Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Sci Rep ; 14(1): 2696, 2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-38302538

RESUMEN

Osteoarthritis is the most common degenerative joint condition, leading to articular cartilage (AC) degradation, chronic pain and immobility. The lack of appropriate therapies that provide tissue restoration combined with the limited lifespan of joint-replacement implants indicate the need for alternative AC regeneration strategies. Differentiation of human pluripotent stem cells (hPSCs) into AC progenitors may provide a long-term regenerative solution but is still limited due to the continued reliance upon growth factors to recapitulate developmental signalling processes. Recently, TTNPB, a small molecule activator of retinoic acid receptors (RARs), has been shown to be sufficient to guide mesodermal specification and early chondrogenesis of hPSCs. Here, we modified our previous differentiation protocol, by supplementing cells with TTNPB and administering BMP2 at specific times to enhance early development (referred to as the RAPID-E protocol). Transcriptomic analyses indicated that activation of RAR signalling significantly upregulated genes related to limb and embryonic skeletal development in the early stages of the protocol and upregulated genes related to AC development in later stages. Chondroprogenitors obtained from RAPID-E could generate cartilaginous pellets that expressed AC-related matrix proteins such as Lubricin, Aggrecan, and Collagen II, but additionally expressed Collagen X, indicative of hypertrophy. This protocol could lay the foundations for cell therapy strategies for osteoarthritis and improve the understanding of AC development in humans.


Asunto(s)
Benzoatos , Cartílago Articular , Osteoartritis , Células Madre Pluripotentes , Retinoides , Humanos , Condrocitos/metabolismo , Tretinoina/farmacología , Condrogénesis/genética , Diferenciación Celular , Cartílago Articular/metabolismo , Colágeno/metabolismo , Osteoartritis/metabolismo
2.
J Ethnopharmacol ; 317: 116784, 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-37321426

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: The treatment of osteoarthritis (OA) patients is a challenging problem. Mesenchymal stem cells (MSCs) are multipotent cells and play key roles in regenerative medicine for cartilage degeneration. GuiLu-ErXian Glue (GLEXG) is an herbal remedy widely used in traditional Chinese medicine to treat joint pain and disability in elderly OA patients. However, the mechanisms of how GLEXG affects MSCs-induced chondrogensis remains to be elucidated. AIM OF THE STUDY: The aim of this study was to investigate the effects of GLEXG on MSC-derived chondrogenesis, both in vitro and in vivo and its potential mechanisms. METHODS: Using human MSC (hMSCs) as in vitro model, the effects of HPLC-profiled GLEXG water extract on chondrogenic differentiation were investigated by 3D spheroid cultures under chondrogenesis-inducing medium (CIM) condition. The chondrogenesis process was evaluated by measuring the sphere sizes, chondrogenesis-related genes expression by reverse transcription real-time PCR that targeted type II/X collagens, SOX9, aggrecan, and protein expression by immunostaining. Anti-TGF-ß1 neutralization antibody was used for mechanistic study. Mono-iodoacetate (MIA) induced OA joint was used to evaluate the effects of GLEXG on in vivo model. MSCs-derived exosomes were purified for proteomics study and senescence process was evaluated by cumulative population doublings and senescence-associated ß-Galactosidase staining. RESULTS: The results showed that GLEXG enhanced hMSCs chondrogenesis and upregulated RNA expression of type II/X collagen, SOX9 and aggrecan at 0.1 µg/mL, 0.3 µg/mL in vitro. In vivo, GLEXG at the dose of 0.3 µg intraarticular (i.a.) injection rescued the MIA-induced cartilage defect. Proteomics and ingenuity pathway analysis obtained from MSCs-released exosomes suggested that senescence pathway was less activated in GLEXG group than in vehicle group. Besides, GLEXG was able to increase cumulative population doubling and delayed hMSCs senescence process after four passages in cultures. CONCLUSION: we conclude that GLEXG promotes in vitro MSC-induced chondrogenesis possibly via exosomes release and delays aging in the MSC senescence process and that treatment with GLEXG (0.3 µg, i.a.) rescued cartilage defects in rat OA knee model.


Asunto(s)
Exosomas , Células Madre Mesenquimatosas , Osteoartritis , Humanos , Ratas , Animales , Anciano , Agrecanos/genética , Agrecanos/metabolismo , Agrecanos/farmacología , Condrogénesis/genética , Exosomas/metabolismo , Diferenciación Celular , Colágeno Tipo II/metabolismo , Colágeno Tipo X/metabolismo , Envejecimiento , Células Cultivadas
3.
Cell Death Dis ; 13(1): 11, 2021 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-34930890

RESUMEN

TMEM165 deficiency leads to skeletal disorder characterized by major skeletal dysplasia and pronounced dwarfism. However, the molecular mechanisms involved have not been fully understood. Here, we uncover that TMEM165 deficiency impairs the synthesis of proteoglycans by producing a blockage in the elongation of chondroitin-and heparan-sulfate glycosaminoglycan chains leading to the synthesis of proteoglycans with shorter glycosaminoglycan chains. We demonstrated that the blockage in elongation of glycosaminoglycan chains is not due to defect in the Golgi elongating enzymes but rather to availability of the co-factor Mn2+. Supplementation of cell with Mn2+ rescue the elongation process, confirming a role of TMEM165 in Mn2+ Golgi homeostasis. Additionally, we showed that TMEM165 deficiency functionally impairs TGFß and BMP signaling pathways in chondrocytes and in fibroblast cells of TMEM165 deficient patients. Finally, we found that loss of TMEM165 impairs chondrogenic differentiation by accelerating the timing of Ihh expression and promoting early chondrocyte maturation and hypertrophy. Collectively, our results indicate that TMEM165 plays an important role in proteoglycan synthesis and underline the critical role of glycosaminoglycan chains structure in the regulation of chondrogenesis. Our data also suggest that Mn2+ supplementation may be a promising therapeutic strategy in the treatment of TMEM165 deficient patients.


Asunto(s)
Antiportadores/deficiencia , Antiportadores/metabolismo , Proteínas de Transporte de Catión/deficiencia , Proteínas de Transporte de Catión/metabolismo , Diferenciación Celular/genética , Condrocitos/metabolismo , Condrocitos/patología , Sulfatos de Condroitina/biosíntesis , Enanismo/metabolismo , Proteoglicanos de Heparán Sulfato/biosíntesis , Transducción de Señal/genética , Animales , Antiportadores/genética , Estudios de Casos y Controles , Proteínas de Transporte de Catión/genética , Línea Celular Tumoral , Condrogénesis/genética , Enanismo/patología , Fibroblastos/metabolismo , Técnicas de Inactivación de Genes/métodos , Glicosilación , Células HEK293 , Humanos , Hipertrofia/metabolismo , Ratones , Transfección
4.
Sci Rep ; 11(1): 6751, 2021 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-33762629

RESUMEN

Bone marrow mesenchymal stem/stromal cells (BMSCs) show great promise for bone repair, however they are isolated by an invasive bone marrow harvest and their regenerative potential decreases with age. Conversely, cord blood can be collected non-invasively after birth and contains MSCs (CBMSCs) that can be stored for future use. However, whether CBMSCs can replace BMSCs targeting bone repair is unknown. This study evaluates the in vitro osteogenic potential of unprimed, osteogenically primed, or chondrogenically primed CBMSCs and BMSCs and their in vivo bone forming capacity following ectopic implantation on biphasic calcium phosphate ceramics in nude mice. In vitro, alkaline phosphatase (intracellular, extracellular, and gene expression), and secretion of osteogenic cytokines (osteoprotegerin and osteocalcin) was significantly higher in BMSCs compared with CBMSCs, while CBMSCs demonstrated superior chondrogenic differentiation and secretion of interleukins IL-6 and IL-8. BMSCs yielded significantly more cell engraftment and ectopic bone formation compared to CBMSCs. However, priming of CBMSCs with either chondrogenic or BMP-4 supplements led to bone formation by CBMSCs. This study is the first direct quantification of the bone forming abilities of BMSCs and CBMSCs in vivo and, while revealing the innate superiority of BMSCs for bone repair, it provides avenues to induce osteogenesis by CBMSCs.


Asunto(s)
Proteína Morfogenética Ósea 4/genética , Diferenciación Celular/genética , Condrogénesis/genética , Sangre Fetal/citología , Hidroxiapatitas , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/genética , Adulto , Biomarcadores , Proteína Morfogenética Ósea 4/metabolismo , Sustitutos de Huesos , Células Cultivadas , Citocinas/metabolismo , Humanos , Inmunohistoquímica , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Ingeniería de Tejidos , Adulto Joven
5.
J Orthop Surg Res ; 16(1): 208, 2021 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-33752715

RESUMEN

BACKGROUND: Deer antler is a zoological exception due to its fantastic characteristics, including amazing growth rate and repeatable regeneration. Deer antler has been used as a key ingredient in traditional Chinese medicine relating to kidney and bone health for centuries. The aim of this study was to dissect the molecular regulation of deer antler extract (DAE) on xiphoid cartilage (XC). METHODS: The DAE used in this experiment was same as the one that was prepared as previously described. The specific pathogen-free (SPF) grade Sprague-Dawley (SD) rats were randomly divided into blank group (n =10) and DAE group (n =10) after 1-week adaptive feeding. The DAE used in this experiment was same as the one that was prepared as previously described. The rats in DAE group were fed with DAE for 3 weeks at a dose of 0.2 g/kg per day according to the body surface area normalization method, and the rats in blank group were fed with drinking water. Total RNA was extracted from XC located in the most distal edge of the sternum. Illumina RNA sequencing (RNA-seq) in combination with quantitative real-time polymerase chain reaction (qRT-PCR) validation assay was carried out to dissect the molecular regulation of DAE on XC. RESULTS: We demonstrated that DAE significantly increased the expression levels of DEGs involved in cartilage growth and regeneration, but decreased the expression levels of DEGs involved in inflammation, and mildly increased the expression levels of DEGs involved in chondrogenesis and chondrocyte proliferation. CONCLUSIONS: Our findings suggest that DAE might serve as a complementary therapeutic regent for cartilage growth and regeneration to treat cartilage degenerative disease, such as osteoarthritis.


Asunto(s)
Cuernos de Venado/química , Regeneración Ósea/genética , Cartílago/crecimiento & desarrollo , Cartílago/fisiología , Condrogénesis/genética , Ciervos/anatomía & histología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Inflamación/prevención & control , Medicina Tradicional China , Extractos de Tejidos/farmacología , Apófisis Xifoides , Animales , Diferenciación Celular/genética , Proliferación Celular/genética , Condrocitos/fisiología , Masculino , Ratas Sprague-Dawley
6.
Phytomedicine ; 81: 153429, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33310311

RESUMEN

BACKGROUND: Phlomis umbrosa Turczaninow root has been traditionally used to treat fractures, rheumatoid arthritis, and arthralgia. However, the effects and mechanisms of P. umbrosa on osteoarthritis (OA) remain poorly understood and a functional genomic approach has not been investigated. AIM: The purpose of this study was to investigate the effects and mechanisms of P. umbrosa extract (PUE) on OA using transcriptomic analysis. METHODS: We performed joint diameter measurements, micro computed tomography, and histopathological analysis of monosodium iodoacetate (MIA)-induced OA rats treated with PUE (200 mg/kg) for 3 weeks. Gene expression profiling in articular cartilage tissue was then performed using RNA sequencing (RNA-seq) followed by signaling pathway analysis of regulatory genes. RESULTS: PUE treatment improved OA based on decreased joint diameter, increased joint morphological parameters, and histopathological features. Many genes involved in multiple signal transduction pathway and collagen activation in OA were differentially regulated by PUE. These included genes related to Wnt/ß-catenin, OA pathway, and sonic hedgehog signaling activity. Furthermore, PUE treatment downregulated cartilage damage factors (MMP-9, MMP-13, ADAMTs4, and ADMATs5) and upregulated chondrogenesis (COL2A1 and SOX-9) by regulating the transcription factors SOX-9, Ctnnb1, and Epas1. CONCLUSION: Based on the results of gene expression profiling, this study highlighted the molecular mechanisms underlying the effects of PUE in MIA-induced OA rats. The findings provide novel insight into the mechanisms by which PUE treatment-induced gene expression changes may influence OA disease progression. Taken together, the results suggest that PUE may be used as a source of therapeutic agents for OA.


Asunto(s)
Osteoartritis/tratamiento farmacológico , Osteoartritis/genética , Phlomis/química , Extractos Vegetales/farmacología , Animales , Cartílago Articular/efectos de los fármacos , Cartílago Articular/patología , Condrogénesis/efectos de los fármacos , Condrogénesis/genética , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Yodoacetatos/toxicidad , Articulaciones/efectos de los fármacos , Articulaciones/patología , Masculino , Osteoartritis/inducido químicamente , Osteoartritis/patología , Extractos Vegetales/química , Ratas Sprague-Dawley , Microtomografía por Rayos X
7.
Tissue Cell ; 56: 107-113, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30736898

RESUMEN

Limited self-restorative ability of the cartilage has necessitated the use of cell and tissue engineering based therapies. Recent advances in the isolation, expansion and characterization of articular cartilage derived chondroprogenitors(CPs) has gained popularity in its role for cartilage repair. Platelet rich plasma (PRP) is a reliable biological scaffold for in-vitro and in-vivo studies with reported therapeutic applications in cartilage and bone pathologies. The aim of this study was to evaluate whether human allogeneic PRP could serve as a biological scaffold for chondroprogenitors (CPs) in cartilage repair. CPs were isolated from the superficial layer of three osteoarthritic knee joints by fibronectin adhesion assay and characterized using flow cytometric analysis. Allogeneic citrated blood was harvested from three subjects to obtain PRP. CPs at a concentration of one million cells per ml were gelled with PRP using calcium chloride. The PRP-CP scaffolds were subjected for adipogeneic, osteogenic, chondrogeneic differentiation and processed for post differentiation-staining studies (Oil Red O, Von Kossa, Alcian blue staining), immunofluorescence (collagen II) and live dead assays (Calcein AM-Ethidium Homodimer). We show that PRP was able to sustain CP cell viability and differentiate towards adipogenic, osteogenic and chondrogenic lineage under appropriate culture conditions. We also noted positive extracellular matrix production in PRP-CP scaffolds cultured without chondrogenic supplementation. Our results suggest that PRP could be a promising bio-active scaffold due to its synergistic effect in supporting cell proliferation, maintaining cell viability and favoring extracellular matrix production. PRP can be used as biological scaffold for the delivery of CPs in cartilage healing.


Asunto(s)
Cartílago Articular/crecimiento & desarrollo , Condrogénesis/genética , Plasma Rico en Plaquetas/citología , Andamios del Tejido , Diferenciación Celular/genética , Condrocitos/citología , Matriz Extracelular/genética , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Plasma Rico en Plaquetas/metabolismo
8.
Lasers Med Sci ; 34(1): 115-126, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30264177

RESUMEN

Tissue engineering aims to take advantage of the ability of undifferentiated stem cells to differentiate into multiple cell types to repair damaged tissue. Photobiomodulation uses either lasers or light-emitting diodes to promote stem cell proliferation and differentiation. The present study aimed to investigate single and dual combinations of laser wavelengths on mesenchymal stem cells (MSCs). MSCs were derived from rabbit iliac bone marrow. One control and eight laser irradiated groups were designated as Infrared (IR, 810 nm), Red (R, 660 nm), Green (G, 532 nm), Blue (B, 485 nm), IR-R, IR-B, R-G, and B-G. Irradiation was repeated daily for 21 days and cell proliferation, osseous, or cartilaginous differentiation was then measured. RT-PCR biomarkers were SOX9, aggrecan, COL 2, and COL 10 expression for cartilage and ALP, COL 1, and osteocalcin expression for bone. Cellular proliferation was increased in all irradiated groups except G. All cartilage markers were significantly increased by IR and IR-B except COL 10 which was suppressed by IR-B combination. ALP expression was highest in R and IR groups during osseous differentiation. ALP was decreased by combinations of IR with B and with R, and also by G alone. R and B-G groups showed stimulated COL 1 expression; however, COL 1 was suppressed in IR-B, IR-R, and G groups. IR significantly increased osteocalcin expression, but in B, B-G, and G groups it was reduced. Cartilage differentiation was stimulated by IR and IR-B laser irradiation. The effects of single or combined laser irradiation were not clear-cut on osseous differentiation. Stimulatory effects on osteogenesis were seen for R and IR lasers, while G laser had inhibitory effects.


Asunto(s)
Huesos/citología , Cartílago/citología , Diferenciación Celular/efectos de la radiación , Rayos Láser , Terapia por Luz de Baja Intensidad , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de la radiación , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Linaje de la Célula/efectos de la radiación , Proliferación Celular/efectos de la radiación , Forma de la Célula/efectos de la radiación , Células Cultivadas , Condrogénesis/genética , Condrogénesis/efectos de la radiación , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Regulación de la Expresión Génica/efectos de la radiación , Osteogénesis/genética , Osteogénesis/efectos de la radiación , Conejos
9.
J Tissue Eng Regen Med ; 12(6): 1402-1411, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29726103

RESUMEN

Bioreactor systems will likely play a key role in establishing regulatory compliant and cost-effective production systems for manufacturing engineered tissue grafts for clinical applications. However, the automation of bioreactor systems could become considerably more complex and costly due to the requirements for additional storage and liquid handling technologies if unstable supplements are added to the culture medium. Ascorbic acid (AA) is a bioactive supplement that is commonly presumed to be essential for the generation of engineered cartilage tissues. However, AA can be rapidly oxidized and degraded. In this work, we addressed whether human nasal chondrocytes can redifferentiate, undergo chondrogenesis, and generate a cartilaginous extracellular matrix when cultured in the absence of AA. We found that when chondrocytes were cultured in 3D micromass pellets either with or without AA, there were no significant differences in their chondrogenic capacity in terms of gene expression or the amount of glycosaminoglycans. Moreover, 3D pellets cultured without AA contained abundant collagen Types II and I extracellular matrix. Although the amounts of Collagens II and I were significantly lower (34% and 50% lower) than in pellets cultured with AA, collagen fibers had similar thicknesses and distributions for both groups, as shown by scanning electron microscopy imaging. Despite the reduced amounts of collagen, if engineered cartilage grafts can be generated with sufficient properties that meet defined quality criteria without the use of unstable supplements such as AA, bioreactor automation requirements can be greatly simplified, thereby facilitating the development of more compact, user-friendly, and cost-effective bioreactor-based manufacturing systems.


Asunto(s)
Ácido Ascórbico/farmacología , Diferenciación Celular/efectos de los fármacos , Condrocitos/citología , Condrogénesis , Adulto , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Condrogénesis/efectos de los fármacos , Condrogénesis/genética , Colágeno/metabolismo , Medios de Cultivo , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glicosaminoglicanos/metabolismo , Humanos , Persona de Mediana Edad , Adulto Joven
10.
Mol Med Rep ; 17(6): 8237-8243, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29693145

RESUMEN

Icariin, a traditional Chinese medicine, has previously been demonstrated to promote chondrogenesis of bone marrow mesenchymal stem cells (BMSCs) in traditional 2D cell culture. The present study investigated whether icariin has the potential to promote stable chondrogenic differentiation of BMSCs without hypertrophy in a 3D microenvironment. BMSCs were cultivated in a self­assembling peptide nanofiber hydrogel scaffold in chondrogenic medium for 3 weeks. Icariin was added to the medium throughout the culture period at concentrations of 1x10­6 M. Chondrogenic differentiation markers, including collagen II and SRY­type high mobility group box 9 (SOX9) were detected by immunofluorescence, reverse transcription­quantitative polymerase chain reaction and toluidine blue staining. Hypertrophic differentiation was further assessed by detecting collagen X and collagen I gene expression levels and alkaline phosphatase activity. The results demonstrated that icariin significantly enhanced cartilage extracellular matrix synthesis and gene expression levels of collagen II and SOX9, and additionally promoted more chondrocyte­like rounded morphology in BMSCs. Furthermore, chondrogenic medium led to hypertrophic differentiation via upregulation of collagen X and collagen I gene expression levels and alkaline phosphatase activity, which was not potentiated by icariin. In conclusion, these results suggested that icariin treatment may promote chondrogenic differentiation of BMSCs, and inhibit the side effect of growth factor activity, thus preventing further hypertrophic differentiation. Therefore, icariin may be a potential compound for cartilage tissue engineering.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Flavonoides/farmacología , Hidrogeles , Células Madre Mesenquimatosas/citología , Andamios del Tejido , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular/genética , Células Cultivadas , Condrogénesis/genética , Matriz Extracelular/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Nanofibras , Ratas
11.
J Tissue Eng Regen Med ; 12(6): 1327-1338, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29522657

RESUMEN

In the current study, the effect of superimposing platelet-rich plasma (PRP) on different culture mediums in a three-dimensional alginate scaffold encapsulated with adipose-derived mesenchymal stem cells for cartilage tissue repair is reported. The three-dimensional alginate scaffolds with co-administration of PRP and/or chondrogenic supplements had a significant effect on the differentiation of adipose mesenchymal stem cells into mature cartilage, as assessed by an evaluation of the expression of cartilage-related markers of Sox9, collagen II, aggrecan and collagen, and glycosaminoglycan assays. For in vivo studies, following induction of osteochondral lesion in a rabbit model, a high degree of tissue regeneration in the alginate plus cell group (treated with PRP plus chondrogenic medium) compared with other groups of cell-free alginate and untreated groups (control) were observed. After 8 weeks, in the alginate plus cell group, functional chondrocytes were observed, which produced immature matrix, and by 16 weeks, the matrix and hyaline-like cartilage became completely homogeneous and integrated with the natural surrounding cartilage in the defect site. Similar effect was also observed in the subchondral bone. The cell-free scaffolds formed fibrocartilage tissue, and the untreated group did not form a continuous cartilage over the defect by 16 weeks.


Asunto(s)
Tejido Adiposo/citología , Alginatos/farmacología , Cartílago/fisiología , Células Inmovilizadas/citología , Plasma Rico en Plaquetas/metabolismo , Regeneración , Células Madre/citología , Andamios del Tejido/química , Adulto , Animales , Cartílago/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Inmovilizadas/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Condrogénesis/genética , Colágeno Tipo II/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Glicosaminoglicanos/metabolismo , Humanos , Masculino , Conejos , Regeneración/efectos de los fármacos , Células Madre/efectos de los fármacos
12.
J Cell Physiol ; 233(9): 6878-6887, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29319178

RESUMEN

Aggrecan is a high molecular weight proteoglycan that plays a critical role in cartilage structure and the function of joints, providing intervertebral disc and cartilage with the ability to resist compressive loads. Aggrecan degradation in articular cartilage is a significant event in early-stage osteoarthritis (OA). Currently, no effective treatment exists for OA other than pain relief. Dextrose (D-glucose) prolotherapy has shown promising activity in the treatment of different musculoskeletal disorders, including OA. However, little is known about the molecular mechanism of the glucose effect in OA and on the regulation of chondrogenesis. We show for the first time that glucose upregulates aggrecan expression and subsequent chondrogenesis in ATDC5 cells. Moreover, we found that glucose-induced aggrecan expression is mediated through the protein kinase Cα (PKCα)- and p38-dependent pathway. As demonstrated by microRNA (miR) and luciferase analyses, the glucose-induced PKCα/p38 signaling axis is responsible for downregulating miR141-3p which targets to the 3'untranslated region of aggrecan. In summary, we show that glucose enhances chondrogenesis by upregulating aggrecan expression via the PKCα-p38-miR141-3p signaling pathway. This result provides new insights into the mechanism of glucose on chondrogenesis.


Asunto(s)
Agrecanos/genética , Condrocitos/fisiología , Glucosa/genética , Sistema de Señalización de MAP Quinasas/genética , MicroARNs/genética , Proteína Quinasa C-alfa/genética , Transducción de Señal/genética , Regiones no Traducidas 3'/genética , Animales , Cartílago Articular/fisiología , Células Cultivadas , Condrogénesis/genética , Regulación hacia Abajo/genética , Ratones , Osteoartritis/genética , Regulación hacia Arriba/genética
13.
Stem Cell Res Ther ; 8(1): 278, 2017 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-29237492

RESUMEN

BACKGROUND: The pharmaceutical agent pentosan polysulfate (PPS) is known to induce proliferation and chondrogenesis of mesenchymal progenitor cells (MPCs) in vitro and in vivo. However, the mechanism(s) of action of PPS in mediating these effects remains unresolved. In the present report we address this issue by investigating the binding and uptake of PPS by MPCs and monitoring gene expression and proteoglycan biosynthesis before and after the cells had been exposed to limited concentrations of PPS and then re-established in culture in the absence of the drug (MPC priming). METHODS: Immuno-selected STRO-1+ mesenchymal progenitor stem cells (MPCs) were prepared from human bone marrow aspirates and established in culture. The kinetics of uptake, shedding, and internalization of PPS by MPCs was determined by monitoring the concentration-dependent loss of PPS media concentrations using an enzyme-linked immunosorbent assay (ELISA) and the uptake of fluorescein isothiocyanate (FITC)-labelled PPS by MPCs. The proliferation of MPCs, following pre-incubation and removal of PPS (priming), was assessed using the Wst-8 assay method, and proteoglycan synthesis was determined by the incorporation of 35SO4 into their sulphated glycosaminoglycans. The changes in expression of MPC-related cell surface antigens of non-primed and PPS-primed MPCs from three donors was determined using flow cytometry. RNA sequencing of RNA isolated from non-primed and PPS-primed MPCs from the same donors was undertaken to identify the genes altered by the PPS priming protocol. RESULTS: The kinetic studies indicated that, in culture, PPS rapidly binds to MPC surface receptors, followed by internalisation and localization within the nucleus of the cells. Following PPS-priming of MPCs and a further 48 h of culture, both cell proliferation and proteoglycan synthesis were enhanced. Reduced expression of MPC-related cell surface antigen expression was promoted by the PPS priming, and RNA sequencing analysis revealed changes in the expression of 42 genes. CONCLUSION: This study has shown that priming of MPCs with low concentrations of PPS enhanced chondrogenesis and MPC proliferation by modifying their characteristic basal gene and protein expression. These findings offer a novel approach to re-programming mesenchymal stem cells for clinical indications which require the repair or regeneration of cartilaginous tissues such as in osteoarthritis and degenerative disc disease.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Condrocitos/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Poliéster Pentosan Sulfúrico/farmacología , Antígenos de Superficie/metabolismo , Transporte Biológico , Biomarcadores/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Condrocitos/citología , Condrocitos/metabolismo , Condrogénesis/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Anotación de Secuencia Molecular , Proteoglicanos/biosíntesis
14.
Curr Stem Cell Res Ther ; 12(3): 260-270, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27306401

RESUMEN

Mechanical stimulation is a key factor in articular cartilage generation and maintenance. Bioreactor systems have been designed and built in order to deliver specific types of mechanical stimulation. The focus has been twofold, applying a type of preconditioning in order to stimulate cell differentiation, and to simulate in vivo conditions in order to gain further insight into how cells respond to different stimulatory patterns. Due to the complex forces at work within joints, it is difficult to simulate mechanical conditions using a bioreactor. The aim of this review is to gain a deeper understanding of the complexities of mechanical stimulation protocols by comparing those employed in bioreactors in the context of tissue engineering for articular cartilage, and to consider their effects on cultured cells. Allied and Complementary Medicine 1985 to 2016, Ovid MEDLINE[R] 1946 to 2016, and Embase 1974 to 2016 were searched using key terms. Results were subject to inclusion and exclusion criteria, key findings summarised into a table and subsequently discussed. Based on this review it is overwhelmingly clear that mechanical stimulation leads to increased chondrogenic properties in the context of bioreactor articular cartilage tissue engineering using human cells. However, given the variability and lack of controlled factors between research articles, results are difficult to compare, and a standardised method of evaluating stimulation protocols proved challenging. With improved standardisation in mechanical stimulation protocol reporting, bioreactor design and building processes, along with a better understanding of joint behaviours, we hope to perform a meta-analysis on stimulation protocols and methods.


Asunto(s)
Condrocitos/metabolismo , Condrogénesis/genética , Mecanotransducción Celular , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Fenómenos Biomecánicos , Reactores Biológicos , Cartílago Articular/lesiones , Cartílago Articular/cirugía , Diferenciación Celular , Proliferación Celular , Condrocitos/citología , Humanos , Estimulación Física
15.
PLoS One ; 11(7): e0159157, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27415616

RESUMEN

Cartilage acidic protein 1 (CRTAC1) was recently identified as an elevated protein in the synovial fluid of patients with osteoarthritis (OA) by a proteomic analysis. This gene is also upregulated in both human and mouse OA by transcriptomic analysis. The objective of this study was to characterize the expression and function of CRTAC1 in OA. Here, we first confirm the increase of CRTAC1 in cartilage biopsies from OA patients undergoing joint replacement by real-time PCR and immunohistochemistry. Furthermore, we report that proinflammatory cytokines interleukin-1beta and tumor necrosis factor alpha upregulate CRTAC1 expression in primary human articular chondrocytes and synovial fibroblasts. Genetic deletion of Crtac1 in mice significantly inhibited cartilage degradation, osteophyte formation and gait abnormalities of post-traumatic OA in female, but not male, animals undergoing the destabilization of medial meniscus (DMM) surgery. Taken together, CRTAC1 is upregulated in the osteoarthritic joint and directly induced in chondrocytes and synovial fibroblasts by pro-inflammatory cytokines. This molecule is necessary for the progression of OA in female mice after DMM surgery and thus represents a potential therapy for this prevalent disease, especially for women who demonstrate higher rates and more severe OA.


Asunto(s)
Artritis Experimental/genética , Artritis Experimental/prevención & control , Proteínas de Unión al Calcio/genética , Osteoartritis de la Rodilla/genética , Osteoartritis de la Rodilla/prevención & control , Animales , Artritis Experimental/fisiopatología , Desarrollo Óseo/genética , Proteínas de Unión al Calcio/deficiencia , Proteínas de Unión al Calcio/fisiología , Células Cultivadas , Condrocitos/fisiología , Condrogénesis/genética , Citocinas/fisiología , Femenino , Eliminación de Gen , Humanos , Masculino , Ratones , Ratones Noqueados , Osteoartritis de la Rodilla/fisiopatología , Caracteres Sexuales , Líquido Sinovial/fisiología , Regulación hacia Arriba
16.
Stem Cells ; 34(4): 1083-96, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26851078

RESUMEN

Calcification of soft tissues, such as heart valves and tendons, is a common clinical problem with limited therapeutics. Tissue specific stem/progenitor cells proliferate to repopulate injured tissues. But some of them become divergent to the direction of ossification in the local pathological microenvironment, thereby representing a cellular target for pharmacological approach. We observed that HIF-2alpha (encoded by EPAS1 inclined form) signaling is markedly activated within stem/progenitor cells recruited at calcified sites of diseased human tendons and heart valves. Proinflammatory microenvironment, rather than hypoxia, is correlated with HIF-2alpha activation and promoted osteochondrogenic differentiation of tendon stem/progenitor cells (TSPCs). Abnormal upregulation of HIF-2alpha served as a key switch to direct TSPCs differentiation into osteochondral-lineage rather than teno-lineage. Notably, Scleraxis (Scx), an essential tendon specific transcription factor, was suppressed on constitutive activation of HIF-2alpha and mediated the effect of HIF-2alpha on TSPCs fate decision. Moreover, pharmacological inhibition of HIF-2alpha with digoxin, which is a widely utilized drug, can efficiently inhibit calcification and enhance tenogenesis in vitro and in the Achilles's tendinopathy model. Taken together, these findings reveal the significant role of the tissue stem/progenitor cells fate decision and suggest that pharmacological regulation of HIF-2alpha function is a promising approach for soft tissue calcification treatment.


Asunto(s)
Tendón Calcáneo/efectos de los fármacos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Calcinosis/tratamiento farmacológico , Tratamiento de Tejidos Blandos , Tendón Calcáneo/crecimiento & desarrollo , Tendón Calcáneo/patología , Anciano , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Calcinosis/genética , Calcinosis/patología , Diferenciación Celular/genética , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Microambiente Celular/efectos de los fármacos , Condrogénesis/genética , Digoxina/administración & dosificación , Humanos , Masculino , Persona de Mediana Edad , Ratas , Cardiopatía Reumática/genética , Cardiopatía Reumática/patología , Células Madre/efectos de los fármacos , Células Madre/patología
17.
Stem Cells Transl Med ; 5(2): 206-17, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26702127

RESUMEN

Bone tissue engineering via endochondral ossification has been explored by chondrogenically priming cells using soluble mediators for at least 3 weeks to produce a hypertrophic cartilage template. Although recapitulation of endochondral ossification has been achieved, long-term in vitro culture is required for priming cells through repeated supplementation of inductive factors in the media. To address this challenge, a microparticle-based growth factor delivery system was engineered to drive endochondral ossification within human bone marrow-derived mesenchymal stem cell (hMSC) aggregates. Sequential exogenous presentation of soluble transforming growth factor-ß1 (TGF-ß1) and bone morphogenetic protein-2 (BMP-2) at various defined time courses resulted in varying degrees of chondrogenesis and osteogenesis as demonstrated by glycosaminoglycan and calcium content. The time course that best induced endochondral ossification was used to guide the development of the microparticle-based controlled delivery system for TGF-ß1 and BMP-2. Gelatin microparticles capable of relatively rapid release of TGF-ß1 and mineral-coated hydroxyapatite microparticles permitting more sustained release of BMP-2 were then incorporated within hMSC aggregates and cultured for 5 weeks following the predetermined time course for sequential presentation of bioactive signals. Compared with cell-only aggregates treated with exogenous growth factors, aggregates with incorporated TGF-ß1- and BMP-2-loaded microparticles exhibited enhanced chondrogenesis and alkaline phosphatase activity at week 2 and a greater degree of mineralization by week 5. Staining for types I and II collagen, osteopontin, and osteocalcin revealed the presence of cartilage and bone. This microparticle-incorporated system has potential as a readily implantable therapy for healing bone defects without the need for long-term in vitro chondrogenic priming. Significance: This study demonstrates the regulation of chondrogenesis and osteogenesis with regard to endochondral bone formation in high-density stem cell systems through the controlled presentation of inductive factors from incorporated microparticles. This work lays the foundation for a rapidly implantable tissue engineering system that promotes bone repair via endochondral ossification, a pathway that can delay the need for a functional vascular network and has an intrinsic ability to promote angiogenesis. The modular nature of this system lends well to using different cell types and/or growth factors to induce endochondral bone formation, as well as the production of other tissue types.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Proteína Morfogenética Ósea 2/farmacología , Condrogénesis/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Factor de Crecimiento Transformador beta1/farmacología , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Biomarcadores/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Proteína Morfogenética Ósea 2/metabolismo , Calcificación Fisiológica/efectos de los fármacos , Calcificación Fisiológica/genética , Calcio/metabolismo , Agregación Celular , Condrogénesis/genética , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Preparaciones de Acción Retardada , Composición de Medicamentos , Durapatita/química , Gelatina/química , Expresión Génica , Glicosaminoglicanos/metabolismo , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Osteocalcina/genética , Osteocalcina/metabolismo , Osteogénesis/genética , Osteopontina/genética , Osteopontina/metabolismo , Cultivo Primario de Células , Ingeniería de Tejidos/métodos , Factor de Crecimiento Transformador beta1/metabolismo
18.
Stem Cell Reports ; 4(4): 712-26, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25818812

RESUMEN

Here we report the successful generation and long-term expansion of SOX9-expressing CD271(+)PDGFRα(+)CD73(+) chondrogenic ectomesenchymal cells from the PAX3/SOX10/FOXD3-expressing MIXL1(-)CD271(hi)PDGFRα(lo)CD73(-) neural crest-like progeny of human pluripotent stem cells in a chemically defined medium supplemented with Nodal/Activin/transforming growth factorß (TGFß) inhibitor and fibroblast growth factor (FGF). When "primed" with TGFß, such cells efficiently formed translucent cartilage particles, which were completely mineralized in 12 weeks in immunocompromized mice. The ectomesenchymal cells were expandable without loss of chondrogenic potential for at least 16 passages. They maintained normal karyotype for at least 10 passages and expressed genes representing embryonic progenitors (SOX4/12, LIN28A/B), cranial mesenchyme (ALX1/3/4), and chondroprogenitors (SOX9, COL2A1) of neural crest origin (SOX8/9, NGFR, NES). Ectomesenchyme is a source of many craniofacial bone and cartilage structures. The method we describe for obtaining a large quantity of human ectomesenchymal cells will help to model craniofacial disorders in vitro and potentially provide cells for the repair of craniofacial damage.


Asunto(s)
Diferenciación Celular , Condrogénesis , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Células Madre Pluripotentes/citología , Factor de Transcripción SOX9/metabolismo , Antígenos de Superficie/metabolismo , Biomarcadores , Cartílago , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Proliferación Celular , Autorrenovación de las Células/genética , Condrogénesis/efectos de los fármacos , Condrogénesis/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunofenotipificación , Cresta Neural/citología , Células Madre Pluripotentes/efectos de los fármacos , Factor de Transcripción SOX9/genética , Transducción de Señal , Factor de Crecimiento Transformador beta/farmacología
19.
Tissue Eng Part A ; 21(9-10): 1695-704, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25669848

RESUMEN

Dedifferentiated fat cells (DFAT cells) isolated from adipose tissue have been demonstrated to differentiate into chondrogenic cells in vitro. Nevertheless, an efficient method to facilitate its chondrogenic differentiation is still unexplored, hampering the extensive application of these cells in cartilage regeneration therapies. Here we provide the evidence that supplementation of strontium ions (Sr) in a chondrogenic medium (CM) significantly promotes early chondrogenic differentiation of DFAT cells. Human DFAT cells and the mesenchymal stem cell line (RCB2153) were subjected to the CM supplemented with/without Sr. After 14 days, alcian blue staining intensity significantly increased in DFAT cells, but not in RCB2153, subjected to CM with Sr. mRNA expression analysis revealed that the CM with 1.5 mM Sr increased the expression of chondrogenic marker, collagen type 2 alpha 1, whereas there was no significant change in osteogenic markers, collagen type 1 alpha 1, runt-related transcription factor 2, and osteocalcin, and hypertrophic chondrogenic marker, collagen type 10 alpha 1. Inhibitors for extracellular signal-regulated kinase 1/2 (ERK1/2), Akt, and calcium-sensing receptor (CaSR) pathways significantly diminished the alcian blue staining intensity, providing the first evidence that these signal pathways are associated with chondrogenic differentiation of DFAT cells. CaSR and ERK1/2 pathways independently induced Sr-mediated early chondrogenic differentiation. These results suggest that Sr supplementation into the CM may provide a powerful platform for preparing chondrogenically differentiated DFAT cells for cartilage regeneration.


Asunto(s)
Adipocitos/citología , Desdiferenciación Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Medios de Cultivo/farmacología , Estroncio/farmacología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Biomarcadores/metabolismo , Western Blotting , Desdiferenciación Celular/genética , Diferenciación Celular/genética , Línea Celular , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Condrocitos/patología , Condrogénesis/genética , Femenino , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Humanos , Hipertrofia , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
20.
Colloids Surf B Biointerfaces ; 112: 177-85, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23974003

RESUMEN

To deliver efficiently osteogenic, chondrogenic or adipogenic induction genes, such as Runx2, SOX9 and C/EBP-α, to adipose tissue-derived stem cells (ADSCs), a photo-mediated nanocomplex internalization gene delivery system was designed using chlorin e6 as a photosensitizer (PS) and polyethyleneimine (PEI) as a gene delivery carrier. In this system, gene delivery efficacy was significantly increased in ADSCs by photo irradiation. The gene transfection efficiency of Runx2, SOX9 and C/EBP-α was increased by 8.6-, 6.7- and 9.3-fold, respectively, by applying 0.7J/cm(2) of irradiation. Osteogenic, chondrogenic and adipogenic differentiation was confirmed by differentiation-related markers and histological analysis. ADSCs transfected with Runx2, SOX9 and C/EBP-α genes via photo irradiation indicated enhanced differentiation in comparison to the non-irradiated cells. These findings demonstrate that photo-mediated internalization is a promising system for efficient gene delivery and differentiation in ADSCs.


Asunto(s)
Tejido Adiposo/citología , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Técnicas de Transferencia de Gen , Fototerapia/métodos , Adipogénesis/genética , Células Madre Adultas/efectos de la radiación , Proteínas Potenciadoras de Unión a CCAAT/genética , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/genética , Diferenciación Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Condrogénesis/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Proteínas Fluorescentes Verdes/genética , Humanos , Nanoestructuras , Osteogénesis/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción SOX9/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA