo de Materiales , Cartílago Articular/fisiología , Cartílago Articular/citología , Pruebas Mecánicas , Glicosaminoglicanos/metabolismo
9.
Stem Cell Rev Rep ; 20(6): 1656-1666, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38837115

RESUMEN

Cell surface marker expression is one of the criteria for defining human mesenchymal stem or stromal cells (MSC) in vitro. However, it is unclear if expression of markers including CD73 and CD90 reflects the in vivo origin of cultured cells. We evaluated expression of 15 putative MSC markers in primary cultured cells from periosteum and cartilage to determine whether expression of these markers reflects either the differentiation state of cultured cells or the self-renewal of in vivo populations. Cultured cells had universal and consistent expression of various putative stem cell markers including > 95% expression CD73, CD90 and PDPN in both periosteal and cartilage cultures. Altering the culture surface with extracellular matrix coatings had minimal effect on cell surface marker expression. Osteogenic differentiation led to loss of CD106 and CD146 expression, however CD73 and CD90 were retained in > 90% of cells. We sorted freshly isolated periosteal populations capable of CFU-F formation on the basis of CD90 expression in combination with CD34, CD73 and CD26. All primary cultures universally expressed CD73 and CD90 and lacked CD34, irrespective of the expression of these markers ex vivo indicating phenotypic convergence in vitro. We conclude that markers including CD73 and CD90 are acquired in vitro in most 'mesenchymal' cells capable of expansion. Overall, we demonstrate that in vitro expression of many cell surface markers in plastic-adherent cultures is unrelated to their expression prior to culture.


Asunto(s)
5'-Nucleotidasa , Biomarcadores , Diferenciación Celular , Células Madre Mesenquimatosas , Osteogénesis , Fenotipo , Antígenos Thy-1 , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Humanos , Biomarcadores/metabolismo , Células Cultivadas , Antígenos Thy-1/metabolismo , 5'-Nucleotidasa/metabolismo , Periostio/citología , Periostio/metabolismo , Cartílago/metabolismo , Cartílago/citología , Proteínas Ligadas a GPI
10.
Int J Biol Macromol ; 272(Pt 1): 132848, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38830491

RESUMEN

Collagen-based (COL) hydrogels could be a promising treatment option for injuries to the articular cartilage (AC) becuase of their similarity to AC native extra extracellular matrix. However, the high hydration of COL hydrogels poses challenges for AC's mechanical properties. To address this, we developed a hydrogel platform that incorporating cellulose nanocrystals (CNCs) within COL and followed by plastic compression (PC) procedure to expel the excessive fluid out. This approach significantly improved the mechanical properties of the hydrogels and enhanced the chondrogenic differentiation of mesenchymal stem cells (MSCs). Radially confined PC resulted in higher collagen fibrillar densities together with reducing fibril-fibril distances. Compressed hydrogels containing CNCs exhibited the highest compressive modulus and toughness. MSCs encapsulated in these hydrogels were initially affected by PC, but their viability improved after 7 days. Furthermore, the morphology of the cells and their secretion of glycosaminoglycans (GAGs) were positively influenced by the compressed COL-CNC hydrogel. Our findings shed light on the combined effects of PC and CNCs in improving the physical and mechanical properties of COL and their role in promoting chondrogenesis.


Asunto(s)
Diferenciación Celular , Celulosa , Condrogénesis , Colágeno , Hidrogeles , Células Madre Mesenquimatosas , Nanopartículas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Celulosa/química , Celulosa/farmacología , Condrogénesis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Nanopartículas/química , Colágeno/química , Colágeno/farmacología , Hidrogeles/química , Hidrogeles/farmacología , Animales , Plásticos/química , Plásticos/farmacología , Supervivencia Celular/efectos de los fármacos , Glicosaminoglicanos/metabolismo , Cartílago/citología , Cartílago/efectos de los fármacos
11.
Int J Biol Macromol ; 273(Pt 2): 133217, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38897519

RESUMEN

Recent research focuses on fabricating scaffolds imitating the extracellular matrix (ECM) in texture, composition, and functionality. Moreover, specific nano-bio-particles can enhance cell differentiation. Decellularized ECM nanoparticles possess all of the mentioned properties. In this research, cartilage ECM, extracted from the cow's femur condyle, was decellularized, and ECM nanoparticles were synthesized. Finally, nanocomposite electrospun fibers containing polyhydroxybutyrate (PHB), chitosan (Cs) nanoparticles, and ECM nanoparticles were fabricated and characterized. TEM and DLS results revealed ECM nanoparticle sizes of 17.51 and 21.6 nm, respectively. Optimal performance was observed in the scaffold with 0.75 wt% ECM nanoparticles (PHB-Cs/0.75E). By adding 0.75 wt% ECM, the ultimate tensile strength and elongation at break increased by about 29 % and 21 %, respectively, while the water contact angle and crystallinity decreased by about 36° and 2 %, respectively. Uneven and rougher surfaces of the PHB-Cs/0.75E were determined by FESEM and AFM images, respectively. TEM images verified the uniform dispersion of nanoparticles within the fibers. After 70 days of degradation in PBS, the PHB-Cs/0.75E and PHB-Cs scaffolds demonstrated insignificant weight loss differences. Eventually, enhanced viability, attachment, and proliferation of the human costal chondrocytes on the PHB-Cs/0.75E scaffold, concluded from MTT, SEM, and DAPI staining, confirmed its potential for cartilage tissue engineering.


Asunto(s)
Cartílago , Quitosano , Matriz Extracelular , Hidroxibutiratos , Nanopartículas , Ingeniería de Tejidos , Andamios del Tejido , Quitosano/química , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Nanopartículas/química , Animales , Hidroxibutiratos/química , Cartílago/citología , Cartílago/metabolismo , Poliésteres/química , Humanos , Bovinos , Condrocitos/citología , Condrocitos/metabolismo , Polihidroxibutiratos
12.
Stem Cell Res Ther ; 15(1): 183, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38902814

RESUMEN

In the realm of studying joint-related diseases, there is a continuous quest for more accurate and representative models. Recently, regenerative medicine and tissue engineering have seen a growing interest in utilizing organoids as powerful tools for studying complex biological systems in vitro. Organoids, three-dimensional structures replicating the architecture and function of organs, provide a unique platform for investigating disease mechanisms, drug responses, and tissue regeneration. The surge in organoid research is fueled by the need for physiologically relevant models to bridge the gap between traditional cell cultures and in vivo studies. Osteochondral organoids have emerged as a promising avenue in this pursuit, offering a better platform to mimic the intricate biological interactions within bone and cartilage. This review explores the significance of osteochondral organoids and the need for their development in advancing our understanding and treatment of bone and cartilage-related diseases. It summarizes osteochondral organoids' insights and research progress, focusing on their composition, materials, cell sources, and cultivation methods, as well as the concept of organoids on chips and application scenarios. Additionally, we address the limitations and challenges these organoids face, emphasizing the necessity for further research to overcome these obstacles and facilitate orthopedic regeneration.


Asunto(s)
Organoides , Ingeniería de Tejidos , Organoides/citología , Organoides/metabolismo , Humanos , Ingeniería de Tejidos/métodos , Animales , Cartílago/citología , Medicina Regenerativa/métodos , Huesos/citología , Huesos/fisiología
13.
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
14.
Int J Biol Macromol ; 273(Pt 1): 132819, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38830498

RESUMEN

The avascular nature of hyaline cartilage results in limited spontaneous self-repair and regenerative capabilities when damaged. Recent advances in three-dimensional bioprinting have enabled the precise dispensing of cell-laden biomaterials, commonly referred to as 'bioinks', which are emerging as promising solutions for tissue regeneration. An effective bioink for cartilage tissue engineering needs to create a micro-environment that promotes cell differentiation and supports neocartilage tissue formation. In this study, we introduced an innovative bioink composed of photocurable acrylated type I collagen (COLMA), thiol-modified hyaluronic acid (THA), and poly(ethylene glycol) diacrylate (PEGDA) for 3D bioprinting cartilage grafts using human nasal chondrocytes. Both collagen and hyaluronic acid, being key components of the extracellular matrix (ECM) in the human body, provide essential biological cues for tissue regeneration. We evaluated three formulations - COLMA, COLMA+THA, and COLMA+THA+PEGDA - for their printability, cell viability, structural integrity, and capabilities in forming cartilage-like ECM. The addition of THA and PEGDA significantly enhanced these properties, showcasing the potential of this bioink in advancing applications in cartilage repair and reconstructive surgery.


Asunto(s)
Ácido Hialurónico , Ingeniería de Tejidos , Andamios del Tejido , Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Ingeniería de Tejidos/métodos , Humanos , Andamios del Tejido/química , Condrocitos/citología , Condrocitos/efectos de los fármacos , Polietilenglicoles/química , Bioimpresión/métodos , Colágeno/química , Impresión Tridimensional , Cartílago/citología , Matriz Extracelular/química , Supervivencia Celular/efectos de los fármacos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Tinta
15.
Adv Healthc Mater ; 13(22): e2400431, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38768997

RESUMEN

The repair and regeneration of cartilage has always been a hot topic in medical research. Cartilage organoids (CORGs) are special cartilage tissue created using tissue engineering techniques outside the body. These engineered organoids tissues provide models that simulate the complex biological functions of cartilage, opening new possibilities for cartilage regenerative medicine and treatment strategies. However, it is crucial to establish suitable matrix scaffolds for the cultivation of CORGs. In recent years, utilizing hydrogel to culture stem cells and induce their differentiation into chondrocytes has emerged as a promising method for the in vitro construction of CORGs. In this review, the methods for establishing CORGs are summarized and an overview of the advantages and limitations of using matrigel in the cultivation of such organoids is provided. Furthermore, the importance of cartilage tissue ECM and alternative hydrogel substitutes for Matrigel, such as alginate, peptides, silk fibroin, and DNA derivatives is discussed, and the pros and cons of using these hydrogels for the cultivation of CORGs are outlined. Finally, the challenges and future directions in hydrogel research for CORGs are discussed. It is hoped that this article provides valuable references for the design and development of hydrogels for CORGs.


Asunto(s)
Cartílago , Hidrogeles , Organoides , Ingeniería de Tejidos , Hidrogeles/química , Organoides/citología , Organoides/metabolismo , Humanos , Cartílago/citología , Ingeniería de Tejidos/métodos , Animales , Proteoglicanos/química , Proteoglicanos/farmacología , Andamios del Tejido/química , Laminina/química , Laminina/farmacología , Condrocitos/citología , Condrocitos/metabolismo , Colágeno , Combinación de Medicamentos
16.
Biofabrication ; 16(3)2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38697073

RESUMEN

Osteochondral tissue (OC) repair remains a significant challenge in the field of musculoskeletal tissue engineering. OC tissue displays a gradient structure characterized by variations in both cell types and extracellular matrix components, from cartilage to the subchondral bone. These functional gradients observed in the native tissue have been replicated to engineer OC tissuein vitro. While diverse fabrication methods have been employed to create these microenvironments, emulating the natural gradients and effective regeneration of the tissue continues to present a significant challenge. In this study, we present the design and development of CMC-silk interpenetrating (IPN) hydrogel with opposing dual biochemical gradients similar to native tissue with the aim to regenerate the complete OC unit. The gradients of biochemical cues were generated using an in-house-built extrusion system. Firstly, we fabricated a hydrogel that exhibits a smooth transition of sulfated carboxymethyl cellulose (sCMC) and TGF-ß1 (SCT gradient hydrogel) from the upper to the lower region of the IPN hydrogel to regenerate the cartilage layer. Secondly, a hydrogel with a hydroxyapatite (HAp) gradient (HAp gradient hydrogel) from the lower to the upper region was fabricated to facilitate the regeneration of the subchondral bone layer. Subsequently, we developed a dual biochemical gradient hydrogel with a smooth transition of sCMC + TGF-ß1 and HAp gradients in opposing directions, along with a blend of both biochemical cues in the middle. The results showed that the dual biochemical gradient hydrogels with biochemical cues corresponding to the three zones (i.e. cartilage, interface and bone) of the OC tissue led to differentiation of bone-marrow-derived mesenchymal stem cells to zone-specific lineages, thereby demonstrating their efficacy in directing the fate of progenitor cells. In summary, our study provided a simple and innovative method for incorporating gradients of biochemical cues into hydrogels. The gradients of biochemical cues spatially guided the differentiation of stem cells and facilitated tissue growth, which would eventually lead to the regeneration of the entire OC tissue with a smooth transition from cartilage (soft) to bone (hard) tissues. This promising approach is translatable and has the potential to generate numerous biochemical and biophysical gradients for regeneration of other interface tissues, such as tendon-to-muscle and ligament-to-bone.


Asunto(s)
Hidrogeles , Ingeniería de Tejidos , Hidrogeles/química , Animales , Andamios del Tejido/química , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Condrogénesis/efectos de los fármacos , Cartílago/citología , Cartílago/fisiología , Diferenciación Celular/efectos de los fármacos , Huesos/citología , Durapatita/química , Durapatita/farmacología
17.
Sci Rep ; 14(1): 11991, 2024 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-38796487

RESUMEN

Physiochemical tissue inducers and mechanical stimulation are both efficient variables in cartilage tissue fabrication and regeneration. In the presence of biomolecules, decellularized extracellular matrix (ECM) may trigger and enhance stem cell proliferation and differentiation. Here, we investigated the controlled release of transforming growth factor beta (TGF-ß1) as an active mediator of mesenchymal stromal cells (MSCs) in a biocompatible scaffold and mechanical stimulation for cartilage tissue engineering. ECM-derived hydrogel with TGF-ß1-loaded alginate-based microspheres (MSs) was created to promote human MSC chondrogenic development. Ex vivo explants and a complicated multiaxial loading bioreactor replicated the physiological conditions. Hydrogels with/without MSs and TGF-ß1 were highly cytocompatible. MSCs in ECM-derived hydrogel containing TGF-ß1/MSs showed comparable chondrogenic gene expression levels as those hydrogels with TGF-ß1 added in culture media or those without TGF-ß1. However, constructs with TGF-ß1 directly added within the hydrogel had inferior properties under unloaded conditions. The ECM-derived hydrogel group including TGF-ß1/MSs under loading circumstances formed better cartilage matrix in an ex vivo osteochondral defect than control settings. This study demonstrates that controlled local delivery of TGF-ß1 using MSs and mechanical loading is essential for neocartilage formation by MSCs and that further optimization is needed to prevent MSC differentiation towards hypertrophy.


Asunto(s)
Alginatos , Reactores Biológicos , Condrogénesis , Hidrogeles , Células Madre Mesenquimatosas , Microesferas , Ingeniería de Tejidos , Alginatos/química , Ingeniería de Tejidos/métodos , Humanos , Hidrogeles/química , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Animales , Cartílago/metabolismo , Cartílago/citología , Andamios del Tejido/química , Matriz Extracelular Descelularizada/química , Factor de Crecimiento Transformador beta1/metabolismo , Diferenciación Celular , Células Cultivadas , Factor de Crecimiento Transformador beta/metabolismo , Matriz Extracelular/metabolismo
18.
J Mater Chem B ; 12(22): 5360-5376, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38700242

RESUMEN

Articular cartilage tissue has limited self-repair capabilities, with damage frequently progressing to irreversible degeneration. Engineered tissues constructed through bioprinting and embedded with stem cell aggregates offer promising therapeutic alternatives. Aggregates of bone marrow mesenchymal stromal cells (BMSCs) demonstrate enhanced and more rapid chondrogenic differentiation than isolated cells, thus facilitating cartilage repair. However, it remains a key challenge to precisely control biochemical microenvironments to regulate cellular adhesion and cohesion within bioprinted matrices simultaneously. Herein, this work reports a bioprintable hydrogel matrix with high cellular adhesion and aggregation properties for cartilage repair. The hydrogel comprises an enhanced cell-adhesive gelatin methacrylate and a cell-cohesive chitosan methacrylate (CHMA), both of which are subjected to photo-initiated crosslinking. By precisely adjusting the CHMA content, the mechanical stability and biochemical cues of the hydrogels are finely tuned to promote cellular aggregation, chondrogenic differentiation and cartilage repair implantation. Multi-layer constructs encapsulated with BMSCs, with high cell viability reaching 91.1%, are bioprinted and photo-crosslinked to support chondrogenic differentiation for 21 days. BMSCs rapidly form aggregates and display efficient chondrogenic differentiation both on the hydrogels and within bioprinted constructs, as evidenced by the upregulated expression of Sox9, Aggrecan and Collagen 2a1 genes, along with high protein levels. Transplantation of these BMSC-laden bioprinted hydrogels into cartilaginous defects demonstrates effective hyaline cartilage repair. Overall, this cell-responsive hydrogel scaffold holds immense promise for applications in cartilage tissue engineering.


Asunto(s)
Bioimpresión , Condrogénesis , Hidrogeles , Células Madre Mesenquimatosas , Regeneración , Condrogénesis/efectos de los fármacos , Hidrogeles/química , Hidrogeles/farmacología , Animales , Células Madre Mesenquimatosas/citología , Regeneración/efectos de los fármacos , Cartílago Articular , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Diferenciación Celular/efectos de los fármacos , Ingeniería de Tejidos , Metacrilatos/química , Supervivencia Celular/efectos de los fármacos , Cartílago/metabolismo , Cartílago/citología , Células Cultivadas , Humanos
20.
Differentiation ; 138: 100782, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38810379

RESUMEN

The mandible is composed of several musculoskeletal tissues including bone, cartilage, and tendon that require precise patterning to ensure structural and functional integrity. Interestingly, most of these tissues are derived from one multipotent cell population called cranial neural crest cells (CNCCs). How CNCCs are properly instructed to differentiate into various tissue types remains nebulous. To better understand the mechanisms necessary for the patterning of mandibular musculoskeletal tissues we utilized the avian mutant talpid2 (ta2) which presents with several malformations of the facial skeleton including dysplastic tendons, mispatterned musculature, and bilateral ectopic cartilaginous processes extending off Meckel's cartilage. We found an ectopic epithelial BMP signaling domain in the ta2 mandibular prominence (MNP) that correlated with the subsequent expansion of SOX9+ cartilage precursors. These findings were validated with conditional murine models suggesting an evolutionarily conserved mechanism for CNCC-derived musculoskeletal patterning. Collectively, these data support a model in which cilia are required to define epithelial signal centers essential for proper musculoskeletal patterning of CNCC-derived mesenchyme.


Asunto(s)
Mandíbula , Cresta Neural , Animales , Embrión de Pollo , Ratones , Proteínas Aviares/genética , Proteínas Aviares/metabolismo , Tipificación del Cuerpo/genética , Cartílago/metabolismo , Cartílago/crecimiento & desarrollo , Cartílago/citología , Diferenciación Celular , Pollos/genética , Cilios/metabolismo , Cilios/genética , Regulación del Desarrollo de la Expresión Génica , Mandíbula/crecimiento & desarrollo , Mandíbula/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Mesodermo/crecimiento & desarrollo , Cresta Neural/citología , Cresta Neural/metabolismo , Transducción de Señal , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA