Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Elife ; 112022 05 03.
Article in English | MEDLINE | ID: mdl-35503090

ABSTRACT

The embryonic extracellular matrix (ECM) undergoes transition to mature ECM as development progresses, yet few mechanisms ensuring ECM proteostasis during this period are known. Fibrillin microfibrils are macromolecular ECM complexes serving structural and regulatory roles. In mice, Fbn1 and Fbn2, encoding the major microfibrillar components, are strongly expressed during embryogenesis, but fibrillin-1 is the major component observed in adult tissue microfibrils. Here, analysis of Adamts6 and Adamts10 mutant mouse embryos, lacking these homologous secreted metalloproteases individually and in combination, along with in vitro analysis of microfibrils, measurement of ADAMTS6-fibrillin affinities and N-terminomics discovery of ADAMTS6-cleaved sites, identifies a proteostatic mechanism contributing to postnatal fibrillin-2 reduction and fibrillin-1 dominance. The lack of ADAMTS6, alone and in combination with ADAMTS10 led to excess fibrillin-2 in perichondrium, with impaired skeletal development defined by a drastic reduction of aggrecan and cartilage link protein, impaired BMP signaling in cartilage, and increased GDF5 sequestration in fibrillin-2-rich tissue. Although ADAMTS6 cleaves fibrillin-1 and fibrillin-2 as well as fibronectin, which provides the initial scaffold for microfibril assembly, primacy of the protease-substrate relationship between ADAMTS6 and fibrillin-2 was unequivocally established by reversal of the defects in Adamts6-/- embryos by genetic reduction of Fbn2, but not Fbn1.


Subject(s)
ADAMTS Proteins , Microfibrils , ADAMTS Proteins/chemistry , ADAMTS Proteins/genetics , ADAMTS Proteins/metabolism , Animals , Fibrillin-1/genetics , Fibrillin-2/metabolism , Fibrillins/metabolism , Mice , Microfibrils/metabolism , Proteolysis
2.
J Biol Chem ; 294(25): 9924-9936, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31085586

ABSTRACT

The secreted metalloprotease ADAMTS9 has dual roles in extracellular matrix (ECM) turnover and biogenesis of the primary cilium during mouse embryogenesis. Its gene locus is associated with several human traits and disorders, but ADAMTS9 has few known interacting partners or confirmed substrates. Here, using a yeast two-hybrid screen for proteins interacting with its C-terminal Gon1 domain, we identified three putative ADAMTS9-binding regions in the ECM glycoprotein fibronectin. Using solid-phase binding assays and surface plasmon resonance experiments with purified proteins, we demonstrate that ADAMTS9 and fibronectin interact. ADAMTS9 constructs, including those lacking Gon1, co-localized with fibronectin fibrils formed by cultured fibroblasts lacking fibrillin-1, which co-localizes with fibronectin and binds several ADAMTSs. We observed no fibrillar ADAMTS9 staining after blockade of fibroblast fibronectin fibrillogenesis with a peptide based on the functional upstream domain of a Staphylococcus aureus adhesin. These findings indicate that ADAMTS9 binds fibronectin dimers and fibrils directly through multiple sites in both molecules. Proteolytically active ADAMTS9, but not a catalytically inactive variant, disrupted fibronectin fibril networks formed by fibroblasts in vitro, and ADAMTS9-deficient RPE1 cells assembled a robust fibronectin fibril network, unlike WT cells. Targeted LC-MS analysis of fibronectin digested by ADAMTS9-expressing cells identified a semitryptic peptide arising from cleavage at Gly2196-Leu2197 We noted that this scissile bond is in the linker between fibronectin modules III17 and I10, a region targeted also by other proteases. These findings, along with stronger fibronectin staining previously observed in Adamts9 mutant embryos, suggest that ADAMTS9 contributes to fibronectin turnover during ECM remodeling.


Subject(s)
ADAMTS9 Protein/metabolism , Fibroblasts/metabolism , Fibronectins/chemistry , Fibronectins/metabolism , Protein Aggregates , ADAMTS9 Protein/genetics , Animals , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Fibronectins/genetics , Humans , Mice , Proteolysis , Retinal Pigment Epithelium/metabolism , Two-Hybrid System Techniques
3.
Nat Commun ; 10(1): 953, 2019 02 27.
Article in English | MEDLINE | ID: mdl-30814516

ABSTRACT

Although hundreds of cytosolic or transmembrane molecules form the primary cilium, few secreted molecules are known to contribute to ciliogenesis. Here, homologous secreted metalloproteases ADAMTS9 and ADAMTS20 are identified as ciliogenesis regulators that act intracellularly. Secreted and furin-processed ADAMTS9 bound heparan sulfate and was internalized by LRP1, LRP2 and clathrin-mediated endocytosis to be gathered in Rab11 vesicles with a unique periciliary localization defined by super-resolution microscopy. CRISPR-Cas9 inactivation of ADAMTS9 impaired ciliogenesis in RPE-1 cells, which was restored by catalytically active ADAMTS9 or ADAMTS20 acting in trans, but not by their proteolytically inactive mutants. Their mutagenesis in mice impaired neural and yolk sac ciliogenesis, leading to morphogenetic anomalies resulting from impaired hedgehog signaling, which is transduced by primary cilia. In addition to their cognate extracellular proteolytic activity, ADAMTS9 and ADAMTS20 thus have an additional proteolytic role intracellularly, revealing an unexpected regulatory dimension in ciliogenesis.


Subject(s)
ADAMTS Proteins/metabolism , ADAMTS9 Protein/metabolism , Cilia/metabolism , Cilia/ultrastructure , ADAMTS Proteins/deficiency , ADAMTS Proteins/genetics , ADAMTS9 Protein/deficiency , ADAMTS9 Protein/genetics , Animals , Cell Line , Endocytosis , Gene Knockout Techniques , Humans , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Electron, Scanning , Models, Biological , Mutation , Neural Tube Defects/embryology , Neural Tube Defects/genetics , Neural Tube Defects/metabolism , Proteolysis , Signal Transduction , Versicans/genetics , Versicans/metabolism , Yolk Sac/embryology , Yolk Sac/metabolism
4.
Matrix Biol ; 77: 117-128, 2019 04.
Article in English | MEDLINE | ID: mdl-30201140

ABSTRACT

Mutations in the secreted metalloproteinase ADAMTS10 cause recessive Weill-Marchesani syndrome (WMS), comprising ectopia lentis, short stature, brachydactyly, thick skin and cardiac valve anomalies. Dominant WMS caused by FBN1 mutations is clinically similar and affects fibrillin-1 microfibrils, which are a major component of the ocular zonule. ADAMTS10 was previously shown to enhance fibrillin-1 assembly in vitro. Here, Adamts10 null mice were analyzed to determine the impact of ADAMTS10 deficiency on fibrillin microfibrils in vivo. An intragenic lacZ reporter identified widespread Adamts10 expression in the eye, musculoskeletal tissues, vasculature, skin and lung. Adamts10-/- mice had reduced viability on the C57BL/6 background, and although surviving mice were slightly smaller and had stiff skin, they lacked brachydactyly and cardiovascular defects. Ectopia lentis was not observed in Adamts10-/- mice, similar to Fbn1-/- mice, most likely because the mouse zonule contains fibrillin-2 in addition to fibrillin-1. Unexpectedly, in contrast to wild-type eyes, Adamts10-/- zonule fibers were thicker and immunostained strongly with fibrillin-2 antibodies into adulthood, whereas fibrillin-1 staining was reduced. Furthermore, fibrillin-2 staining of hyaloid vasculature remnants persisted post-natally in Adamts10-/- eyes. ADAMTS10 was found to cleave fibrillin-2, providing an explanation for persistence of fibrillin-2 at these sites. Thus, analysis of Adamts10-/- mice led to identification of fibrillin-2 as a novel ADAMTS10 substrate and defined a proteolytic mechanism for clearance of ocular fibrillin-2 at the end of the juvenile period.


Subject(s)
ADAMTS Proteins/genetics , Eye/metabolism , Fibrillin-1/genetics , Fibrillin-2/genetics , Microfibrils/metabolism , Weill-Marchesani Syndrome/genetics , ADAMTS Proteins/deficiency , Animals , Blood Vessels/growth & development , Blood Vessels/metabolism , Blood Vessels/pathology , Disease Models, Animal , Eye/growth & development , Eye/pathology , Female , Fibrillin-1/metabolism , Fibrillin-2/metabolism , Gene Expression Regulation, Developmental , Genes, Reporter , HEK293 Cells , Humans , Lac Operon , Lung/growth & development , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfibrils/pathology , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Proteolysis , Signal Transduction , Skin/growth & development , Skin/metabolism , Skin/pathology , Weill-Marchesani Syndrome/metabolism , Weill-Marchesani Syndrome/pathology
5.
Sci Rep ; 6: 33974, 2016 Sep 30.
Article in English | MEDLINE | ID: mdl-27687499

ABSTRACT

Peters Plus syndrome (PPS), a congenital disorder of glycosylation, results from recessive mutations affecting the glucosyltransferase B3GLCT, leading to congenital corneal opacity and diverse extra-ocular manifestations. Together with the fucosyltransferase POFUT2, B3GLCT adds Glucoseß1-3Fucose disaccharide to a consensus sequence in thrombospondin type 1 repeats (TSRs) of several proteins. Which of these target proteins is functionally compromised in PPS is unknown. We report here that haploinsufficiency of murine Adamts9, encoding a secreted metalloproteinase with 15 TSRs, leads to congenital corneal opacity and Peters anomaly (persistent lens-cornea adhesion), which is a hallmark of PPS. Mass spectrometry of recombinant ADAMTS9 showed that 9 of 12 TSRs with the O-fucosylation consensus sequence carried the Glucoseß1-3Fucose disaccharide and B3GLCT knockdown reduced ADAMTS9 secretion in HEK293F cells. Together, the genetic and biochemical findings imply a dosage-dependent role for ADAMTS9 in ocular morphogenesis. Reduced secretion of ADAMTS9 in the absence of B3GLCT is proposed as a mechanism of Peters anomaly in PPS. The functional link between ADAMTS9 and B3GLCT established here also provides credence to their recently reported association with age-related macular degeneration.

6.
Dis Model Mech ; 8(5): 487-99, 2015 May.
Article in English | MEDLINE | ID: mdl-25762570

ABSTRACT

Mutations in the secreted glycoprotein ADAMTSL2 cause recessive geleophysic dysplasia (GD) in humans and Musladin-Lueke syndrome (MLS) in dogs. GD is a severe, often lethal, condition presenting with short stature, brachydactyly, stiff skin, joint contractures, tracheal-bronchial stenosis and cardiac valve anomalies, whereas MLS is non-lethal and characterized by short stature and severe skin fibrosis. Although most mutations in fibrillin-1 (FBN1) cause Marfan syndrome (MFS), a microfibril disorder leading to transforming growth factor-ß (TGFß) dysregulation, domain-specific FBN1 mutations result in dominant GD. ADAMTSL2 has been previously shown to bind FBN1 and latent TGFß-binding protein-1 (LTBP1). Here, we investigated mice with targeted Adamtsl2 inactivation as a new model for GD (Adamtsl2(-/-) mice). An intragenic lacZ reporter in these mice showed that ADAMTSL2 was produced exclusively by bronchial smooth muscle cells during embryonic lung development. Adamtsl2(-/-) mice, which died at birth, had severe bronchial epithelial dysplasia with abnormal glycogen-rich inclusions in bronchial epithelium resembling the cellular anomalies described previously in GD. An increase in microfibrils in the bronchial wall was associated with increased FBN2 and microfibril-associated glycoprotein-1 (MAGP1) staining, whereas LTBP1 staining was increased in bronchial epithelium. ADAMTSL2 was shown to bind directly to FBN2 with an affinity comparable to FBN1. The observed extracellular matrix (ECM) alterations were associated with increased bronchial epithelial TGFß signaling at 17.5 days of gestation; however, treatment with TGFß-neutralizing antibody did not correct the epithelial dysplasia. These investigations reveal a new function of ADAMTSL2 in modulating microfibril formation, and a previously unsuspected association with FBN2. Our studies suggest that the bronchial epithelial dysplasia accompanying microfibril dysregulation in Adamtsl2(-/-) mice cannot be reversed by TGFß neutralization, and thus might be mediated by other mechanisms.


Subject(s)
Bone Diseases, Developmental/pathology , Bronchi/pathology , Epithelium/pathology , Extracellular Matrix Proteins/metabolism , Gene Deletion , Limb Deformities, Congenital/pathology , Microfibrils/metabolism , Microfilament Proteins/metabolism , ADAMTS Proteins , Animals , Animals, Newborn , Bone Diseases, Developmental/metabolism , Bronchi/ultrastructure , Cellular Microenvironment , Disease Models, Animal , Epithelium/metabolism , Epithelium/ultrastructure , Extracellular Matrix/metabolism , Fibrillin-1 , Fibrillin-2 , Fibrillins , Glycogen/metabolism , Limb Deformities, Congenital/metabolism , Mice, Inbred C57BL , Protein Binding , Signal Transduction , Transforming Growth Factor beta/metabolism
7.
Mol Cancer Ther ; 13(5): 1259-69, 2014 May.
Article in English | MEDLINE | ID: mdl-24634412

ABSTRACT

Hedgehog (Hh) pathway inhibition in cancer has been evaluated in both the ligand-independent and ligand-dependent settings, where Hh signaling occurs either directly within the cancer cells or within the nonmalignant cells of the tumor microenvironment. Chondrosarcoma is a malignant tumor of cartilage in which there is ligand-dependent activation of Hh signaling. IPI-926 is a potent, orally delivered small molecule that inhibits Hh pathway signaling by binding to Smoothened (SMO). Here, the impact of Hh pathway inhibition on primary chondrosarcoma xenografts was assessed. Mice bearing primary human chondrosarcoma xenografts were treated with IPI-926. The expression levels of known Hh pathway genes, in both the tumor and stroma, and endpoint tumor volumes were measured. Gene expression profiling of tumors from IPI-926-treated mice was conducted to identify potential novel Hh target genes. Hh target genes were studied to determine their contribution to the chondrosarcoma neoplastic phenotype. IPI-926 administration results in downmodulation of the Hh pathway in primary chondrosarcoma xenografts, as demonstrated by evaluation of the Hh target genes GLI1 and PTCH1, as well as inhibition of tumor growth. Chondrosarcomas exhibited autocrine and paracrine Hh signaling, and both were affected by IPI-926. Decreased tumor growth is accompanied by histopathologic changes, including calcification and loss of tumor cells. Gene profiling studies identified genes differentially expressed in chondrosarcomas following IPI-926 treatment, one of which, ADAMTSL1, regulates chondrosarcoma cell proliferation. These studies provide further insight into the role of the Hh pathway in chondrosarcoma and provide a scientific rationale for targeting the Hh pathway in chondrosarcoma.


Subject(s)
Chondrosarcoma/metabolism , Hedgehog Proteins/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction/drug effects , Veratrum Alkaloids/pharmacology , ADAMTS Proteins , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Calcinosis/drug therapy , Calcinosis/genetics , Calcinosis/metabolism , Cell Line, Tumor , Cell Proliferation , Chondrosarcoma/genetics , Chondrosarcoma/pathology , Disease Models, Animal , Extracellular Matrix Proteins/genetics , Humans , Mice , Smoothened Receptor , Tumor Burden/drug effects , Veratrum Alkaloids/administration & dosage , Xenograft Model Antitumor Assays
8.
Mol Cell Proteomics ; 13(2): 580-93, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24281761

ABSTRACT

Proteolysis is an irreversible post-translational modification that affects intra- and intercellular communication by modulating the activity of bioactive mediators. Key to understanding protease function is the system-wide identification of cleavage events and their dynamics in physiological contexts. Despite recent advances in mass spectrometry-based proteomics for high-throughput substrate screening, current approaches suffer from high false positive rates and only capture single states of protease activity. Here, we present a workflow based on multiplexed terminal amine isotopic labeling of substrates for time-resolved substrate degradomics in complex proteomes. This approach significantly enhances confidence in substrate identification and categorizes cleavage events by specificity and structural accessibility of the cleavage site. We demonstrate concomitant quantification of cleavage site spanning peptides and neo-N and/or neo-C termini to estimate relative ratios of noncleaved and cleaved forms of substrate proteins. By applying this strategy to dissect the matrix metalloproteinase 10 (MMP10) substrate degradome in fibroblast secretomes, we identified the extracellular matrix protein ADAMTS-like protein 1 (ADAMTSL1) as a direct MMP10 substrate and revealed MMP10-dependent ectodomain shedding of platelet-derived growth factor receptor alpha (PDGFRα) as well as sequential processing of type I collagen. The data have been deposited to the ProteomeXchange Consortium with identifier PXD000503.


Subject(s)
Isotope Labeling/methods , Matrix Metalloproteinase 10/metabolism , Proteolysis , Proteome/metabolism , Proteomics/methods , Animals , BALB 3T3 Cells , Catalytic Domain , Cells, Cultured , Embryo, Mammalian , Matrix Metalloproteinase 10/chemistry , Mice , Mice, Knockout , Models, Molecular , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Proteome/analysis , Substrate Specificity , Time Factors
9.
Invest Ophthalmol Vis Sci ; 54(13): 8337-44, 2013 Dec 23.
Article in English | MEDLINE | ID: mdl-24265020

ABSTRACT

PURPOSE: Fibrillins are the major constituent of tissue microfibrils, which form the ocular zonule. In Marfan syndrome (MFS), FBN1 mutations lead to ectopia lentis. The goal of this work was to investigate zonule composition and formation in fibrillin-deficient and wild-type mice. METHODS: Immunofluorescence staining of eyes from wild-type, Fbn1-deficient, and Fbn2-deficient mice, as well as other species, was performed using monospecific fibrillin 1 and fibrillin 2 antibodies. The zonule of Fbn1-deficient and Fbn2-deficient mice was studied by electron microscopy. Microfibril formation in vitro was evaluated by immunofluorescence microscopy of cultured nonpigmented ciliary epithelial cells and fibroblasts. RESULTS: A zonule was present in both Fbn1-deficient and Fbn2-deficient mouse eyes. Immunofluorescence demonstrated that the zonule of Fbn1-deficient mice, wild-type mice, rats, and hamsters contained fibrillin 2. The zonule of Fbn2(-/-) mice contained fibrillin 1. Fibrillin 1 and fibrillin 2 colocalized in microfibrils formed in human nonpigmented ciliary epithelium cultures. Like fibrillin 1, fibrillin 2 microfibril assembly was fibronectin dependent and initiated by cell surface punctate deposits that elongated to form microfibrils. CONCLUSIONS: These data suggest that fibrillin 1 assembly and fibrillin 2 assembly share similar mechanisms. Microfibril composition depends substantially on the local levels of fibrillin isoforms and is not highly selective in regard to the isoform. This raises the intriguing possibility that the zonule could be strengthened in MFS by inducing fibrillin 2 expression in ciliary epithelium. The presence of fibrillin 2 in the murine zonule and an intact zonule in Fbn1-knockout mice may limit the utility of rodent models for studying ectopia lentis in MFS.


Subject(s)
Ciliary Body/metabolism , Lens, Crystalline/metabolism , Ligaments/metabolism , Marfan Syndrome/prevention & control , Microfilament Proteins/metabolism , Aged , Animals , Cattle , Cells, Cultured , Ciliary Body/cytology , Cricetinae , Ectopia Lentis/metabolism , Ectopia Lentis/prevention & control , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/metabolism , Fibrillin-1 , Fibrillin-2 , Fibrillins , Fibroblasts/metabolism , Fluorescent Antibody Technique, Indirect , Gene Expression , Genetic Therapy , Humans , Ligaments/ultrastructure , Marfan Syndrome/metabolism , Mesocricetus , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfibrils/metabolism , Microfibrils/ultrastructure , Microfilament Proteins/genetics , Microscopy, Electron , Microscopy, Fluorescence , Rats , Rats, Sprague-Dawley
10.
Matrix Biol ; 31(7-8): 398-411, 2012.
Article in English | MEDLINE | ID: mdl-23010571

ABSTRACT

ADAMTS-like proteins are related to ADAMTS metalloproteases by their similarity to ADAMTS ancillary domains. Here, we have characterized ADAMTSL5, a novel member of the superfamily with a unique modular organization that includes a single C-terminal netrin-like (NTR) module. Alternative splicing of ADAMTSL5 at its 5' end generates two transcripts that encode different signal peptides, but the same mature protein. These transcripts differ in their translational efficiency. Recombinant ADAMTSL5 is a secreted, N-glycosylated 60kDa glycoprotein located in the subcellular matrix, on the cell-surface, and in the medium of transfected cells. RT-PCR and western blot analysis of adult mouse tissues showed broad expression. Western blot analysis suggested proteolytic release of the NTR module in transfected cells as well as in some mouse tissues. Immunostaining during mouse organogenesis identified ADAMTSL5 in musculoskeletal tissues such as skeletal muscle, cartilage and bone, as well as in many epithelia. Affinity-chromatography demonstrated heparin-binding of ADAMTSL5 through its NTR-module. Recombinant ADAMTSL5 bound to both fibrillin-1 and fibrillin-2, and co-localized with fibrillin microfibrils in the extracellular matrix of cultured fibroblasts, but without discernible effect on microfibril assembly. ADAMTSL5 is the first family member shown to bind both fibrillin-1 and fibrillin-2. Like other ADAMTS proteins implicated in microfibril biology through identification of human and animal mutations, ADAMTSL5 could have a role in modulating microfibril functions.


Subject(s)
ADAM Proteins/metabolism , Heparin/metabolism , Microfibrils/metabolism , Microfilament Proteins/metabolism , Recombinant Proteins/metabolism , Thrombospondin 1/metabolism , ADAM Proteins/genetics , ADAMTS Proteins , Alternative Splicing/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , Fibrillin-1 , Fibrillin-2 , Fibrillins , Fluorescent Antibody Technique , HEK293 Cells , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Molecular Sequence Data , Muscle, Skeletal/metabolism , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Netrin-1 , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Thrombospondin 1/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
11.
Invest Ophthalmol Vis Sci ; 53(1): 461-9, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-21989719

ABSTRACT

PURPOSE: ADAMTSL4 mutations cause autosomal recessive isolated ectopia lentis (IEL) and ectopia lentis et pupillae. Dominant FBN1 mutations cause IEL or syndromic ectopia lentis (Marfan syndrome and Weill-Marchesani syndrome). The authors sought to characterize recombinant ADAMTSL4 and the ocular distribution of ADAMTSL4 and to investigate whether ADAMTSL4 influences the biogenesis of fibrillin-1 microfibrils, which compose the zonule. METHODS: ADAMTSL4 was expressed by the transfection of HEK293F cells. Protein extracts and paraffin sections from human eyes were analyzed by Western blot analysis and by immunoperoxidase staining, respectively. Immunofluorescence was used to evaluate fibrillin-1 deposition in the ECM of fetal bovine nuchal ligament cells after culture in ADAMTSL4-conditioned medium or control medium. Confocal microscopy was performed to investigate ADAMTSL4 and fibrillin-1 colocalization in these cultures. RESULTS: Western blot analysis identified ADAMTSL4 as a glycoprotein in HEK293F cells and as a major band of 150 kDa in ocular tissues including ciliary body, sclera, cornea, and retina. Immunoperoxidase staining showed a broad ocular distribution of ADAMTSL4, associated with both cells and fibrillar ECM. When cultured in ADAMTSL4-containing medium, fetal bovine nuchal ligament cells showed accelerated fibrillin-1 deposition in ECM. ADAMTSL4 colocalized with fibrillin-1 microfibrils in the ECM of these cells. CONCLUSIONS: ADAMTSL4 is a secreted glycoprotein that is widely distributed in the human eye. Enhanced fibrillin-1 deposition in the presence of ADAMTSL4 and colocalization of ADAMTSL4 with fibrillin-1 in the ECM of cultured fibroblasts suggest a potential role for ADAMTSL4 in the formation or maintenance of the zonule.


Subject(s)
Eye/metabolism , Gene Expression Regulation , Microfibrils/genetics , Microfilament Proteins/metabolism , RNA/genetics , Thrombospondins/genetics , ADAMTS Proteins , Animals , Binding Sites , Blotting, Western , Cattle , Cells, Cultured , Ectopia Lentis/genetics , Ectopia Lentis/metabolism , Ectopia Lentis/pathology , Extracellular Matrix/metabolism , Extracellular Matrix Proteins , Eye/pathology , Fibrillin-1 , Fibrillins , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Microfibrils/metabolism , Microscopy, Confocal , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Thrombospondins/biosynthesis , Thrombospondins/metabolism
12.
Am J Hum Genet ; 89(1): 7-14, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21683322

ABSTRACT

Geleophysic (GD) and acromicric dysplasia (AD) belong to the acromelic dysplasia group and are both characterized by severe short stature, short extremities, and stiff joints. Although AD has an unknown molecular basis, we have previously identified ADAMTSL2 mutations in a subset of GD patients. After exome sequencing in GD and AD cases, we selected fibrillin 1 (FBN1) as a candidate gene, even though mutations in this gene have been described in Marfan syndrome, which is characterized by tall stature and arachnodactyly. We identified 16 heterozygous FBN1 mutations that are all located in exons 41 and 42 and encode TGFß-binding protein-like domain 5 (TB5) of FBN1 in 29 GD and AD cases. Microfibrillar network disorganization and enhanced TGFß signaling were consistent features in GD and AD fibroblasts. Importantly, a direct interaction between ADAMTSL2 and FBN1 was demonstrated, suggesting a disruption of this interaction as the underlying mechanism of GD and AD phenotypes. Although enhanced TGFß signaling caused by FBN1 mutations can trigger either Marfan syndrome or GD and AD, our findings support the fact that TB5 mutations in FBN1 are responsible for short stature phenotypes.


Subject(s)
Bone Diseases, Developmental/genetics , Dwarfism/genetics , Eye Abnormalities/genetics , Limb Deformities, Congenital/genetics , Microfilament Proteins/genetics , Mutation , Adolescent , Adult , Child , Child, Preschool , Connective Tissue/abnormalities , DNA Mutational Analysis , Exons , Extracellular Matrix Proteins/metabolism , Fibrillin-1 , Fibrillins , Fluorescent Antibody Technique , Heterozygote , Humans , Inclusion Bodies/genetics , Marfan Syndrome/genetics , Microfibrils/ultrastructure , Microfilament Proteins/metabolism , Middle Aged , Phenotype , Protein Structure, Tertiary , Signal Transduction , Transforming Growth Factor beta1/metabolism , Young Adult
13.
J Biol Chem ; 286(19): 17156-67, 2011 May 13.
Article in English | MEDLINE | ID: mdl-21402694

ABSTRACT

Autosomal recessive and autosomal dominant forms of Weill-Marchesani syndrome, an inherited connective tissue disorder, are caused by mutations in ADAMTS10 (encoding a secreted metalloprotease) and FBN1 (encoding fibrillin-1, which forms tissue microfibrils), respectively, yet they are clinically indistinguishable. This genetic connection prompted investigation of a potential functional relationship between ADAMTS10 and fibrillin-1. Specifically, fibrillin-1 was investigated as a potential ADAMTS10 binding partner and substrate, and the role of ADAMTS10 in influencing microfibril biogenesis was addressed. Using ligand affinity blotting and surface plasmon resonance, recombinant ADAMTS10 was found to bind to fibrillin-1 with a high degree of specificity and with high affinity. Two sites of ADAMTS10 binding to fibrillin-1 were identified, one toward the N terminus and another in the C-terminal half of fibrillin-1. Confocal microscopy and immunoelectron microscopy localized ADAMTS10 to fibrillin-1-containing microfibrils in human tissues. Furin-activated ADAMTS10 could cleave fibrillin-1, but innate resistance of ADAMTS10 zymogen to propeptide excision by furin was observed, suggesting that, unless activated, ADAMTS10 is an inefficient fibrillinase. To investigate the role of ADAMTS10 in microfibril biogenesis, fetal bovine nuchal ligament cells were cultured in the presence or absence of ADAMTS10. Exogenously added ADAMTS10 led to accelerated fibrillin-1 microfibril biogenesis. Conversely, fibroblasts obtained from a Weill-Marchesani syndrome patient with ADAMTS10 mutations deposited fibrillin-1 microfibrils sparsely compared with unaffected control cells. Taken together, these findings suggest that ADAMTS10 participates in microfibril biogenesis rather than in fibrillin-1 turnover.


Subject(s)
ADAM Proteins/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Microfilament Proteins/metabolism , ADAMTS Proteins , Binding Sites , Fibrillin-1 , Fibrillins , Gene Expression Regulation , Humans , Microscopy, Confocal , Microscopy, Immunoelectron , Models, Biological , Mutagenesis, Site-Directed , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Surface Plasmon Resonance
14.
PLoS One ; 5(9)2010 Sep 17.
Article in English | MEDLINE | ID: mdl-20862248

ABSTRACT

BACKGROUND: Musladin-Lueke Syndrome (MLS) is a hereditary disorder affecting Beagle dogs that manifests with extensive fibrosis of the skin and joints. In this respect, it resembles human stiff skin syndrome and the Tight skin mouse, each of which is caused by gene defects affecting fibrillin-1, a major component of tissue microfibrils. The objective of this work was to determine the genetic basis of MLS and the molecular consequence of the identified mutation. METHODOLOGY AND PRINCIPAL FINDINGS: We mapped the locus for MLS by genome-wide association to a 3.05 Mb haplotype on canine chromosome 9 (CFA9 (50.11-54.26; p(raw) <10(-7))), which was homozygous and identical-by-descent among all affected dogs, consistent with recessive inheritance of a founder mutation. Sequence analysis of a candidate gene at this locus, ADAMTSL2, which is responsible for the human TGFß dysregulation syndrome, Geleophysic Dysplasia (GD), uncovered a mutation in exon 7 (c.660C>T; p.R221C) perfectly associated with MLS (p-value=10(-12)). Murine ADAMTSL2 containing the p.R221C mutation formed anomalous disulfide-bonded dimers when transiently expressed in COS-1, HEK293F and CHO cells, and was present in the medium of these cells at lower levels than wild-type ADAMTSL2 expressed in parallel. CONCLUSIONS/SIGNIFICANCE: The genetic basis of MLS is a founder mutation in ADAMTSL2, previously shown to interact with latent TGF-ß binding protein, which binds fibrillin-1. The molecular effect of the founder mutation on ADAMTSL2 is formation of disulfide-bonded dimers. Although caused by a distinct mutation, and having a milder phenotype than human GD, MLS nevertheless offers a new animal model for study of GD, and for prospective insights on mechanisms and pathways of skin fibrosis and joint contractures.


Subject(s)
Dog Diseases/congenital , Dog Diseases/genetics , Extracellular Matrix Proteins/genetics , Joint Diseases/veterinary , Mutation, Missense , Skin Abnormalities/veterinary , Animals , Base Sequence , Cell Line , Chromosome Mapping , Dog Diseases/metabolism , Dog Diseases/physiopathology , Dogs , Exons , Extracellular Matrix Proteins/metabolism , Humans , Joint Diseases/genetics , Joint Diseases/metabolism , Joint Diseases/physiopathology , Mice , Molecular Sequence Data , Skin Abnormalities/genetics , Skin Abnormalities/metabolism , Skin Abnormalities/physiopathology
15.
Am J Pathol ; 176(3): 1494-504, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20093484

ABSTRACT

The metalloprotease ADAMTS9 participates in melanoblast development and is a tumor suppressor in esophageal and nasopharyngeal cancer. ADAMTS9 null mice die before gastrulation, but, ADAMTS9+/- mice were initially thought to be normal. However, when congenic with the C57Bl/6 strain, 80% of ADAMTS9+/- mice developed spontaneous corneal neovascularization. beta-Galactosidase staining enabled by a lacZ cassette targeted to the ADAMTS9 locus showed that capillary endothelial cells (ECs) in embryonic and adult tissues and in capillaries growing into heterotopic tumors expressed ADAMTS9. Heterotopic B.16-F10 melanomas elicited greater vascular induction in ADAMTS9+/- mice than in wild-type littermates, suggesting a potential inhibitory role in tumor angiogenesis. Treatment of cultured human microvascular ECs with ADAMTS9 small-interfering RNA resulted in enhanced filopodial extension, decreased cell adhesion, increased cell migration, and enhanced formation of tube-like structures on Matrigel. Conversely, overexpression of catalytically active, but not inactive, ADAMTS9 in ECs led to fewer tube-like structures, demonstrating that the proteolytic activity of ADAMTS9 was essential. However, unlike the related metalloprotease ADAMTS1, which exerts anti-angiogenic effects by cleavage of thrombospondins and sequestration of vascular endothelial growth factor165, ADAMTS9 neither cleaved thrombospondins 1 and 2, nor bound vascular endothelial growth factor165. Taken together, these data identify ADAMTS9 as a novel, constitutive, endogenous angiogenesis inhibitor that operates cell-autonomously in ECs via molecular mechanisms that are distinct from those used by ADAMTS1.


Subject(s)
ADAM Proteins/metabolism , Endothelial Cells/enzymology , Endothelial Cells/pathology , Microvessels/enzymology , Microvessels/pathology , Neovascularization, Pathologic/enzymology , ADAM Proteins/genetics , ADAMTS9 Protein , Aging/metabolism , Animals , Biocatalysis , Cell Movement , Corneal Neovascularization/enzymology , Corneal Neovascularization/pathology , Embryo, Mammalian/enzymology , Embryo, Mammalian/pathology , Enzyme Activation , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Neoplasms/blood supply , Neoplasms/enzymology , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Organ Specificity , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Thrombospondin 1/metabolism , Thrombospondins/metabolism , Vascular Endothelial Growth Factor A/metabolism
16.
J Biol Chem ; 284(44): 30004-15, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19671700

ABSTRACT

Protein C-mannosylation is the attachment of alpha-mannopyranose to tryptophan via a C-C linkage. This post-translational modification typically occurs within the sequence motif WXXW, which is frequently present in thrombospondin type-1 repeats (TSRs). TSRs are especially numerous in and a defining feature of the ADAMTS superfamily. We investigated the presence and functional significance of C-mannosylation of ADAMTS-like 1/punctin-1, which contains four TSRs (two with predicted C-mannosylation sites), using mass spectrometry, metabolic labeling, site-directed mutagenesis, and expression in C-mannosylation-defective Chinese hamster ovary cell variants. Analysis of tryptic fragments of recombinant human punctin-1 by mass spectrometry identified a peptide derived from TSR1 containing the (36)WDAWGPWSECSRTC(49) sequence of interest modified with two mannose residues and a Glc-Fuc disaccharide (O-fucosylation). Tandem mass spectrometry (MS/MS) and MS/MS/MS analysis demonstrated the characteristic cross-ring cleavage of C-mannose and identified the modified residues as Trp(39) and Trp(42). C-Mannosylation of TSR1 of the related protease ADAMTS5 was also identified. Metabolic labeling of CHO-K1 cells or Lec35.1 cells demonstrated incorporation of d-[2,6-(3)H]mannose in secreted punctin-1 from CHO-K1 cells but not Lec35.1 cells. Quantitation of punctin-1 secretion in Lec35.1 cells versus CHO-K1 cells suggested decreased secretion in Lec35.1 cells. Replacement of mannosylated Trp residues in TSR1 with either Ala or Phe affected punctin secretion efficiency. These data demonstrate that TSR1 from punctin-1 carries C-mannosylation in close proximity to O-linked fucose. Together, these modifications appear to provide a quality control mechanism for punctin-1 secretion.


Subject(s)
Extracellular Matrix Proteins/metabolism , Mannose/metabolism , Protein Processing, Post-Translational , Thrombospondin 1/metabolism , Tryptophan/metabolism , ADAMTS Proteins , Amino Acid Substitution , Animals , Cell Line , Extracellular Matrix Proteins/genetics , Fucose , Glycosylation , Humans , Repetitive Sequences, Nucleic Acid , Thrombospondin 1/genetics
17.
Nat Genet ; 40(9): 1119-23, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18677313

ABSTRACT

Geleophysic dysplasia is an autosomal recessive disorder characterized by short stature, brachydactyly, thick skin and cardiac valvular anomalies often responsible for an early death. Studying six geleophysic dysplasia families, we first mapped the underlying gene to chromosome 9q34.2 and identified five distinct nonsense and missense mutations in ADAMTSL2 (a disintegrin and metalloproteinase with thrombospondin repeats-like 2), which encodes a secreted glycoprotein of unknown function. Functional studies in HEK293 cells showed that ADAMTSL2 mutations lead to reduced secretion of the mutated proteins, possibly owing to the misfolding of ADAMTSL2. A yeast two-hybrid screen showed that ADAMTSL2 interacts with latent TGF-beta-binding protein 1. In addition, we observed a significant increase in total and active TGF-beta in the culture medium as well as nuclear localization of phosphorylated SMAD2 in fibroblasts from individuals with geleophysic dysplasia. These data suggest that ADAMTSL2 mutations may lead to a dysregulation of TGF-beta signaling and may be the underlying mechanism of geleophysic dysplasia.


Subject(s)
Abnormalities, Multiple/genetics , Extracellular Matrix Proteins/genetics , Growth Disorders/genetics , Heart Valves/abnormalities , Transforming Growth Factor beta/metabolism , Biological Availability , Cell Line , Child , Child, Preschool , Hand Deformities, Congenital/genetics , Heart Defects, Congenital/genetics , Humans , Mutation
18.
Hum Mutat ; 29(12): 1425-34, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18567016

ABSTRACT

We report the identification and functional analysis of the first missense ADAMTS10 mutation (c.73G>A; p.Ala25Thr) causing recessive Weill-Marchesani syndrome (WMS). The Ala25 residue affected by the missense mutation is at the -1 position relative to the ADAMTS10 signal peptidase cleavage site. p.Ala25Thr substituted full-length ADAMTS10 showed consistent and significantly diminished secretion in both HEK293F and Cos-1 cells. However, a C-terminally truncated construct lacking the ancillary domain and containing only the signal peptide, the propeptide and the catalytic domain (p.Ala25Thr Pro-Cat) was efficiently secreted in both HEK293F cells and Cos-1 cells. Edman degradation of purified p.Ala25Thr Pro-Cat and p.Ala25Thr substituted full-length ADAMTS10 from HEK293F cells demonstrated correct signal peptide processing. Thus, the p.Ala25Thr substitution hinders secretion of full-length ADAMTS10, but not Pro-Cat from cells, yet permits signal peptide removal. We infer that folding of the complex C-terminal ancillary domain is the rate-limiting step in biosynthesis of ADAMTS10, and that it (but not Pro-Cat) is sensitive to subtle changes in efficiency of signal peptide cleavage. These observations represent an unprecedented effect of a signal peptide mutation and support a model in which the initial cotranslational processing events during protein biosynthesis can have long-range effects on protein folding and secretion.


Subject(s)
ADAM Proteins/genetics , Growth Disorders/genetics , Mutation , Protein Sorting Signals , ADAM Proteins/chemistry , ADAMTS Proteins , Aged, 80 and over , Animals , COS Cells , Cell Line , Chlorocebus aethiops , DNA Mutational Analysis , Humans , Protein Structure, Tertiary , Syndrome
19.
J Biol Chem ; 282(23): 17024-31, 2007 Jun 08.
Article in English | MEDLINE | ID: mdl-17395588

ABSTRACT

The ADAMTS superfamily contains several metalloproteases (ADAMTS proteases) as well as ADAMTS-like molecules that lack proteolytic activity. Their common feature is the presence of one or more thrombospondin type-1 repeats (TSRs) within a characteristic modular organization. ADAMTS like-1/punctin-1 has four TSRs. Previously, O-fucosylation on Ser or Thr mediated by the endoplasmic reticulum-localized enzyme protein-O-fucosyltransferase 2 (POFUT2) was described for TSRs of thrombospondin-1, properdin, and F-spondin within the sequence Cys-Xaa(1)-Xaa(2)-(Ser/Thr)-Cys-Xaa-Xaa-Gly (where the fucosylated residue is underlined). On mass spectrometric analysis of tryptic peptides from recombinant secreted human punctin-1, the appropriate peptides from TSR2, TSR3, and TSR4 were found to bear either a fucose monosaccharide (TSR3, TSR4) or a fucose-glucose disaccharide (TSR2, TSR3, TSR4). Although mass spectral analysis did not unambiguously identify the relevant peptide from TSR1, metabolic labeling of cells expressing TSR1 and the cysteine-rich module led to incorporation of [(3)H]fucose into this construct. Mutation of the putative modified Ser/Thr residues in TSR2, TSR3, and TSR4 led to significantly decreased levels of secreted punctin-1. Similarly, expression of punctin-1 in Lec-13 cells that are deficient in conversion of GDP-mannose to GDP-fucose substantially decreased the levels of secreted protein, which were restored upon culture in the presence of exogenous l-fucose. In addition, mutation of the single N-linked oligosaccharide in punctin-1 led to decreased levels of secreted punctin-1. Taken together, the data define a critical role for N-glycosylation and O-fucosylation in the biosynthesis of punctin-1. From a broad perspective, these data suggest that O-fucosylation may be a widespread post-translational modification in members of the ADAMTS superfamily with possible regulatory consequences.


Subject(s)
Extracellular Matrix Proteins/metabolism , Fucose/metabolism , Thrombospondin 1/metabolism , ADAMTS Proteins , Amino Acid Sequence , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/genetics , Glycosylation , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Plasmids , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
20.
J Biol Chem ; 279(34): 35159-75, 2004 Aug 20.
Article in English | MEDLINE | ID: mdl-15192113

ABSTRACT

We have characterized ADAMTS7B, the authentic full-length protein product of the ADAMTS7 gene. ADAMTS7B has a domain organization similar to that of ADAMTS12, with a total of eight thrombospondin type 1 repeats in its ancillary domain. Of these, seven are arranged in two distinct clusters that are separated by a mucin domain. Unique to the ADAMTS family, ADAMTS7B is modified by attachment of the glycosaminoglycan chondroitin sulfate within the mucin domain, thus rendering it a proteoglycan. Glycosaminoglycan addition has potentially important implications for ADAMTS7B cellular localization and for substrate recognition. Although not an integral membrane protein, ADAMTS7B is retained near the cell surface of HEK293F cells via interactions involving both the ancillary domain and the prodomain. ADAMTS7B undergoes removal of the prodomain by a multistep furin-dependent mechanism. At least part of the final processing event, i.e. cleavage following Arg(220) (mouse sequence annotation), occurs at the cell surface. ADAMTS7B is an active metalloproteinase as shown by its ability to cleave alpha(2)-macroglobulin, but it does not cleave specific peptide bonds in versican and aggrecan attacked by ADAMTS proteases. Together with ADAMTS12, whose primary structure also predicts a mucin domain, ADAMTS7B constitutes a unique subgroup of the ADAMTS family.


Subject(s)
Chondroitin Sulfate Proteoglycans/genetics , Metalloendopeptidases/genetics , Metalloproteases/genetics , ADAM Proteins , ADAMTS Proteins , ADAMTS7 Protein , Amino Acid Sequence , Animals , Base Sequence , Chondroitin Sulfate Proteoglycans/chemistry , Cloning, Molecular , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Humans , Metalloproteases/chemistry , Mice , Molecular Sequence Data , Mucins/genetics , Protein Structure, Tertiary/genetics , Repetitive Sequences, Nucleic Acid , Sequence Alignment , Thrombospondin 1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL