Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
PLoS Genet ; 17(10): e1009848, 2021 10.
Article in English | MEDLINE | ID: mdl-34662339

ABSTRACT

Patients with inherited retinal dystrophies (IRDs) were recruited from two understudied populations: Mexico and Pakistan as well as a third well-studied population of European Americans to define the genetic architecture of IRD by performing whole-genome sequencing (WGS). Whole-genome analysis was performed on 409 individuals from 108 unrelated pedigrees with IRDs. All patients underwent an ophthalmic evaluation to establish the retinal phenotype. Although the 108 pedigrees in this study had previously been examined for mutations in known IRD genes using a wide range of methodologies including targeted gene(s) or mutation(s) screening, linkage analysis and exome sequencing, the gene mutations responsible for IRD in these 108 pedigrees were not determined. WGS was performed on these pedigrees using Illumina X10 at a minimum of 30X depth. The sequence reads were mapped against hg19 followed by variant calling using GATK. The genome variants were annotated using SnpEff, PolyPhen2, and CADD score; the structural variants (SVs) were called using GenomeSTRiP and LUMPY. We identified potential causative sequence alterations in 61 pedigrees (57%), including 39 novel and 54 reported variants in IRD genes. For 57 of these pedigrees the observed genotype was consistent with the initial clinical diagnosis, the remaining 4 had the clinical diagnosis reclassified based on our findings. In seven pedigrees (12%) we observed atypical causal variants, i.e. unexpected genotype(s), including 4 pedigrees with causal variants in more than one IRD gene within all affected family members, one pedigree with intrafamilial genetic heterogeneity (different affected family members carrying causal variants in different IRD genes), one pedigree carrying a dominant causative variant present in pseudo-recessive form due to consanguinity and one pedigree with a de-novo variant in the affected family member. Combined atypical and large structural variants contributed to about 20% of cases. Among the novel mutations, 75% were detected in Mexican and 50% found in European American pedigrees and have not been reported in any other population while only 20% were detected in Pakistani pedigrees and were not previously reported. The remaining novel IRD causative variants were listed in gnomAD but were found to be very rare and population specific. Mutations in known IRD associated genes contributed to pathology in 63% Mexican, 60% Pakistani and 45% European American pedigrees analyzed. Overall, contribution of known IRD gene variants to disease pathology in these three populations was similar to that observed in other populations worldwide. This study revealed a spectrum of mutations contributing to IRD in three populations, identified a large proportion of novel potentially causative variants that are specific to the corresponding population or not reported in gnomAD and shed light on the genetic architecture of IRD in these diverse global populations.


Subject(s)
Ethnicity/genetics , Retinal Degeneration/genetics , Consanguinity , DNA Mutational Analysis/methods , Exome/genetics , Eye Proteins/genetics , Female , Genetic Association Studies/methods , Genetic Linkage/genetics , Genotype , Humans , Male , Mexico , Mutation/genetics , Pakistan , Pedigree , Retina/pathology , Exome Sequencing/methods , Whole Genome Sequencing/methods
2.
Sci Transl Med ; 12(535)2020 03 18.
Article in English | MEDLINE | ID: mdl-32188720

ABSTRACT

Danon disease (DD) is a rare X-linked autophagic vacuolar myopathy associated with multiorgan dysfunction, including the heart, skeletal muscle, and liver. There are no specific treatments, and most male patients die from advanced heart failure during the second or third decade of life. DD is caused by mutations in the lysosomal-associated membrane protein 2 (LAMP2) gene, a key mediator of autophagy. LAMP2 has three isoforms: LAMP2A, LAMP2B, and LAMP2C. LAMP2B is the predominant isoform expressed in cardiomyocytes. This study evaluates the efficacy of human LAMP2B gene transfer using a recombinant adeno-associated virus 9 carrying human LAMP2B (AAV9.LAMP2B) in a Lamp2 knockout (KO) mouse, a DD model. AAV9.LAMP2B was intravenously injected into 2- and 6-month-old Lamp2 KO male mice to assess efficacy in adolescent and adult phenotypes. Lamp2 KO mice receiving AAV9.LAMP2B demonstrated dose-dependent restoration of human LAMP2B protein in the heart, liver, and skeletal muscle tissue. Impaired autophagic flux, evidenced by increased LC3-II, was abrogated by LAMP2B gene transfer in all tissues in both cohorts. Cardiac function was also improved, and transaminases were reduced in AAV9.LAMP2B-treated KO mice, indicating favorable effects on the heart and liver. Survival was also higher in the older cohort receiving high vector doses. No anti-LAMP2 antibodies were detected in mice that received AAV9.LAMP2B. In summary, LAMP2B gene transfer improves metabolic and physiologic function in a DD murine model, suggesting that a similar therapeutic approach may be effective for treating patients with this highly morbid disease.


Subject(s)
Glycogen Storage Disease Type IIb , Adolescent , Animals , Disease Models, Animal , Glycogen Storage Disease Type IIb/genetics , Humans , Lysosomal-Associated Membrane Protein 2/genetics , Male , Mice , Mice, Knockout , Phenotype
3.
Aging Cell ; 18(6): e13011, 2019 12.
Article in English | MEDLINE | ID: mdl-31385385

ABSTRACT

Late-onset retinal degeneration (L-ORD) is an autosomal dominant macular degeneration characterized by the formation of sub-retinal pigment epithelium (RPE) deposits and neuroretinal atrophy. L-ORD results from mutations in the C1q-tumor necrosis factor-5 protein (CTRP5), encoded by the CTRP5/C1QTNF5 gene. To understand the mechanism underlying L-ORD pathology, we used a human cDNA library yeast two-hybrid screen to identify interacting partners of CTRP5. Additionally, we analyzed the Bruch's membrane/choroid (BM-Ch) from wild-type (Wt), heterozygous S163R Ctrp5 mutation knock-in (Ctrp5S163R/wt ), and homozygous knock-in (Ctrp5S163R/S163R ) mice using mass spectrometry. Both approaches showed an association between CTRP5 and HTRA1 via its C-terminal PDZ-binding motif, stimulation of the HTRA1 protease activity by CTRP5, and CTRP5 serving as an HTRA1 substrate. The S163R-CTRP5 protein also binds to HTRA1 but is resistant to HTRA1-mediated cleavage. Immunohistochemistry and proteomic analysis showed significant accumulation of CTRP5 and HTRA1 in BM-Ch of Ctrp5S163R/S163R and Ctrp5S163R/wt mice compared with Wt. Additional extracellular matrix (ECM) components that are HTRA1 substrates also accumulated in these mice. These results implicate HTRA1 and its interaction with CTRP5 in L-ORD pathology.


Subject(s)
Collagen/genetics , High-Temperature Requirement A Serine Peptidase 1/genetics , Mutation , Retinal Degeneration/genetics , Animals , Cellular Senescence/genetics , Collagen/metabolism , High-Temperature Requirement A Serine Peptidase 1/metabolism , Humans , Mass Spectrometry , Mice , Retinal Degeneration/metabolism , Retinal Degeneration/pathology
4.
Hum Gene Ther ; 30(5): 632-650, 2019 05.
Article in English | MEDLINE | ID: mdl-30499344

ABSTRACT

Patients harboring homozygous c.498_499insC mutations in MFRP demonstrate hyperopia, microphthalmia, retinitis pigmentosa, retinal pigment epithelial atrophy, variable degrees of foveal edema, and optic disc drusen. The disease phenotype is variable, however, with some patients maintaining good central vision and cone function till late in the disease. A knock-in mouse model with the c.498_499insC mutation in Mfrp (Mfrp KI/KI) was developed to understand the effects of these mutations in the retina. The model shares many of the features of human clinical disease, including reduced axial length, hyperopia, retinal degeneration, retinal pigment epithelial atrophy, and decreased electrophysiological responses. In addition, the eyes of these mice had a significantly greater refractive error (p < 0.01) when compared to age-matched wild-type control animals. Administration of recombinant adeno-associated virus-mediated Mfrp gene therapy significantly prevented thinning from retinal neurodegeneration (p < 0.005) and preserved retinal electrophysiology (p < 0.001) when treated eyes were compared to contralateral sham-treated control eyes. The Mfrp KI/KI mice will serve as a useful tool to model human disease and point to a potential gene therapeutic approach for patients with preserved vision and electrophysiological responses in MFRP-related retinopathy.


Subject(s)
Genetic Predisposition to Disease , Genetic Therapy , Membrane Proteins/genetics , Retinal Diseases/genetics , Animals , Biomarkers , Dependovirus/genetics , Disease Models, Animal , Electroretinography , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Immunohistochemistry , Mice , Mice, Knockout , Phenotype , Retinal Diseases/diagnosis , Retinal Pigment Epithelium/metabolism , Tomography, Optical Coherence
5.
Hum Genet ; 137(6-7): 447-458, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29978320

ABSTRACT

Whole genome sequencing (WGS) was performed to identify the variants responsible for inherited retinal degeneration (IRD) in a Caucasian family. Segregation analysis of selected rare variants with pathogenic potential identified a set of compound heterozygous changes p.Arg266*:c.796C>T and p.Ala568Thr:c.1702G>A in the intraflagellar transport protein-88 (IFT88) gene segregating with IRD. Expression of IFT88 with the p.Arg266* and p.Ala568Thr mutations in mIMDC3 cells by transient transfection and in HeLa cells by introducing the mutations using CRISPR-cas9 system suggested that both mutations result in the formation of abnormal ciliary structures. The introduction of the IFT88 p.Arg266* variant in the homozygous state in HeLa cells by CRISPR-Cas9 genome-editing revealed that the mutant transcript undergoes nonsense-mediated decay leading to a significant depletion of IFT88 transcript. Additionally, abnormal ciliogenesis was observed in these cells. These observations suggest that the rare and unique combination of IFT88 alleles observed in this study provide insight into the physiological role of IFT88 in humans and the likely mechanism underlying retinal pathology in the pedigree with IRD.


Subject(s)
Ciliopathies/genetics , Retinal Degeneration/genetics , Tumor Suppressor Proteins/genetics , Whole Genome Sequencing , Alleles , CRISPR-Cas Systems/genetics , Ciliopathies/physiopathology , Female , Gene Editing , Genetic Predisposition to Disease , HeLa Cells , Homozygote , Humans , Male , Middle Aged , Mutation , Pedigree , Retina/pathology , Retinal Degeneration/physiopathology
7.
Genes (Basel) ; 8(9)2017 08 24.
Article in English | MEDLINE | ID: mdl-28837078

ABSTRACT

Retinitis pigmentosa (RP) causes progressive photoreceptor loss resulting from mutations in over 80 genes. This study identified the genetic cause of RP in three members of a non-consanguineous pedigree. Detailed ophthalmic evaluation was performed in the three affected family members. Whole exome sequencing (WES) and whole genome sequencing (WGS) were performed in the three affected and the two unaffected family members and variants were filtered to detect rare, potentially deleterious variants segregating with disease. WES and WGS did not identify potentially pathogenic variants shared by all three affected members. However, WES identified a previously reported homozygous nonsense mutation in KIZ (c.226C>T, p.Arg76*) in two affected sisters, but not in their affected second cousin. WGS revealed a novel 1.135 kb homozygous deletion in a retina transcript of C21orf2 and a novel 30.651 kb heterozygous deletion in CACNA2D4 in the affected second cousin. The sisters with the KIZ mutation carried no copies of the C21orf2 or CACNA2D4 deletions, while the second cousin with the C21orf2 and CACNA2D4 deletions carried no copies of the KIZ mutation. This study identified two independent, homozygous mutations in genes previously reported in autosomal recessive RP in a non-consanguineous family, and demonstrated the value of WGS when WES fails to identify likely disease-causing mutations.

8.
Rheumatology (Oxford) ; 56(2): 263-270, 2017 02.
Article in English | MEDLINE | ID: mdl-27864563

ABSTRACT

OBJECTIVE: To conduct a replication study and meta-analysis involving the study of mtDNA variants in the radiographic progression of OA in different cohorts worldwide, including Cohort Hip and Cohort Knee (CHECK), the OA Initiative and a cohort from Spain. METHODS: The influence of the haplogroups in the rate of radiographic progression at 96 months in 431 subjects from CHECK was assessed in terms of Kellgren and Lawrence (KL) grade. Progression was defined as a change from KL ⩾ 1 at baseline to any higher grade during the follow-up. Extended Cox proportional hazard models were used to analyse the influence of mtDNA variants in the rate of radiographic knee OA progression. A subsequent meta-analysis of 1603 subjects following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines was conducted to combine the data of individual studies. A sensitivity analysis was performed to validate the stability of the results. RESULTS: CHECK subjects carrying the haplogroup T showed the lowest rate of radiographic knee OA progression [hazard ratio (HR) 0.645 (95% CI 0.419, 0.978); P < 0.05]. When pooled, subjects within the superhaplogroup JT showed the same trend [HR 0.707 (95% CI 0.501, 0.965); P < 0.05]. BMI [HR 1.046 (95% CI 1.018, 1.073); P < 0.05] and bilateral OA [HR 2.266 (95% CI 1.733, 2.954); P < 0.05] at baseline are risk factors for radiographic knee OA progression as well. In the meta-analysis there was a reduced rate of radiographic progression in subjects with haplogroup T [HR 0.612 (95% CI 0.454, 0.824); P = 0.001] or in the superhaplogroup JT [HR 0.765 (95% CI 0.624, 0.938); P = 0.009]. Sensitivity analysis revealed that the results were robust. CONCLUSION: The mtDNA variants in the superhaplogroup JT associate with a reduced rate of radiographic OA progression. The mtDNA polymorphisms in the superhaplogroup JT emerge as potential complementary genetic biomarkers for disease progression.


Subject(s)
DNA, Mitochondrial/genetics , Osteoarthritis, Knee/genetics , Aged , Body Mass Index , Cohort Studies , Disease Progression , Female , Haplotypes , Humans , Longitudinal Studies , Male , Middle Aged , Osteoarthritis, Knee/diagnostic imaging , Osteoarthritis, Knee/physiopathology , Polymorphism, Genetic , Proportional Hazards Models , Prospective Studies , Radiography , Reproducibility of Results , Risk Factors , Spain
9.
Ann Rheum Dis ; 76(6): 1114-1122, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27919866

ABSTRACT

OBJECTIVE: To evaluate the influence of the mitochondrial DNA (mtDNA) haplogroups in the risk of incident knee osteoarthritis (OA) and to explain the functional consequences of this association to identify potential diagnostic biomarkers and therapeutic targets. METHODS: Two prospective cohorts contributed participants. The osteoarthritis initiative (OAI) included 2579 subjects of the incidence subcohort, and the cohort hip and cohort knee (CHECK) included 635, both with 8-year follow-up. The analysis included the association of mtDNA haplogroups with the rate of incident knee OA in subjects from both cohorts followed by a subsequent meta-analysis. Transmitochondrial cybrids harbouring haplogroup J or H were constructed to detect differences between them in relation to physiological features including specific mitochondrial metabolic parameters, reactive oxygen species production, oxidative stress and apoptosis. RESULTS: Compared with H, the haplogroup J associates with decreased risk of incident knee OA in subjects from OAI (HR=0.680; 95% CI 0.470 to 0.968; p<0.05) and CHECK (HR=0.728; 95% CI 0.469 to 0.998; p<0.05). The subsequent meta-analysis including 3214 cases showed that the haplogroup J associates with a lower risk of incident knee OA (HR=0.702; 95% CI 0.541 to 0.912; p=0.008). J cybrids show a lower free radical production, higher cell survival under oxidative stress conditions, lower grade of apoptosis as well as lower expression of the mitochondrially related pro-apoptotic gene BCL2 binding component 3 (BBC3). In addition, J cybrids also show a lower mitochondrial respiration and glycolysis leading to decreased ATP production. CONCLUSIONS: The physiological effects of the haplogroup J are beneficial to have a lower rate of incident knee OA over time. Potential drugs to treat OA could focus on emulating the mitochondrial behaviour of this haplogroup.


Subject(s)
DNA, Mitochondrial , Osteoarthritis, Knee/epidemiology , Osteoarthritis, Knee/genetics , Apoptosis/genetics , Biomarkers , DNA, Mitochondrial/metabolism , Haplotypes , Humans , Incidence , Oxidative Stress/genetics , Reactive Oxygen Species/metabolism
10.
Rheumatol Int ; 35(2): 337-44, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25086630

ABSTRACT

Not all patients with osteoarthritis (OA) show the same disease progression, as some of them remain relatively stable over time, while others progress to severe structural deterioration of the joint. In this sense, the main goal of both genetic and protein biomarkers in OA is to predict not only the risk of OA at an earlier stage of the disease but also which OA patients are more likely to progress to severe disease. Taking into account the incidence of the mitochondria and the mtDNA haplogroups in the pathogenesis of OA, the main objective of this work was to evaluate the incidence of the mtDNA haplogroups in the radiographic progression of the OA disease in a well-characterized follow-up cohort of Spanish patients. DNA from 281 OA patients from Hospital Universitario A Coruña was isolated to determine the European mtDNA haplogroups. Knee or hip radiographs from all affected joints were obtained at two time points with at least 36 months apart. Radiographs were evaluated using the Kellgren/Lawrence (K/L) scale; radiographic OA progression was defined as any radiographic worsening of the K/L joint score. Statistical analyses included Kaplan-Meier survival curves and Cox regression models. Patients belonging to the cluster TJ showed a slower radiographic OA progression than patients in the cluster KU (p = 0.036). Moreover, patients carrying the most common mtDNA haplogroup H are more apt to require total joint replacement surgery than non-H patients (p = 0.049). The inherited mitochondrial variants influence the radiographic progression of OA and could be considered among the genetic variants taken into account when the radiographic progression of OA is analyzed.


Subject(s)
Cartilage, Articular/diagnostic imaging , DNA, Mitochondrial/genetics , Hip Joint/diagnostic imaging , Knee Joint/diagnostic imaging , Osteoarthritis, Hip/genetics , Osteoarthritis, Knee/genetics , White People/genetics , Adult , Aged , Aged, 80 and over , Cohort Studies , Disease Progression , Female , Haplotypes , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Osteoarthritis, Hip/diagnostic imaging , Osteoarthritis, Knee/diagnostic imaging , Proportional Hazards Models , Radiography , Spain
11.
PLoS One ; 9(11): e112735, 2014.
Article in English | MEDLINE | ID: mdl-25390621

ABSTRACT

OBJECTIVE: To evaluate the influence of the mtDNA haplogroups on knee osteoarthritis progression in Osteoarthritis Initiative (OAI) participants through longitudinal data from radiographs and magnetic resonance imaging (MRI). METHODS: Four-year knee osteoarthritis progression was analyzed as increase in Kellgren and Lawrence (KL) grade, in addition to increase in OARSI atlas grade for joint space narrowing (JSN), osteophytes and subchondral sclerosis in the tibia medial compartment of 891 Caucasian individuals from the progression subcohort. The influence of the haplogroups on the rate of structural progression was also assessed as the four-year change in minimum joint space width (mJSW in millimetres) in both knees of (n = 216) patients with baseline unilateral medial-tibiofemoral JSN. Quantitative cartilage measures from longitudinal MRI data were those related to cartilage thickness and volume with a 24 month follow-up period (n = 381). RESULTS: During the four-year follow-up period, knee OA patients with the haplogroup T showed the lowest increase in KL grade (Hazard Risk [HR] = 0.499; 95% Confidence Interval [CI]: 0.261-0.819; p<0.05) as well as the lowest cumulative probability of progression for JSN (HR = 0.547; 95% CI: 0.280-0.900; p<0.05), osteophytes (HR = 0.573; 95% CI: 0.304-0.893; p<0.05) and subchondral sclerosis (HR = 0.549; 95% CI: 0.295-0.884; p<0.05). They also showed the lowest decline in mJSW (standardized response means (SRM) = -0.39; p = 0.037) in those knees without baseline medial JSN (no-JSN knees). Normalized cartilage volume loss was significantly lower in patients carrying the haplogroup T at medial tibia femoral (SRM = -0.33; p = 0.023) and central medial femoral (SRM = -0.27; p = 0.031) compartments. Cartilage thickness loss was significantly lower in carriers of haplogroup T at central medial tibia-femoral (SRM = -0.42; p = 0.011), medial tibia femoral (SRM = -0.32; p = 0.018), medial tibia anterior (SRM = +0.31; p = 0.013) and central medial femoral (SRM = -0.19; p = 0.013) compartments. CONCLUSIONS: Mitochondrial genome seems to play a role in the progression of knee osteoarthritis. mtDNA variation could improve identification of patients predisposed to faster or severe progression of the disease.


Subject(s)
DNA, Mitochondrial , Genome, Mitochondrial , Haplotypes , Osteoarthritis, Knee/genetics , Aged , Cartilage, Articular/diagnostic imaging , Cartilage, Articular/pathology , Disease Progression , Female , Humans , Knee Joint/diagnostic imaging , Knee Joint/pathology , Magnetic Resonance Imaging , Male , Middle Aged , Osteoarthritis, Knee/diagnostic imaging , Osteoarthritis, Knee/pathology , Radiography
12.
Ann Rheum Dis ; 73(4): 668-77, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23505229

ABSTRACT

OBJECTIVE: Alterations in DNA methylation patterns have been found to correlate with several diseases including osteoarthritis (OA). The aim of this study was to identify, for the first time, the genome-wide DNA methylation profiles of human articular chondrocytes from OA cartilage and healthy control cartilage samples. METHODS: DNA methylation profiling was performed using Illumina Infinium HumanMethylation27 in 25 patients with OA and 20 healthy controls. Subsequent validation was performed by genome-wide expression analysis using the Affymetrix Human Gene 1.1 ST array in an independent cohort of 24 patients with OA. Finally, the most consistent genes in both assays were amplified by quantitative reverse transcriptase PCR in a validation cohort of 48 patients using microfluidic real-time quantitative PCR. Appropriate bioinformatics analyses were carried out using R bioconductor software packages and qBase plus software from Biogazelle. RESULTS: We found 91 differentially methylated (DM) probes, which permitted us to separate patients with OA from healthy controls. Among the patients with OA, we detected 1357 DM probes that identified a tight cluster of seven patients who were different from the rest. This cluster was also identified by genome-wide expression in which 450 genes were differentially expressed. Further validation of the most consistent genes in an independent cohort of patients with OA permitted us to identify this cluster, which was characterised by increased inflammatory processes. CONCLUSIONS: We were able to identify a tight subgroup of patients with OA, characterised by an increased inflammatory response that could be regulated by epigenetics. The identification and isolation of this subgroup may be critical for the development of effective treatment and disease prevention.


Subject(s)
Cartilage, Articular/metabolism , Chondrocytes/metabolism , DNA Methylation , Osteoarthritis, Knee/genetics , Aged , Aged, 80 and over , Case-Control Studies , Cluster Analysis , Female , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Genome-Wide Association Study/methods , Humans , Male , Middle Aged , Osteoarthritis, Knee/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods
13.
Front Biosci (Schol Ed) ; 5(1): 360-8, 2013 01 01.
Article in English | MEDLINE | ID: mdl-23277056

ABSTRACT

The genetic contribution is one of the most notable factors that play a main role in the risk of OA. Despite the genetics of this disease is complex and the finding of risk-related genes has been very challenging, evidence for genetic predisposition has been reported. Besides, in the last years recent evidences indicate that the mitochondrion is implicated in OA. In this context, the mtDNA haplogroups, defined as individual groups characterized by the presence of a particular set of single nucleotide polymorphisms (SNPs) in the mtDNA sequence, emerged as new genetic variants involved in this pathology. Moreover, it has been described that mtDNA damage not only accumulates in OA chondrocytes, but also that OA chondrocytes have limited mtDNA repair capacity. In this review we will focus on the influence of mitochondrial genetics and the mtDNA haplogroups in the prevalence, severity and progression of the OA disease, as well as their incidence on many OA-related features, such as serum levels of OA-related molecular markers, Nitric Oxide production or telomere length.


Subject(s)
Mitochondria/genetics , Osteoarthritis/genetics , DNA, Mitochondrial/genetics , Genetic Predisposition to Disease , Humans , Mitochondria/pathology , Osteoarthritis/pathology
14.
Front Physiol ; 3: 129, 2012.
Article in English | MEDLINE | ID: mdl-22593743

ABSTRACT

OBJECTIVE: To assess a mitochondrion-related phenotype in patients with osteoarthritis (OA). METHODS: Serum levels of the following OA-related biomarkers: matrix metalloproteinase-1 (MMP-1); MMP-3; MMP-13; myeloperoxidase (MPO); a peptide of the alpha-helical region of type II collagen, Coll2-1, and its nitrated form Coll2-1NO(2); a C-terminal neoepitope generated by the collagenase-mediated cleavage of collagen type II triple helix, C2C; the C-propeptide of collagen type II, CPII; hyaluronic acid (HA); human cartilage glycoprotein 39, YKL-40; cartilage oligomeric matrix protein; and cathepsin K were analyzed in 48 OA patients and 52 healthy controls carrying the haplogroups H and J. Logistic regression models and receiver operating characteristic (ROC) curves were performed to predict the onset of OA. RESULTS: MMP-13 was the only biomarker significantly increased in OA patients compared to healthy controls in both haplogroups H and J. The collagen type II biomarkers, Coll2-1, Coll2-1NO(2), the Coll2-1NO(2)/Coll2-1 ratio, C2C, CPII, and the C2C:CPII ratio were significantly increased in OA patients carrying haplogroup H compared to OA carriers of the haplogroup J. Two logistic regression models for diagnosis were constructed and adjusted for age, gender, and body mass index. For haplogroup H, the biomarkers significantly associated with OA were MMP-13 and Coll2-1; the area under the curve (AUC) of the ROC curve for this model was 0.952 (95% CI = 0.892-1.012). For haplogroup J, the only biomarker significantly associated with OA was MMP-13; the AUC for this model was 0.895 (95% CI = 0.801-0.989). CONCLUSION: The mitochondrial DNA haplogroups are potential complementary candidates for biomarkers of OA; their genotyping in conjunction with the assessment of classical protein molecular markers is recommended.

15.
BMC Musculoskelet Disord ; 12: 283, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22171676

ABSTRACT

BACKGROUND: Oxidative stress due to the overproduction of nitric oxide (NO) and other oxygen reactive species (ROS), play a main role in the initiation and progression of the OA disease and leads to the degeneration of mitochondria. Therefore, the goal of this work is to describe the difference in telomere length of peripheral blood leukocytes (PBLs) and Nitric Oxide (NO) production between mitochondrial DNA (mtDNA) haplogroup J and non-J carriers, as indirect approaches of oxidative stress. METHODS: The telomere length of PBL was analyzed in DNA samples from 166 healthy controls (114 J and 52 non-J) and 79 OA patients (41 J and 38 non-J) by means of a validated qPCR method. The NO production was assessed in 7 carriers of the haplogroup J and 27 non-J carriers, by means of the colorimetric reaction of the Griess reagent in supernatants of cultured chondrocytes. Inducible nitric oxide synthase (iNOS) mRNA from these samples was analyzed by qPCR. Appropiated statistical analyses were performed RESULTS: Carriers of the haplogroup J showed a significantly longer telomere length of PBLs than non-J carriers, regardless of age, gender and diagnosis (p = 0.025). Cultured chondrocytes carrying the mtDNA haplogroup J also showed a lower NO production than non-J carriers (p = 0.043). No significant correlations between age and telomore length of PBLs were detected neither for carriers of the haplogroup J nor for non-J carriers. A strong positive correlation between NO production and iNOS expression was also observed (correlation coefficient = 0.791, p < 0.001). CONCLUSION: The protective effect of the mtDNA haplogroup J in the OA disease arise from a lower oxidative stress in carriers of this haplogroup, since this haplogroup is related to lower NO production and hence longer telomere length of PBLs too.


Subject(s)
Chondrocytes/metabolism , DNA, Mitochondrial/genetics , Haplotypes , Leukocytes/metabolism , Nitric Oxide/metabolism , Osteoarthritis, Hip/genetics , Osteoarthritis, Hip/metabolism , Oxidative Stress , Telomere/metabolism , Adult , Aged , Aged, 80 and over , Case-Control Studies , Cells, Cultured , Colorimetry , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Multivariate Analysis , Nitric Oxide Synthase Type II/metabolism , Osteoarthritis, Hip/diagnosis , Phenotype , Polymerase Chain Reaction , Regression Analysis , Spain , Telomere Shortening , Young Adult
16.
BMC Musculoskelet Disord ; 12: 264, 2011 Nov 22.
Article in English | MEDLINE | ID: mdl-22108257

ABSTRACT

BACKGROUND: Oxidative stress play a main role in the initiation and progression of the OA disease and leads to the degeneration of mitochondria. To prevent this, the chondrocytes possess a well-coordinated enzymatic antioxidant system. Besides, the mitochondrial DNA (mtDNA) haplogroups are associated with the OA disease. Thus, the main goal of this work is to assess the incidence of the mtDNA haplogroups on serum levels of two of the main antioxidant enzymes, Manganese Superoxide Dismutase (Mn-SOD or SOD2) and catalase, and to test the suitability of these two proteins for potential OA-related biomarkers. METHODS: We analyzed the serum levels of SOD2 and catalase in 73 OA patients and 77 healthy controls carrying the haplogroups J, U and H, by ELISA assay. Knee and hip radiographs were classified according to Kellgren and Lawrence (K/L) scoring from Grade 0 to Grade IV. Appropriate statistical analyses were performed to test the effects of clinical variables, including gender, body mass index (BMI), age, smoking status, diagnosis, haplogroups and radiologic K/L grade on serum levels of these enzymes. RESULTS: Serum levels of SOD2 appeared statistically increased in OA patients when compared with healthy controls (p < 0.001). Even in those OA patients with higher OA severity (K/L grade IV), the serum levels of this antioxidant enzyme appeared more significantly increased than in OA patients with lower K/L grade (p < 0.001). The mtDNA haplogroups showed an influence on serum levels of catalase (p = 0.054), being carriers of the mtDNA haplogroup J those who showed higher serum levels than non-J carriers (p = 0.057). CONCLUSIONS: The increased levels of SOD2 in OA patients indicate an increased oxidative stress OA-related, therefore this antioxidant enzyme could be a suitable candidate biomarker for diagnosis of OA. Mitochondrial haplogroups significantly correlates with serum levels of catalase.


Subject(s)
Antioxidants/metabolism , Catalase/blood , DNA, Mitochondrial/genetics , Osteoarthritis, Hip/blood , Osteoarthritis, Hip/enzymology , Osteoarthritis, Knee/blood , Osteoarthritis, Knee/enzymology , Superoxide Dismutase/blood , Adult , Aged , Aged, 80 and over , Antioxidants/physiology , Biomarkers/blood , Catalase/genetics , Female , Haplotypes/physiology , Heterozygote , Humans , Male , Middle Aged , Osteoarthritis, Hip/genetics , Osteoarthritis, Knee/genetics , Oxidative Stress/genetics , Superoxide Dismutase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...