Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Int J Mol Sci ; 22(7)2021 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-33916312

RESUMEN

Cartilage is a non-innervated and non-vascularized tissue. It is composed of one main cell type, the chondrocyte, which governs homeostasis within the cartilage tissue, but has low metabolic activity. Articular cartilage undergoes substantial stresses that lead to chondral defects, and inevitably osteoarthritis (OA) due to the low intrinsic repair capacity of cartilage. OA remains an incurable degenerative disease. In this context, several dietary supplements have shown promising results, notably in the relief of OA symptoms. In this study, we investigated the effects of collagen hydrolysates derived from fish skin (Promerim®30 and Promerim®60) and fish cartilage (Promerim®40) on the phenotype and metabolism of human articular chondrocytes (HACs). First, we demonstrated the safety of Promerim® hydrolysates on HACs cultured in monolayers. Then we showed that, Promerim® hydrolysates can increase the HAC viability and proliferation, while decreasing HAC SA-ß-galactosidase activity. To evaluate the effect of Promerim® on a more relevant model of culture, HAC were cultured as organoids in the presence of Promerim® hydrolysates with or without IL-1ß to mimic an OA environment. In such conditions, Promerim® hydrolysates led to a decrease in the transcript levels of some proteases that play a major role in the development of OA, such as Htra1 and metalloproteinase-1. Promerim® hydrolysates downregulated HtrA1 protein expression. In contrast, the treatment of cartilage organoids with Promerim® hydrolysates increased the neosynthesis of type I collagen (Promerim®30, 40 and 60) and type II collagen isoforms (Promerim®30 and 40), the latter being the major characteristic component of the cartilage extracellular matrix. Altogether, our results demonstrate that the use of Promerim® hydrolysates hold promise as complementary dietary supplements in combination with the current classical treatments or as a preventive therapy to delay the occurrence of OA in humans.


Asunto(s)
Condrocitos/efectos de los fármacos , Osteoartritis/tratamiento farmacológico , Cartílago Articular/citología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Condrocitos/metabolismo , Evaluación Preclínica de Medicamentos , Humanos , Cultivo Primario de Células
2.
Lab Invest ; 100(1): 64-71, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31409892

RESUMEN

Osteoarthritis (OA) is characterized by cartilage degradation, inflammation, and hypertrophy. Therapies are mainly symptomatic and aim to manage pain. Consequently, medical community is waiting for new treatments able to reduce OA process. This study aims to develop an in vitro simple OA model useful to predict drug ability to reduce cartilage hypertrophy. Human primary OA chondrocytes were incubated with transforming growth factor beta 1 (TGF-ß1). Hypertrophy was evaluated by Runx2, type X collagen, MMP13, and VEGF expression. Cartilage anabolism was investigated by Sox9, aggrecan, type II collagen, and glycosaminoglycan expression. In chondrocytes, TGF-ß1 increased expression of hypertrophic genes and activated canonical WNT pathway, while it decreased dramatically cartilage anabolism, suggesting that this treatment could mimic some OA features in vitro. Additionally, EZH2 inhibition, that has been previously reported to decrease cartilage hypertrophy and reduce OA development in vivo, attenuated COL10A1 and MMP13 upregulation and SOX9 downregulation induced by TGF-ß1 treatment. Similarly, pterosin B (an inhibitor of Sik3), and DMOG (a hypoxia-inducible factor prolyl hydroxylase which mimicks hypoxia), repressed the expression of hypertrophy markers in TGF-ß stimulated chondrocytes. In conclusion, we established an innovative OA model in vitro. This cheap and simple model will be useful to quickly screen new drugs with potential anti-arthritic effects, in complementary to current inflammatory models, and should permit to accelerate development of efficient treatments against OA able to reduce cartilage hypertrophy.


Asunto(s)
Cartílago Articular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Modelos Biológicos , Osteoartritis/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , Aminoácidos Dicarboxílicos , Benzamidas , Compuestos de Bifenilo , Evaluación Preclínica de Medicamentos , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Humanos , Hipertrofia/tratamiento farmacológico , Indanos , Persona de Mediana Edad , Morfolinas , Cultivo Primario de Células , Piridonas , Factor de Crecimiento Transformador beta1 , Vía de Señalización Wnt
3.
IUBMB Life ; 68(9): 756-63, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27521280

RESUMEN

Cartilage engineering is one challenging issue in regenerative medicine. Low oxygen tension or hypoxia inducible factor-1 (HIF-1α) gene therapy are promising strategies in the field of cartilage repair. Previously, we showed that hypoxia and its mediator HIF-1 regulate matrix genes expression (collagens and aggrecan). Here, we investigated the molecular mechanism involved in the regulation of type I collagen (COL1A1) by HIF-1 in human articular chondrocytes. We show that HIF-1α reduces COL1A1 transcription, through a distal promoter (-2300 to -1816 bp upstream transcription initiation site), containing two GC boxes that bind Sp transcription factors (Sp1/Sp3). Sp1 acts as a positive regulator but is not induced by HIF-1. COL1A1 inhibition caused by HIF-1 implies only Sp3, which accumulates and competes Sp1 binding on COL1A1 promoter. Additionally, Sp3 ectopic expression inhibits COL1A1, while Sp3 knockdown counteracts the downregulation of COL1A1 induced by HIF-1. In conclusion, we established a new regulatory model of COL1A1 regulation by HIF-1, and bring out its relationship with Sp3 transcription factor. In a fundamental level, these findings give insights in the mechanisms controlling COL1A1 gene expression. This may be helpful to improve strategies to impair type I collagen expression during chondrocyte differentiation for cartilage engineering. © 2016 IUBMB Life, 68(9):756-763, 2016.


Asunto(s)
Enfermedades de los Cartílagos/genética , Colágeno Tipo I/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Factor de Transcripción Sp3/genética , Enfermedades de los Cartílagos/patología , Enfermedades de los Cartílagos/terapia , Diferenciación Celular/genética , Condrocitos/metabolismo , Condrocitos/patología , Colágeno Tipo I/biosíntesis , Cadena alfa 1 del Colágeno Tipo I , Regulación del Desarrollo de la Expresión Génica , Terapia Genética , Humanos , Regiones Promotoras Genéticas , Factor de Transcripción Sp3/metabolismo
4.
BMC Complement Altern Med ; 15: 217, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-26156631

RESUMEN

BACKGROUND: Tendinopathies are tendon conditions associated with degeneration and disorganization of the matrix collagen fibers, tendon cells apoptosis and inflammation through up-regulation of proinflammatory cytokines, matrix metalloproteinase (MMP) expression, and prostaglandin E2 (PGE2) production. Currently, the pharmacological treatment is mainly based on non-steroidal anti-inflammatory drugs (NSAIDs) use and corticosteroid injections, which both can lead to numerous side effects for patients. TOL19-001 is a diet supplementary composed mostly of spirulina and glucosamine sulfate whose antioxidant properties could be helpful to treat tendinopathies while avoiding taking NSAIDs. In this study we developed an in vitro model of tendinopathy in order to evaluate the therapeutic potential of TOL19-001. METHODS: Tendon cells were cultured on monolayer and treated with interleukin-1ß (IL-1ß) or ciprofloxacin (CIP), and then, MMPs, PGE2 and collagen expression was evaluated by RT-PCR or Elisa. In addition, a cotreatment with increased doses of TOL19-001 was done. Toxicity of TOL19-001 was evaluated using a metabolic activity assay. RESULTS: This study demonstrates that IL-1ß mimics some aspects of tendinopathies with PGE2 induction, MMP expression (mostly MMP1 and MMP3), and increases of type III/I collagen ratio. CIP, meanwhile, leads to an increase of MMP2 and p65 mRNA, whereas it reduces TIMP1 expression. Scleraxis expression was also increased by CIP whereas it was decreased by IL-1ß treatment. Besides, TOL19-001 cotreatment suppresses tendon cell inflammation in vitro, marked by the downregulation of PGE2, MMPs and type III collagen in IL-1ß stimulated-cells. TOL19-001 also represses CIP induced-changes. CONCLUSIONS: These findings indicate that TOL19-001 exerts anti-inflammatory effects on tendon cells, which might explain why TOL19-001 diet may improve tendon function in patients with tendon injury. Future research is required to determine TOL19-001 effect on injured or overused tendons in vivo.


Asunto(s)
Suplementos Dietéticos , Metaloproteinasas de la Matriz/metabolismo , Tendones , Células Cultivadas , Humanos , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Tendones/citología , Tendones/efectos de los fármacos , Tendones/inmunología
5.
Biochim Biophys Acta ; 1823(5): 983-6, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22425785

RESUMEN

Interleukin-1ß (IL-1ß), a key-cytokine in osteoarthritis, impairs TGFß signaling through TßRII down-regulation by increasing its degradation. Here, we investigated the molecular mechanism that controls TßRII fate in IL-1ß treated cells. Chondrocytes were treated with IL-1ß in the presence of different inhibitors. TßRII and Cav-1 expression were assayed by Western blot and RT-PCR. We showed that IL-1ß-induced degradation of TßRII is dependent on proteasome and on its internalization in caveolae. In addition, IL-1ß enhances Cav-1 expression, a major constituent of lipid raft. In conclusion, we enlighten a new mechanism by which IL-1ß antagonizes TGFß pathway and propose a model of TßRII turnover regulation upon IL-1ß treatment.


Asunto(s)
Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Interleucina-1beta/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteolisis/efectos de los fármacos , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Anciano , Anciano de 80 o más Años , Transporte Biológico/efectos de los fármacos , Ácidos Borónicos/farmacología , Bortezomib , Caveolas/metabolismo , Caveolina 1/metabolismo , Células Cultivadas , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Persona de Mediana Edad , Nistatina/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Pirazinas/farmacología , Receptor Tipo II de Factor de Crecimiento Transformador beta
6.
Int J Stem Cells ; 16(3): 304-314, 2023 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-37105555

RESUMEN

Background and Objectives: Ear cartilage malformations are commonly encountered problems in reconstructive surgery, since cartilage has low self-regenerating capacity. Malformations that impose psychological and social burden on one's life are currently treated using ear prosthesis, synthetic implants or autologous flaps from rib cartilage. These approaches are challenging because not only they request high surgical expertise, but also they lack flexibility and induce severe donor-site morbidity. Through the last decade, tissue engineering gained attention where it aims at regenerating human tissues or organs in order to restore normal functions. This technique consists of three main elements, cells, growth factors, and above all, a scaffold that supports cells and guides their behavior. Several studies have investigated different scaffolds prepared from both synthetic or natural materials and their effects on cellular differentiation and behavior. Methods and Results: In this study, we investigated a natural scaffold (alginate) as tridimensional hydrogel seeded with progenitors from different origins such as bone marrow, perichondrium and dental pulp. In contact with the scaffold, these cells remained viable and were able to differentiate into chondrocytes when cultured in vitro. Quantitative and qualitative results show the presence of different chondrogenic markers as well as elastic ones for the purpose of ear cartilage, upon different culture conditions. Conclusions: We confirmed that auricular perichondrial cells outperform other cells to produce chondrogenic tissue in normal oxygen levels and we report for the first time the effect of hypoxia on these cells. Our results provide updates for cartilage engineering for future clinical applications.

7.
Mol Med ; 17(7-8): 816-23, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21528154

RESUMEN

A significant association between a polymorphism in the D repeat of the gene encoding asporin and osteoarthritis, the most frequent of articular diseases, has been recently reported. The goal of the present study was to investigate the expression of this new class I small leucine-rich proteoglycan (SLRP) in human articular chondrocytes. First, we studied the modulation of asporin (ASPN) expression by cytokines by Western blot and reverse transcription-polymerase chain reaction. Interleukin-1ß and tumor necrosis factor-α downregulated ASPN, whereas transforming growth factor-ß1 (when incubated in a serum-free medium) upregulated it. Similarly to proinflammatory cytokines, chondrocyte dedifferentiation induced by a successive passages of cells was accompanied by a decreased asporin expression, whereas their redifferentiation by three-dimensional culture restored its expression. Finally, we found an important role of the transcription factor Sp1 in the regulation of ASPN expression. Sp1 ectopic expression increased ASPN mRNA level and promoter activity. In addition, using gene reporter assay and electrophoretic mobility shift assay, we showed that Sp1 mediated its effect through a region located between -473 and -140 bp upstream of the transcription start site in ASPN gene. In conclusion, this report is the first study on the regulation of asporin expression by different cytokines in human articular chondrocytes. Our data indicate that the expression of this gene is finely regulated in cartilage and suggest a major role of Sp1.


Asunto(s)
Condrocitos/metabolismo , Proteínas de la Matriz Extracelular/genética , Expresión Génica/genética , Anciano , Anciano de 80 o más Años , Sitios de Unión/genética , Western Blotting , Cartílago Articular/citología , Desdiferenciación Celular/genética , Diferenciación Celular/genética , Células Cultivadas , Condrocitos/citología , Condrocitos/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Ensayo de Cambio de Movilidad Electroforética , Proteínas de la Matriz Extracelular/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Interleucina-1beta/farmacología , Persona de Mediana Edad , Cultivo Primario de Células , Regiones Promotoras Genéticas/genética , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Transfección , Factor de Crecimiento Transformador beta1/farmacología , Factor de Necrosis Tumoral alfa/farmacología
8.
Biomolecules ; 11(10)2021 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-34680093

RESUMEN

The role of genetics in the development of osteoarthritis is well established but the molecular bases are not fully understood. Here, we describe a family carrying a germline mutation in COMP (Cartilage Oligomeric Matrix Protein) associated with three distinct phenotypes. The index case was enrolled for a familial form of idiopathic early-onset osteoarthritis. By screening potential causal genes for osteoarthritis, we identified a heterozygous missense mutation of COMP (c.1358C>T, p.Asn453Ser), absent from genome databases, located on a highly conserved residue and predicted to be deleterious. Molecular dynamics simulation suggests that the mutation destabilizes the overall COMP protein structure and consequently the calcium releases from neighboring calcium binding sites. This mutation was once reported in the literature as causal for severe multiple epiphyseal dysplasia (MED). However, no sign of dysplasia was present in the index case. The mutation was also identified in one of her brothers diagnosed with MED and secondary osteoarthritis, and in her sister affected by an atypical syndrome including peripheral inflammatory arthritis of unknown cause, without osteoarthritis nor dysplasia. This article suggests that this mutation of COMP is not only causal for idiopathic early-onset osteoarthritis or severe MED, but can also be associated to a broad phenotypic variability with always joint alterations.


Asunto(s)
Proteína de la Matriz Oligomérica del Cartílago/genética , Predisposición Genética a la Enfermedad , Osteoartritis/genética , Osteocondrodisplasias/genética , Adulto , Femenino , Variación Genética/genética , Mutación de Línea Germinal/genética , Humanos , Articulaciones/patología , Masculino , Persona de Mediana Edad , Simulación de Dinámica Molecular , Mutación Missense/genética , Osteoartritis/patología , Osteocondrodisplasias/patología , Conformación Proteica , Relación Estructura-Actividad
9.
Sci Rep ; 10(1): 19577, 2020 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-33177650

RESUMEN

Histone methyltransferase EZH2 is upregulated during osteoarthritis (OA), which is the most widespread rheumatic disease worldwide, and a leading cause of disability. This study aimed to assess the impact of EZH2 inhibition on cartilage degradation, inflammation and functional disability. In vitro, gain and loss of EZH2 function were performed in human articular OA chondrocytes stimulated with IL-1ß. In vivo, the effects of EZH2 inhibition were investigated on medial meniscectomy (MMX) OA mouse model. The tissue alterations were assayed by histology and the functional disabilities of the mice by actimetry and running wheel. In vitro, EZH2 overexpression exacerbated the action of IL-1ß in chondrocytes increasing the expression of genes involved in inflammation, pain (NO, PGE2, IL6, NGF) and catabolism (MMPs), whereas EZH2 inhibition by a pharmacological inhibitor, EPZ-6438, reduced IL-1ß effects. Ex vivo, EZH2 inhibition decreased IL-1ß-induced degradation of cartilage. In vivo, intra-articular injections of the EZH2 inhibitor reduced cartilage degradation and improved motor functions of OA mice. This study demonstrates that the pharmacological inhibition of the histone methyl-transferase EZH2 slows the progression of osteoarthritis and improves motor functions in an experimental OA model, suggesting that EZH2 could be an effective target for the treatment of OA by reducing catabolism, inflammation and pain.


Asunto(s)
Cartílago Articular/patología , Proteína Potenciadora del Homólogo Zeste 2/genética , Osteoartritis/patología , Anciano , Anciano de 80 o más Años , Animales , Benzamidas/farmacología , Compuestos de Bifenilo/farmacología , Cartílago Articular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Condrocitos/fisiología , Modelos Animales de Enfermedad , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Regulación de la Expresión Génica , Humanos , Interleucina-1beta/farmacología , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Morfolinas/farmacología , Factor de Crecimiento Nervioso/metabolismo , Técnicas de Cultivo de Órganos , Piridonas/farmacología
10.
Mar Biotechnol (NY) ; 20(4): 436-450, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29627869

RESUMEN

The shells of the bivalve mollusks are organo-mineral structures predominantly composed of calcium carbonate, but also of a minor organic matrix, a mixture of proteins, glycoproteins, and polysaccharides. These proteins are involved in mineral deposition and, more generally, in the spatial organization of the shell crystallites in well-defined microstructures. In this work, we extracted different organic shell extracts (acid-soluble matrix, acid-insoluble matrix, water-soluble matrix, guanidine HCl/EDTA-extracted matrix, referred as ASM, AIM, WSM, and EDTAM, respectively) from the shell of the scallop Pecten maximus and studied their biological activities on human articular chondrocytes (HACs). We found that these extracts differentially modulate the biological activities of HACs, depending on the type of extraction and the concentration used. Furthermore, we showed that, unlike ASM and AIM, WSM promotes maintenance of the chondrocyte phenotype in monolayer culture. WSM increased the expression of chondrocyte-specific markers (aggrecan and type II collagen), without enhancing that of the main chondrocyte dedifferentiation marker (type I collagen). We also demonstrated that WSM could favor redifferentiation of chondrocyte in collagen sponge scaffold in hypoxia. Thus, this study suggests that the organic matrix of Pecten maximus, particularly WSM, may contain interesting molecules with chondrogenic effects. Our research emphasizes the potential use of WSM of Pecten maximus for cell therapy of cartilage.


Asunto(s)
Exoesqueleto/química , Condrocitos/efectos de los fármacos , Matriz Extracelular , Pecten/química , Anciano , Anciano de 80 o más Años , Agrecanos/genética , Agrecanos/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Condrocitos/citología , Condrocitos/metabolismo , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Perfilación de la Expresión Génica , Humanos , Persona de Mediana Edad , Fenotipo
11.
Sci Rep ; 7(1): 6483, 2017 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-28744016

RESUMEN

3-Deazaneplanocin A (DZNep) is an inhibitor of S-Adenosyl-L-Homocysteine Hydrolase (SAHH) known to inhibit EZH2, a histone methylase upregulated during osteoarthritis. In this study, we assessed its effects in human articular chondrocytes. Anti-inflammatory effects were assessed by Nitric Oxide (NO), Prostaglandin E2 (PGE2) and Metalloprotease (MMP) release in IL-1ß-stimulated chondrocytes. MAPK and NFκB activation was analyzed by western blotting. Differentially expressed genes (DEG) regulated by DZNep were identified by whole-transcriptome microarray. DZNep inhibited SAHH activity and was not toxic. It counteracted NO, PGE2 and MMP release, and reduced MAPK activation induced by IL-1ß. By whole-transcriptome analysis, we identified that DNZep counteracts the effect of IL-1ß on the expression of 81 protein-coding genes, including CITED2, an MMP inhibitor. These genes are organized in a protein-protein network centred on EGR1, which is known to functionally interact with EZH2. Gene ontologies enrichment analysis confirmed that DZNep counteracts IL-1ß-induced expression of genes involved in cartilage matrix breakdown (MMPs and ADAMTS). In addition, DZNep up-regulated cartilage specific genes, such as COL2A1 and SOX9, suggesting a chondroprotective effect of DZNep. DZNep exhibits anti-inflammatory effects, and regulates genes implicated in chondroprotective response in human articular chondrocytes, suggesting that inhibitors of S-adenosylmethionine-dependent methyltransferases could be effective treatments for OA.


Asunto(s)
Adenosina/análogos & derivados , Adenosilhomocisteinasa/antagonistas & inhibidores , Antiinflamatorios/farmacología , Cartílago Articular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Sustancias Protectoras/farmacología , Adenosina/farmacología , Cartílago Articular/citología , Cartílago Articular/metabolismo , Células Cultivadas , Condrocitos/citología , Condrocitos/metabolismo , Citoprotección , Dinoprostona/metabolismo , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Interleucina-1beta/metabolismo , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Osteoartritis/metabolismo , Osteoartritis/patología , Osteoartritis/prevención & control , Mapas de Interacción de Proteínas
12.
Sci Rep ; 7(1): 3406, 2017 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-28611369

RESUMEN

Mesenchymal stem cells (MSCs) hold promise for cartilage engineering. Here, we aimed to determine the best culture conditions to induce chondrogenesis of MSCs isolated from bone marrow (BM) of aged osteoarthritis (OA) patients. We showed that these BM-MSCs proliferate slowly, are not uniformly positive for stem cell markers, and maintain their multilineage potential throughout multiple passages. The chondrogenic lineage of BM-MSCs was induced in collagen scaffolds, under normoxia or hypoxia, by BMP-2 and/or TGF-ß1. The best chondrogenic induction, with the least hypertrophic induction, was obtained with the combination of BMP-2 and TGF-ß1 under hypoxia. Differentiated BM-MSCs were then transfected with siRNAs targeting two markers overexpressed in OA chondrocytes, type I collagen and/or HtrA1 protease. siRNAs significantly decreased mRNA and protein levels of type I collagen and HtrA1, resulting in a more typical chondrocyte phenotype, but with frequent calcification of the subcutaneously implanted constructs in a nude mouse model. Our 3D culture model with BMP-2/TGF-ß1 and COL1A1/HtrA1 siRNAs was not effective in producing a cartilage-like matrix in vivo. Further optimization is needed to stabilize the chondrocyte phenotype of differentiated BM-MSCs. Nevertheless, this study offers the opportunity to develop a combinatory cellular therapy strategy for cartilage tissue engineering.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Condrogénesis , Hipoxia , Células Madre Mesenquimatosas/citología , Osteoartritis/terapia , ARN Interferente Pequeño/genética , Ingeniería de Tejidos , Anciano , Anciano de 80 o más Años , Animales , Médula Ósea/crecimiento & desarrollo , Médula Ósea/metabolismo , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Diferenciación Celular , Células Cultivadas , Condrocitos/citología , Condrocitos/fisiología , Colágeno Tipo I/antagonistas & inhibidores , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Femenino , Serina Peptidasa A1 que Requiere Temperaturas Altas/antagonistas & inhibidores , Serina Peptidasa A1 que Requiere Temperaturas Altas/genética , Serina Peptidasa A1 que Requiere Temperaturas Altas/metabolismo , Humanos , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Desnudos , Persona de Mediana Edad , Osteoartritis/metabolismo , Osteoartritis/patología , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
13.
Trends Mol Med ; 22(1): 38-52, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26691295

RESUMEN

Osteoarthritis (OA) is the most common joint disease worldwide. A minority of cases correspond to familial presentation characterized by early-onset forms which are genetically heterogeneous. This review brings a new point of view on the molecular basis of OA by focusing on gene mutations causing early-onset OA (EO-OA). Recently, thanks to whole-exome sequencing, a gain-of-function mutation in the TNFRSF11B gene was identified in two distant family members with EO-OA, opening new therapeutic perspectives for OA. Indeed, unraveling the molecular basis of rare Mendelian OA forms will improve our understanding of molecular processes involved in OA pathogenesis and will contribute to better patient diagnosis, management, and therapy.


Asunto(s)
Predisposición Genética a la Enfermedad , Osteoartritis/genética , Edad de Inicio , Animales , Cartílago/metabolismo , Colágeno/genética , Humanos , Mutación , Osteoprotegerina/genética
14.
Age (Dordr) ; 35(4): 1105-16, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22653295

RESUMEN

Aging is one of the major risk factors of osteoarthritis. This pathology during which chondrocytes undergo modifications of their phenotype may result from alteration of transforming growth factor ß (TGFß) signaling. This study investigates the role of TGFß response in the process of chondrocyte dedifferentiation/redifferentiation. Dedifferentiation was induced by successive passages of human articular chondrocytes, whereas their redifferentiation was performed by three-dimensional culture in alginate. Human mesenchymal stem cells were obtained from bone marrow and differentiated into chondrocyte-like phenotype by three-dimensional culture, embedded in the same scaffold. Protein and mRNA levels were analyzed by Western blot and real-time reverse transcription PCR. Regulatory mechanism was investigated using specific inhibitors (mithramycin), mRNA silencing or decoy oligonucleotides, and expression vectors. Chondrocyte dedifferentiation interfered with TGFß signaling by decreasing TßRII mRNA and protein levels and subsequent TGFß response. TßRII ectopic expression in passaged chondrocytes permitted to increase the expression of several matrix genes, such as aggrecan or type II collagen. Redifferentiation of passaged chondrocytes permitted to restore, at least in part, TßRII expression and was related to differentiation of human bone marrow mesenchymal stem cells toward chondrocytes, where both specific protein 1 (Sp1) and TßRII mRNA levels were increased. Moreover, Sp1 manipulation by silencing or ectopic expression and pharmacologic inhibition revealed a link between expression levels of this transcriptional factor, which is crucial for constitutive expression of TßRII in cartilage, and TGFß response. Therefore, these data permit us to suggest an important role of TßRII expression in the maintenance of chondrocyte phenotype, which is altered with age, and bring new insights in our understanding of chondrogenesis process.


Asunto(s)
Envejecimiento/genética , Cartílago Articular/metabolismo , Condrocitos/metabolismo , Regulación de la Expresión Génica , ARN Mensajero/genética , Factor de Crecimiento Transformador beta2/genética , Anciano , Anciano de 80 o más Años , Envejecimiento/metabolismo , Envejecimiento/patología , Western Blotting , Cartílago Articular/patología , Diferenciación Celular/genética , Células Cultivadas , Condrocitos/patología , Progresión de la Enfermedad , Humanos , Persona de Mediana Edad , Osteoartritis de la Cadera/genética , Osteoartritis de la Cadera/metabolismo , Osteoartritis de la Cadera/patología , Fenotipo , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética , Factor de Crecimiento Transformador beta2/biosíntesis
15.
Biomaterials ; 33(26): 6042-51, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22677190

RESUMEN

Cartilage engineering is one of the most challenging issue in regenerative medicine, due to its limited self-ability to repair. Here, we assessed engineering of cartilage tissue starting from human bone marrow (hBM) stem cells under hypoxic environment and delineated the mechanism whereby chondrogenesis could be conducted without addition of exogenous growth factors. hBM stem cells were cultured in alginate beads and chondrogenesis was monitored by chondrocyte phenotypic markers. Activities and roles of Sox and HIF-1α transcription factors were investigated with complementary approaches of gain and loss of function and provided evidences that HIF-1α is essential for hypoxic induction of chondrogenesis. Thereafter, hBM cells and human articular chondrocytes (HAC) underwent chondrogenesis by 3D and hypoxic culture for 7 days or by ectopic expression of HIF-1α. After subcutaneous implantation of 3 weeks into athymic mice, tissue analysis showed that hypoxia or HIF-1α overexpression is effective and sufficient to induce chondrocyte phenotype in hBM cells, without use of exogenous growth factors. Therefore, this study brings interesting data for a simple and affordable system in biotechnology of cartilage engineering.


Asunto(s)
Cartílago/citología , Hipoxia de la Célula/fisiología , Condrogénesis/fisiología , Ingeniería de Tejidos/métodos , Anciano , Animales , Western Blotting , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Hipoxia de la Célula/genética , Células Cultivadas , Condrogénesis/genética , Ensayo de Cambio de Movilidad Electroforética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Desnudos , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Arthritis Res Ther ; 13(1): R23, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21324108

RESUMEN

INTRODUCTION: Transforming growth factor beta (TGFß) plays a central role in morphogenesis, growth, and cell differentiation. This cytokine is particularly important in cartilage where it regulates cell proliferation and extracellular matrix synthesis. While the action of TGFß on chondrocyte metabolism has been extensively catalogued, the modulation of specific genes that function as mediators of TGFß signalling is poorly defined. In the current study, elements of the Smad component of the TGFß intracellular signalling system and TGFß receptors were characterised in human chondrocytes upon TGFß1 treatment. METHODS: Human articular chondrocytes were incubated with TGFß1. Then, mRNA and protein levels of TGFß receptors and Smads were analysed by RT-PCR and western blot analysis. The role of specific protein 1 (Sp1) was investigated by gain and loss of function (inhibitor, siRNA, expression vector). RESULTS: We showed that TGFß1 regulates mRNA levels of its own receptors, and of Smad3 and Smad7. It modulates TGFß receptors post-transcriptionally by affecting their mRNA stability, but does not change the Smad-3 and Smad-7 mRNA half-life span, suggesting a potential transcriptional effect on these genes. Moreover, the transcriptional factor Sp1, which is downregulated by TGFß1, is involved in the repression of both TGFß receptors but not in the modulation of Smad3 and Smad7. Interestingly, Sp1 ectopic expression permitted also to maintain a similar expression pattern to early response to TGFß at 24 hours of treatment. It restored the induction of Sox9 and COL2A1 and blocked the late response (repression of aggrecan, induction of COL1A1 and COL10A1). CONCLUSIONS: These data help to better understand the negative feedback loop in the TGFß signalling system, and enlighten an interesting role of Sp1 to regulate TGFß response.


Asunto(s)
Condrocitos/metabolismo , Regulación de la Expresión Génica/genética , Transducción de Señal/genética , Factor de Transcripción Sp1/genética , Factor de Crecimiento Transformador beta1/genética , Western Blotting , Semivida , Humanos , ARN Mensajero/análisis , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Factores de Crecimiento Transformadores beta/biosíntesis , Receptores de Factores de Crecimiento Transformadores beta/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína smad3/biosíntesis , Proteína smad3/genética , Proteína smad7/biosíntesis , Proteína smad7/genética , Factor de Transcripción Sp1/metabolismo
17.
Arthritis Rheum ; 60(3): 760-70, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19248106

RESUMEN

OBJECTIVE: To uncover the mechanism by which chondroitin sulfate (CS) enhances hyaluronan (HA) production by human osteoarthritic (OA) fibroblast-like synoviocytes (FLS). METHODS: The production of HA was investigated by exposing human OA FLS to CS in the presence or absence of interleukin-1beta (IL-1beta). HA levels were determined by enzyme-linked immunosorbent assay, and levels of messenger RNA (mRNA) for HA synthase 1 (HAS-1), HAS-2, and HAS-3 were determined by real-time polymerase chain reaction analysis. The effect of CS and IL-1beta on signaling pathways was assessed by Western blotting. Specific inhibitors were used to determine their effects on both HA production and HAS expression. The molecular size of HA was analyzed by high-pressure liquid chromatography. RESULTS: CS increased HA production by FLS through up-regulation of the expression of HAS1 and HAS2. This was associated with activation of ERK-1/2, p38, and Akt, although to a lesser extent. Both p38 and Akt were involved in CS-induced HA accumulation. IL-1beta increased HA production and levels of mRNA for HAS1, HAS2, and HAS3. CS enhanced the IL-1beta-induced level of HAS2 mRNA and reduced the level of HAS3 mRNA. IL-1beta-induced activation of p38 and JNK was slightly decreased by CS, whereas that of ERK-1/2 and Akt was enhanced. More high molecular weight HA was found in CS plus IL-1beta-treated FLS than in FLS treated with IL-1beta alone. CONCLUSION: CS stimulates the synthesis of high molecular weight HA in OA FLS through up-regulation of HAS1 and HAS2. It reduces the IL-1beta-enhanced transcription of HAS3 and increases the production of HA of large molecular sizes. These effects may be beneficial for maintaining viscosity and antiinflammatory properties in the joint.


Asunto(s)
Sulfatos de Condroitina/farmacología , Glucuronosiltransferasa/metabolismo , Ácido Hialurónico/metabolismo , Osteoartritis de la Rodilla/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Membrana Sinovial/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Células Cultivadas , Humanos , Hialuronano Sintasas , Interleucina-1beta/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Osteoartritis de la Rodilla/patología , ARN Mensajero/metabolismo , Transducción de Señal , Membrana Sinovial/efectos de los fármacos , Membrana Sinovial/patología , Regulación hacia Arriba
18.
Arthritis Rheum ; 60(10): 3038-48, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19790048

RESUMEN

OBJECTIVE: Autologous chondrocyte implantation requires expansion of cells ex vivo, leading to dedifferentiation of chondrocytes (loss of aggrecan and type II collagen to the profit of type I and type III collagens). Several approaches have been described for redifferentiation of these cells. Among them, low oxygen tension has been exploited to restore the differentiated chondrocyte phenotype, but molecular mechanisms of this process remain unclear. However, under conditions of hypoxia, one of the major factors involved is hypoxia-inducible factor 1alpha (HIF-1alpha). The purpose of this study was to investigate the role of HIF-1alpha during human chondrocyte redifferentiation. METHODS: We used complementary approaches to achieving HIF-1alpha loss (inhibition by cadmium ions and dominant-negative expression) or gain (ectopic expression and cobalt ion treatment) of function. Expression of chondrocyte, as well as fibroblast-like, phenotype markers was determined using real-time reverse transcription-polymerase chain reaction and Western blot analyses. Binding activities of HIF-1alpha and SOX9, a pivotal transcription factor of chondrogenesis, were evaluated by electrophoretic mobility shift assays and by chromatin immunoprecipitation assay. RESULTS: We found that hypoxia and HIF-1alpha not only induced the expression of SOX9, COL2A1, and aggrecan, but they simultaneously inhibited the expression of COL1A1, COL1A2, and COL3A1. In addition, we identified the binding of HIF-1alpha to the aggrecan promoter, the first such reported demonstration of this binding. CONCLUSION: This study is the first to show a bimodal role of HIF-1alpha in cartilage homeostasis, since HIF-1alpha was shown to favor specific markers and to impair dedifferentiation. This suggests that manipulation of HIF-1alpha could represent a promising approach to the treatment of osteoarthritis.


Asunto(s)
Agrecanos/metabolismo , Diferenciación Celular/fisiología , Hipoxia de la Célula/fisiología , Condrocitos/metabolismo , Colágeno Tipo III/antagonistas & inhibidores , Colágeno Tipo II/metabolismo , Colágeno Tipo I/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Células Cultivadas , Condrocitos/patología , Colágeno/antagonistas & inhibidores , Colágeno/metabolismo , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Colágeno Tipo III/metabolismo , Humanos , Persona de Mediana Edad , Factor de Transcripción SOX9/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA