Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
BMJ Open ; 10(7): e035101, 2020 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-32723735

RESUMO

PURPOSE: The Applied Public-Private Research enabling OsteoArthritis Clinical Headway (APPROACH) consortium intends to prospectively describe in detail, preselected patients with knee osteoarthritis (OA), using conventional and novel clinical, imaging, and biochemical markers, to support OA drug development. PARTICIPANTS: APPROACH is a prospective cohort study including 297 patients with tibiofemoral OA, according to the American College of Rheumatology classification criteria. Patients were (pre)selected from existing cohorts using machine learning models, developed on data from the CHECK cohort, to display a high likelihood of radiographic joint space width (JSW) loss and/or knee pain progression. FINDINGS TO DATE: Selection appeared logistically feasible and baseline characteristics of the cohort demonstrated an OA population with more severe disease: age 66.5 (SD 7.1) vs 68.1 (7.7) years, min-JSW 2.5 (1.3) vs 2.1 (1.0) mm and Knee injury and Osteoarthritis Outcome Score pain 31.3 (19.7) vs 17.7 (14.6), except for age, all: p<0.001, for selected versus excluded patients, respectively. Based on the selection model, this cohort has a predicted higher chance of progression. FUTURE PLANS: Patients will visit the hospital again at 6, 12 and 24 months for physical examination, pain and general health questionnaires, collection of blood and urine, MRI scans, radiographs of knees and hands, CT scan of the knee, low radiation whole-body CT, HandScan, motion analysis and performance-based tests.After two years, data will show whether those patients with the highest probabilities for progression experienced disease progression as compared to those wit lower probabilities (model validation) and whether phenotypes/endotypes can be identified and predicted to facilitate targeted drug therapy. TRIAL REGISTRATION NUMBER: NCT03883568.


Assuntos
Progressão da Doença , Osteoartrite do Joelho/diagnóstico por imagem , Osteoartrite do Joelho/patologia , Idoso , Artralgia , Biomarcadores/sangue , Estudos de Coortes , Europa (Continente) , Feminino , Humanos , Articulação do Joelho/diagnóstico por imagem , Articulação do Joelho/patologia , Aprendizado de Máquina , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Osteoartrite do Joelho/sangue , Fenótipo , Estudos Prospectivos , Radiografia , Tomografia Computadorizada por Raios X
2.
Nat Commun ; 10(1): 2434, 2019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-31164647

RESUMO

To date, genome-wide association studies have implicated at least 35 loci in osteoarthritis but, due to linkage disequilibrium, the specific variants underlying these associations and the mechanisms by which they contribute to disease risk have yet to be pinpointed. Here, we functionally test 1,605 single nucleotide variants associated with osteoarthritis for regulatory activity using a massively parallel reporter assay. We identify six single nucleotide polymorphisms (SNPs) with differential regulatory activity between the major and minor alleles. We show that the most significant SNP, rs4730222, exhibits differential nuclear protein binding in electrophoretic mobility shift assays and drives increased expression of an alternative isoform of HBP1 in a heterozygote chondrosarcoma cell line, in a CRISPR-edited osteosarcoma cell line, and in chondrocytes derived from osteoarthritis patients. This study provides a framework for prioritization of GWAS variants and highlights a role of HBP1 and Wnt signaling in osteoarthritis pathogenesis.


Assuntos
Condrócitos/metabolismo , Redes Reguladoras de Genes , Proteínas de Grupo de Alta Mobilidade/genética , Osteoartrite/genética , Proteínas Repressoras/genética , Alelos , Cartilagem Articular/citologia , Linhagem Celular Tumoral , Ensaio de Desvio de Mobilidade Eletroforética , Técnicas de Introdução de Genes , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Haplótipos , Proteínas de Grupo de Alta Mobilidade/metabolismo , Humanos , Desequilíbrio de Ligação , Proteínas Nucleares/metabolismo , Polimorfismo de Nucleotídeo Único , Ligação Proteica , Isoformas de Proteínas , Proteínas Repressoras/metabolismo , Via de Sinalização Wnt
3.
Commun Biol ; 1: 56, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30273415

RESUMO

Developmental dysplasia of the hip (DDH) is the most common skeletal developmental disease. However, its genetic architecture is poorly understood. We conduct the largest DDH genome-wide association study to date and replicate our findings in independent cohorts. We find the heritable component of DDH attributable to common genetic variants to be 55% and distributed equally across the autosomal and X-chromosomes. We identify replicating evidence for association between GDF5 promoter variation and DDH (rs143384, effect allele A, odds ratio 1.44, 95% confidence interval 1.34-1.56, P = 3.55 × 10-22). Gene-based analysis implicates GDF5 (P = 9.24 × 10-12), UQCC1 (P = 1.86 × 10- 10), MMP24 (P = 3.18 × 10-9), RETSAT (P = 3.70 × 10- 8) and PDRG1 (P = 1.06 × 10- 7) in DDH susceptibility. We find shared genetic architecture between DDH and hip osteoarthritis, but no predictive power of osteoarthritis polygenic risk score on DDH status, underscoring the complex nature of the two traits. We report a scalable, time-efficient recruitment strategy and establish for the first time to our knowledge a robust DDH genetic association locus at GDF5.

4.
Hum Mol Genet ; 27(19): 3464-3474, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30010910

RESUMO

Osteoarthritis (OA) is a common, multifactorial and polygenic skeletal disease that, in its severest form, requires joint replacement surgery to restore mobility and to relieve chronic pain. Using tissues from the articulating joints of 260 patients with OA and a range of in vitro experiments, including CRISPR-Cas9, we have characterized an intergenic regulatory element. Here, genotype at an OA risk locus correlates with differential DNA methylation, with altered gene expression of both a transcriptional regulator (RUNX2), and a chromatin remodelling protein (SUPT3H). RUNX2 is a strong candidate for OA susceptibility, with its encoded protein being essential for skeletogenesis and healthy joint function. The OA risk locus includes single nucleotide polymorphisms (SNPs) located within and flanking the differentially methylated region (DMR). The OA association SNP, rs10948172, demonstrates particularly strong correlation with methylation, and two intergenic SNPs falling within the DMR (rs62435998 and rs62435999) demonstrate genetic and epigenetic effects on the regulatory activity of this region. We therefore posit that the OA signal mediates its effect by modulating the methylation of the regulatory element, which then impacts on gene expression, with RUNX2 being the principal target. Our study highlights the interplay between DNA methylation, OA genetic risk and the downstream regulation of genes critical to normal joint function.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/genética , Metilação de DNA/genética , Osteoartrite/genética , Fatores de Transcrição/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Sistemas CRISPR-Cas , Feminino , Regulação da Expressão Gênica/genética , Predisposição Genética para Doença , Genótipo , Humanos , Articulações/fisiopatologia , Masculino , Pessoa de Meia-Idade , Osteoartrite/fisiopatologia , Polimorfismo de Nucleotídeo Único , Sequências Reguladoras de Ácido Nucleico , Fatores de Risco
5.
Arthritis Rheumatol ; 70(10): 1577-1587, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29732726

RESUMO

OBJECTIVE: To identify the functional single-nucleotide polymorphisms (SNPs) and mechanisms conferring increased risk of hand osteoarthritis (OA) at the ALDH1A2 locus, which is a retinoic acid regulatory gene. METHODS: Tissue samples from 247 patients with knee, hip, or hand OA who had undergone joint surgery were included. RNA-sequencing analysis was used to investigate differential expression of ALDH1A2 and other retinoic acid signaling pathway genes in cartilage. Expression of ALDH1A2 in joint tissues obtained from multiple sites was quantified using quantitative reverse transcription-polymerase chain reaction. Allelic expression imbalance (AEI) was measured by pyrosequencing. The consequences of ALDH1A2 depletion by RNA interference were assessed in primary human chondrocytes. In silico and in vitro analyses were used to pinpoint which, among 62 highly correlated SNPs, could account for the association at the locus. RESULTS: ALDH1A2 expression was observed across multiple joint tissue samples, including osteochondral tissue from the hand. The expression of ALDH1A2 and of several retinoic acid signaling genes was different in diseased cartilage compared to non-diseased cartilage, with ALDH1A2 showing lower levels in OA cartilage. Experimental depletion of ALDH1A2 resulted in changes in the expression levels of a number of chondrogenic markers, including SOX9. In addition, reduced expression of the OA risk-conferring allele was witnessed in a number of joint tissues, with the strongest effect in cartilage. The intronic SNP rs12915901 recapitulated the AEI observed in patient tissues, while the Ets transcription factors were identified as potential mediators of this effect. CONCLUSION: The ALDH1A2 locus seems to increase the risk of hand OA through decreased expression of ALDH1A2 in joint tissues, with the effect dependent on rs12915901. These findings indicate a mechanism that may now be targeted to modulate OA risk.


Assuntos
Osteoartrite/genética , Polimorfismo de Nucleotídeo Único/genética , Retinal Desidrogenase/metabolismo , Família Aldeído Desidrogenase 1 , Alelos , Cartilagem Articular/metabolismo , Condrócitos/metabolismo , Feminino , Expressão Gênica , Predisposição Genética para Doença , Mãos/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Osteoartrite/patologia , Fatores de Risco , Transdução de Sinais/genética
6.
Eur J Obstet Gynecol Reprod Biol ; 222: 7-12, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29328940

RESUMO

OBJECTIVES: Screening and diagnosis of gestational diabetes (GDM) has been a source of controversy. The prevalence has increased in line with an obesity epidemic and a trend towards delayed child-bearing. Treatment of even modest glycaemic impairment in pregnancy has been shown to be beneficial in preventing its clinical sequalae. However the cumbersome nature and timing of the oral glucose tolerance test coupled with debate around universal versus risk factor based screening have been problematic. This group aimed to investigate a panel of biomarkers which have shown promise in the literature to predict GDM from the first trimester in a group of high risk women. METHODS: Serum samples were drawn on 248 women deemed at risk of GDM before 15 weeks' gestation to measure C-reactive protein, sex hormone binding globulin, adiponectin and 1,5 anhydroglucitol. Patients underwent an oral glucose tolerance test as per IADPSG criteria at 28 weeks' gestation. Multiple logistic regression was used to examine the link between incidence of GDM and early pregnancy serum biomarkers. RESULTS: Adiponectin levels in the first trimester are independently linked to the risk of GDM. Serum adiponectin <8.9 µg/ml gives an odds ratio of 3.3 for GDM.Mean 1,5 AG levels are significantly lower in those that go on to develop GDM. SHBG levels measured in the first trimester were linked to the risk of GDM. However, this was no longer statistically significant once BMI, ethnicity and family history were taken into consideration. First trimester measurement of CRP is not a useful indicator of GDM risk. CONCLUSIONS: First trimester measurement of Adiponectin and 1,5 Anhydroglucitol are potential early biomarkers for the later onset of GDM. Risk stratification using these biomarkers may facilitate early diagnosis and management of GDM to mitigate against its complications.


Assuntos
Adiponectina/sangue , Desoxiglucose/sangue , Diabetes Gestacional/sangue , Regulação para Baixo , Testes para Triagem do Soro Materno , Adulto , Biomarcadores/sangue , Proteína C-Reativa/análise , Estudos de Coortes , Diabetes Gestacional/diagnóstico , Diabetes Gestacional/epidemiologia , Diagnóstico Precoce , Feminino , Teste de Tolerância a Glucose , Humanos , Incidência , Irlanda/epidemiologia , Gravidez , Trimestres da Gravidez , Fatores de Risco , Sensibilidade e Especificidade , Globulina de Ligação a Hormônio Sexual/análise
7.
PLoS One ; 12(5): e0176523, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28481944

RESUMO

Osteoarthritis (OA) is a common joint disease characterised by the focal loss of the protective cartilage layer at the ends of the bones. It is painful, disabling, multifactorial and polygenic. The growth differentiation factor 5 gene GDF5 was one of the first reported OA susceptibility signals that showed consistent association to OA, with the transcript single nucleotide polymorphism (SNP) rs143383 demonstrating association in Asians and Europeans. The functional effect of the signal is reduced expression of the gene. The GDF5 protein is an extracellular matrix signalling molecule that is active during chondrogenesis and in mature chondrocytes. Due to the functional impact of the susceptibility, we previously assessed the effect of supplementing chondrocytes from OA patients with exogenous GDF5. Their response was highly discordant, precluding the application of GDF5 as a simple means of attenuating the genetic deficit. Since GDF5 is also active during development, we have now assessed the effect of exogenous GDF5 on bone marrow derived mesenchymal stem cells (MSCs) that are undergoing chondrogenesis during cartilage disc formation. MSCs from healthy donors and OA patients were studied and the effect of GDF5 was assessed by measuring the wet mass of the discs, by histological staining, and by monitoring the change in expression of anabolic, catabolic and hypertrophic protein-coding genes. The MSCs expressed the three principal GDF5 receptor genes and responded in a significantly anabolic manner (increase in wet mass, p = 0.0022; Bonferroni corrected p = 0.018) to a variant form of GDF5 that targets the most abundantly expressed receptor, BMPR-IA. GDF5 elicited significant (p < 0.05) changes in the expression of anabolic, catabolic and hypertrophic genes with several consistent effects in healthy donors and in OA patients. Our data implies that, unlike OA chondrocytes, OA MSCs do respond in a predictable, anabolic manner to GDF5, which could therefore provide a route to modulate the genetic deficit mediated by the rs143383 association signal.


Assuntos
Condrogênese , Predisposição Genética para Doença , Fator 5 de Diferenciação de Crescimento/genética , Células-Tronco Mesenquimais/patologia , Osteoartrite/patologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Osteoartrite/genética , Adulto Jovem
8.
Stem Cell Res Ther ; 7(1): 133, 2016 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-27624072

RESUMO

BACKGROUND: Mesenchymal stem cells (MSCs) have prospective applications in regenerative medicine and tissue engineering but to what extent phenotype and differentiation capacity alter with ageing is uncertain. Consequently, any loss in functionality with age would have profound consequences for the maintenance of tissue viability and the quality of tissues. Proteomics enables the set of proteins responsible for a particular cell phenotype to be identified, as well as enabling insights into mechanisms responsible for age-related alterations in musculoskeletal tissues. Few proteomic studies have been undertaken regarding age-related effects on tissue engineered into cartilage and bone, and none for tendon. This study provides a proteome inventory for chondrogenic, osteogenic and tenogenic constructs synthesised from human MSCs, and elucidates proteomic alterations as a consequence of donor age. METHODS: Human bone-marrow derived MSCs from young (n = 4, 21.8 years ± 2.4SD) and old (n = 4, 65.5 years ± 8.3SD) donors were used to make chondrogenic, osteogenic and tenogenic tissue-engineered constructs. We utilised an analytical method relying on extracted peptide intensities as a label-free approach for peptide quantitation by liquid chromatography-mass spectrometry. Results were validated using western blotting. RESULTS: We identified proteins that were differentially expressed with ageing; 128 proteins in chondrogenic constructs, 207 in tenogenic constructs and four in osteogenic constructs. Differentially regulated proteins were subjected to bioinformatic analysis to ascertain their molecular functions and the signalling pathways. For all construct types, age-affected proteins were involved in altered cell survival and death, and antioxidant and cytoskeletal changes. Energy and protein metabolism were the principle pathways affected in tenogenic constructs, whereas lipid metabolism was strongly affected in chondrogenic constructs and mitochondrial dysfunction in osteogenic constructs. CONCLUSIONS: Our results imply that further work on MSC-based therapeutics for the older population needs to focus on oxidative stress protection. The differentially regulated proteome characterised by this study can potentially guide translational research specifically aimed at effective clinical interventions.


Assuntos
Envelhecimento/metabolismo , Células da Medula Óssea/metabolismo , Condrogênese/fisiologia , Células-Tronco Mesenquimais/metabolismo , Osteogênese/fisiologia , Tendões/metabolismo , Adulto , Idoso , Envelhecimento/fisiologia , Células da Medula Óssea/fisiologia , Osso e Ossos/metabolismo , Osso e Ossos/fisiologia , Cartilagem/metabolismo , Cartilagem/fisiologia , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Condrócitos/metabolismo , Condrócitos/fisiologia , Humanos , Células-Tronco Mesenquimais/fisiologia , Estudos Prospectivos , Proteômica/métodos , Medicina Regenerativa/métodos , Tendões/fisiologia , Engenharia Tecidual/métodos , Adulto Jovem
9.
PLoS One ; 11(8): e0160517, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27533049

RESUMO

Mesenchymal stem cells (MSC) are capable of multipotent differentiation into connective tissues and as such are an attractive source for autologous cell-based regenerative medicine and tissue engineering. Epigenetic mechanisms, like DNA methylation, contribute to the changes in gene expression in ageing. However there was a lack of sufficient knowledge of the role that differential methylation plays during chondrogenic, osteogenic and tenogenic differentiation from ageing MSCs. This study undertook genome level determination of the effects of DNA methylation on expression in engineered tissues from chronologically aged MSCs. We compiled unique DNA methylation signatures from chondrogenic, osteogenic, and tenogenic engineered tissues derived from young; n = 4 (21.8 years ± 2.4 SD) and old; n = 4 (65.5 years±8.3SD) human MSCs donors using the Illumina HumanMethylation 450 Beadchip arrays and compared these to gene expression by RNA sequencing. Unique and common signatures of global DNA methylation were identified. There were 201, 67 and 32 chondrogenic, osteogenic and tenogenic age-related DE protein-coding genes respectively. Findings inferred the nature of the transcript networks was predominantly for 'cell death and survival', 'cell morphology', and 'cell growth and proliferation'. Further studies are required to validate if this gene expression effect translates to cell events. Alternative splicing (AS) was dysregulated in ageing with 119, 21 and 9 differential splicing events identified in chondrogenic, osteogenic and tenogenic respectively, and enrichment in genes associated principally with metabolic processes. Gene ontology analysis of differentially methylated loci indicated age-related enrichment for all engineered tissue types in 'skeletal system morphogenesis', 'regulation of cell proliferation' and 'regulation of transcription' suggesting that dynamic epigenetic modifications may occur in genes associated with shared and distinct pathways dependent upon engineered tissue type. An altered phenotype in engineered tissues was observed with ageing at numerous levels. These changes represent novel insights into the ageing process, with implications for stem cell therapies in older patients. In addition we have identified a number of tissue-dependant pathways, which warrant further studies.


Assuntos
Envelhecimento/genética , Metilação de DNA , Sistema Musculoesquelético/metabolismo , Idoso , Envelhecimento/patologia , Processamento Alternativo , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Condrogênese/genética , Epigênese Genética , Redes Reguladoras de Genes , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Pessoa de Meia-Idade , Sistema Musculoesquelético/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Osteogênese/genética , Medicina Regenerativa , Análise de Sequência de RNA , Tendões/citologia , Tendões/metabolismo , Engenharia Tecidual/métodos , Adulto Jovem
10.
PLoS One ; 11(7): e0159024, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27391021

RESUMO

The single nucleotide polymorphism (SNP) rs835487 is associated with hip osteoarthritis (OA) at the genome-wide significance level and is located within CHST11, which codes for carbohydrate sulfotransferase 11. This enzyme post-translationally modifies proteoglycan prior to its deposition in the cartilage extracellular matrix. Using bioinformatics and experimental analyses, our aims were to characterise the rs835487 association signal and to identify the causal functional variant/s. Database searches revealed that rs835487 resides within a linkage disequilibrium (LD) block of only 2.7 kb and is in LD (r2 ≥ 0.8) with six other SNPs. These are all located within intron 2 of CHST11, in a region that has predicted enhancer activity and which shows a high degree of conservation in primates. Luciferase reporter assays revealed that of the seven SNPs, rs835487 and rs835488, which have a pairwise r2 of 0.962, are the top functional candidates; the haplotype composed of the OA-risk conferring G allele of rs835487 and the corresponding T allele of rs835488 (the G-T haplotype) demonstrated significantly different enhancer activity relative to the haplotype composed of the non-risk A allele of rs835487 and the corresponding C allele of rs835488 (the A-C haplotype) (p < 0.001). Electrophoretic mobility shift assays and supershifts identified several transcription factors that bind more strongly to the risk-conferring G and T alleles of the two SNPs, including SP1, SP3, YY1 and SUB1. CHST11 was found to be upregulated in OA versus non-OA cartilage (p < 0.001) and was expressed dynamically during chondrogenesis. Its expression in adult cartilage did not however correlate with rs835487 genotype. Our data demonstrate that the OA susceptibility is mediated by differential protein binding to the alleles of rs835487 and rs835488, which are located within an enhancer whose target may be CHST11 during chondrogenesis or an alternative gene.


Assuntos
Condrogênese/genética , Predisposição Genética para Doença , Íntrons , Osteoartrite do Quadril/genética , Polimorfismo de Nucleotídeo Único , Sulfotransferases/genética , Alelos , Animais , Cartilagem/enzimologia , Cartilagem/patologia , Linhagem Celular Tumoral , Biologia Computacional , Bases de Dados de Ácidos Nucleicos , Estudo de Associação Genômica Ampla , Haplótipos , Humanos , Osteoartrite do Quadril/enzimologia , Osteoartrite do Quadril/patologia , Sulfotransferases/metabolismo
11.
Arthritis Rheumatol ; 67(11): 2923-32, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26211391

RESUMO

OBJECTIVE: To functionally characterize the osteoarthritis (OA) susceptibility variants that map to a region of high linkage disequilibrium (LD) on chromosome 20q13 marked by the single-nucleotide polymorphism (SNP) rs6094710 and encompassing NCOA3 and SULF2. METHODS: Nucleic acids were extracted from the cartilage of OA patients. Overall and allelic expression of NCOA3 and SULF2 were measured by quantitative reverse transcription-polymerase chain reaction and pyrosequencing, respectively. The functional effect of SNPs within the 20q13 locus was assessed in vitro using luciferase reporter constructs and electrophoretic mobility shift assays (EMSAs). The in vivo effect of nuclear receptor coactivator 3 (NCOA3) protein depletion on primary human OA articular cartilage chondrocytes was assessed using RNA interference. RESULTS: Expression of NCOA3 correlated with the genotype at rs6094710 (P = 0.006), and the gene demonstrated allelic expression imbalance (AEI) in individuals heterozygous for the SNP (mean AEI 1.21; P < 0.0001). In both instances, expression of the OA-associated allele was reduced. In addition, there was reduced enhancer activity of the OA-associated allele of rs116855380, a SNP in perfect LD with rs6094710 in luciferase assays (P < 0.001). EMSAs demonstrated a protein complex binding with reduced affinity to this allele. Depletion of NCOA3 led to significant changes (all P < 0.05) in the expression of genes involved in cartilage homeostasis. CONCLUSION: NCOA3 is subject to a cis-acting expression quantitative trait locus in articular cartilage, which correlates with the OA association signal and with the OA-associated allele of the functional SNP rs116855380, a SNP that is located only 10.3 kb upstream of NCOA3. These findings elucidate the effect of the association of the 20q13 region on OA cartilage and provide compelling evidence of a potentially causal candidate SNP.


Assuntos
Cromossomos Humanos Par 20 , Predisposição Genética para Doença , Coativador 3 de Receptor Nuclear/genética , Osteoartrite/genética , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas , Alelos , Cartilagem Articular/metabolismo , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Regulação da Expressão Gênica , Genótipo , Humanos , Metaloproteinase 13 da Matriz/genética , Metaloproteinase 13 da Matriz/metabolismo , Coativador 3 de Receptor Nuclear/metabolismo , Osteoartrite/metabolismo , Transcrição Gênica
12.
Ann Rheum Dis ; 74(9): 1778-82, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25854584

RESUMO

OBJECTIVES: We have previously shown that the cartilage DNA methylome delineates two clusters of osteoarthritic (OA) hip patients, characterised by differential methylation of inflammatory genes, while others have demonstrated a link between zinc homeostasis and inflammation in OA. We aimed to investigate these effects at the methylation and gene expression level. METHODS: We used our previously generated methylation data while quantitative PCR was used to measure gene expression using RNA from the hip cartilage of members of both clusters and from control individuals without hip OA. RESULTS: One of the OA clusters is characterised by the promoter hypomethylation and increased expression of inflammation-associated genes including IL1A and TNF. Furthermore, we show that the increase in expression of these genes is accompanied by increased expression of several zinc transporter genes. In addition, the zinc responsive transcription factor MTF1 is also upregulated, which is accompanied by an increase in the expression of its targets the metalloproteinases MMP13 and ADAMTS5. CONCLUSIONS: We have identified a subgroup of OA hip patients that are epigenetically and transcriptiomically characterised by a cartilage inflammatory phenotype with concurrent differential regulation of zinc regulators. The identification of subgroups enhances stratified phenotyping of OA patients and has important implications for future therapeutic applications.


Assuntos
Cartilagem Articular/metabolismo , Metilação de DNA , Osteoartrite do Quadril/genética , RNA Mensageiro/metabolismo , Proteínas ADAM/genética , Proteína ADAMTS5 , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Proteínas de Ligação a DNA/genética , Epigênese Genética , Feminino , Regulação da Expressão Gênica , Humanos , Interleucina-1alfa/genética , Masculino , Metaloproteinase 13 da Matriz/genética , Pessoa de Meia-Idade , Fatores de Transcrição/genética , Fator de Necrose Tumoral alfa/genética , Regulação para Cima , Fator MTF-1 de Transcrição
13.
BMC Med Genet ; 15: 53, 2014 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-24886551

RESUMO

BACKGROUND: An osteoarthritis (OA) susceptibility locus has been mapped to chromosome 3p21, to a region of high linkage disequilibrium encompassing twelve genes. Six of these genes are expressed in joint tissues and we therefore assessed whether any of the six were subject to cis-acting regulatory polymorphisms active in these tissues and which could therefore account for the association signal. METHODS: We measured allelic expression using pyrosequencing assays that can distinguish mRNA output from each allele of a transcript single nucleotide polymorphism. We assessed RNA extracted from the cartilage and other joint tissues of OA patients who had undergone elective joint replacement surgery. A two-tailed Mann-Whitney exact test was used to test the significance of any allelic differences. RESULTS: GNL3 and SPCS1 demonstrated significant allelic expression imbalance (AEI) in OA cartilage (GNL3, mean AEI = 1.04, p = 0.0002; SPCS1, mean AEI = 1.07, p < 0.0001). Similar results were observed in other tissues. Expression of the OA-associated allele was lower than that of the non-associated allele for both genes. CONCLUSIONS: cis-acting regulatory polymorphisms acting on GNL3 and SPCS1 contribute to the OA association signal at chromosome 3p21, and these genes therefore merit further investigation.


Assuntos
Alelos , Cromossomos Humanos Par 3 , Proteínas de Ligação ao GTP/genética , Expressão Gênica , Predisposição Genética para Doença , Proteínas de Membrana/genética , Proteínas Nucleares/genética , Osteoartrite/genética , Locos de Características Quantitativas , Idoso , Idoso de 80 Anos ou mais , Desequilíbrio Alélico , Cartilagem/metabolismo , Cartilagem/patologia , Proteínas de Ciclo Celular , Proteínas do Citoesqueleto , Proteínas de Ligação a DNA , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Proteínas/genética , Fatores de Transcrição/genética
14.
Hum Genet ; 133(8): 1059-73, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24861163

RESUMO

GDF5 encodes an extracellular signalling molecule that is essential for normal skeletal development. The rs144383 C to T SNP located in the 5'UTR of this gene is functional and has a pleiotropic effect on the musculoskeletal system, being a risk factor for knee-osteoarthritis (OA), congenital hip dysplasia, lumbar disc degeneration and Achilles tendon pathology. rs143383 exerts a joint-wide effect on GDF5 expression, with expression of the OA-associated T allele being significantly reduced relative to the C allele, termed allelic expression imbalance. We have previously reported that the GDF5 locus is subject to DNA methylation and that allelic imbalance of rs143383 is mediated by SP1, SP3 and DEAF1 transcriptional repressors. In this study, we have assayed GDF5 methylation in normal and osteoarthritic cartilage, and investigated the effect of methylation on the allelic imbalance of rs143383. We observed demethylation of the GDF5 5'UTR in OA knee cartilage relative to both OA (p = 0.009) and non-OA (p = 0.001) hip cartilage, with the most significant demethylation observed at the highly conserved +37 CpG site located 4 bp upstream of rs143383. Methylation modulates the level and direction of allelic imbalance of rs143383, with methylation of the +37 CpG dinucleotide within the SP1/SP3 binding site having an allele-specific effect on SP1 and SP3 binding. Furthermore, methylation attenuated the repressive effects of SP1, SP3 and DEAF1 on GDF5 promoter activity. This data suggest that the differential methylation of the +37 CpG site between osteoarthritic hip and knee cartilage may be responsible for the knee-specific effect of rs143383 on OA susceptibility.


Assuntos
Ilhas de CpG/genética , Metilação de DNA , Fator 5 de Diferenciação de Crescimento/genética , Osteoartrite/genética , Polimorfismo de Nucleotídeo Único/genética , Fator de Transcrição Sp1/metabolismo , Fator de Transcrição Sp3/metabolismo , Alelos , Cartilagem Articular/citologia , Cartilagem Articular/metabolismo , Estudos de Casos e Controles , Células Cultivadas , Ensaio de Desvio de Mobilidade Eletroforética , Predisposição Genética para Doença , Fator 5 de Diferenciação de Crescimento/metabolismo , Humanos , Osteoartrite/metabolismo , Osteoartrite/patologia , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Arthritis Rheumatol ; 66(4): 940-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24757145

RESUMO

OBJECTIVE: To assess candidate genes for association with osteoarthritis (OA) and identify promising genetic factors and, secondarily, to assess the candidate gene approach in OA. METHODS: A total of 199 candidate genes for association with OA were identified using Human Genome Epidemiology (HuGE) Navigator. All of their single-nucleotide polymorphisms (SNPs) with an allele frequency of >5% were assessed by fixed-effects meta-analysis of 9 genome-wide association studies (GWAS) that included 5,636 patients with knee OA and 16,972 control subjects and 4,349 patients with hip OA and 17,836 control subjects of European ancestry. An additional 5,921 individuals were genotyped for significantly associated SNPs in the meta-analysis. After correction for the number of independent tests, P values less than 1.58 × 10(-5) were considered significant. RESULTS: SNPs at only 2 of the 199 candidate genes (COL11A1 and VEGF) were associated with OA in the meta-analysis. Two SNPs in COL11A1 showed association with hip OA in the combined analysis: rs4907986 (P = 1.29 × 10(-5) , odds ratio [OR] 1.12, 95% confidence interval [95% CI] 1.06-1.17) and rs1241164 (P = 1.47 × 10(-5) , OR 0.82, 95% CI 0.74-0.89). The sex-stratified analysis also showed association of COL11A1 SNP rs4908291 in women (P = 1.29 × 10(-5) , OR 0.87, 95% CI 0.82-0.92); this SNP showed linkage disequilibrium with rs4907986. A single SNP of VEGF, rs833058, showed association with hip OA in men (P = 1.35 × 10(-5) , OR 0.85, 95% CI 0.79-0.91). After additional samples were genotyped, association at one of the COL11A1 signals was reinforced, whereas association at VEGF was slightly weakened. CONCLUSION: Two candidate genes, COL11A1 and VEGF, were significantly associated with OA in this focused meta-analysis. The remaining candidate genes were not associated.


Assuntos
Predisposição Genética para Doença , Osteoartrite do Quadril/genética , Osteoartrite do Joelho/genética , Polimorfismo de Nucleotídeo Único , Colágeno Tipo XI/genética , Frequência do Gene , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Fator A de Crescimento do Endotélio Vascular/genética
16.
Eur J Hum Genet ; 21(5): 517-21, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-22929025

RESUMO

Osteoarthritis (OA) is a polygenic disease characterized by cartilage loss, with the single-nucleotide polymorphism (SNP) rs143383 (C/T) influencing OA susceptibility across a range of ethnic groups. The SNP resides within the 5'-UTR of the growth and differentiation factor 5 gene (GDF5), with the OA-associated T-allele mediating reduced GDF5 expression. As GDF5 codes for a cartilage anabolic protein, this reduced expression may explain why the T-allele of rs143383 is an OA risk factor. Our deep sequencing of GDF5 identified a C/A transversion located -41 bp relative to the gene's transcription start site. This promoter variant is predicted to affect transcription factor binding and it may therefore highlight a regulatory site that could be exploited to manipulate GDF5 expression and alleviate the detrimental effect mediated by the T-allele of rs143383. Here, we describe our functional assessment of the -41 bp variant. Using reporter constructs we demonstrated that the transversion leads to increased gene expression to such a degree that the A-allele is able to compensate for the reduced expression mediated by the T-allele of rs143383. Using electrophoretic mobility shift assays we identified YY1 as a trans-acting factor that differentially binds to the alleles of the -41 bp variant, with more avid binding to allele A. Knockdown of YY1 led to a significant reduction in GDF5 expression, supporting YY1 as a GDF5 activator. In conclusion, we demonstrated that the -41 bp variant is functional and we have identified a regulatory region of GDF5 that can be exploited to overcome the OA genetic deficit mediated by the T-allele of rs143383.


Assuntos
Regulação da Expressão Gênica/genética , Fator 5 de Diferenciação de Crescimento/genética , Osteoartrite/genética , Polimorfismo de Nucleotídeo Único/genética , Linhagem Celular Tumoral , Condrócitos/metabolismo , Primers do DNA/genética , Ensaio de Desvio de Mobilidade Eletroforética , Técnicas de Silenciamento de Genes , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Luciferases , Plasmídeos/genética , Regiões Promotoras Genéticas/genética , Fator de Transcrição YY1/genética , Fator de Transcrição YY1/metabolismo
17.
Mov Disord ; 27(6): 789-93, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22508347

RESUMO

BACKGROUND: Neurodegeneration with brain iron accumulation is clinically and genetically heterogeneous because of mutations in at least 7 nuclear genes. METHODS: We performed homozygosity mapping and whole-exome sequencing in 2 brothers with brain iron accumulation from a consanguineous family. RESULTS: We identified a homozygous missense mutation in both brothers in the very recently identified chromosome 19 open-reading frame 12 gene. The disease presented before age 10 with slowly progressive tremor, dystonia, and spasticity. Additional features were optic atrophy, peripheral neuropathy, and learning difficulties. A raised serum creatine kinase indicated neuromuscular involvement, and compensatory mitochondrial proliferation implicated mitochondrial dysfunction as a pathological mechanism. CONCLUSIONS: Further studies are needed to explore the function of the chromosome 19 open-reading frame 12 gene, and extended genetic analysis on larger patient cohorts will provide more information about the presentation and frequency of this disease.


Assuntos
Encéfalo/metabolismo , Distúrbios Distônicos/genética , Ferro/metabolismo , Degeneração Neural/genética , Atrofia Óptica/genética , Doenças do Sistema Nervoso Periférico/genética , Adolescente , Encéfalo/patologia , Criança , Consanguinidade , Distúrbios Distônicos/metabolismo , Distúrbios Distônicos/patologia , Humanos , Masculino , Mutação de Sentido Incorreto , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Atrofia Óptica/metabolismo , Atrofia Óptica/patologia , Linhagem , Doenças do Sistema Nervoso Periférico/metabolismo , Doenças do Sistema Nervoso Periférico/patologia , Síndrome
18.
BMC Med Genet ; 13: 12, 2012 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-22385522

RESUMO

BACKGROUND: A genome-wide association scan with subsequent replication study that involved over 67,000 individuals of European ancestry has produced evidence of association of single nucleotide polymorphism rs2277831 to primary osteoarthritis (OA) with a P-value of 2.9 × 10(-5). rs2277831, an A/G transition, is located in an intron of MICAL3. This gene is located on chromosome 22q11.21 and the association signal encompasses two additional genes, BCL2L13 and BID. It is becoming increasingly apparent that many common complex traits are mediated by cis-acting regulatory polymorphisms that influence, in a tissue-specific manner, gene expression or transcript stability. METHODS: We used total and allelic expression analysis to assess whether the OA association to rs2277831 is mediated by an influence on MICAL3, BCL2L13 or BID expression. Using RNA extracted from joint tissues of 60 patients who had undergone elective joint replacement surgery, we assessed whether rs2277831 correlated with allelic expression of either of the three genes by: 1) measuring the expression of each gene by quantitative PCR and then stratifying the data by genotype at rs2277831 and 2) accurately discriminating and quantifying the mRNA synthesised from the alleles of OA patients using allelic-quantitative PCR. RESULTS: We found no evidence for a correlation between gene expression and genotype at rs2277831, with P-values of 0.09 for BCL2L13, 0.07 for BID and 0.33 for MICAL3. In the allelic expression analysis we observed several examples of significant (p < 0.05) allelic imbalances, with an allelic expression ratio of 2.82 observed in BCL2L13 (P = 0.004), 2.09 at BID (P = 0.001) and the most extreme case being at MICAL3, with an allelic expression ratio of 5.47 (P = 0.001). However, there was no correlation observed between the pattern of allelic expression and the genotype at rs2277831. CONCLUSIONS: In the tissues that we have studied, our data do not support our hypothesis that the association between rs2277831 and OA is due to the effect this SNP has on MICAL3, BCL2L13 or BID gene expression. Instead, our data point towards other functional effects accounting for the OA associated signal.


Assuntos
Mapeamento Cromossômico , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Oxigenases de Função Mista/genética , Osteoartrite/genética , Alelos , Perfilação da Expressão Gênica , Humanos , Íntrons , Polimorfismo de Nucleotídeo Único , População Branca/genética
19.
Blood ; 118(10): 2656-8, 2011 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-21765025

RESUMO

The human syndrome of dendritic cell, monocyte, B and natural killer lymphoid deficiency presents as a sporadic or autosomal dominant trait causing susceptibility to mycobacterial and other infections, predisposition to myelodysplasia and leukemia, and, in some cases, pulmonary alveolar proteinosis. Seeking a genetic cause, we sequenced the exomes of 4 unrelated persons, 3 with sporadic disease, looking for novel, heterozygous, and probably deleterious variants. A number of genes harbored novel variants in person, but only one gene, GATA2, was mutated in all 4 persons. Each person harbored a different mutation, but all were predicted to be highly deleterious and to cause loss or mutation of the C-terminal zinc finger domain. Because GATA2 is the only common mutated gene in 4 unrelated persons, it is highly probable to be the cause of dendritic cell, monocyte, B, and natural killer lymphoid deficiency. This disorder therefore constitutes a new genetic form of heritable immunodeficiency and leukemic transformation.


Assuntos
Linfócitos B/patologia , Células Dendríticas/patologia , Suscetibilidade a Doenças/etiologia , Éxons/genética , Fator de Transcrição GATA2/genética , Células Matadoras Naturais/patologia , Tecido Linfoide/patologia , Monócitos/patologia , Mutação/genética , Fator de Transcrição GATA2/química , Humanos , Conformação Proteica
20.
Hum Mol Genet ; 20(17): 3450-60, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21642387

RESUMO

GDF5 is involved in synovial joint development, maintenance and repair, and the rs143383 C/T single nucleotide polymorphism (SNP) located in the 5'UTR of GDF5 is associated, at the genome-wide significance level, with osteoarthritis susceptibility, and with other musculoskeletal phenotypes including height, congenital hip dysplasia and Achilles tendinopathy. There is a significant reduction in the expression of the disease-associated T allele relative to the C allele in synovial joint tissues, an effect influenced by a second SNP (rs143384, C/T) also within the 5'UTR. The differential allelic expression (DAE) imbalance of the C and T alleles of rs143383 varies intra- and inter-individually, suggesting that DAE may be modulated epigenetically. The C alleles of both SNPs form CpG dinucleotides that are potentially amenable to regulation by methylation. Here, we have examined whether DNA methylation regulates GDF5 expression and the allelic imbalance caused by rs143383. We observed methylation of the GDF5 promoter and 5'UTR in cell lines and joint tissues, with demethylation correlating with increased GDF5 expression. The CpG sites created by the C alleles at rs143383 and rs143384 were variably methylated, and treatment of a heterozygous cell line with a demethylating agent further increased the allelic expression imbalance between the C and T alleles. This demonstrates that the genetic effect of the rs143383 SNP on GDF5 expression is modulated epigenetically by DNA methylation. The variability in DAE of rs143383 is therefore partly accounted for by differences in DNA methylation that could influence the penetrance of this allele in susceptibility to common musculoskeletal diseases.


Assuntos
Metilação de DNA/genética , Fator 5 de Diferenciação de Crescimento/genética , Osteoartrite/genética , Regiões 5' não Traduzidas/genética , Linhagem Celular , Linhagem Celular Tumoral , Predisposição Genética para Doença/genética , Humanos , Polimorfismo de Nucleotídeo Único/genética , Reação em Cadeia da Polimerase em Tempo Real
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA