Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Development ; 144(19): 3562-3577, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28851708

ABSTRACT

Cartilage originates from mesenchymal cell condensations that differentiate into chondrocytes of transient growth plate cartilage or permanent cartilage of the articular joint surface and trachea. MicroRNAs fine-tune the activation of entire signaling networks and thereby modulate complex cellular responses, but so far only limited data are available on miRNAs that regulate cartilage development. Here, we characterize a miRNA that promotes the biosynthesis of a key component in the RAF/MEK/ERK pathway in cartilage. Specifically, by transcriptome profiling we identified miR-322 to be upregulated during chondrocyte differentiation. Among the various miR-322 target genes in the RAF/MEK/ERK pathway, only Mek1 was identified as a regulated target in chondrocytes. Surprisingly, an increased concentration of miR-322 stabilizes Mek1 mRNA to raise protein levels and dampen ERK1/2 phosphorylation, while cartilage-specific inactivation of miR322 in mice linked the loss of miR-322 to decreased MEK1 levels and to increased RAF/MEK/ERK pathway activation. Such mice died perinatally due to tracheal growth restriction and respiratory failure. Hence, a single miRNA can stimulate the production of an inhibitory component of a central signaling pathway to impair cartilage development.


Subject(s)
Cartilage/embryology , Cartilage/enzymology , MAP Kinase Kinase 1/metabolism , MAP Kinase Signaling System , MicroRNAs/metabolism , Animals , Animals, Newborn , Binding Sites/genetics , CRISPR-Cas Systems/genetics , Chondrocytes/metabolism , Gene Deletion , Gene Expression Regulation, Developmental , Gene Silencing , Growth Plate/metabolism , Hemizygote , Homeostasis , MAP Kinase Kinase 1/genetics , Male , Mice, Transgenic , MicroRNAs/genetics , Organogenesis/genetics , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Transfection
2.
Stem Cells ; 34(5): 1297-309, 2016 05.
Article in English | MEDLINE | ID: mdl-26934179

ABSTRACT

microRNAs (miRNAs) can regulate the interplay between perivascular cells (PVC) and endothelial cells (EC) during angiogenesis, but the relevant PVC-specific miRNAs are not yet defined. Here, we identified miR-126-3p and miR-146a to be exclusively upregulated in PVC upon interaction with EC, determined their influence on the PVC phenotype and elucidate their molecular mechanisms of action. Specifically the increase of miR-126-3p strongly promoted the motility of PVC on the basement membrane-like composite and stabilized networks of EC. Subsequent miRNA target analysis showed that miR-126-3p inhibits SPRED1 and PLK2 expression, induces ERK1/2 phosphorylation and stimulates TLR3 expression to modulate cell-cell and cell-matrix contacts of PVC. Gain of expression experiments in vivo demonstrated that miR-126-3p stimulates PVC coverage of newly formed vessels and transform immature into mature, less permeable vessels. In conclusion we showed that miR-126-3p regulates matrix-dependent PVC migration and intercellular interaction to modulate vascular integrity. Stem Cells 2016;34:1297-1309.


Subject(s)
Blood Vessels/cytology , Cell Communication/genetics , Cell Movement/genetics , Extracellular Matrix/metabolism , MicroRNAs/metabolism , Animals , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Communication/drug effects , Cell Movement/drug effects , Cell Shape/drug effects , Chemokines/metabolism , Coculture Techniques , Collagen/pharmacology , Drug Combinations , Extracellular Matrix/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Silencing/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Laminin/pharmacology , MAP Kinase Signaling System/drug effects , Mice , MicroRNAs/genetics , Neovascularization, Physiologic/genetics , Proteoglycans/pharmacology , Transcriptome/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
3.
Arthritis Rheum ; 65(6): 1547-60, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23436205

ABSTRACT

OBJECTIVE: To identify changes in gene expression in mice with osteoarthritis (OA) in order to explore the mechanisms of the disease. METHODS: Gene expression profiling was performed in cartilage from mice with surgically induced OA. We used wild-type (WT) mice and Adamts5Δcat mice, in which ADAMTS-5 activity is lacking and aggrecan loss and cartilage erosion are inhibited, to distinguish gene expression changes that are independent of ADAMTS-5 activity and cartilage breakdown. Mechanical instability was introduced into the knee joints of 10-week-old male mice via surgical destabilization of the medial meniscus (DMM). Cartilage from the developing lesion in the destabilized medial meniscus and corresponding regions in sham-operated joints was harvested by microdissection at 1, 2, and 6 weeks postsurgery, and RNA was extracted, amplified, and hybridized to whole-genome microarrays. RESULTS: Several previously identified OA-related genes, including Ptgs2, Crlf1, and Inhba, and novel genes, such as Phdla2 and Il11, were up-regulated in both WT mice and Adamts5Δcat mice, indicating that they are independent of ADAMTS-5 activity. The altered expression of other genes, including Col10a1, the sentinel marker of cartilage hypertrophy, and Wnt/ß-catenin pathway genes, required ADAMTS-5 activity. Cell death pathway genes were dysregulated, and Tp53, Foxo4, and Xbp1 endoplasmic reticulum-stress transcriptional networks were activated. Analysis of degradome genes identified up-regulation of many proteases, including Mmp3, Capn2, and the novel cartilage proteases Prss46 and Klk8. Comparison with other studies identified 16 genes also dysregulated in rat and human OA as priorities for study. CONCLUSION: We have identified, for the first time, several genes that have an ADAMTS-5-independent role in OA, identifying them as possible OA initiation candidates. This work provides new insights into the sequence of gene dysregulation and the molecular basis of cartilage destruction in OA.


Subject(s)
ADAM Proteins/deficiency , Cartilage, Articular/pathology , Osteoarthritis/genetics , Osteoarthritis/pathology , Transcriptome , ADAM Proteins/genetics , ADAMTS5 Protein , Animals , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL
4.
J Biol Chem ; 286(43): 37758-67, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-21880736

ABSTRACT

Using transcriptome profiling to determine differential gene expression between the permanent mouse articular cartilage and the transient growth plate cartilage, we identified a highly expressed gene, Cilp2, which is expressed differentially by articular chondrocytes. CILP-2 is highly homologous to CILP-1 (cartilage intermediate layer protein 1), which is expressed in the intermediate zone of articular cartilage and has been linked to cartilage degenerative diseases. We demonstrated that Cilp2 has a restricted mRNA distribution at the surface of the mouse articular cartilage during development, becoming localized to the intermediate zone of articular cartilage and meniscal cartilage with maturity. Although the extracellular CILP-2 protein localization is broadly similar to CILP-1, CILP-2 appears to be more localized in the deeper intermediate zone of the articular cartilage extracellular matrix at maturity. CILP-2 was shown to be proteolytically processed, N-glycosylated, and present in human articular cartilage. In surgically induced osteoarthritis in mice, Cilp1 and Cilp2 gene expression was dysregulated. However, whereas Cilp1 expression was increased, Cilp2 gene expression was down-regulated demonstrating a differential response to mechanically induced joint destabilization. CILP-2 protein was reduced in the mouse osteoarthritic cartilage. Ultrastructural analysis also suggested that CILP-2 may be associated with collagen VI microfibrils and thus may mediate interactions between matrix components in the territorial and inter-territorial articular cartilage matrix. mRNA expression analysis indicated that whereas Cilp1 and Cilp2 are expressed most abundantly in cartilaginous tissues, expression can be detected in muscle and heart.


Subject(s)
Cartilage, Articular/metabolism , Down-Regulation , Extracellular Matrix Proteins/metabolism , Extracellular Matrix/metabolism , Menisci, Tibial/metabolism , Osteoarthritis/metabolism , Pyrophosphatases/metabolism , Aged , Animals , Cartilage, Articular/ultrastructure , Collagen Type VI/genetics , Collagen Type VI/metabolism , Disease Models, Animal , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Extracellular Matrix Proteins/genetics , Female , Humans , Male , Menisci, Tibial/ultrastructure , Mice , Osteoarthritis/genetics , Osteoarthritis/pathology , Pyrophosphatases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
Mol Cell Endocrinol ; 546: 111570, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35051551

ABSTRACT

Complete androgen insensitivity syndrome (CAIS), where 46,XY individuals present as female, is caused by variants in the androgen receptor gene (AR). We analyzed the DNA of a patient with suspected CAIS using a targeted gene sequencing panel and whole exome sequencing (WES) but did not detect any small nucleotide variants in AR. Analysis of WES data using our bioinformatics pipeline designed to detect copy number variations (CNV) uncovered a rare duplication of exon 2 of AR. Using array comparative genomic hybridization, the duplication was found to span 43.6 kb and is predicted to cause a frameshift and loss of AR protein. We confirmed the power of our WES-CNV detection protocol by identifying pathogenic CNVs in FSHR and NR5A1 in previously undiagnosed patients with disorders of sex development. Our findings illustrate the usefulness of CNV analysis in WES data to detect pathogenic genomic changes that may go undetected using only standard analysis protocols.


Subject(s)
Androgen-Insensitivity Syndrome , DNA Copy Number Variations , Androgen-Insensitivity Syndrome/genetics , Comparative Genomic Hybridization , DNA Copy Number Variations/genetics , Exome/genetics , Female , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Exome Sequencing/methods
6.
Biochim Biophys Acta ; 1779(5): 330-40, 2008 May.
Article in English | MEDLINE | ID: mdl-18374667

ABSTRACT

In vertebrates, longitudinal bone growth is the consequence of a complex series of events that take place in a specialized structure, the growth plate cartilage. Within the growth plate chondrocytes undergo a sequential maturation program from resting cells to proliferative, pre-hypertrophic, and ultimately hypertrophic end-stage chondrocytes. This process of chondrocyte maturation is under the control of the temporally and spatially regulated expression of a myriad of signaling molecules, transmembrane receptors, transcription factors, and structural extracellular matrix (ECM) proteins. One approach to the comprehensive definition of the key components of such complex interrelated pathways is the use of microarray expression profiling to catalogue transcriptome changes during chondrocyte maturation in the individual developmental zones of the mouse growth plate cartilage. However, this has not been achieved because of the difficulty in obtaining sufficient quantities of the individual growth plate cartilage zones to all microarray analysis. In this study we describe the development of microdissection methods for the isolation of tissue from the proliferative, pre-hypertrophic, and proliferative zone from one single mouse femur, RNA extraction and linear amplification of the RNA to allow interrogation of NIA 15k microarrays to generate comparative expression profiles. Verification of a subset of differentially expressed genes by RT-PCR and by in situ hybridization confirmed the reliability of this approach.


Subject(s)
Cartilage/metabolism , Chondrocytes/metabolism , Gene Expression Profiling/methods , Growth Plate/metabolism , Oligonucleotide Array Sequence Analysis/methods , RNA, Messenger/metabolism , Animals , Cartilage/cytology , Cartilage/growth & development , Chondrocytes/cytology , Growth Plate/cytology , Growth Plate/growth & development , In Situ Hybridization , Mice , Microdissection , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
7.
BMC Dev Biol ; 9: 20, 2009 Mar 10.
Article in English | MEDLINE | ID: mdl-19272164

ABSTRACT

BACKGROUND: During vertebrate embryogenesis the initial stages of bone formation by endochondral ossification involve the aggregation and proliferation of mesenchymal cells into condensations. Continued growth of the condensations and differentiation of the mesenchymal cells into chondrocytes results in the formation of cartilage templates, or anlagen, which prefigure the shape of the future bones. The chondrocytes in the anlagen further differentiate by undergoing a complex sequence of maturation and hypertrophy, and are eventually replaced by mineralized bone. Regulation of the onset of chondrogenesis is incompletely understood, and would be informed by comprehensive analyses of in vivo gene expression. RESULTS: Tibial and fibular pre-condensed mesenchyme was microdissected from mouse hind limbs at 11.5 dpc, and the corresponding condensations at 12.5 dpc and cartilage anlagen at 13.5 dpc. Total RNA was isolated, and cRNA generated by linear amplification was interrogated using mouse whole genome microarrays. Differential expression was validated by quantitative PCR for Agc1, Bmp8a, Col2a1, Fgfr4, Foxa3, Gdf5, Klf2, Klf4, Lepre1, Ncad, Sox11, and Trpv4. Further, independent validation of the microarray data was achieved by in situ hybridization to analyse the expression of Lepre1, Pcdh8, Sox11, and Trpv4 from 11.5 dpc to 13.5 dpc during mouse hind limb development. We found significant differential expression of 931 genes during these early stages of chondrogenesis. Of these, 380 genes were down-regulated and 551 up-regulated. Our studies characterized the expression pattern of gene families previously associated with chondrogenesis, such as adhesion molecules, secreted signalling molecules, transcription factors, and extracellular matrix components. Gene ontology approaches identified 892 differentially expressed genes not previously identified during the initiation of chondrogenesis. These included several Bmp, Gdf, Wnt, Sox and Fox family members. CONCLUSION: These data represent the first global gene expression profiling analysis of chondrogenic tissues during in vivo development. They identify genes for further study on their functional roles in chondrogenesis, and provide a comprehensive and important resource for future studies on cartilage development and disease.


Subject(s)
Chondrogenesis , Embryonic Development , Gene Expression Regulation, Developmental , Limb Buds/embryology , Animals , Gene Expression Profiling , Kruppel-Like Factor 4 , Organogenesis
8.
Bone ; 116: 67-77, 2018 11.
Article in English | MEDLINE | ID: mdl-30010080

ABSTRACT

OBJECTIVE: Osteoarthritis (OA) is a joint disease characterized by progressive degeneration of articular cartilage. Some features of OA, including chondrocyte hypertrophy and focal calcification of articular cartilage, resemble the endochondral ossification processes. Alterations in transforming growth factor ß (TGFß) signaling have been associated with OA as well as with chondrocyte hypertrophy. Our aim was to identify novel candidate genes implicated in chondrocyte hypertrophy during OA pathogenesis by determining which TGFß-related genes are regulated during murine OA and endochondral ossification. METHODS: A list of 580 TGFß-related genes, including TGFß signaling pathway components and TGFß-target genes, was generated. Regulation of these TGFß-related genes was assessed in a microarray of murine OA cartilage: 1, 2 and 6 weeks after destabilization of the medial meniscus (DMM). Subsequently, genes regulated in the DMM model were studied in two independent murine microarray datasets on endochondral ossification: the growth plate and transient embryonic cartilage (joint development). RESULTS: A total of 106 TGFß-related genes were differentially expressed in articular cartilage of DMM-operated mice compared to sham-control. From these genes, 43 were similarly regulated during chondrocyte hypertrophy in the growth plate or embryonic joint development. Among these 43 genes, 18 genes have already been associated with OA. The remaining 25 genes were considered as novel candidate genes involved in OA pathogenesis and endochondral ossification. In supplementary data of published human OA microarrays we found indications that 15 of the 25 novel genes are indeed regulated in articular cartilage of human OA patients. CONCLUSION: By focusing on TGFß-related genes during OA and chondrocyte hypertrophy in mice, we identified 18 known and 25 new candidate genes potentially implicated in phenotypical changes in chondrocytes leading to OA. We propose that 15 of these candidates warrant further investigation as therapeutic target for OA as they are also regulated in articular cartilage of OA patients.


Subject(s)
Chondrocytes/metabolism , Chondrocytes/pathology , Databases, Genetic , Gene Expression Regulation , Oligonucleotide Array Sequence Analysis , Osteoarthritis/genetics , Osteoarthritis/pathology , Transforming Growth Factor beta/metabolism , Animals , Cell Line , Disease Models, Animal , Hypertrophy , Joints/metabolism , Male , Mice, Inbred C57BL , Reproducibility of Results
9.
J Mol Diagn ; 19(4): 602-612, 2017 07.
Article in English | MEDLINE | ID: mdl-28502726

ABSTRACT

The sensitivity and specificity of next-generation sequencing laboratory developed tests (LDTs) are typically determined by an analyte-specific approach. Analyte-specific validations use disease-specific controls to assess an LDT's ability to detect known pathogenic variants. Alternatively, a methods-based approach can be used for LDT technical validations. Methods-focused validations do not use disease-specific controls but use benchmark reference DNA that contains known variants (benign, variants of unknown significance, and pathogenic) to assess variant calling accuracy of a next-generation sequencing workflow. Recently, four whole-genome reference materials (RMs) from the National Institute of Standards and Technology (NIST) were released to standardize methods-based validations of next-generation sequencing panels across laboratories. We provide a practical method for using NIST RMs to validate multigene panels. We analyzed the utility of RMs in validating a novel newborn screening test that targets 70 genes, called NEO1. Despite the NIST RM variant truth set originating from multiple sequencing platforms, replicates, and library types, we discovered a 5.2% false-negative variant detection rate in the RM truth set genes that were assessed in our validation. We developed a strategy using complementary non-RM controls to demonstrate 99.6% sensitivity of the NEO1 test in detecting variants. Our findings have implications for laboratories or proficiency testing organizations using whole-genome NIST RMs for testing.


Subject(s)
Genome, Human , High-Throughput Nucleotide Sequencing/methods , Multigene Family , DNA Copy Number Variations , Gene Deletion , Genetic Variation , High-Throughput Nucleotide Sequencing/standards , Humans , Infant, Newborn , Infant, Newborn, Diseases/genetics , Mutagenesis, Insertional , Polymorphism, Single Nucleotide , Reference Standards , Validation Studies as Topic
10.
Med Sci (Basel) ; 5(4)2017 Oct 10.
Article in English | MEDLINE | ID: mdl-29099038

ABSTRACT

Sudden cardiac death (SCD) in people before the age of 35 years is a devastating event for any family. The causes of SCD in the young can be broadly divided into two groups: heritable cardiac disorders that affect the heart structure (cardiomyopathies) and primary electrical disorders (cardiac ion channelopathies). Genetic testing is vital as those suffering from cardiac ion channelopathies have structurally normal hearts, and those with cardiomyopathies may only show subtle abnormalities in the heart and these signs may not be detected during an autopsy. Post-mortem genetic testing of SCD victims is important to identify the underlying genetic cause. This is important as family cascade screening may be undertaken to identify those who may be at risk and provide vital information about risk stratification and clinical management. The development of massively parallel sequencing (MPS) has made it possible for the simultaneous screening of multiple patients for hundreds of genes. In light of this, we opted to develop an MPS approach for SCD analysis that would allow us to screen for mutations in genes implicated in cardiomyopathies and cardiac ion channelopathies. The rationale behind this panel was to limit it to genes carrying the greatest mutation load. If no likely pathogenic gene variant were found then testing could cascade to whole exome/genome sequencing as a gene-discovery exercise. The overarching aim was to design and validate a custom-cardiac panel that satisfies the diagnostic requirements of LabPLUS (Auckland City Hospital, Auckland, NZ) and the guidelines provided by the Royal College of Pathologists of Australasia and the Association for Clinical Genetic Science.

11.
PLoS One ; 8(3): e58154, 2013.
Article in English | MEDLINE | ID: mdl-23505463

ABSTRACT

Biomineralization of the extracellular matrix occurs inappropriately in numerous pathological conditions such as cancer and vascular disease, but during normal mammalian development calcification is restricted to the formation of the skeleton and dentition. The comprehensive study of gene expression in mineralized skeletal tissues has been compromized by the traditional decalcification/fixation methods that result in significant mRNA degradation. In this study we developed a novel RNAlater/EDTA decalcification method that protects the integrity of the mRNA in mature mouse tibial epiphyses. Furthermore, this method preserves the tissue structure to allow histological sectioning and microdissection to determine region-specific gene expression, in addition to immuno- and in situ histology. This method will be widely applicable to the molecular analysis of calcified tissues in various pathological conditions, and will be of particular importance in dissection of the gene expression in mouse bone and joint tissues during development and in important clinical conditions such as arthritis.


Subject(s)
Calcification, Physiologic , RNA, Messenger/metabolism , Animals , Cartilage/metabolism , Gene Expression , Immunohistochemistry , Male , Mice , RNA Stability
12.
Nat Genet ; 43(11): 1142-6, 2011 Oct 02.
Article in English | MEDLINE | ID: mdl-21964574

ABSTRACT

Familial digital arthropathy-brachydactyly (FDAB) is a dominantly inherited condition that is characterized by aggressive osteoarthropathy of the fingers and toes and consequent shortening of the middle and distal phalanges. Here we show in three unrelated families that FDAB is caused by mutations encoding p.Gly270Val, p.Arg271Pro and p.Phe273Leu substitutions in the intracellular ankyrin-repeat domain of the cation channel TRPV4. Functional testing of mutant TRPV4 in HEK-293 cells showed that the mutant proteins have poor cell-surface localization. Calcium influx in response to the synthetic TRPV4 agonists GSK1016790A and 4αPDD was significantly reduced, and mutant channels did not respond to hypotonic stress. Others have shown that gain-of-function TRPV4 mutations cause skeletal dysplasias and peripheral neuropathies. Our data indicate that TRPV4 mutations that reduce channel activity cause a third phenotype, inherited osteoarthropathy, and show the importance of TRPV4 activity in articular cartilage homeostasis. Our data raise the possibility that TRPV4 may also have a role in age- or injury-related osteoarthritis.


Subject(s)
Mutation , TRPV Cation Channels/genetics , Cell Line , Humans , TRPV Cation Channels/physiology
13.
Arthritis Res Ther ; 12(1): R16, 2010.
Article in English | MEDLINE | ID: mdl-20105291

ABSTRACT

INTRODUCTION: The objective was to evaluate the changes in S100A8 S100A9, and their complex (S100A8/S100A9) in cartilage during the onset of osteoarthritis (OA) as opposed to inflammatory arthritis. METHODS: S100A8 and S100A9 protein localization were determined in antigen-induced inflammatory arthritis in mice, mouse femoral head cartilage explants stimulated with interleukin-1 (IL-1), and in surgically-induced OA in mice. Microarray expression profiling of all S100 proteins in cartilage was evaluated at different times after initiation of degradation in femoral head explant cultures stimulated with IL-1 and surgically-induced OA. The effect of S100A8, S100A9 or the complex on the expression of aggrecan (Acan), collagen II (Col2a1), disintegrin and metalloproteases with thrombospondin motifs (Adamts1, Adamts 4 &Adamts 5), matrix metalloproteases (Mmp1, Mmp3, Mmp13 &Mmp14) and tissue inhibitors of metalloproteinases (Timp1, Timp2 &Timp3), by primary adult ovine articular chondrocytes was determined using real time quantitative reverse transcription polymerase chain reaction (qRT-PCR). RESULTS: Stimulation with IL-1 increased chondrocyte S100a8 and S100a9 mRNA and protein levels. There was increased chondrocyte mRNA expression of S100a8 and S100a9 in early but not late mouse OA. However, loss of the S100A8 staining in chondrocytes occurred as mouse OA progressed, in contrast to the positive reactivity for both S100A8 and S100A9 in chondrocytes in inflammatory arthritis in mice. Homodimeric S100A8 and S100A9, but not the heterodimeric complex, significantly upregulated chondrocyte Adamts1, Adamts4 and Adamts 5, Mmp1, Mmp3 and Mmp13 gene expression, while collagen II and aggrecan mRNAs were significantly decreased. CONCLUSIONS: Chondrocyte derived S100A8 and S100A9 may have a sustained role in cartilage degradation in inflammatory arthritis. In contrast, while these proteins may have a role in initiating early cartilage degradation in OA by upregulating MMPs and aggrecanases, their reduced expression in late stages of OA suggests they do not have an ongoing role in cartilage degradation in this non-inflammatory arthropathy.


Subject(s)
Arthritis, Experimental/metabolism , Calgranulin B/biosynthesis , Osteoarthritis/metabolism , S100 Proteins/biosynthesis , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Calgranulin A , Cartilage/metabolism , Cartilage/pathology , Chondrocytes/metabolism , Gene Expression , Gene Expression Profiling , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Osteoarthritis/genetics , Osteoarthritis/pathology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
14.
J Bone Miner Res ; 25(1): 141-53, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19580468

ABSTRACT

Initiation of mineralization during endochondral ossification is a multistep process and has been assumed to correlate with specific interactions of annexins A5 and A6 and collagens. However, skeletal development appears to be normal in mice deficient for either A5 or A6, and the highly conserved structures led to the assumption that A5 and A6 may fulfill redundant functions. We have now generated mice deficient of both proteins. These mice were viable and fertile and showed no obvious abnormalities. Assessment of skeletal elements using histologic, ultrastructural, and peripheral quantitative computed tomographic methods revealed that mineralization and development of the skeleton were not significantly affected in mutant mice. Otherwise, global gene expression analysis showed subtle changes at the transcriptome level of genes involved in cell growth and intermediate metabolism. These results indicate that annexins A5 and A6 may not represent the essential annexins that promote mineralization in vivo.


Subject(s)
Annexin A5/deficiency , Annexin A6/deficiency , Calcification, Physiologic/genetics , Cartilage/metabolism , Gene Expression Profiling , Growth Plate/metabolism , Animals , Animals, Newborn , Annexin A5/genetics , Annexin A5/metabolism , Annexin A6/genetics , Annexin A6/metabolism , Antibody Specificity , Bone Development/genetics , Bone Matrix/metabolism , Bone Matrix/ultrastructure , Cartilage/ultrastructure , Cell Proliferation , Collagen/metabolism , Cytoplasmic Vesicles/metabolism , Cytoplasmic Vesicles/ultrastructure , Femur/growth & development , Femur/metabolism , Femur/ultrastructure , Gene Expression Regulation, Developmental , Growth Plate/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Mutant Strains
15.
J Bone Miner Res ; 25(6): 1267-81, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20200945

ABSTRACT

Axial growth of long bones occurs through a coordinated process of growth plate chondrocyte proliferation and differentiation. This maturation of chondrocytes is reflected in a zonal change in gene expression and cell morphology from resting to proliferative, prehypertrophic, and hypertrophic chondrocytes of the growth plate followed by ossification. A major experimental limitation in understanding growth plate biology and pathophysiology is the lack of a robust technique to isolate cells from the different zones, particularly from small animals. Here, we report on a new strategy for separating distinct chondrocyte populations from mouse growth plates. By transcriptome profiling of microdissected zones of growth plates, we identified novel, zone-specific cell surface markers and used these for flow cytometry and immunomagnetic cell separation to quantify, enrich, and characterize chondrocytes populations with respect to their differentiation status. This approach provides a novel platform to study cartilage development and characterize mouse growth plate chondrocytes to reveal unique cellular phenotypes of the distinct subpopulations within the growth plate.


Subject(s)
Cell Differentiation , Cell Separation/methods , Chondrocytes/cytology , Flow Cytometry/methods , Growth Plate/cytology , Animals , Antigens, Surface/metabolism , Biomarkers/metabolism , Cell Death , Cell Membrane/metabolism , Chondrocytes/metabolism , Growth Plate/growth & development , Immunomagnetic Separation , Mice , Mice, Inbred C57BL , Microspheres , Organ Specificity , Phenotype , Reproducibility of Results
16.
Methods ; 45(1): 22-31, 2008 May.
Article in English | MEDLINE | ID: mdl-18442702

ABSTRACT

While the analysis of the cartilage proteome is important for our comprehensive understanding of the development and disease of this important tissue, several unique features of cartilage present some technical obstacles. Firstly, cartilage is difficult to obtain in adequate quantities for many protein analyses, especially from mice which are otherwise powerful experimental models. Furthermore, the cartilage extracellular matrix contains an insoluble network of collagen II-containing fibrils that are integrated within an abundant anionic network of aggrecan and hyaluronan aggregates. These interacting networks provide a structural scaffold for the covalent and non-covalent attachment of other proteins and glycoproteins. Consequently, proteomic analysis of cartilage requires extraction of proteins with chaotropic agents to achieve and significant protein solubilization. Finally, isolated chondrocytes are phenotypically unstable, which requires rapid isolation of cells or the use of specific culture conditions. Despite these problems, recent improvements in the sensitivity and reproducibility of two-dimensional electrophoresis (2-DE) and tandem mass spectrometry (MS/MS) techniques, combined with improved tissue preparation and sample pre-fractionation approaches, have made the proteomic characterization of cartilage tissues possible. Here we review the approaches that have been used and describe in detail protocols for the proteomic analysis of cartilage tissues and cells.


Subject(s)
Cartilage/chemistry , Proteins/analysis , Proteomics/methods , Animals , Mice , Proteins/chemistry
17.
Arthritis Rheum ; 58(10): 3120-31, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18821673

ABSTRACT

OBJECTIVE: To develop proteomics to analyze mouse cartilage degradation and correlate transcriptional and translational responses to catabolic stimuli. METHODS: Proteomic techniques were used to analyze catabolism in mouse femoral head cartilage. Using specific methods to prepare cartilage extracts and conditioned media for 2-dimensional polyacrylamide gel electrophoresis and subsequent tandem mass spectrometry, we identified novel proteins and fragments released into the media of control, interleukin-1alpha (IL-1alpha)-treated, and all-trans-retinoic acid (RetA)-treated explants. Fluorescence 2-dimensional difference gel electrophoresis was used to quantify protein expression changes. We also measured changes in messenger RNA (mRNA) expression to distinguish transcriptional and posttranslational regulation of released proteins. RESULTS: Differentially abundant proteins in the media of control and treated explants included fragments of thrombospondin 1 and connective tissue growth factor. IL-1alpha stimulated release of the cartilage degeneration marker matrix metalloproteinase 3, as well as proteins with uncharacterized roles in cartilage pathology, such as neutrophil gelatinase-associated lipocalin. RetA stimulated release of the extracellular matrix proteins cartilage oligomeric matrix protein, link protein, and matrilin-3 into the media, which was accompanied by a dramatic reduction in the corresponding mRNA transcript levels. Gelsolin, which has been implicated in cytoskeletal reorganization in arthritis synovial fibroblasts but has not been previously associated with cartilage pathology, was regulated by IL-1alpha and RetA. CONCLUSION: In this first analysis of mouse cartilage degradation and protein release using proteomics, we identified proteins and fragments, some of which represent novel candidate biomarkers for cartilage degradation. Applying these proteomic techniques to wild-type and genetically modified mouse cartilage will provide insights into the mechanisms of cartilage degeneration.


Subject(s)
Cartilage/metabolism , Femur Head/metabolism , Gene Expression Profiling , Proteomics , Animals , Mice , Tissue Culture Techniques
18.
Proteomics ; 6(24): 6549-53, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17163436

ABSTRACT

Cartilage is a highly specialized load-bearing tissue with a small number of cells and a high proportion of extracellular matrix (ECM). The abundance of heavily sulfated proteoglycans and a poorly soluble collagenous ECM presents a major technical challenge to 2-DE. Here we report proteomic analysis of mouse growth plate cartilage using novel methodology for tissue dissection and sample prefractionation. We have successfully resolved cartilage tissue extracts by 2-DE for the first time and identified cartilage ECM proteins by Western blotting and MS/MS.


Subject(s)
Growth Plate/metabolism , Proteomics/methods , Animals , Blotting, Western , Electrophoresis, Gel, Two-Dimensional , Extracellular Matrix Proteins/isolation & purification , Extracellular Matrix Proteins/metabolism , Growth Plate/chemistry , Male , Mice , Proteome/isolation & purification , Proteome/metabolism , Tandem Mass Spectrometry
19.
Arthritis Rheum ; 52(5): 1461-72, 2005 May.
Article in English | MEDLINE | ID: mdl-15880348

ABSTRACT

OBJECTIVE: To determine the role of the proteinase ADAMTS-1 in normal and accelerated catabolism of aggrecan in articular and growth plate cartilage of mice. METHODS: Expression of ADAMTS-1 was determined using reverse transcriptase-polymerase chain reaction (RT-PCR) analysis of RNA isolated from microdissected chondrocytes from different zones of mouse growth plate and articular cartilage. Real-time RT-PCR for ADAMTS-4, ADAMTS-5, and ADAMTS-9 was performed on femoral head cartilage of wild-type (WT) and ADAMTS-1-knockout (KO) mice. Histologic and immunohistologic evaluation of growth plate and articular cartilage was performed in WT and KO mice from birth to 12 weeks of age. The effect of ADAMTS-1 ablation on cartilage proteoglycan loss was studied in antigen-induced arthritis (AIA). Aggrecan catabolism in WT and KO mice was studied in an in vitro model of cartilage degradation, by quantitation of glycosaminoglycan loss and histologic, immunohistologic, and Western immunoblot analyses. RESULTS: ADAMTS-1 messenger RNA (mRNA) was expressed in normal mouse articular and growth plate cartilage and was up-regulated in terminal hypertrophic differentiation of growth plate chondrocytes. There was no difference in mRNA levels in the cartilage of WT compared with KO mice for the other potential aggrecanases ADAMTS-4, ADAMTS-5, or ADAMTS-9. ADAMTS-1-KO mice were significantly smaller than their WT littermates; however, no morphologic differences between the genotypes were evident in growth plate or articular cartilage from birth to skeletal maturity (12-16 weeks). Similarly, no difference in cartilage aggrecan content or presence of aggrecan degradation products was detected between WT and KO mice. There was no difference between WT and KO mice in the degree of synovial inflammation or depletion of cartilage aggrecan in AIA. There was no difference between WT and KO cartilage in either basal or stimulated aggrecan loss in vitro; however, subtle changes in the aggrecanase-generated aggrecan catabolites were observed in interleukin-1-treated cartilage. CONCLUSION: Although ADAMTS-1 is expressed in articular and growth plate cartilage and is able to cleave aggrecan at physiologically relevant sites, our results indicate that it does not play a significant nonredundant role in normal cartilage and bone development and growth. Similarly, ablation of ADAMTS-1 offered no protection from accelerated aggrecanolysis in an inflammatory model of arthritis or in an in vitro model of early cartilage degradation. ADAMTS-1 does not appear to be a viable target for treatment of cartilage destruction in arthritis.


Subject(s)
Cartilage/metabolism , Extracellular Matrix Proteins/metabolism , Proteoglycans/metabolism , ADAM Proteins , ADAMTS1 Protein , Aggrecans , Animals , Arthritis/etiology , Bone Development , Disintegrins/genetics , Extracellular Matrix Proteins/biosynthesis , Growth Plate/metabolism , In Vitro Techniques , Joints/growth & development , Lectins, C-Type , Metalloendopeptidases/genetics , Mice , Mice, Knockout , Proteoglycans/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL