Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros













Intervalo de año de publicación
1.
Biol Res ; 56(1): 9, 2023 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-36890579

RESUMEN

BACKGROUND: Knowledge about regulating transcription factors (TFs) for osteoblastogenesis from mesenchymal stem cells (MSCs) is limited. Therefore, we investigated the relationship between genomic regions subject to DNA-methylation changes during osteoblastogenesis and the TFs known to directly interact with these regulatory regions. RESULTS: The genome-wide DNA-methylation signature of MSCs differentiated to osteoblasts and adipocytes was determined using the Illumina HumanMethylation450 BeadChip array. During adipogenesis no CpGs passed our test for significant methylation changes. Oppositely, during osteoblastogenesis we identified 2462 differently significantly methylated CpGs (adj. p < 0.05). These resided outside of CpGs islands and were significantly enriched in enhancer regions. We confirmed the correlation between DNA-methylation and gene expression. Accordingly, we developed a bioinformatic tool to analyse differentially methylated regions and the TFs interacting with them. By overlaying our osteoblastogenesis differentially methylated regions with ENCODE TF ChIP-seq data we obtained a set of candidate TFs associated to DNA-methylation changes. Among them, ZEB1 TF was highly related with DNA-methylation. Using RNA interference, we confirmed that ZEB1, and ZEB2, played a key role in adipogenesis and osteoblastogenesis processes. For clinical relevance, ZEB1 mRNA expression in human bone samples was evaluated. This expression positively correlated with weight, body mass index, and PPARγ expression. CONCLUSIONS: In this work we describe an osteoblastogenesis-associated DNA-methylation profile and, using these data, validate a novel computational tool to identify key TFs associated to age-related disease processes. By means of this tool we identified and confirmed ZEB TFs as mediators involved in the MSCs differentiation to osteoblasts and adipocytes, and obesity-related bone adiposity.


Asunto(s)
Células Madre Mesenquimatosas , Osteogénesis , Humanos , Osteogénesis/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Metilación de ADN , Diferenciación Celular/genética
2.
Biol. Res ; 56: 9-9, 2023. ilus, tab, graf
Artículo en Inglés | LILACS | ID: biblio-1429910

RESUMEN

BACKGROUND: Knowledge about regulating transcription factors (TFs) for osteoblastogenesis from mesenchymal stem cells (MSCs) is limited. Therefore, we investigated the relationship between genomic regions subject to DNA-methylation changes during osteoblastogenesis and the TFs known to directly interact with these regulatory regions. RESULTS: The genome-wide DNA-methylation signature of MSCs differentiated to osteoblasts and adipocytes was determined using the Illumina HumanMethylation450 BeadChip array. During adipogenesis no CpGs passed our test for significant methylation changes. Oppositely, during osteoblastogenesis we identified 2462 differently significantly methylated CpGs (adj. p < 0.05). These resided outside of CpGs islands and were significantly enriched in enhancer regions. We confirmed the correlation between DNA-methylation and gene expression. Accordingly, we developed a bioinformatic tool to analyse differentially methylated regions and the TFs interacting with them. By overlaying our osteoblastogenesis differentially methylated regions with ENCODE TF ChIP-seq data we obtained a set of candidate TFs associated to DNA-methylation changes. Among them, ZEB1 TF was highly related with DNA-methylation. Using RNA interference, we confirmed that ZEB1, and ZEB2, played a key role in adipogenesis and osteoblastogenesis processes. For clinical relevance, ZEB1 mRNA expression in human bone samples was evaluated. This expression positively correlated with weight, body mass index, and PPARγ expression. CONCLUSIONS: In this work we describe an osteoblastogenesis-associated DNA-methylation profile and, using these data, validate a novel computational tool to identify key TFs associated to age-related disease processes. By means of this tool we identified and confirmed ZEB TFs as mediators involved in the MSCs differentiation to osteoblasts and adipocytes, and obesity-related bone adiposity.


Asunto(s)
Humanos , Osteogénesis/genética , Células Madre Mesenquimatosas , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Diferenciación Celular/genética , Metilación de ADN
3.
Sci Rep ; 12(1): 6640, 2022 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-35459919

RESUMEN

Elevated pro-inflammatory signalling coupled with catabolic metalloproteinase expression is a common feature of arthritis, leading to cartilage damage, deterioration of the joint architecture and the associated pain and immobility. Countering these processes, histone deacetylase inhibitors (HDACi) have been shown to suppress matrix metalloproteinase (MMP) expression, block cytokine-induced signalling and reduce the cartilage degradation in animal models of the arthritis. In order to establish which specific HDACs account for these chondro-protective effects an HDAC1-11 RNAi screen was performed. HDAC6 was required for both the interleukin (IL)-1 induction of MMP expression and pro-inflammatory interleukin expression in chondrocytes, implicating an effect on NF-κB signalling. Depletion of HDAC6 post-transcriptionally up-regulated inhibitor of κB (IκB), prevented the nuclear translocation of NF-κB subunits and down-regulated NF-κB reporter activation. The pharmacological inhibition of HDAC6 reduced MMP expression in chondrocytes and cartilage collagen release. This work highlights the important role of HDAC6 in pro-inflammatory signalling and metalloproteinase gene expression, and identifies a part for HDAC6 in the NF-κB signalling pathway. By confirming the protection of cartilage this work supports the inhibition of HDAC6 as a possible therapeutic strategy in arthritis.


Asunto(s)
Artritis , Condrocitos , Animales , Artritis/genética , Artritis/metabolismo , Células Cultivadas , Condrocitos/metabolismo , Expresión Génica , Interleucina-1beta/metabolismo , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasas de la Matriz/metabolismo , FN-kappa B/metabolismo
4.
Sci Rep ; 11(1): 10452, 2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-34001919

RESUMEN

MicroRNAs are non-coding RNAs that act to downregulate the expression of target genes by translational repression and degradation of messenger RNA molecules. Individual microRNAs have the ability to specifically target a wide array of gene transcripts, therefore allowing each microRNA to play key roles in multiple biological pathways. miR-324 is a microRNA predicted to target thousands of RNA transcripts and is expressed far more highly in the brain than in any other tissue, suggesting that it may play a role in one or multiple neurological pathways. Here we present data from the first global miR-324-null mice, in which increased excitability and interictal discharges were identified in vitro in the hippocampus. RNA sequencing was used to identify differentially expressed genes in miR-324-null mice which may contribute to this increased hippocampal excitability, and 3'UTR luciferase assays and western blotting revealed that two of these, Suox and Cd300lf, are novel direct targets of miR-324. Characterisation of microRNAs that produce an effect on neurological activity, such as miR-324, and identification of the pathways they regulate will allow a better understanding of the processes involved in normal neurological function and in turn may present novel pharmaceutical targets in treating neurological disease.


Asunto(s)
Excitabilidad Cortical/genética , Hipocampo/fisiología , MicroARNs/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/genética , Receptores Inmunológicos/genética , Animales , Línea Celular , Femenino , Masculino , Ratones , Ratones Noqueados , MicroARNs/genética , Neocórtex/fisiología , RNA-Seq , Transducción de Señal/genética
5.
Epigenetics ; 16(1): 106-119, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32741307

RESUMEN

Dynamic modifications of chromatin allow rapid access of the gene regulatory machinery to condensed genomic regions facilitating subsequent gene expression. Inflammatory cytokine stimulation of cells can cause rapid gene expression changes through direct signalling pathway-mediated transcription factor activation and regulatory element binding. Here we used the Assay for Transposase Accessible Chromatin with high-throughput sequencing (ATAC-seq) to assess regions of the genome that are differentially accessible following treatment of cells with interleukin-1 (IL-1). We identified 126,483 open chromatin regions, with 241 regions significantly differentially accessible following stimulation, with 64 and 177 more or less accessible, respectively. These differentially accessible regions predominantly correspond to regions of the genome marked as enhancers. Motif searching identified an overrepresentation of a number of transcription factors, most notably RelA, in the regions becoming more accessible, with analysis of ChIP-seq data confirmed RelA binding to these regions. A significant correlation in differential chromatin accessibility and gene expression was also observed. Functionality in regulating gene expression was confirmed using CRISPR/Cas9 genome-editing to delete regions that became more accessible following stimulation in the genes MMP13, IKBKE and C1QTNF1. These same regions were also accessible for activation using a dCas9-transcriptional activator and showed enhancer activity in a cellular model. Together, these data describe and functionally validate a number of dynamically accessible chromatin regions involved in inflammatory signalling.


Asunto(s)
Ensamble y Desensamble de Cromatina/efectos de los fármacos , Interleucina-1/farmacología , Línea Celular Tumoral , Secuenciación de Inmunoprecipitación de Cromatina , Elementos de Facilitación Genéticos , Células HEK293 , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Unión Proteica , Proteínas/genética , Factor de Transcripción ReIA/metabolismo
6.
RNA ; 26(11): 1575-1588, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32660984

RESUMEN

miR-140 is selectively expressed in cartilage. Deletion of the entire Mir140 locus in mice results in growth retardation and early-onset osteoarthritis-like pathology; however, the relative contribution of miR-140-5p or miR-140-3p to the phenotype remains to be determined. An unbiased small RNA sequencing approach identified miR-140-3p as significantly more abundant (>10-fold) than miR-140-5p in human cartilage. Analysis of these data identified multiple miR-140-3p isomiRs differing from the miRBase annotation at both the 5' and 3' end, with >99% having one of two seed sequences (5' bases 2-8). Canonical (miR-140-3p.2) and shifted (miR-140-3p.1) seed isomiRs were overexpressed in chondrocytes and transcriptomics performed to identify targets. miR-140-3p.1 and miR-140-3p.2 significantly down-regulated 694 and 238 genes, respectively, of which only 162 genes were commonly down-regulated. IsomiR targets were validated using 3'UTR luciferase assays. miR-140-3p.1 targets were enriched within up-regulated genes in rib chondrocytes of Mir140-null mice and within down-regulated genes during human chondrogenesis. Finally, through imputing the expression of miR-140 from the expression of the host gene WWP2 in 124 previously published data sets, an inverse correlation with miR-140-3p.1 predicted targets was identified. Together these data suggest the novel seed containing isomiR miR-140-3p.1 is more functional than original consensus miR-140-3p seed containing isomiR.


Asunto(s)
Cartílago/química , MicroARNs/genética , Análisis de Secuencia de ARN/métodos , Regiones no Traducidas 3' , Regiones no Traducidas 5' , Animales , Condrogénesis , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Ratones , Anotación de Secuencia Molecular , Especificidad de Órganos , Regulación hacia Arriba
7.
Sci Rep ; 10(1): 1169, 2020 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-31980739

RESUMEN

Regulation of transcription occurs in a cell type specific manner orchestrated by epigenetic mechanisms including DNA methylation. Methylation changes may also play a key role in lineage specification during stem cell differentiation. To further our understanding of epigenetic regulation in chondrocytes we characterised the DNA methylation changes during chondrogenesis of mesenchymal stem cells (MSCs) by Infinium 450 K methylation array. Significant DNA hypomethylation was identified during chondrogenic differentiation including changes at many key cartilage gene loci. Integration with chondrogenesis gene expression data revealed an enrichment of significant CpGs in upregulated genes, while characterisation of significant CpG loci indicated their predominant localisation to enhancer regions. Comparison with methylation profiles of other tissues, including healthy and diseased adult cartilage, identified chondrocyte-specific regions of hypomethylation and the overlap with differentially methylated CpGs in osteoarthritis. Taken together we have associated DNA methylation levels with the chondrocyte phenotype. The consequences of which has potential to improve cartilage generation for tissue engineering purposes and also to provide context for observed methylation changes in cartilage diseases such as osteoarthritis.


Asunto(s)
Condrogénesis/genética , Metilación de ADN , Elementos de Facilitación Genéticos/genética , Células Madre Mesenquimatosas/metabolismo , Adolescente , Adulto , Células de la Médula Ósea/metabolismo , Cartílago Articular/citología , Cartílago Articular/metabolismo , Células Cultivadas , Condrocitos/metabolismo , Cromatina/ultraestructura , Islas de CpG , Regulación de la Expresión Génica , Humanos , Especificidad de Órganos , Adulto Joven
8.
F1000Res ; 82019.
Artículo en Inglés | MEDLINE | ID: mdl-30828429

RESUMEN

Metalloproteinases remain important players in arthritic disease, in part because members of this large enzymatic family, namely matrix metalloproteinase-1 (MMP-1) and MMP-13, are responsible for the irreversible degradation of articular cartilage collagen. Although direct inhibition of MMPs fell out of vogue with the initial clinical disappointment of the first generation of compounds, interest in other mechanisms that control these important enzymes has always been maintained. Since these enzymes are critically important for tissue homeostasis, their expression and activity are tightly regulated at many levels, not just by direct inhibition by their endogenous inhibitors the tissue inhibitors of metalloproteinases (TIMPs). Focussing on MMP-13, we discuss recent work that highlights new discoveries in the transcriptional regulation of this enzyme, from defined promoter functional analysis to how more global technologies can provide insight into the enzyme's regulation, especially by epigenetic mechanisms, including non-coding RNAs. In terms of protein regulation, we highlight recent findings into enzymatic cascades involved in MMP-13 regulation and activation. Importantly, we highlight a series of recent studies that describe how MMP-13 activity, and in fact that of other metalloproteinases, is in part controlled by receptor-mediated endocytosis. Together, these new discoveries provide a plethora of novel regulatory mechanisms, besides direct inhibition, which with renewed vigour could provide further therapeutic opportunities for regulating the activity of this class of important enzymes.


Asunto(s)
Metaloproteinasa 13 de la Matriz/fisiología , Inhibidores Tisulares de Metaloproteinasas/fisiología , Colágeno , Endocitosis , Regulación de la Expresión Génica , Humanos
9.
Matrix Biol ; 77: 87-100, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30193893

RESUMEN

The Hedgehog (Hh) signalling pathway plays important roles during embryonic development and in adult tissue homeostasis, for example cartilage, where its deregulation can lead to osteoarthritis (OA). microRNAs (miRNAs) are important regulators of gene expression, and have been implicated in the regulation of signalling pathways, including Hh, thereby impacting upon development and disease. Our aim was to identify the function of miRNAs whose expression is altered in OA cartilage. Here we identified an increase in miR-324-5p expression in OA cartilage and hypothesised that, as in glioma, miR-324-5p would regulate Hh signalling. We determined that miR-324-5p regulates osteogenesis in human mesenchymal stem cells (MSCs) and in mouse C3H10T1/2 cells. Luciferase reporter assays demonstrated that miR-324-5p directly regulated established targets GLI1 and SMO in human but not in mouse, suggesting species-dependent mechanism of Hh pathway regulation. Stable Isotope Labelling with Amino acids in Cell culture (SILAC), mass spectrometry and whole genome transcriptome analysis identified Glypican 1 (Gpc1) as a novel miR-324-5p target in mouse, which was confirmed by real-time RT-PCR, immunoblotting and 3'UTR-luciferase reporters. Knockdown of Gpc1 reduced Hh pathway activity, and phenocopied the effect of miR-324-5p on osteogenesis, indicating that miR-324-5p regulates Hh signalling in mouse via direct targeting of Gpc1. Finally, we showed that human GPC1 is not a direct target of miR-324-5p. Importantly, as well as identifying novel regulation of Indian Hedgehog (Ihh) signalling, this study demonstrates how a miRNA can show conserved pathway regulation in two species but by distinct mechanisms and highlights important differences between human diseases and mouse models.


Asunto(s)
Cartílago/metabolismo , Glipicanos/genética , Proteínas Hedgehog/genética , MicroARNs/genética , Osteoartritis/genética , Receptor Smoothened/genética , Proteína con Dedos de Zinc GLI1/genética , Regiones no Traducidas 3' , Adulto , Animales , Cartílago/patología , Línea Celular , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Genes Reporteros , Glipicanos/antagonistas & inhibidores , Glipicanos/metabolismo , Proteínas Hedgehog/metabolismo , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , MicroARNs/metabolismo , Osteoartritis/metabolismo , Osteoartritis/patología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Receptor Smoothened/metabolismo , Especificidad de la Especie , Proteína con Dedos de Zinc GLI1/metabolismo
10.
Development ; 144(24): 4510-4521, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29084806

RESUMEN

Long non-coding RNAs (lncRNAs) are expressed in a highly tissue-specific manner and function in various aspects of cell biology, often as key regulators of gene expression. In this study, we established a role for lncRNAs in chondrocyte differentiation. Using RNA sequencing we identified a human articular chondrocyte repertoire of lncRNAs from normal hip cartilage donated by neck of femur fracture patients. Of particular interest are lncRNAs upstream of the master chondrocyte transcription factor SOX9 locus. SOX9 is an HMG-box transcription factor that plays an essential role in chondrocyte development by directing the expression of chondrocyte-specific genes. Two of these lncRNAs are upregulated during chondrogenic differentiation of mesenchymal stem cells (MSCs). Depletion of one of these lncRNAs, LOC102723505, which we termed ROCR (regulator of chondrogenesis RNA), by RNA interference disrupted MSC chondrogenesis, concomitant with reduced cartilage-specific gene expression and incomplete matrix component production, indicating an important role in chondrocyte biology. Specifically, SOX9 induction was significantly ablated in the absence of ROCR, and overexpression of SOX9 rescued the differentiation of MSCs into chondrocytes. Our work sheds further light on chondrocyte-specific SOX9 expression and highlights a novel method of chondrocyte gene regulation involving a lncRNA.


Asunto(s)
Cartílago Articular/crecimiento & desarrollo , Diferenciación Celular/genética , Condrogénesis/genética , Células Madre Mesenquimatosas/citología , ARN Largo no Codificante/genética , Factor de Transcripción SOX9/biosíntesis , Anciano , Secuencia de Bases , Cartílago Articular/citología , Células Cultivadas , Condrocitos/citología , Femenino , Cadera/fisiología , Humanos , ARN Largo no Codificante/biosíntesis , Análisis de Secuencia de ARN
11.
Stem Cells ; 33(11): 3266-80, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26175215

RESUMEN

microRNAs (miRNAs) are abundantly expressed in development where they are critical determinants of cell differentiation and phenotype. Accordingly miRNAs are essential for normal skeletal development and chondrogenesis in particular. However, the question of which miRNAs are specific to the chondrocyte phenotype has not been fully addressed. Using microarray analysis of miRNA expression during mesenchymal stem cell chondrogenic differentiation and detailed examination of the role of essential differentiation factors, such as SOX9, TGF-ß, and the cell condensation phase, we characterize the repertoire of specific miRNAs involved in chondrocyte development, highlighting in particular miR-140 and miR-455. Further with the use of mRNA microarray data we integrate miRNA expression and mRNA expression during chondrogenesis to underline the particular importance of miR-140, especially the -5p strand. We provide a detailed identification and validation of direct targets of miR-140-5p in both chondrogenesis and adult chondrocytes with the use of microarray and 3'UTR analysis. This emphasizes the diverse array of targets and pathways regulated by miR-140-5p. We are also able to confirm previous experimentally identified targets but, additionally, identify a novel positive regulation of the Wnt signaling pathway by miR-140-5p. Wnt signaling has a complex role in chondrogenesis and skeletal development and these findings illustrate a previously unidentified role for miR-140-5p in regulation of Wnt signaling in these processes. Together these developments further highlight the role of miRNAs during chondrogenesis to improve our understanding of chondrocyte development and guide cartilage tissue engineering.


Asunto(s)
Condrogénesis/fisiología , Perfilación de la Expresión Génica/métodos , Marcación de Gen/métodos , Estudio de Asociación del Genoma Completo/métodos , Células Madre Mesenquimatosas/fisiología , MicroARNs/fisiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Células Cultivadas , Humanos , Persona de Mediana Edad , Adulto Joven
12.
Ann Rheum Dis ; 74(8): 1580-7, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24713355

RESUMEN

OBJECTIVES: To determine whether the process of ubiquitination and/or activity of the 26S proteasome are involved in the induction of osteoarthritis (OA). METHODS: Bovine cartilage resorption assays, chondrocyte cell-line SW1353 and primary human articular chondrocytes were used with the general proteasome inhibitor MG132 or vehicle to identify a role of the ubiquitin-proteasome system (UPS) in cartilage destruction and matrix metalloproteinase-13 (MMP13) expression. In vivo, MG132 or vehicle, were delivered subcutaneously to mice following destabilisation of the medial meniscus (DMM)-induced OA. Subsequently, DMM was induced in Lys-to-Arg (K48R and K63R) mutant ubiquitin (Ub) transgenic mice. Cytokine signalling in SW1353s was monitored by immunoblotting and novel ubiquitinated substrates identified using Tandem Ubiquitin Binding Entities purification followed by mass spectrometry. The ubiquitination of TRAFD1 was assessed via immunoprecipitation and immunoblotting and its role in cytokine signal-transduction determined using RNA interference and real-time RT-PCR for MMP13 and interleukin-6 (IL6). RESULTS: Supplementation with the proteasome inhibitor MG132 protected cartilage from cytokine-mediated resorption and degradation in vivo in mice following DMM-induced OA. Using transgenic animals only K48R-mutated Ub partially protected against OA compared to wild-type or wild-type Ub transgenic mice, and this was only evident on the medial femoral condyle. After confirming ubiquitination was vital for NF-κB signalling and MMP13 expression, a screen for novel ubiquitinated substrates involved in cytokine-signalling identified TRAFD1; the depletion of which reduced inflammatory mediator-induced MMP13 and IL6 expression. CONCLUSIONS: Our data for the first time identifies a role for ubiquitination and the proteasome in the induction of OA via regulation of inflammatory mediator-induced MMP13 expression. These data open avenues of research to determine whether the proteasome, or K48-linked ubiquitination, are potential therapeutic targets in OA.


Asunto(s)
Inhibidores de Cisteína Proteinasa/farmacología , Leupeptinas/farmacocinética , Metaloproteinasa 13 de la Matriz/metabolismo , Osteoartritis/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitinación/fisiología , Animales , Modelos Animales de Enfermedad , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisina/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal/fisiología , Dedos de Zinc/fisiología
13.
Arthritis Rheumatol ; 66(9): 2450-60, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24838673

RESUMEN

OBJECTIVE: The aim of this study was to characterize the genome-wide DNA methylation profile of chondrocytes from knee and hip cartilage obtained from patients with osteoarthritis (OA) and hip cartilage obtained from patients with femoral neck fracture, providing the first comparison of DNA methylation between OA and non-OA hip cartilage, and between OA hip and OA knee cartilage. METHODS: The study was performed using the Illumina Infinium HumanMethylation450 BeadChip array, which allows the annotation of ∼480,000 CpG sites. Genome-wide methylation was assessed in chondrocyte DNA extracted from 23 hip OA patients, 73 knee OA patients, and 21 healthy hip control patients with femoral neck fracture. RESULTS: Analysis revealed that chondrocytes from the hip cartilage of OA patients and healthy controls have unique methylation profiles, with 5,322 differentially methylated loci (DMLs) identified between the 2 groups. In addition, a comparison between hip and knee OA chondrocytes revealed 5,547 DMLs between the 2 groups, including DMLs in several genes known to be involved in the pathogenesis of OA. Hip OA samples were found to cluster into 2 groups. A total of 15,239 DMLs were identified between the 2 clusters, with an enrichment of genes involved in inflammation and immunity. Similarly, we confirmed a previous report of knee OA samples that also clustered into 2 groups. CONCLUSION: We demonstrated that global DNA methylation using a high-density array can be a powerful tool in the characterization of OA at the molecular level. Identification of pathways enriched in DMLs between OA and OA-free cartilage highlight potential etiologic mechanisms that are involved in the initiation and/or progression of the disease and that could be therapeutically targeted.


Asunto(s)
Cartílago Articular/metabolismo , Metilación de ADN , Osteoartritis de la Cadera/metabolismo , Osteoartritis de la Rodilla/metabolismo , Anciano , Anciano de 80 o más Años , Condrocitos/metabolismo , Femenino , Fracturas del Cuello Femoral/genética , Fracturas del Cuello Femoral/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Osteoartritis de la Cadera/genética , Osteoartritis de la Rodilla/genética
14.
Curr Rheumatol Rep ; 15(9): 353, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23888362

RESUMEN

Osteoarthritis (OA) is a disease typified by the loss of cartilage, the normal integrity of which is maintained by the resident cell, the chondrocyte. Alterations in chondrocyte gene expression with age, injury, loading or predisposing genetics, underpin OA cartilage loss. Cell- and tissue-specific gene expression is determined by epigenetic mechanisms, including DNA methylation, chromatin modifications and non-coding RNAs, including microRNAs and long-non-coding RNAs. A number of epigenetic changes have been identified between OA and normal cartilage, and the enzymes which impart the epigenetic code are increasingly seen as important players in a number of pathologies, including OA. Here, we will describe current and potential new epigenetic studies that are likely to reveal novel aspects of chondrocyte and cartilage biology and potentially help sub-characterise OA phenotypes. Importantly, many of these epigenetic modifiers or non-coding RNAs are proposed drug targets and could represent a therapeutic opportunity for this currently untreatable disease.


Asunto(s)
Epigénesis Genética , Osteoartritis/genética , ARN no Traducido/genética , Condrocitos/metabolismo , Cromatina/genética , Metilación de ADN , Predisposición Genética a la Enfermedad , Humanos , Osteoartritis/patología
15.
Arthritis Rheum ; 65(7): 1822-30, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23575963

RESUMEN

OBJECTIVE: To examine the ability of a broad-spectrum histone deacetylase (HDAC) inhibitor to protect cartilage in vivo, and to explore the effects of class-selective HDAC inhibitors and small interfering RNA (siRNA)-induced knockdown of HDACs on metalloproteinase expression and cartilage degradation in vitro. METHODS: A destabilization of the medial meniscus (DMM) model was used to assess the in vivo activity of the HDAC inhibitor trichostatin A (TSA). Human articular chondrocytes (HACs) and SW-1353 chondrosarcoma cells were treated with cytokines and TSA, valproic acid, MS-275, or siRNA, and quantitative reverse transcription-polymerase chain reaction was performed to determine the effect of treatment on metalloproteinase expression. HDAC inhibitor activity was detected by Western blotting. A bovine nasal cartilage (BNC) explant assay was performed to measure cartilage resorption in vitro. RESULTS: Systemically administered TSA protected cartilage in the DMM model. TSA, valproic acid, and MS-275 repressed cytokine-induced MMP1 and MMP13 expression in HACs. Knockdown of each class I HDAC diminished interleukin-1-induced MMP13 expression. All of the HDAC inhibitors prevented degradation of BNC, in which TSA and MS-275 repressed cytokine-induced MMP expression. CONCLUSION: Inhibition of class I HDACs (HDAC-1, HDAC-2, HDAC-3) by MS-275 or by specific depletion of HDACs is capable of repressing cytokine-induced metalloproteinase expression in cartilage cells and BNC explants, resulting in inhibition of cartilage resorption. These observations indicate that specific inhibition of class I HDACs is a possible therapeutic strategy in the arthritides.


Asunto(s)
Benzamidas/farmacología , Condrocitos/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Metaloproteasas/efectos de los fármacos , Cartílagos Nasales/efectos de los fármacos , Osteoartritis de la Rodilla , Piridinas/farmacología , Animales , Bovinos , Línea Celular Tumoral , Células Cultivadas , Condrocitos/metabolismo , Modelos Animales de Enfermedad , Histonas/efectos de los fármacos , Histonas/metabolismo , Humanos , Metaloproteasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Cartílagos Nasales/metabolismo , ARN Interferente Pequeño/farmacología , Tubulina (Proteína)/efectos de los fármacos , Tubulina (Proteína)/metabolismo
16.
Nat Med ; 18(9): 1369-77, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22941276

RESUMEN

We investigated whether ancestral liver damage leads to heritable reprogramming of hepatic wound healing in male rats. We found that a history of liver damage corresponds with transmission of an epigenetic suppressive adaptation of the fibrogenic component of wound healing to the male F1 and F2 generations. Underlying this adaptation was less generation of liver myofibroblasts, higher hepatic expression of the antifibrogenic factor peroxisome proliferator-activated receptor γ (PPAR-γ) and lower expression of the profibrogenic factor transforming growth factor ß1 (TGF-ß1) compared to rats without this adaptation. Remodeling of DNA methylation and histone acetylation underpinned these alterations in gene expression. Sperm from rats with liver fibrosis were enriched for the histone variant H2A.Z and trimethylation of histone H3 at Lys27 (H3K27me3) at PPAR-γ chromatin. These modifications to the sperm chromatin were transmittable by adaptive serum transfer from fibrotic rats to naive rats and similar modifications were induced in mesenchymal stem cells exposed to conditioned media from cultured rat or human myofibroblasts. Thus, it is probable that a myofibroblast-secreted soluble factor stimulates heritable epigenetic signatures in sperm so that the resulting offspring better adapt to future fibrogenic hepatic insults. Adding possible relevance to humans, we found that people with mild liver fibrosis have hypomethylation of the PPARG promoter compared to others with severe fibrosis.


Asunto(s)
Adaptación Biológica/fisiología , Metilación de ADN , Histonas/metabolismo , Hepatopatías/patología , Espermatozoides/química , Cicatrización de Heridas/fisiología , Acetilación , Actinas , Adaptación Biológica/genética , Animales , Western Blotting , Humanos , Inmunohistoquímica , Hepatopatías/genética , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Miofibroblastos/fisiología , PPAR gamma/metabolismo , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Crecimiento Transformador beta1/metabolismo , Cicatrización de Heridas/genética
17.
FASEB J ; 26(7): 3000-11, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22505473

RESUMEN

Osteoarthritis is a degenerative joint disease characterized by a progressive and irreversible loss of the articular cartilage, due in main part to the cleavage of type II collagen within the matrix by the enzyme matrix metalloproteinase (MMP)13. Here, we examined the methylation status of MMP13 promoter and report the demethylation of specific CpG dinucleotides within its promoter in osteoarthritic compared to normal cartilage, which correlates with increased MMP13 expression. Of the promoter CpG sites examined, the -104 CpG was consistently demethylated following treatment of human articular chondrocytes with 10 µM DNA-methyltransferase inhibitor 5-aza-2'-deoxycytidine, again correlating with increased MMP13 expression. Methylation of the -104 CpG site resulted in reduced promoter activity in the chondrosarcoma cell line SW1353 as shown by CpG-free luciferase reporter. Using electrophoretic mobility shift assays, we identified CREB as the regulating factor able to only bind to the MMP13 promoter when the -104 CpG is demethylated, and confirmed this binding by chromatin immunoprecipitation. Finally, we demonstrated that CREB induces MMP13 expression only following treatment of SW1353 with 0.5 µM Ca(2+) ionophore A23187. In summary, the -104 CpG is demethylated in osteoarthritic cartilage, correlating with the elevated MMP13 expression and cartilage destruction, providing a highly novel link between epigenetic status and arthritic disease.


Asunto(s)
Condrocitos/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Metaloproteinasa 13 de la Matriz/genética , Osteoartritis/genética , Osteoartritis/metabolismo , Azacitidina/análogos & derivados , Azacitidina/farmacología , Secuencia de Bases , Calcio/metabolismo , Condrocitos/efectos de los fármacos , Islas de CpG , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/antagonistas & inhibidores , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Metilación de ADN , Metilasas de Modificación del ADN/antagonistas & inhibidores , Cartilla de ADN/genética , Decitabina , Inhibidores Enzimáticos/farmacología , Humanos , Técnicas In Vitro , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética , Regulación hacia Arriba/efectos de los fármacos
18.
Ann Rheum Dis ; 69(12): 2189-98, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20693272

RESUMEN

OBJECTIVE: To investigate if statins prevent cartilage degradation and the production of collagenases and gelatinases in bovine nasal and human articular cartilage after proinflammatory cytokine stimulation. METHODS: In a cartilage degradation model, the effects of several statins were assessed by measuring proteoglycan degradation and collagen degradation, while collagenolytic and gelatinolytic activity in culture supernatants were determined by collagen bioassay and gelatin zymography. The production of matrix metalloproteinases (MMPs) in cartilage and chondrocytes were analysed by real-time reverse transcriptase PCR and immunoassay. Cytokine-induced signalling pathway activation was studied by immunoblotting. RESULTS: Simvastatin and mevastatin significantly inhibited interleukin 1 (IL-1)+oncostatin M (OSM)-induced collagen degradation; this was accompanied with a marked decrease in collagenase and gelatinase activity from bovine nasal cartilage. The cholesterol pathway intermediate mevalonic acid reversed the simvastatin-mediated protection of cartilage degradation, and the expression and production of collagenase (MMP-1 and MMP-13) and gelatinase (MMP-2 and MMP-9). Statins also significantly decreased MMP-1 and MMP-13 expression in human articular cartilage and chondrocytes stimulated with IL-1+OSM, and blocked the activation of critical proinflammatory signalling pathways required for MMP expression. The loss of the isoprenoid intermediate geranylgeranyl pyrophosphate due to statin treatment accounted for the inhibition of MMP expression and signalling pathway activation. CONCLUSIONS: This study shows, for the first time, that lipophilic statins are able to block cartilage collagen breakdown induced by proinflammatory cytokines, by downregulating key cartilage-degrading enzymes. This demonstrates a possible therapeutic role for statins in acting as anti-inflammatory agents and in protecting cartilage from damage in joint diseases.


Asunto(s)
Cartílago Articular/efectos de los fármacos , Colágeno/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Metaloproteinasas de la Matriz/fisiología , Cartílagos Nasales/efectos de los fármacos , Animales , Cartílago Articular/metabolismo , Bovinos , Células Cultivadas , Colagenasas/biosíntesis , Regulación hacia Abajo/efectos de los fármacos , Gelatinasas/biosíntesis , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-1alfa/farmacología , Lovastatina/análogos & derivados , Lovastatina/farmacología , Metaloproteinasas de la Matriz/genética , Ácido Mevalónico/farmacología , Cartílagos Nasales/metabolismo , Oncostatina M/farmacología , Transducción de Señal/efectos de los fármacos , Simvastatina/antagonistas & inhibidores , Simvastatina/farmacología , Terpenos/metabolismo , Técnicas de Cultivo de Tejidos
19.
Rheumatology (Oxford) ; 49(11): 2043-53, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20634235

RESUMEN

OBJECTIVES: To determine the effects and mechanism of action of lithium chloride (LiCl) on cartilage destruction induced by the pro-inflammatory cytokines IL-1, IL-1 + oncostatin M and TNF-α. METHODS: The release of collagen was assessed in bovine cartilage explant cultures, whereas collagenolytic activities (active and total) in conditioned culture supernatants were determined by bioassay. The expression and production of MMP from chondrocytes were analysed by real-time RT-PCR and ELISA. Signalling pathway analysis was performed using a phospho-antibody array and standard immunoblotting. RESULTS: LiCl, but not selective glycogen synthase kinase 3 (GSK-3) inhibitor compounds SB-415286 and TDZD-8, significantly decreased pro-inflammatory cytokine-induced collagen release from bovine cartilage via the down-regulation of collagenolytic activity. Furthermore, MMP-1 and MMP-13 expression was reduced in both bovine and human chondrocytes. Pathway analysis revealed that LiCl selectively inhibited activation of the p38 mitogen-activated protein kinase pathway; effects that were recapitulated by specific p38 pathway inhibition. CONCLUSIONS: This study demonstrates for the first time that LiCl can protect against cartilage damage induced by pro-inflammatory cytokines, and indicates that LiCl-mediated cartilage protection is not via a GSK-3-dependent mechanism, but potentially via inhibition of the p38 pathway. These data indicate that lithium administration may represent a potential therapy for arthritis.


Asunto(s)
Cartílago/efectos de los fármacos , Condrocitos/efectos de los fármacos , Cloruro de Litio/farmacología , Metaloproteinasas de la Matriz/metabolismo , Osteoartritis/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Anciano , Animales , Cartílago/metabolismo , Bovinos , Condrocitos/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Humanos , Cloruro de Litio/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
J Biol Chem ; 285(29): 22414-25, 2010 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-20463008

RESUMEN

The protein kinase C (PKC) signaling pathway is a major regulator of cellular functions and is implicated in pathologies involving extracellular matrix remodeling. Inflammatory joint disease is characterized by excessive extracellular matrix catabolism, and here we assess the role of PKC in the induction of the collagenases, matrix metalloproteinase (MMP)-1 and MMP-13, in human chondrocytes by the potent cytokine stimulus interleukin-1 (IL-1) in combination with oncostatin M (OSM). IL-1 + OSM-stimulated collagenolysis and gelatinase activity were ameliorated by pharmacological PKC inhibition in bovine cartilage, as was collagenase gene induction in human chondrocytes. Small interfering RNA-mediated silencing of PKC gene expression showed that both novel (nPKC delta, nPKC eta) and atypical (aPKC zeta, aPKC iota) isoforms were involved in collagenase induction by IL-1. However, MMP1 and MMP13 induction by IL-1 + OSM was inhibited only by aPKC silencing, suggesting that only atypical isoforms play a significant role in complex inflammatory milieus. Silencing of either aPKC led to diminished IL-1 + OSM-dependent extracellular signal-regulated kinase (ERK) and signal transducer and activator of transcription (STAT) 3 phosphorylation, and c-fos expression. STAT3 gene silencing or ERK pathway inhibition also resulted in loss of IL-1 + OSM-stimulated c-fos and collagenase expression. Silencing of c-fos and c-jun expression was sufficient to abrogate IL-1 + OSM-stimulated collagenase gene induction, and overexpression of both c-fos and c-jun was sufficient to drive transcription from the MMP1 promoter in the absence of a stimulus. Our data identify atypical PKC isozymes as STAT and ERK activators that mediate c-fos and collagenase expression during IL-1 + OSM synergy in human chondrocytes. aPKCs may constitute potential therapeutic targets for inflammatory joint diseases involving increased collagenase expression.


Asunto(s)
Cartílago Articular/patología , Colagenasas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Isoenzimas/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Cartílago Articular/efectos de los fármacos , Cartílago Articular/enzimología , Bovinos , Condrocitos/efectos de los fármacos , Condrocitos/enzimología , Condrocitos/patología , Colagenasas/biosíntesis , Colagenasas/genética , Inducción Enzimática/efectos de los fármacos , Gelatinasas/metabolismo , Silenciador del Gen/efectos de los fármacos , Humanos , Interleucina-1/farmacología , Isoenzimas/antagonistas & inhibidores , Oncostatina M/farmacología , Fosforilación/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Factor de Transcripción STAT3/deficiencia , Transducción de Señal/efectos de los fármacos , Especificidad por Sustrato/efectos de los fármacos , Factor de Transcripción AP-1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA