Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters

Publication year range
1.
Int J Mol Sci ; 24(2)2023 Jan 12.
Article in English | MEDLINE | ID: mdl-36675026

ABSTRACT

The intracellular retention of mutant cartilage matrix proteins and pathological endoplasmic reticulum (ER) stress disrupts ossification and has been identified as a shared disease mechanism in a range of skeletal dysplasias including short limbed-dwarfism, multiple epiphyseal dysplasia type 5 (EDM5). Although targeting ER stress is an attractive avenue for treatment and has proven successful in the treatment of a related skeletal dysplasia, to date no drugs have proven successful in reducing ER stress in EDM5 caused by the retention of mutant matrilin-3. Our exciting findings show that by using our established luciferase ER stress screening assay, we can identify a "natural" chemical, curcumin, which is able to reduce pathological ER stress in a cell model of EDM5 by promoting the proteasomal degradation mutant matrilin-3. Therefore, this is an important in vitro study in which we describe, for the first time, the success of a naturally occurring chemical as a potential treatment for this currently incurable rare skeletal disease. As studies show that curcumin can be used as a potential treatment for range of diseases in vitro, current research is focused on developing novel delivery strategies to enhance its bioavailability. This is an important and exciting area of research that will have significant clinical impact on a range of human diseases including the rare skeletal disease, EDM5.


Subject(s)
Chondrocytes , Curcumin , Matrilin Proteins , Humans , Chondrocytes/drug effects , Chondrocytes/metabolism , Curcumin/pharmacology , Curcumin/metabolism , Endoplasmic Reticulum Stress , Matrilin Proteins/metabolism , Proteolysis
2.
PLoS Genet ; 15(7): e1008215, 2019 07.
Article in English | MEDLINE | ID: mdl-31260448

ABSTRACT

The unfolded protein response (UPR) is a conserved cellular response to the accumulation of proteinaceous material in endoplasmic reticulum (ER), active both in health and disease to alleviate cellular stress and improve protein folding. Multiple epiphyseal dysplasia (EDM5) is a genetic skeletal condition and a classic example of an intracellular protein aggregation disease, whereby mutant matrilin-3 forms large insoluble aggregates in the ER lumen, resulting in a specific 'disease signature' of increased expression of chaperones and foldases, and alternative splicing of the UPR effector XBP1. Matrilin-3 is expressed exclusively by chondrocytes thereby making EDM5 a perfect model system to study the role of protein aggregation in disease. In order to dissect the role of XBP1 signalling in aggregation-related conditions we crossed a p.V194D Matn3 knock-in mouse model of EDM5 with a mouse line carrying a cartilage specific deletion of XBP1 and analysed the resulting phenotype. Interestingly, the growth of mice carrying the Matn3 p.V194D mutation compounded with the cartilage specific deletion of XBP1 was severely retarded. Further phenotyping revealed increased intracellular retention of amyloid-like aggregates of mutant matrilin-3 coupled with dramatically decreased cell proliferation and increased apoptosis, suggesting a role of XBP1 signalling in protein accumulation and/or degradation. Transcriptomic analysis of chondrocytes extracted from wild type, EDM5, Xbp1-null and compound mutant lines revealed that the alternative splicing of Xbp1 is crucial in modulating levels of protein aggregation. Moreover, through detailed transcriptomic comparison with a model of metaphyseal chondrodysplasia type Schmid (MCDS), an UPR-related skeletal condition in which XBP1 was removed without overt consequences, we show for the first time that the differentiation-state of cells within the cartilage growth plate influences the UPR resulting from retention of a misfolded mutant protein and postulate that modulation of XBP1 signalling pathway presents a therapeutic target for aggregation related conditions in cells undergoing proliferation.


Subject(s)
Mutation , Osteochondrodysplasias/genetics , Osteochondrodysplasias/pathology , X-Box Binding Protein 1/genetics , Alternative Splicing , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Disease Models, Animal , Endoplasmic Reticulum Stress , Gene Expression Profiling , Humans , Matrilin Proteins/chemistry , Matrilin Proteins/genetics , Mice , Osteochondrodysplasias/metabolism , Protein Aggregates , Signal Transduction , Unfolded Protein Response , X-Box Binding Protein 1/metabolism
3.
Dev Dyn ; 250(3): 345-359, 2021 03.
Article in English | MEDLINE | ID: mdl-32633442

ABSTRACT

For the vast majority of the 6000 known rare disease the pathogenic mechanisms are poorly defined and there is little treatment, leading to poor quality of life and high healthcare costs. Genetic skeletal diseases (skeletal dysplasias) are archetypal examples of rare diseases that are chronically debilitating, often life-threatening and for which no treatments are currently available. There are more than 450 unique phenotypes that, although individually rare, have an overall prevalence of at least 1 per 4000 children. Multiple epiphyseal dysplasia (MED) is a clinically and genetically heterogeneous disorder characterized by disproportionate short stature, joint pain, and early-onset osteoarthritis. MED is caused by mutations in the genes encoding important cartilage extracellular matrix proteins, enzymes, and transporter proteins. Recently, through the use of various cell and mouse models, disease mechanisms underlying this diverse phenotypic spectrum are starting to be elucidated. For example, ER stress induced as a consequence of retained misfolded mutant proteins has emerged as a unifying disease mechanisms for several forms of MED in particular and skeletal dysplasia in general. Moreover, targeting ER stress through drug repurposing has become an attractive therapeutic avenue.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Extracellular Matrix Proteins , Mutation , Osteochondrodysplasias , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Humans , Osteochondrodysplasias/drug therapy , Osteochondrodysplasias/genetics , Osteochondrodysplasias/metabolism , Osteochondrodysplasias/pathology , Quality of Life
4.
Hum Mol Genet ; 22(25): 5262-75, 2013 Dec 20.
Article in English | MEDLINE | ID: mdl-23956175

ABSTRACT

Mutant matrilin-3 (V194D) forms non-native disulphide bonded aggregates in the rER of chondrocytes from cell and mouse models of multiple epiphyseal dysplasia (MED). Intracellular retention of mutant matrilin-3 causes endoplasmic reticulum (ER) stress and induces an unfolded protein response (UPR) including the upregulation of two genes recently implicated in ER stress: Armet and Creld2. Nothing is known about the role of Armet and Creld2 in human genetic diseases. In this study, we used a variety of cell and mouse models of chondrodysplasia to determine the genotype-specific expression profiles of Armet and Creld2. We also studied their interactions with various mutant proteins and investigated their potential roles as protein disulphide isomerases (PDIs). Armet and Creld2 were up-regulated in cell and/or mouse models of chondrodysplasias caused by mutations in Matn3 and Col10a1, but not Comp. Intriguingly, both Armet and Creld2 were also secreted into the ECM of these disease models following ER stress. Armet and Creld2 interacted with mutant matrilin-3, but not with COMP, thereby validating the genotype-specific expression. Substrate-trapping experiments confirmed Creld2 processed PDI-like activity, thus identifying a putative functional role. Finally, alanine substitution of the two terminal cysteine residues from the A-domain of V194D matrilin-3 prevented aggregation, promoted mutant protein secretion and reduced the levels of Armet and Creld2 in a cell culture model. We demonstrate that Armet and Creld2 are genotype-specific ER stress response proteins with substrate specificities, and that aggregation of mutant matrilin-3 is a key disease trigger in MED that could be exploited as a potential therapeutic target.


Subject(s)
Cell Adhesion Molecules/genetics , Endoplasmic Reticulum Stress/genetics , Extracellular Matrix Proteins/genetics , Nerve Growth Factors/genetics , Osteochondrodysplasias/genetics , Animals , Apoptosis/genetics , Chondrocytes/metabolism , Collagen Type X/genetics , Disease Models, Animal , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Humans , Matrilin Proteins/genetics , Mice , Osteochondrodysplasias/pathology
5.
Am J Hum Genet ; 89(6): 767-72, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22152678

ABSTRACT

Spondyloepimetaphyseal dysplasia with joint laxity, leptodactylic type (lepto-SEMDJL, aka SEMDJL, Hall type), is an autosomal dominant skeletal disorder that, in spite of being relatively common among skeletal dysplasias, has eluded molecular elucidation so far. We used whole-exome sequencing of five unrelated individuals with lepto-SEMDJL to identify mutations in KIF22 as the cause of this skeletal condition. Missense mutations affecting one of two adjacent amino acids in the motor domain of KIF22 were present in 20 familial cases from eight families and in 12 other sporadic cases. The skeletal and connective tissue phenotype produced by these specific mutations point to functions of KIF22 beyond those previously ascribed functions involving chromosome segregation. Although we have found Kif22 to be strongly upregulated at the growth plate, the precise pathogenetic mechanisms remain to be elucidated.


Subject(s)
Abnormalities, Multiple/genetics , DNA-Binding Proteins/genetics , Genes, Dominant , Joint Dislocations/congenital , Joint Instability/genetics , Kinesins/genetics , Mutation, Missense , Osteochondrodysplasias/genetics , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , Child , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Exome , Gene Expression , Genetic Association Studies , Growth Plate/metabolism , Humans , Joint Dislocations/genetics , Kinesins/chemistry , Kinesins/metabolism , Male , Mice , Protein Structure, Tertiary , Sequence Analysis, DNA , Tibia/metabolism
6.
Arthritis Rheum ; 64(5): 1529-39, 2012 May.
Article in English | MEDLINE | ID: mdl-22083516

ABSTRACT

OBJECTIVE: Mutations in matrilin 3 can result in multiple epiphyseal dysplasia (MED), a disease characterized by delayed and irregular bone growth and early-onset osteoarthritis. Although intracellular retention of the majority of mutant matrilin 3 was previously observed in a murine model of MED caused by a Matn3 V194D mutation, some mutant protein was secreted into the extracellular matrix. Thus, it was proposed that secretion of mutant matrilin 3 may be dependent on the formation of hetero-oligomers with matrilin 1. The aim of this study was to investigate the hypothesis that deletion of matrilin 1 would abolish the formation of matrilin 1/matrilin 3 hetero-oligomers, eliminate the secretion of mutant matrilin 3, and influence disease severity. METHODS: Mice with a Matn3 V194D mutation were crossed with Matn1-null mice, generating mice that were homozygous for V194D and null for matrilin 1. This novel mouse was used for in-depth phenotyping, while cartilage and chondrocytes were studied both histochemically and biochemically. RESULTS: Endochondral ossification was not disrupted any further in mice with a double V194D mutation compared with mice with a single mutation. A similar proportion of mutant matrilin 3 was present in the extracellular matrix, and the amount of retained mutant matrilin 3 was not noticeably increased. Retained mutant matrilin 3 formed disulfide-bonded aggregates and caused the co-retention of matrilin 1. CONCLUSION: We showed that secretion of matrilin 3 V194D mutant protein is not dependent on hetero-oligomerization with matrilin 1, and that the total ablation of matrilin 1 expression has no impact on disease severity in mice with MED. Mutant matrilin 3 oligomers form non-native disulfide-bonded aggregates through the misfolded A domain.


Subject(s)
Bone and Bones/pathology , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/genetics , Glycoproteins/deficiency , Mutation , Osteochondrodysplasias/genetics , Osteochondrodysplasias/pathology , Animals , Apoptosis , Bone and Bones/diagnostic imaging , Bone and Bones/metabolism , Cartilage/metabolism , Cartilage/pathology , Cell Proliferation , Dimerization , Disease Models, Animal , Extracellular Matrix Proteins/metabolism , Female , Glycoproteins/genetics , Glycoproteins/metabolism , Growth Plate/metabolism , Growth Plate/pathology , Male , Matrilin Proteins , Mice , Mice, Knockout , Osteochondrodysplasias/metabolism , Phenotype , Radiography
7.
Hum Mutat ; 33(1): 218-31, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22006726

ABSTRACT

Pseudoachondroplasia (PSACH) results from mutations in cartilage oligomeric matrix protein (COMP) and the p.D469del mutation within the type III repeats of COMP accounts for approximately 30% of PSACH. To determine disease mechanisms of PSACH in vivo, we introduced the Comp D469del mutation into the mouse genome. Mutant animals were normal at birth but grew slower than their wild-type littermates and developed short-limb dwarfism. In the growth plates of mutant mice chondrocyte columns were reduced in number and poorly organized, while mutant COMP was retained within the endoplasmic reticulum (ER) of cells. Chondrocyte proliferation was reduced and apoptosis was both increased and spatially dysregulated. Previous studies on COMP mutations have shown mutant COMP is co-localized with chaperone proteins, and we have reported an unfolded protein response (UPR) in mouse models of PSACH-MED (multiple epiphyseal dysplasia) harboring mutations in Comp (T585M) and Matn3, Comp etc (V194D). However, we found no evidence of UPR in this mouse model of PSACH. In contrast, microarray analysis identified expression changes in groups of genes implicated in oxidative stress, cell cycle regulation, and apoptosis, which is consistent with the chondrocyte pathology. Overall, these data suggest that a novel form of chondrocyte stress triggered by the expression of mutant COMP is central to the pathogenesis of PSACH.


Subject(s)
Achondroplasia/genetics , Apoptosis/genetics , Cell Cycle Checkpoints/genetics , Chondrocytes/metabolism , Extracellular Matrix Proteins/genetics , Glycoproteins/genetics , Mutation , Achondroplasia/metabolism , Achondroplasia/pathology , Animals , Cell Proliferation , Chondrocytes/pathology , Disease Models, Animal , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Extracellular Matrix Proteins/metabolism , Gene Expression , Gene Expression Profiling , Glycoproteins/metabolism , Growth Plate/metabolism , Growth Plate/pathology , Matrilin Proteins , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Oxidative Stress , Phenotype
8.
Hum Mutat ; 33(1): 144-57, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21922596

ABSTRACT

Pseudoachondroplasia (PSACH) and multiple epiphyseal dysplasia (MED) are relatively common skeletal dysplasias resulting in short-limbed dwarfism, joint pain, and stiffness. PSACH and the largest proportion of autosomal dominant MED (AD-MED) results from mutations in cartilage oligomeric matrix protein (COMP); however, AD-MED is genetically heterogenous and can also result from mutations in matrilin-3 (MATN3) and type IX collagen (COL9A1, COL9A2, and COL9A3). In contrast, autosomal recessive MED (rMED) appears to result exclusively from mutations in sulphate transporter solute carrier family 26 (SLC26A2). The diagnosis of PSACH and MED can be difficult for the nonexpert due to various complications and similarities with other related diseases and often mutation analysis is requested to either confirm or exclude the diagnosis. Since 2003, the European Skeletal Dysplasia Network (ESDN) has used an on-line review system to efficiently diagnose cases referred to the network prior to mutation analysis. In this study, we present the molecular findings in 130 patients referred to ESDN, which includes the identification of novel and recurrent mutations in over 100 patients. Furthermore, this study provides the first indication of the relative contribution of each gene and confirms that they account for the majority of PSACH and MED.


Subject(s)
Achondroplasia/genetics , Anion Transport Proteins/genetics , Collagen Type IX/genetics , Extracellular Matrix Proteins/genetics , Glycoproteins/genetics , Osteochondrodysplasias/genetics , Amino Acid Sequence , Cartilage Oligomeric Matrix Protein , Child , Child, Preschool , DNA Mutational Analysis , Exons , Female , Genetic Heterogeneity , Humans , Longitudinal Studies , Male , Matrilin Proteins , Molecular Sequence Data , Mutation , Pedigree , Phenotype , Practice Guidelines as Topic , Sulfate Transporters
9.
Hum Mol Genet ; 19(1): 52-64, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19808781

ABSTRACT

Pseudoachondroplasia (PSACH) and multiple epiphyseal dysplasia (MED) are relatively common skeletal dysplasias belonging to the same bone dysplasia family. PSACH is characterized by generalized epi-metaphyseal dysplasia, short-limbed dwarfism, joint laxity and early onset osteoarthritis. MED is a milder disease with radiographic features often restricted to the epiphyses of the long bones. PSACH and some forms of MED result from mutations in cartilage oligomeric matrix protein (COMP), a pentameric glycoprotein found in cartilage, tendon, ligament and muscle. PSACH-MED patients often have a mild myopathy characterized by mildly increased plasma creatine kinase levels, a variation in myofibre size and/or small atrophic fibres. In some instances, patients are referred to neuromuscular clinics prior to the diagnosis of an underlying skeletal dysplasia; however, the myopathy associated with PSACH-MED has not previously been studied. In this study, we present a detailed study of skeletal muscle, tendon and ligament from a mouse model of mild PSACH harbouring a COMP mutation. Mutant mice exhibited a progressive muscle weakness associated with an increased number of muscle fibres with central nuclei at the perimysium and at the myotendinous junction. Furthermore, the distribution of collagen fibril diameters in the mutant tendons and ligaments was altered towards thicker collagen fibrils, and the tendons became more lax in cyclic strain tests. We hypothesize that the myopathy in PSACH-MED originates from an underlying tendon and ligament pathology that is a direct result of structural abnormalities to the collagen fibril architecture. This is the first comprehensive characterization of the musculoskeletal phenotype of PSACH-MED and is directly relevant to the clinical management of these patients.


Subject(s)
Osteochondrodysplasias/complications , Osteochondrodysplasias/pathology , Tendinopathy/complications , Tendinopathy/pathology , Achilles Tendon/metabolism , Achilles Tendon/pathology , Achilles Tendon/ultrastructure , Animals , Apoptosis , Biomechanical Phenomena , Disease Models, Animal , Disease Progression , Endoplasmic Reticulum/pathology , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/metabolism , Fibrillar Collagens/metabolism , Fibrillar Collagens/ultrastructure , Glycoproteins/deficiency , Glycoproteins/metabolism , Immunohistochemistry , Ligaments/metabolism , Ligaments/pathology , Matrilin Proteins , Mice , Mice, Mutant Strains , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Weakness/complications , Muscle Weakness/pathology
10.
Am J Hum Genet ; 84(1): 72-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19110214

ABSTRACT

Analysis of a nuclear family with three affected offspring identified an autosomal-recessive form of spondyloepimetaphyseal dysplasia characterized by severe short stature and a unique constellation of radiographic findings. Homozygosity for a haplotype that was identical by descent between two of the affected individuals identified a locus for the disease gene within a 17.4 Mb interval on chromosome 15, a region containing 296 genes. These genes were assessed and ranked by cartilage selectivity with whole-genome microarray data, revealing only two genes, encoding aggrecan and chondroitin sulfate proteoglycan 4, that were selectively expressed in cartilage. Sequence analysis of aggrecan complementary DNA from an affected individual revealed homozygosity for a missense mutation (c.6799G --> A) that predicts a p.D2267N amino acid substitution in the C-type lectin domain within the G3 domain of aggrecan. The D2267 residue is predicted to coordinate binding of a calcium ion, which influences the conformational binding loops of the C-type lectin domain that mediate interactions with tenascins and other extracellular-matrix proteins. Expression of the normal and mutant G3 domains in mammalian cells showed that the mutation created a functional N-glycosylation site but did not adversely affect protein trafficking and secretion. Surface-plasmon-resonance studies showed that the mutation influenced the binding and kinetics of the interactions between the aggrecan G3 domain and tenascin-C. These findings identify an autosomal-recessive skeletal dysplasia and a significant role for the aggrecan C-type lectin domain in regulating endochondral ossification and, thereby, height.


Subject(s)
Aggrecans/genetics , Antigens/genetics , Genetic Predisposition to Disease , Lectins, C-Type/genetics , Mutation, Missense , Osteochondrodysplasias/genetics , Proteoglycans/genetics , Adolescent , Adult , Aggrecans/metabolism , Amino Acid Sequence , Antigens/metabolism , Cartilage/metabolism , Cell Line , Child , Female , Humans , Lectins, C-Type/metabolism , Male , Molecular Sequence Data , Osteochondrodysplasias/metabolism , Pedigree , Protein Binding , Protein Structure, Tertiary , Proteoglycans/metabolism , Tenascin/metabolism , Young Adult
11.
PLoS Genet ; 5(10): e1000691, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19834559

ABSTRACT

Pathologies caused by mutations in extracellular matrix proteins are generally considered to result from the synthesis of extracellular matrices that are defective. Mutations in type X collagen cause metaphyseal chondrodysplasia type Schmid (MCDS), a disorder characterised by dwarfism and an expanded growth plate hypertrophic zone. We generated a knock-in mouse model of an MCDS-causing mutation (COL10A1 p.Asn617Lys) to investigate pathogenic mechanisms linking genotype and phenotype. Mice expressing the collagen X mutation had shortened limbs and an expanded hypertrophic zone. Chondrocytes in the hypertrophic zone exhibited endoplasmic reticulum (ER) stress and a robust unfolded protein response (UPR) due to intracellular retention of mutant protein. Hypertrophic chondrocyte differentiation and osteoclast recruitment were significantly reduced indicating that the hypertrophic zone was expanded due to a decreased rate of VEGF-mediated vascular invasion of the growth plate. To test directly the role of ER stress and UPR in generating the MCDS phenotype, we produced transgenic mouse lines that used the collagen X promoter to drive expression of an ER stress-inducing protein (the cog mutant of thyroglobulin) in hypertrophic chondrocytes. The hypertrophic chondrocytes in this mouse exhibited ER stress with a characteristic UPR response. In addition, the hypertrophic zone was expanded, gene expression patterns were disrupted, osteoclast recruitment to the vascular invasion front was reduced, and long bone growth decreased. Our data demonstrate that triggering ER stress per se in hypertrophic chondrocytes is sufficient to induce the essential features of the cartilage pathology associated with MCDS and confirm that ER stress is a central pathogenic factor in the disease mechanism. These findings support the contention that ER stress may play a direct role in the pathogenesis of many connective tissue disorders associated with the expression of mutant extracellular matrix proteins.


Subject(s)
Cartilage/metabolism , Cartilage/pathology , Chondrodysplasia Punctata/metabolism , Chondrodysplasia Punctata/pathology , Collagen Type X/metabolism , Endoplasmic Reticulum/metabolism , Stress, Physiological , Animals , Base Sequence , Cell Differentiation , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrodysplasia Punctata/genetics , Collagen Type X/genetics , Disease Models, Animal , Mice , Unfolded Protein Response , Vascular Endothelial Growth Factor A/metabolism
12.
Sci Rep ; 12(1): 13884, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35974042

ABSTRACT

Cysteine rich with epidermal growth factor (EGF)-like domains 2 (CRELD2) is an endoplasmic reticulum (ER) resident chaperone protein with calcium binding properties. CRELD2 is an ER-stress regulated gene that has been implicated in the pathogenesis of skeletal dysplasias and has been shown to play an important role in the differentiation of chondrocytes and osteoblasts. Despite CRELD2 having an established role in skeletal development and bone formation, its role in osteoclasts is currently unknown. Here we show for the first time that CRELD2 plays a novel role in trafficking transforming growth factor beta 1 (TGF-ß1), which is linked to an upregulation in the expression of Nfat2, the master regulator of osteoclast differentiation in early osteoclastogenesis. Despite this finding, we show that overexpressing CRELD2 impaired osteoclast differentiation due to a reduction in the activity of the calcium-dependant phosphatase, calcineurin. This in turn led to a subsequent block in the dephosphorylation of nuclear factor of activated T cells 1 (NFATc1), preventing its nuclear localisation and activation as a pro-osteoclastogenic transcription factor. Our exciting results show that the overexpression of Creld2 in osteoclasts impaired calcium release from the ER which is essential for activating calcineurin and promoting osteoclastogenesis. Therefore, our data proposes a novel inhibitory role for this calcium-binding ER-resident chaperone in modulating calcium flux during osteoclast differentiation which has important implications in our understanding of bone remodelling and the pathogenesis of skeletal diseases.


Subject(s)
Calcium , Osteoclasts , Calcineurin/metabolism , Calcium/metabolism , Cell Differentiation/physiology , Endoplasmic Reticulum/metabolism , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , RANK Ligand/metabolism
13.
J Biol Chem ; 285(44): 34048-61, 2010 Oct 29.
Article in English | MEDLINE | ID: mdl-20729554

ABSTRACT

Matrilin-1 is expressed predominantly in cartilage and co-localizes with matrilin-3 with which it can form hetero-oligomers. We recently described novel structural and functional features of the matrilin-3 A-domain (M3A) and demonstrated that it bound with high affinity to type II and IX collagens. Interactions preferentially occurred in the presence of Zn(2+) suggesting that matrilin-3 has acquired a requirement for specific metal ions for activation and/or molecular associations. To understand the interdependence of matrilin-1/-3 hetero-oligomers in extracellular matrix (ECM) interactions, we have extended these studies to include the two matrilin-1 A-domains (i.e. M1A1 and M1A2 respectively). In this study we have identified new characteristics of the matrilin-1 A-domains by describing their glycosylation state and the effect of N-glycan chains on their structure, thermal stability, and protein-protein interactions. Initial characterization revealed that N-glycosylation did not affect secretion of these two proteins, nor did it alter their folding characteristics. However, removal of the glycosylation decreased their thermal stability. We then compared the effect of different cations on binding between both M1A domains and type II and IX collagens and showed that Zn(2+) also supports their interactions. Finally, we have demonstrated that both M1A1 domains and biglycan are essential for the association of the type II·VI collagen complex. We predict that a potential role of the matrilin-1/-3 hetero-oligomer might be to increase multivalency, and therefore the ability to connect various ECM components. Differing affinities could act to regulate the integrated network, thus coordinating the organization of the macromolecular structures in the cartilage ECM.


Subject(s)
Cartilage/metabolism , Extracellular Matrix Proteins/chemistry , Extracellular Matrix/metabolism , Glycoproteins/chemistry , Biglycan , Cartilage Oligomeric Matrix Protein , Cell Line , Cloning, Molecular , Collagen/chemistry , Extracellular Matrix Proteins/physiology , Glycoproteins/physiology , Glycosylation , Humans , Kinetics , Matrilin Proteins , Mutagenesis, Site-Directed , Protein Interaction Mapping , Protein Structure, Tertiary , Proteoglycans/chemistry , Zinc/chemistry
14.
Sci Rep ; 11(1): 10452, 2021 05 17.
Article in English | MEDLINE | ID: mdl-34001919

ABSTRACT

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.


Subject(s)
Cortical Excitability/genetics , Hippocampus/physiology , MicroRNAs/metabolism , Oxidoreductases Acting on Sulfur Group Donors/genetics , Receptors, Immunologic/genetics , Animals , Cell Line , Female , Male , Mice , Mice, Knockout , MicroRNAs/genetics , Neocortex/physiology , RNA-Seq , Signal Transduction/genetics
15.
Cell Tissue Res ; 339(1): 197-211, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19851784

ABSTRACT

The unfolded protein response (UPR) has evolved to counter the stresses that occur in the endoplasmic reticulum (ER) as a result of misfolded proteins. This sophisticated quality control system attempts to restore homeostasis through the action of a number of different pathways that are coordinated in the first instance by the ER stress-senor proteins IRE1, ATF6 and PERK. However, prolonged ER-stress-related UPR can have detrimental effects on cell function and, in the longer term, may induce apoptosis. Connective tissue cells such as fibroblasts, osteoblasts and chondrocytes synthesise and secrete large quantities of proteins and mutations in many of these gene products give rise to heritable disorders of connective tissues. Until recently, these mutant gene products were thought to exert their effect through the assembly of a defective extracellular matrix that ultimately disrupted tissue structure and function. However, it is now becoming clear that ER stress and UPR, because of the expression of a mutant gene product, is not only a feature of, but may be a key mediator in the initiation and progression of a whole range of different connective tissue diseases. This review focuses on ER stress and the UPR that characterises an increasing number of connective tissue diseases and highlights novel therapeutic opportunities that may arise.


Subject(s)
Connective Tissue Diseases/metabolism , Endoplasmic Reticulum/metabolism , Unfolded Protein Response , Activating Transcription Factor 6/biosynthesis , Activating Transcription Factor 6/genetics , Animals , Apoptosis/genetics , Chondrocytes/metabolism , Chondrocytes/pathology , Connective Tissue Diseases/genetics , Connective Tissue Diseases/pathology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Endoribonucleases/biosynthesis , Endoribonucleases/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation/genetics , Homeostasis/genetics , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Osteoblasts/metabolism , Osteoblasts/pathology , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , eIF-2 Kinase/biosynthesis , eIF-2 Kinase/genetics
16.
Am J Med Genet A ; 152A(4): 863-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20358595

ABSTRACT

Multiple epiphyseal dysplasia (MED) is a clinically variable and genetically heterogeneous disease that is characterized by mild short stature and early onset osteoarthritis. Autosomal dominant forms are caused by mutations in the genes that encode type IX collagen, cartilage oligomeric matrix protein, and matrilin-3: COL9A1, COL9A2, COL9A3, COMP, and MATN3, respectively. Splicing mutations have been identified in all three genes encoding type IX collagen and are restricted to specific exons encoding an equivalent region of the COL3 domain in all three alpha(IX) chains. MED has been associated with mild myopathy in some families, in particular one family with a COL9A3 mutation and two families with C-terminal COMP mutations. In this study we have identified COL9A2 mutations in two families with MED that also have osteochondritis dissecans and mild myopathy. This study therefore extends the range of gene-mutations that can cause MED-related myopathy. (c) 2010 Wiley-Liss, Inc.


Subject(s)
Collagen Type IX/genetics , Muscular Diseases/complications , Muscular Diseases/genetics , Mutation/genetics , Osteochondritis Dissecans/genetics , Osteochondrodysplasias/complications , Osteochondrodysplasias/genetics , Adult , Biopsy , Child , Child, Preschool , Family , Female , Humans , Infant, Newborn , Male , Muscles/pathology , Muscular Diseases/diagnostic imaging , Osteochondritis Dissecans/complications , Osteochondritis Dissecans/diagnostic imaging , Osteochondrodysplasias/diagnostic imaging , Pedigree , Pregnancy , Radiography
17.
J Biomed Biotechnol ; 2010: 686457, 2010.
Article in English | MEDLINE | ID: mdl-20508815

ABSTRACT

Musculoskeletal system is a complex assembly of tissues which acts as scaffold for the body and enables locomotion. It is often overlooked that different components of this system may biomechanically interact and affect each other. Skeletal dysplasias are diseases predominantly affecting the development of the osseous skeleton. However, in some cases skeletal dysplasia patients are referred to neuromuscular clinics prior to the correct skeletal diagnosis. The muscular complications seen in these cases are usually mild and may stem directly from the muscle defect and/or from the altered interactions between the individual components of the musculoskeletal system. A correct early diagnosis may enable better management of the patients and a better quality of life. This paper attempts to summarise the different components of the musculoskeletal system which are affected in skeletal dysplasias and lists several interesting examples of such diseases in order to enable better understanding of the complexity of human musculoskeletal system.

18.
F1000Res ; 92020.
Article in English | MEDLINE | ID: mdl-32399188

ABSTRACT

Cartilage comprises a single cell type, the chondrocyte, embedded in a highly complex extracellular matrix. Disruption to the cartilage growth plate leads to reduced bone growth and results in a clinically diverse group of conditions known as genetic skeletal diseases (GSDs). Similarly, long-term degradation of articular cartilage can lead to osteoarthritis (OA), a disease characterised by joint pain and stiffness. As professionally secreting cells, chondrocytes are particularly susceptible to endoplasmic reticulum (ER) stress and this has been identified as a core disease mechanism in a group of clinically and pathologically related GSDs. If unresolved, ER stress can lead to chondrocyte cell death. Recent interest has focused on ER stress as a druggable target for GSDs and this has led to the first clinical trial for a GSD by repurposing an antiepileptic drug. Interestingly, ER stress markers have also been associated with OA in multiple cell and animal models and there is increasing interest in it as a possible therapeutic target for treatment. In summary, chondrocyte ER stress has been identified as a core disease mechanism in GSDs and as a contributory factor in OA. Thus, chondrocyte ER stress is a unifying factor for both common and rare cartilage-related diseases and holds promise as a novel therapeutic target.


Subject(s)
Chondrocytes/pathology , Endoplasmic Reticulum Stress , Animals , Cells, Cultured , Growth Plate/pathology , Osteoarthritis/pathology
19.
J Bone Miner Res ; 35(8): 1452-1469, 2020 08.
Article in English | MEDLINE | ID: mdl-32181934

ABSTRACT

Cysteine-rich with epidermal growth factor (EGF)-like domains 2 (CRELD2) is an endoplasmic reticulum (ER)-resident chaperone highly activated under ER stress in conditions such as chondrodysplasias; however, its role in healthy skeletal development is unknown. We show for the first time that cartilage-specific deletion of Creld2 results in disrupted endochondral ossification and short limbed dwarfism, whereas deletion of Creld2 in bone results in osteopenia, with a low bone density and altered trabecular architecture. Our study provides the first evidence that CRELD2 promotes the differentiation and maturation of skeletal cells by modulating noncanonical WNT4 signaling regulated by p38 MAPK. Furthermore, we show that CRELD2 is a novel chaperone for the receptor low-density lipoprotein receptor-related protein 1 (LRP1), promoting its transport to the cell surface, and that LRP1 directly regulates WNT4 expression in chondrocytes through TGF-ß1 signaling. Therefore, our data provide a novel link between an ER-resident chaperone and the essential WNT signaling pathways active during skeletal differentiation that could be applicable in other WNT-responsive tissues. © 2020 American Society for Bone and Mineral Research. © 2020 The Authors. Journal of Bone and Mineral Research published by American Society for Bone and Mineral Research..


Subject(s)
Cell Adhesion Molecules , Extracellular Matrix Proteins , Cell Differentiation , Chondrocytes , Endoplasmic Reticulum , Endoplasmic Reticulum Stress , Wnt Signaling Pathway
20.
Arthritis Res Ther ; 21(1): 206, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31511053

ABSTRACT

BACKGROUND: Osteoarthritis has been associated with a plethora of pathological factors and one which has recently emerged is chondrocyte endoplasmic reticulum (ER) stress. ER stress is sensed by key ER-resident stress sensors, one of which is activating transcription factor 6 (ATF6). The purpose of this study is to determine whether increased ER stress plays a role in OA. METHODS: OA was induced in male wild-type (+/+), ColIITgcog (c/c) and Atf6α-/- mice by destabilisation of the medial meniscus (DMM). c/c mice have increased ER stress in chondrocytes via the collagen II promoter-driven expression of ER stress-inducing Tgcog. Knee joints were scored histologically for OA severity. RNA-seq was performed on laser-micro-dissected RNA from cartilage of +/+ and c/c DMM-operated mice. RESULTS: In situ hybridisation demonstrated a correlation between the upregulation of ER stress marker, BiP, and early signs of proteoglycan loss and cartilage damage in DMM-operated +/+ mice. Histological analysis revealed a significant reduction in OA severity in c/c mice compared with +/+ at 2 weeks post-DMM. This chondroprotective effect in c/c mice was associated with a higher ambient level of BiP protein prior to DMM and a delay in chondrocyte apoptosis. RNA-seq analysis suggested Xbp1-regulated networks to be significantly enriched in c/c mice at 2 weeks post-DMM. Compromising the ER through genetically ablating Atf6α, a key ER stress sensor, had no effect on DMM-induced OA severity. CONCLUSION: Our studies indicate that an increased capacity to effectively manage increases in ER stress in articular cartilage due either to pre-conditioning as a result of prior exposure to ER stress or to genetic pre-disposition may be beneficial in delaying the onset of OA, but once established, ER stress plays no significant role in disease progression.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Osteoarthritis/metabolism , RNA/genetics , Animals , Apoptosis , Biomarkers/metabolism , Cartilage, Articular , Disease Models, Animal , Disease Progression , Gene Expression Regulation , Immunohistochemistry , Male , Mice , Osteoarthritis/genetics , Osteoarthritis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL