Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters

Publication year range
1.
Am J Hum Genet ; 109(12): 2230-2252, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36351433

ABSTRACT

EMILIN1 (elastin-microfibril-interface-located-protein-1) is a structural component of the elastic fiber network and localizes to the interface between the fibrillin microfibril scaffold and the elastin core. How EMILIN1 contributes to connective tissue integrity is not fully understood. Here, we report bi-allelic EMILIN1 loss-of-function variants causative for an entity combining cutis laxa, arterial tortuosity, aneurysm formation, and bone fragility, resembling autosomal-recessive cutis laxa type 1B, due to EFEMP2 (FBLN4) deficiency. In both humans and mice, absence of EMILIN1 impairs EFEMP2 extracellular matrix deposition and LOX activity resulting in impaired elastogenesis, reduced collagen crosslinking, and aberrant growth factor signaling. Collagen fiber ultrastructure and histopathology in EMILIN1- or EFEMP2-deficient skin and aorta corroborate these findings and murine Emilin1-/- femora show abnormal trabecular bone formation and strength. Altogether, EMILIN1 connects elastic fiber network with collagen fibril formation, relevant for both bone and vascular tissue homeostasis.


Subject(s)
Bone Diseases, Metabolic , Cutis Laxa , Animals , Humans , Mice , Collagen/genetics , Cutis Laxa/genetics , Elastin/metabolism , Extracellular Matrix Proteins/metabolism
2.
Am J Hum Genet ; 108(6): 1095-1114, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33991472

ABSTRACT

Latent transforming growth factor ß (TGFß)-binding proteins (LTBPs) are microfibril-associated proteins essential for anchoring TGFß in the extracellular matrix (ECM) as well as for correct assembly of ECM components. Variants in LTBP2, LTBP3, and LTBP4 have been identified in several autosomal recessive Mendelian disorders with skeletal abnormalities with or without impaired development of elastin-rich tissues. Thus far, the human phenotype associated with LTBP1 deficiency has remained enigmatic. In this study, we report homozygous premature truncating LTBP1 variants in eight affected individuals from four unrelated consanguineous families. Affected individuals present with connective tissue features (cutis laxa and inguinal hernia), craniofacial dysmorphology, variable heart defects, and prominent skeletal features (craniosynostosis, short stature, brachydactyly, and syndactyly). In vitro studies on proband-derived dermal fibroblasts indicate distinct molecular mechanisms depending on the position of the variant in LTBP1. C-terminal variants lead to an altered LTBP1 loosely anchored in the microfibrillar network and cause increased ECM deposition in cultured fibroblasts associated with excessive TGFß growth factor activation and signaling. In contrast, N-terminal truncation results in a loss of LTBP1 that does not alter TGFß levels or ECM assembly. In vivo validation with two independent zebrafish lines carrying mutations in ltbp1 induce abnormal collagen fibrillogenesis in skin and intervertebral ligaments and ectopic bone formation on the vertebrae. In addition, one of the mutant zebrafish lines shows voluminous and hypo-mineralized vertebrae. Overall, our findings in humans and zebrafish show that LTBP1 function is crucial for skin and bone ECM assembly and homeostasis.


Subject(s)
Collagen/metabolism , Cutis Laxa/etiology , Genetic Variation , Latent TGF-beta Binding Proteins/genetics , Adolescent , Alleles , Animals , Cells, Cultured , Child , Child, Preschool , Cutis Laxa/pathology , Extracellular Matrix/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Infant , Male , Pedigree , Skin/metabolism , Skin/pathology , Zebrafish
3.
FASEB J ; 37(1): e22717, 2023 01.
Article in English | MEDLINE | ID: mdl-36563024

ABSTRACT

Bone morphogenetic proteins (BMP) are powerful regulators of cellular processes such as proliferation, differentiation, and apoptosis. However, the specific molecular requirements controlling the bioavailability of BMPs in the extracellular matrix (ECM) are not yet fully understood. Our previous work showed that BMPs are targeted to the ECM as growth factor-prodomain (GF-PD) complexes (CPLXs) via specific interactions of their PDs. We showed that BMP-7 PD binding to the extracellular microfibril component fibrillin-1 renders the CPLXs from an open, bioactive V-shape into a closed, latent ring shape. Here, we show that specific PD interactions with heparin/heparan sulfate glycosaminoglycans (GAGs) allow to target and spatially concentrate BMP-7 and BMP-9 CPLXs in bioactive V-shape conformation. However, targeting to GAGs may be BMP specific, since BMP-10 GF and CPLX do not interact with heparin. Bioactivity assays on solid phase in combination with interaction studies showed that the BMP-7 PD protects the BMP-7 GF from inactivation by heparin. By using transmission electron microscopy, molecular docking, and site-directed mutagenesis, we determined the BMP-7 PD-binding site for heparin. Further, fine-mapping of the fibrillin-1-binding site within the BMP-7 PD and molecular modeling showed that both binding sites are mutually exclusive in the open V- versus closed ring-shape conformation. Together, our data suggest that targeting exquisite BMP PD-binding sites by extracellular protein and GAG scaffolds integrates BMP GF bioavailability in a contextual manner in development, postnatal life, and connective tissue disease.


Subject(s)
Bone Morphogenetic Protein 7 , Glycosaminoglycans , Bone Morphogenetic Protein 7/metabolism , Heparin/metabolism , Fibrillin-1/metabolism , Molecular Docking Simulation , Bone Morphogenetic Proteins/metabolism , Heparitin Sulfate/metabolism , Protein Binding , Bone Morphogenetic Protein 2/metabolism
4.
Int J Mol Sci ; 24(5)2023 Mar 03.
Article in English | MEDLINE | ID: mdl-36902380

ABSTRACT

Dedifferentiated vascular smooth muscle cells (vSMCs) play an essential role in neointima formation, and we now aim to investigate the role of the bone morphogenetic protein (BMP) modulator BMPER (BMP endothelial cell precursor-derived regulator) in neointima formation. To assess BMPER expression in arterial restenosis, we used a mouse carotid ligation model with perivascular cuff placement. Overall BMPER expression after vessel injury was increased; however, expression in the tunica media was decreased compared to untreated control. Consistently, BMPER expression was decreased in proliferative, dedifferentiated vSMC in vitro. C57BL/6_Bmper+/- mice displayed increased neointima formation 21 days after carotid ligation and enhanced expression of Col3A1, MMP2, and MMP9. Silencing of BMPER increased the proliferation and migration capacity of primary vSMCs, as well as reduced contractibility and expression of contractile markers, whereas stimulation with recombinant BMPER protein had the opposite effect. Mechanistically, we showed that BMPER binds insulin-like growth factor-binding protein 4 (IGFBP4), resulting in the modulation of IGF signaling. Furthermore, perivascular application of recombinant BMPER protein prevented neointima formation and ECM deposition in C57BL/6N mice after carotid ligation. Our data demonstrate that BMPER stimulation causes a contractile vSMC phenotype and suggest that BMPER has the potential for a future therapeutic agent in occlusive cardiovascular diseases.


Subject(s)
Carrier Proteins , Neointima , Vascular Remodeling , Animals , Mice , Bone Morphogenetic Proteins/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima/metabolism , Phenotype , Carrier Proteins/metabolism
5.
FASEB J ; 35(3): e21353, 2021 03.
Article in English | MEDLINE | ID: mdl-33629769

ABSTRACT

Since their discovery as pluripotent cytokines extractable from bone matrix, it has been speculated how bone morphogenetic proteins (BMPs) become released and activated from the extracellular matrix (ECM). In contrast to TGF-ßs, most investigated BMPs are secreted as bioactive prodomain (PD)-growth factor (GF) complexes (CPLXs). Recently, we demonstrated that PD-dependent targeting of BMP-7 CPLXs to the extracellular fibrillin microfibril (FMF) components fibrillin-1 and -2 represents a BMP sequestration mechanism by rendering the GF latent. Understanding how BMPs become activated from ECM scaffolds such as FMF is crucial to elucidate pathomechanisms characterized by aberrant BMP activation and ECM destruction. Here, we describe a new MMP-dependent BMP-7 activation mechanism from ECM-targeted pools via specific PD degradation. Using Edman sequencing and mutagenesis, we identified a new and conserved MMP-13 cleavage site within the BMP-7 PD. A degradation screen with different BMP family PDs and representative MMP family members suggested utilization of the identified site in a general MMP-driven BMP activation mechanism. Furthermore, sandwich ELISA and solid phase cleavage studies in combination with bioactivity assays, single particle TEM, and in silico molecular docking experiments provided evidence that PD cleavage by MMP-13 leads to BMP-7 CPLX disintegration and bioactive GF release.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Extracellular Matrix/metabolism , Matrix Metalloproteinases/physiology , Amino Acid Motifs , Animals , Bone Morphogenetic Protein 7/chemistry , Bone Morphogenetic Protein 7/metabolism , Bone Morphogenetic Proteins/chemistry , HEK293 Cells , Humans , Matrix Metalloproteinase 13/physiology , Mice , Molecular Docking Simulation , Protein Domains
6.
PLoS Biol ; 17(12): e3000557, 2019 12.
Article in English | MEDLINE | ID: mdl-31826007

ABSTRACT

Balanced transforming growth factor-beta (TGFß)/bone morphogenetic protein (BMP)-signaling is essential for tissue formation and homeostasis. While gain in TGFß signaling is often found in diseases, the underlying cellular mechanisms remain poorly defined. Here we show that the receptor BMP type 2 (BMPR2) serves as a central gatekeeper of this balance, highlighted by its deregulation in diseases such as pulmonary arterial hypertension (PAH). We show that BMPR2 deficiency in endothelial cells (ECs) does not abolish pan-BMP-SMAD1/5 responses but instead favors the formation of mixed-heteromeric receptor complexes comprising BMPR1/TGFßR1/TGFßR2 that enable enhanced cellular responses toward TGFß. These include canonical TGFß-SMAD2/3 and lateral TGFß-SMAD1/5 signaling as well as formation of mixed SMAD complexes. Moreover, BMPR2-deficient cells express genes indicative of altered biophysical properties, including up-regulation of extracellular matrix (ECM) proteins such as fibrillin-1 (FBN1) and of integrins. As such, we identified accumulation of ectopic FBN1 fibers remodeled with fibronectin (FN) in junctions of BMPR2-deficient ECs. Ectopic FBN1 deposits were also found in proximity to contractile intimal cells in pulmonary artery lesions of BMPR2-deficient heritable PAH (HPAH) patients. In BMPR2-deficient cells, we show that ectopic FBN1 is accompanied by active ß1-integrin highly abundant in integrin-linked kinase (ILK) mechano-complexes at cell junctions. Increased integrin-dependent adhesion, spreading, and actomyosin-dependent contractility facilitates the retrieval of active TGFß from its latent fibrillin-bound depots. We propose that loss of BMPR2 favors endothelial-to-mesenchymal transition (EndMT) allowing cells of myo-fibroblastic character to create a vicious feed-forward process leading to hyperactivated TGFß signaling. In summary, our findings highlight a crucial role for BMPR2 as a gatekeeper of endothelial homeostasis protecting cells from increased TGFß responses and integrin-mediated mechano-transduction.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/metabolism , Endothelial Cells/metabolism , Transforming Growth Factor beta/metabolism , Bone Morphogenetic Protein Receptors, Type II/physiology , Cell Line , Endothelium, Vascular/metabolism , Fibrillin-1/metabolism , Gene Expression Regulation/genetics , Humans , Lung/pathology , Protein Serine-Threonine Kinases/metabolism , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Artery/metabolism , Receptors, Transforming Growth Factor beta , Signal Transduction , Smad Proteins
7.
Exp Dermatol ; 30(1): 25-37, 2021 01.
Article in English | MEDLINE | ID: mdl-32920888

ABSTRACT

Supramolecular networks composed of fibrillins (fibrillin-1 and fibrillin-2) and associated ligands form intricate cellular microenvironments which balance skin homoeostasis and direct remodelling. Fibrillins assemble into microfibrils which are not only indispensable for conferring elasticity to the skin, but also control the bioavailability of growth factors targeted to the extracellular matrix architecture. Fibrillin microfibrils (FMF) represent the core scaffolds for elastic fibre formation, and they also decorate the surface of elastic fibres and form independent networks. In normal dermis, elastic fibres are suspended in a three-dimensional basket-like lattice of FMF intersecting basement membranes at the dermal-epidermal junction and thus conferring pliability to the skin. The importance of FMF for skin homoeostasis is illustrated by the clinical features caused by mutations in the human fibrillin genes (FBN1, FBN2), summarized as "fibrillinopathies." In skin, fibrillin mutations result in phenotypes ranging from thick, stiff and fibrotic skin to thin, lax and hyperextensible skin. The most plausible explanation for this spectrum of phenotypic outcomes is that FMF regulate growth factor signalling essential for proper growth and homoeostasis of the skin. Here, we will give an overview about the current understanding of the underlying pathomechanisms leading to fibrillin-dependent fibrosis as well as forms of cutis laxa caused by mutational inactivation of FMF-associated ligands.


Subject(s)
Connective Tissue Diseases/genetics , Elastic Tissue/metabolism , Fibrillins/genetics , Fibrillins/metabolism , Homeostasis , Skin/metabolism , Animals , Bone Diseases, Developmental/genetics , Elastic Tissue/ultrastructure , Elasticity , Fibrillins/ultrastructure , Fibrosis , Humans , Limb Deformities, Congenital/genetics , Microfibrils/metabolism , Microfibrils/ultrastructure , Molecular Conformation , Signal Transduction , Skin/pathology , Skin/ultrastructure , Skin Physiological Phenomena , Transforming Growth Factor beta/metabolism
8.
FASEB J ; 34(9): 12040-12052, 2020 09.
Article in English | MEDLINE | ID: mdl-32716577

ABSTRACT

Although collagens are the most abundant proteins implicated in various disease pathways, essential mechanisms required for their proper folding and assembly are poorly understood. Heat-shock protein 47 (HSP47), an ER-resident chaperone, was mainly reported to fulfill key functions in folding and secretion of fibrillar collagens by stabilizing pro-collagen triple-helices. In this study, we demonstrate unique functions of HSP47 for different collagen subfamilies. Our results show that HSP47 binds to the N-terminal region of procollagen I and is essential for its secretion. However, HSP47 ablation does not majorly impact collagen VI secretion, but its lateral assembly. Moreover, specific ablation of Hsp47 in murine keratinocytes revealed a new role for the transmembrane collagen XVII triple-helix formation. Incompletely folded collagen XVII C-termini protruding from isolated HSP47 null keratinocyte membrane vesicles could be fully restored upon the application of recombinant HSP47. Thus, our study expands the current view regarding the client repertoire and function of HSP47, as well as emphasizes its importance for transmembrane collagen folding.


Subject(s)
HSP47 Heat-Shock Proteins/metabolism , Keratinocytes/metabolism , Procollagen/metabolism , Protein Folding , Animals , HSP47 Heat-Shock Proteins/genetics , Mice , Procollagen/genetics
9.
J Biol Chem ; 294(37): 13769-13780, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31346034

ABSTRACT

The assembly of collagen VI microfibrils is a multistep process in which proteolytic processing within the C-terminal globular region of the collagen VI α3 chain plays a major role. However, the mechanisms involved remain elusive. Moreover, C5, the short and most C-terminal domain of the α3 chain, recently has been proposed to be released as an adipokine that enhances tumor progression, fibrosis, inflammation, and insulin resistance and has been named "endotrophin." Serum endotrophin could be a useful biomarker to monitor the progression of such disorders as chronic obstructive pulmonary disease, systemic sclerosis, and kidney diseases. Here, using biochemical and isotopic MS-based analyses, we found that the extracellular metalloproteinase bone morphogenetic protein 1 (BMP-1) is involved in endotrophin release and determined the exact BMP-1 cleavage site. Moreover, we provide evidence that several endotrophin-containing fragments are present in various tissues and body fluids. Among these, a large C2-C5 fragment, which contained endotrophin, was released by furin-like proprotein convertase cleavage. By using immunofluorescence microscopy and EM, we also demonstrate that these proteolytic maturations occur after secretion of collagen VI tetramers and during microfibril assembly. Differential localization of N- and C-terminal regions of the collagen VI α3 chain revealed that cleavage products are deposited in tissue and cell cultures. The detailed information on the processing of the collagen VI α3 chain reported here provides a basis for unraveling the function of endotrophin (C5) and larger endotrophin-containing fragments and for refining their use as biomarkers of disease progression.


Subject(s)
Bone Morphogenetic Protein 1/metabolism , Collagen Type VI/metabolism , Proprotein Convertases/metabolism , Fibrosis , Furin/metabolism , HEK293 Cells , Humans , Insulin Resistance , Microfibrils/metabolism , Peptide Fragments/metabolism , Proteolysis
10.
Hum Mol Genet ; 27(21): 3675-3687, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30060141

ABSTRACT

Fibrillin microfibrils are extracellular matrix assemblies that form the template for elastic fibres, endow blood vessels, skin and other elastic tissues with extensible properties. They also regulate the bioavailability of potent growth factors of the TGF-ß superfamily. A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)10 is an essential factor in fibrillin microfibril function. Mutations in fibrillin-1 or ADAMTS10 cause Weill-Marchesani syndrome (WMS) characterized by short stature, eye defects, hypermuscularity and thickened skin. Despite its importance, there is poor understanding of the role of ADAMTS10 and its function in fibrillin microfibril assembly. We have generated an ADAMTS10 WMS mouse model using Clustered Regularly Spaced Interspaced Short Palindromic Repeats and CRISPR associated protein 9 (CRISPR-Cas9) to introduce a truncation mutation seen in WMS patients. Homozygous WMS mice are smaller and have shorter long bones with perturbation to the zones of the developing growth plate and changes in cell proliferation. Furthermore, there are abnormalities in the ciliary apparatus of the eye with decreased ciliary processes and abundant fibrillin-2 microfibrils suggesting perturbation of a developmental expression switch. WMS mice have increased skeletal muscle mass and more myofibres, which is likely a consequence of an altered skeletal myogenesis. These results correlated with expression data showing down regulation of Growth differentiation factor (GDF8) and Bone Morphogenetic Protein (BMP) growth factor genes. In addition, the mitochondria in skeletal muscle are larger with irregular shape coupled with increased phospho-p38 mitogen-activated protein kinase (MAPK) suggesting muscle remodelling. Our data indicate that decreased SMAD1/5/8 and increased p38/MAPK signalling are associated with ADAMTS10-induced WMS. This model will allow further studies of the disease mechanism to facilitate the development of therapeutic interventions.


Subject(s)
ADAMTS Proteins/genetics , Disease Models, Animal , Microfibrils/metabolism , Mutation , Signal Transduction , Weill-Marchesani Syndrome/metabolism , ADAMTS Proteins/metabolism , Animals , MAP Kinase Signaling System , Mice , Mice, Transgenic , Smad Proteins, Receptor-Regulated/metabolism , Weill-Marchesani Syndrome/genetics
11.
J Immunol ; 201(2): 663-674, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29898959

ABSTRACT

Myeloid cells can be beneficial as well as harmful in tissue regenerative responses. The molecular mechanisms by which myeloid cells control this critical decision of the immune system are not well understood. Using two different models of physiological acute or pathological chronic skin damage, in this study we identified myeloid cell-restricted STAT3 signaling as important and an injury context-dependent regulator of skin fibrosis. Targeted disruption of STAT3 signaling in myeloid cells significantly accelerated development of pathological skin fibrosis in a model of chronic bleomycin-induced tissue injury, whereas the impact on wound closure dynamics and quality of healing after acute excision skin injury was minor. Chronic bleomycin-mediated tissue damage in control mice provoked an antifibrotic gene signature in macrophages that was characterized by upregulated expression of IL-10, SOCS3, and decorin. In contrast, in STAT3-deficient macrophages this antifibrotic repair program was abolished whereas TGF-ß1 expression was increased. Notably, TGF-ß1 synthesis in cultured control bone marrow-derived macrophages (BMDMs) was suppressed after IL-10 exposure, and this suppressive effect was alleviated by STAT3 deficiency. Accordingly, coculture of IL-10-stimulated control BMDMs with fibroblasts suppressed expression of the TGF-ß1 downstream target connective tissue growth factor in fibroblasts, whereas this suppressive effect was lost by STAT3 deficiency in BMDMs. Our findings highlight a previously unrecognized protective role of myeloid cell-specific STAT3 signaling in immune cell-mediated skin fibrosis, and its regulatory pathway could be a potential target for therapy.


Subject(s)
Macrophages/immunology , Myeloid Cells/physiology , STAT3 Transcription Factor/metabolism , Skin Diseases/immunology , Skin/pathology , Acute Disease , Animals , Cells, Cultured , Chronic Disease , Disease Models, Animal , Fibrosis , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Regeneration , STAT3 Transcription Factor/genetics , Signal Transduction , Skin Diseases/chemically induced , Transcriptome , Transforming Growth Factor beta/metabolism , Wound Healing
13.
Eur Respir J ; 53(3)2019 03.
Article in English | MEDLINE | ID: mdl-30578393

ABSTRACT

Epithelial tubes, comprised of polarised epithelial cells around a lumen, are crucial for organ function. However, the molecular mechanisms underlying tube formation remain largely unknown. Here, we report on the function of fibrillin (FBN)2, an extracellular matrix (ECM) glycoprotein, as a critical regulator of tracheal tube formation.We performed a large-scale forward genetic screen in mouse to identify regulators of respiratory organ development and disease. We identified Fbn2 mutants which exhibit shorter and narrowed tracheas as well as defects in tracheal smooth muscle cell alignment and polarity.We found that FBN2 is essential for elastic fibre formation and Fibronectin accumulation around tracheal smooth muscle cells. These processes appear to be regulated at least in part through inhibition of p38-mediated upregulation of matrix metalloproteinases (MMPs), as pharmacological decrease of p38 phosphorylation or MMP activity partially attenuated the Fbn2 mutant tracheal phenotypes. Analysis of human tracheal tissues indicates that a decrease in ECM proteins, including FBN2 and Fibronectin, is associated with tracheomalacia.Our findings provide novel insights into the role of ECM homeostasis in mesenchymal cell polarisation during tracheal tubulogenesis.


Subject(s)
Extracellular Matrix/metabolism , Fibrillin-2/metabolism , Muscle, Smooth/embryology , Myocytes, Smooth Muscle/cytology , Trachea/embryology , Animals , Embryo, Mammalian , Female , Fibrillin-2/genetics , Fibronectins/metabolism , Gene Expression Regulation, Developmental , Homeostasis , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth/cytology , Phenotype , Phosphorylation , Signal Transduction , Trachea/cytology
14.
Biochem Biophys Res Commun ; 503(3): 1273-1277, 2018 09 10.
Article in English | MEDLINE | ID: mdl-30001809

ABSTRACT

We investigated the presence of autoantibodies against the extracellular matrix proteins thrombospondin-4 (TSP-4), cartilage oligomeric matrix protein (COMP), C-type lectin domain family 3 member A (CLEC3A), collagen II, collagen VI, matrilin-3, and fibrillin-2 in the serum of osteoarthritis (OA) patients. We compared those results with the presence of such antibodies in rheumatoid arthritis (RA) patients and in healthy donors (HD). Our study examines whether antibodies against extracellular proteins can be used as potential biomarkers to support the clinical diagnosis of OA. 10 OA, 10 RA patients and 10 HD were enrolled in this explorative cross-sectional study. SDS-PAGE and immunoblot were used to investigate the presence of antibodies against extracellular matrix proteins. The serum of 5/10 OA patients but 0/10 HD exhibited TSP-4 IgG isotype antibodies (P = 0.033). The serum of 8/10 OA patients but only 1/10 HD exhibited IgG isotype antibodies against TSP-4 or COMP (P = 0.005). The serum of 9/10 OA patients but only 1/10 HD exhibited IgG isotype antibodies against TSP-4, COMP or CLEC3A (P = 0.005). We found strong evidence for the presence of IgG isotype autoantibodies against the cartilage extracellular matrix proteins TSP-4, COMP and CLEC3A in OA. The detection of IgG isotype autoantibodies against TSP-4, COMP and CLEC3A may support the clinical diagnosis of OA. OA with autoantibodies against cartilage extracellular matrix proteins defines a new OA subgroup suggesting that patients with high concentrations of autoantibodies may benefit from an immune suppressive therapy.


Subject(s)
Arthritis, Rheumatoid/immunology , Autoantibodies/immunology , Osteoarthritis/immunology , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/therapy , Biomarkers/blood , Cartilage Oligomeric Matrix Protein/blood , Cartilage Oligomeric Matrix Protein/immunology , Collagen Type II/blood , Collagen Type II/immunology , Collagen Type VI/blood , Collagen Type VI/immunology , Fibrillin-2/blood , Fibrillin-2/immunology , Humans , Lectins, C-Type/blood , Lectins, C-Type/immunology , Matrilin Proteins/blood , Matrilin Proteins/immunology , Middle Aged , Osteoarthritis/diagnosis , Osteoarthritis/therapy , Thrombospondins/blood , Thrombospondins/immunology
15.
PLoS Genet ; 11(6): e1005340, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26114882

ABSTRACT

Fibrillins are large extracellular macromolecules that polymerize to form the backbone structure of connective tissue microfibrils. Mutations in the gene for fibrillin-1 cause the Marfan syndrome, while mutations in the gene for fibrillin-2 cause Congenital Contractural Arachnodactyly. Both are autosomal dominant disorders, and both disorders affect musculoskeletal tissues. Here we show that Fbn2 null mice (on a 129/Sv background) are born with reduced muscle mass, abnormal muscle histology, and signs of activated BMP signaling in skeletal muscle. A delay in Myosin Heavy Chain 8, a perinatal myosin, was found in Fbn2 null forelimb muscle tissue, consistent with the notion that muscle defects underlie forelimb contractures in these mice. In addition, white fat accumulated in the forelimbs during the early postnatal period. Adult Fbn2 null mice are already known to demonstrate persistent muscle weakness. Here we measured elevated creatine kinase levels in adult Fbn2 null mice, indicating ongoing cycles of muscle injury. On a C57Bl/6 background, Fbn2 null mice showed severe defects in musculature, leading to neonatal death from respiratory failure. These new findings demonstrate that loss of fibrillin-2 results in phenotypes similar to those found in congenital muscular dystrophies and that FBN2 should be considered as a candidate gene for recessive congenital muscular dystrophy. Both in vivo and in vitro evidence associated muscle abnormalities and accumulation of white fat in Fbn2 null mice with abnormally activated BMP signaling. Genetic rescue of reduced muscle mass and accumulation of white fat in Fbn2 null mice was accomplished by deleting a single allele of Bmp7. In contrast to other reports that activated BMP signaling leads to muscle hypertrophy, our findings demonstrate the exquisite sensitivity of BMP signaling to the fibrillin-2 extracellular environment during early postnatal muscle development. New evidence presented here suggests that fibrillin-2 can sequester BMP complexes in a latent state.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Microfilament Proteins/genetics , Muscular Diseases/genetics , Animals , Animals, Newborn , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , Bone Morphogenetic Proteins/genetics , Creatine Kinase/blood , Female , Fibrillin-1 , Fibrillin-2 , Fibrillins , Gene Expression Regulation , Limb Deformities, Congenital/genetics , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Microfilament Proteins/metabolism , Muscle, Skeletal/abnormalities , Muscle, Skeletal/pathology , Muscular Diseases/physiopathology , Muscular Dystrophies/genetics , Organ Culture Techniques , Signal Transduction/genetics
16.
J Biol Chem ; 291(24): 12732-12746, 2016 Jun 10.
Article in English | MEDLINE | ID: mdl-27059954

ABSTRACT

Since the discovery of bone morphogenetic proteins (BMPs) as pluripotent cytokines extractable from bone matrix, it has been speculated how targeting of BMPs to the extracellular matrix (ECM) modulates their bioavailability. Understanding these processes is crucial for elucidating pathomechanisms of connective tissue disorders characterized by ECM deficiency and growth factor dysregulation. Here, we provide evidence for a new BMP targeting and sequestration mechanism that is controlled by the ECM molecule fibrillin-1. We present the nanoscale structure of the BMP-7 prodomain-growth factor complex using electron microscopy, small angle x-ray scattering, and circular dichroism spectroscopy, showing that it assumes an open V-like structure when it is bioactive. However, upon binding to fibrillin-1, the BMP-7 complex is rendered into a closed ring shape, which also confers latency to the growth factor, as demonstrated by bioactivity measurements. BMP-7 prodomain variants were used to map the critical epitopes for prodomain-growth factor and prodomain-prodomain binding. Together, these data show that upon prodomain binding to fibrillin-1, the BMP-7 complex undergoes a conformational change, which denies access of BMP receptors to the growth factor.


Subject(s)
Bone Morphogenetic Protein 7/metabolism , Extracellular Matrix/metabolism , Fibrillin-1/metabolism , Microfibrils/metabolism , Amino Acid Sequence , Animals , Bone Morphogenetic Protein 7/chemistry , Bone Morphogenetic Protein 7/genetics , Cell Line , Circular Dichroism , Fibrillin-1/chemistry , Fibrillin-1/genetics , HEK293 Cells , Humans , Microscopy, Electron, Transmission , Models, Molecular , Nanostructures/chemistry , Nanostructures/ultrastructure , Protein Binding , Protein Conformation , Protein Domains , Protein Structure, Secondary , Scattering, Small Angle , Surface Plasmon Resonance , X-Ray Diffraction
17.
J Biol Chem ; 291(3): 1103-14, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26601954

ABSTRACT

MFAP4 (microfibrillar-associated protein 4) is an extracellular glycoprotein found in elastic fibers without a clearly defined role in elastic fiber assembly. In the present study, we characterized molecular interactions between MFAP4 and elastic fiber components. We established that MFAP4 primarily assembles into trimeric and hexameric structures of homodimers. Binding analysis revealed that MFAP4 specifically binds tropoelastin and fibrillin-1 and -2, as well as the elastin cross-linking amino acid desmosine, and that it co-localizes with fibrillin-1-positive fibers in vivo. Site-directed mutagenesis disclosed residues Phe(241) and Ser(203) in MFAP4 as being crucial for type I collagen, elastin, and tropoelastin binding. Furthermore, we found that MFAP4 actively promotes tropoelastin self-assembly. In conclusion, our data identify MFAP4 as a new ligand of microfibrils and tropoelastin involved in proper elastic fiber organization.


Subject(s)
Carrier Proteins/metabolism , Desmosine/metabolism , Elastic Tissue/metabolism , Extracellular Matrix Proteins/metabolism , Glycoproteins/metabolism , Microfibrils/metabolism , Microfilament Proteins/metabolism , Models, Molecular , Tropoelastin/metabolism , Amino Acid Substitution , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/genetics , Fibrillin-1 , Fibrillins , Glycoproteins/chemistry , Glycoproteins/genetics , Humans , Ligands , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Mutation , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Multimerization , Protein Transport , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Tropoelastin/chemistry , Tropoelastin/genetics
18.
Exp Dermatol ; 26(5): 435-438, 2017 05.
Article in English | MEDLINE | ID: mdl-27892605

ABSTRACT

EMILIN3 is an extracellular matrix glycoprotein that displays a dynamic and restricted expression pattern in connective tissues during post-natal life. In this study, we report the characterization of EMILIN3 deposition in the skin. In addition, to unravel the functions of this protein in skin homeostasis, we generated Emilin3 null mice and provide evidence that EMILIN3 is dispensable for hair follicle growth and maintenance throughout adult life.


Subject(s)
Extracellular Matrix Proteins/metabolism , Glycoproteins/metabolism , Hair Follicle/growth & development , Animals , Hair Follicle/metabolism , Mice
19.
Proc Natl Acad Sci U S A ; 111(36): 13063-8, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25157165

ABSTRACT

Bone morphogenetic proteins (BMPs) orchestrate key cellular events, such as proliferation and differentiation, in development and homeostasis. Extracellular antagonists, such as chordin, are essential regulators of BMP signaling. Chordin binds to BMPs blocking interaction with receptors, and cleavage by tolloid proteinases is thought to relieve this inhibition. A model has been previously proposed where chordin adopts a horseshoe-like arrangement enabling BMP binding cooperatively by terminal domains (1). Here, we present the nanoscale structure of human chordin using electron microscopy, small angle X-ray scattering, and solution-based biophysical techniques, which together show that chordin indeed has a compact horseshoe-shaped structure. Chordin variants were used to map domain locations within the chordin molecule. The terminal BMP-binding domains protrude as prongs from the main body of the chordin structure, where they are well positioned to interact with the growth factor. The spacing provided by the chordin domains supports the principle of a cooperative BMP-binding arrangement that the original model implied in which growth factors bind to both an N- and C-terminal von Willebrand factor C domain of chordin. Using binding and bioactivity assays, we compared full-length chordin with two truncated chordin variants, such as those produced by partial tolloid cleavage. Cleavage of either terminal domain has little effect on the affinity of chordin for BMP-4 and BMP-7 but C-terminal cleavage increases the efficacy of chordin as a BMP-4 inhibitor. Together these data suggest that partial tolloid cleavage is insufficient to ablate BMP inhibition and the C-terminal chordin domains play an important role in BMP regulation.


Subject(s)
Bone Morphogenetic Proteins/antagonists & inhibitors , Glycoproteins/chemistry , Intercellular Signaling Peptides and Proteins/chemistry , Nanoparticles/chemistry , Animals , Bone Morphogenetic Proteins/chemistry , Glycoproteins/ultrastructure , HEK293 Cells , Humans , Hydrodynamics , Imaging, Three-Dimensional , Mice , Models, Molecular , Mutant Proteins/chemistry , Nanoparticles/ultrastructure , Protein Binding , Protein Structure, Tertiary , Scattering, Small Angle , Solutions , Surface Plasmon Resonance , X-Ray Diffraction
20.
Biochem Soc Trans ; 43(5): 795-800, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26517884

ABSTRACT

Chordin-mediated regulation of bone morphogenetic protein (BMP) family growth factors is essential in early embryogenesis and adult homoeostasis. Chordin binds to BMPs through cysteine-rich von Willebrand factor type C (vWC) homology domains and blocks them from interacting with their cell surface receptors. These domains also self-associate and enable chordin to target related proteins to fine-tune BMP regulation. The chordin-BMP inhibitory complex is strengthened by the secreted glycoprotein twisted gastrulation (Tsg); however, inhibition is relieved by cleavage of chordin at two specific sites by tolloid family metalloproteases. As Tsg enhances this cleavage process, it serves a dual role as both promoter and inhibitor of BMP signalling. Recent developments in chordin research suggest that rather than simply being by-products, the cleavage fragments of chordin continue to play a role in BMP regulation. In particular, chordin cleavage at the C-terminus potentiates its anti-BMP activity in a type-specific manner.


Subject(s)
Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Bone Morphogenetic Proteins/antagonists & inhibitors , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Models, Biological , Proteins/metabolism , Signal Transduction , Tolloid-Like Metalloproteinases/metabolism , Animals , Bone Morphogenetic Protein Receptors/agonists , Bone Morphogenetic Protein Receptors/chemistry , Bone Morphogenetic Protein Receptors/metabolism , Bone Morphogenetic Proteins/chemistry , Bone Morphogenetic Proteins/metabolism , Glycoproteins/chemistry , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Stability , Proteins/chemistry , Proteolysis , Tolloid-Like Metalloproteinases/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL