Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Cell ; 139(2): 380-92, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19818485

ABSTRACT

Synapses are asymmetric cellular adhesions that are critical for nervous system development and function, but the mechanisms that induce their formation are not well understood. We have previously identified thrombospondin as an astrocyte-secreted protein that promotes central nervous system (CNS) synaptogenesis. Here, we identify the neuronal thrombospondin receptor involved in CNS synapse formation as alpha2delta-1, the receptor for the anti-epileptic and analgesic drug gabapentin. We show that the VWF-A domain of alpha2delta-1 interacts with the epidermal growth factor-like repeats common to all thrombospondins. alpha2delta-1 overexpression increases synaptogenesis in vitro and in vivo and is required postsynaptically for thrombospondin- and astrocyte-induced synapse formation in vitro. Gabapentin antagonizes thrombospondin binding to alpha2delta-1 and powerfully inhibits excitatory synapse formation in vitro and in vivo. These findings identify alpha2delta-1 as a receptor involved in excitatory synapse formation and suggest that gabapentin may function therapeutically by blocking new synapse formation.


Subject(s)
CD36 Antigens/metabolism , Calcium Channels/metabolism , Neurogenesis , Synapses , Amines/pharmacology , Animals , Calcium Channels, L-Type , Cyclohexanecarboxylic Acids/pharmacology , Gabapentin , Mice , Neuronal Plasticity , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Synapses/drug effects , gamma-Aminobutyric Acid/pharmacology
2.
EMBO J ; 38(1)2019 01 03.
Article in English | MEDLINE | ID: mdl-30396995

ABSTRACT

Control of synapse number and function in the developing central nervous system is critical to the formation of neural circuits. Astrocytes play a key role in this process by releasing factors that promote the formation of excitatory synapses. Astrocyte-secreted thrombospondins (TSPs) induce the formation of structural synapses, which however remain post-synaptically silent, suggesting that completion of early synaptogenesis may require a two-step mechanism. Here, we show that the humoral innate immune molecule Pentraxin 3 (PTX3) is expressed in the developing rodent brain. PTX3 plays a key role in promoting functionally-active CNS synapses, by increasing the surface levels and synaptic clustering of AMPA glutamate receptors. This process involves tumor necrosis factor-induced protein 6 (TSG6), remodeling of the perineuronal network, and a ß1-integrin/ERK pathway. Furthermore, PTX3 activity is regulated by TSP1, which directly interacts with the N-terminal region of PTX3. These data unveil a fundamental role of PTX3 in promoting the first wave of synaptogenesis, and show that interplay of TSP1 and PTX3 sets the proper balance between synaptic growth and synapse function in the developing brain.


Subject(s)
C-Reactive Protein/physiology , Extracellular Matrix/metabolism , Integrin beta1/metabolism , Nerve Tissue Proteins/physiology , Receptors, AMPA/metabolism , Synapses/physiology , Animals , Astrocytes/metabolism , Brain/growth & development , Brain/metabolism , C-Reactive Protein/genetics , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Extracellular Matrix/genetics , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Neuronal Plasticity/genetics , Protein Transport/genetics , Thrombospondin 1/metabolism
3.
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
4.
Mol Cell Proteomics ; 17(7): 1410-1425, 2018 07.
Article in English | MEDLINE | ID: mdl-29669734

ABSTRACT

Secreted and cell-surface proteases are major mediators of extracellular matrix (ECM) turnover, but their mechanisms and regulatory impact are poorly understood. We developed a mass spectrometry approach using a cell-free ECM produced in vitro to identify fibronectin (FN) as a novel substrate of the secreted metalloprotease ADAMTS16. ADAMTS16 cleaves FN between its (I)5 and (I)6 modules, releasing the N-terminal 30 kDa heparin-binding domain essential for FN self-assembly. ADAMTS16 impairs FN fibrillogenesis as well as fibrillin-1 and tenascin-C assembly, thus inhibiting formation of a mature ECM by cultured fibroblasts. Furthermore ADAMTS16 has a marked morphogenetic impact on spheroid formation by renal tubule-derived MDCKI cells. The N-terminal FN domain released by ADAMTS16 up-regulates MMP3, which cleaves the (I)5-(I)6 linker of FN similar to ADAMTS16, therefore creating a proteolytic feed-forward mechanism. Thus, FN proteolysis not only regulates FN turnover, but also FN assembly, with potential long-term consequences for ECM assembly and morphogenesis.


Subject(s)
ADAMTS Proteins/metabolism , Extracellular Matrix/metabolism , Fibronectins/metabolism , Morphogenesis , Proteolysis , Proteomics/methods , Spheroids, Cellular/metabolism , 3T3 Cells , ADAMTS Proteins/chemistry , Amino Acid Sequence , Animals , Collagen/metabolism , Dogs , Fibroblasts/metabolism , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Matrix Metalloproteinase 3/metabolism , Mice , Peptides/chemistry , Peptides/metabolism , Protein Binding , Protein Domains , Substrate Specificity , Up-Regulation
5.
J Biol Chem ; 293(34): 13166-13175, 2018 08 24.
Article in English | MEDLINE | ID: mdl-29954947

ABSTRACT

Human myeloid-derived growth factor (MYDGF; also known as C19orf10) is named based on its identification as a secreted monocyte/macrophage-derived mediator of cardiac repair following myocardial infarction in mice. Homologs of MYDGF, however, are present in organisms throughout and outside of the animal kingdom, some of which lack hematopoietic and circulatory systems. Moreover, the UPF0556 protein domain, which defines these homologs, lacks a known structure. As a result, the functions and properties of MYDGF are unclear. Our current work was initiated to test whether MYDGF is present in secretory vesicles of eosinophils as it was recently reported to be abundant in these cells. However, we could not demonstrate secretion and unexpectedly discovered that MYDGF colocalizes with P4HB in the nuclear envelope, which comprises the bulk of endoplasmic reticulum (ER) in eosinophils, and with P4HB and RCAS1 in Golgi. We noted a ubiquitous C-terminal sequence, BXEL (B, basic; X, variable residue; E, Glu; L, Leu), that has the potential to retain human MYDGF and its homologs in the ER. To test the functionality of this sequence, we expressed full-length human MYDGF or MYDGF lacking the C-terminal Glu-Leu residues in monolayers of human embryonic kidney 293 (HEK293) cells. Full-length MYDGF accumulated in cells, whereas truncated MYDGF appeared in the medium. These observations reveal that MYDGF resides in the ER and Golgi and provide a new framework for investigating and understanding this intriguing protein.


Subject(s)
Antigens, Neoplasm/metabolism , Endoplasmic Reticulum/metabolism , Eosinophils/metabolism , Golgi Apparatus/metabolism , Interleukins/metabolism , Procollagen-Proline Dioxygenase/metabolism , Protein Disulfide-Isomerases/metabolism , Amino Acid Sequence , Antigens, Neoplasm/genetics , Biological Transport , Humans , Interleukins/genetics , Procollagen-Proline Dioxygenase/genetics , Protein Disulfide-Isomerases/genetics , Sequence Homology
6.
Angiogenesis ; 22(1): 133-144, 2019 02.
Article in English | MEDLINE | ID: mdl-30168023

ABSTRACT

Thrombospondin (TSP)-1 and TSP-2 share similar structures and functions, including a remarkable antiangiogenic activity. We have previously demonstrated that a mechanism of the antiangiogenic activity of TSP-1 is the interaction of its type III repeats domain with fibroblast growth factor-2 (FGF2), affecting the growth factor bioavailability and angiogenic activity. Since the type III repeats domain is conserved in TSP-2, this study aimed at investigating whether also TSP-2 retained the ability to interact with FGF2. The FGF2 binding properties of TSP-1 and TSP-2 and their recombinant domains were analyzed by solid-phase binding and surface plasmon resonance assays. TSP-2 bound FGF2 with high affinity (Kd = 1.3 nM). TSP-2/FGF2 binding was inhibited by calcium and heparin. The FGF2-binding domain of TSP-2 was located in the type III repeats and the minimal interacting sequence was identified as the GVTDEKD peptide in repeat 3C, corresponding to KIPDDRD, the active sequence of TSP-1. A second putative FGF2 binding sequence was also identified in repeat 11C of both TSPs. Computational docking analysis predicted that both the TSP-2 and TSP-1-derived heptapeptides interacted with FGF2 with comparable binding properties. Accordingly, small molecules based on the TSP-1 active sequence blocked TSP-2/FGF2 interaction. Binding of TSP-2 to FGF2 impaired the growth factor ability to interact with its cellular receptors, since TSP-2-derived fragments prevented the binding of FGF2 to both heparin (used as a structural analog of heparan sulfate proteoglycans) and FGFR-1. These findings identify TSP-2 as a new FGF2 ligand that shares with TSP-1 the same molecular requirements for interaction with the growth factor and a comparable capacity to block FGF2 interaction with proangiogenic receptors. These features likely contribute to TSP-2 antiangiogenic and antineoplastic activity, providing the rationale for future therapeutic applications.


Subject(s)
Fibroblast Growth Factor 2/chemistry , Surface Plasmon Resonance , Thrombospondins/chemistry , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/chemistry , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Fibroblast Growth Factor 2/metabolism , Humans , Protein Binding , Protein Domains , Repetitive Sequences, Amino Acid , Thrombospondins/metabolism
7.
Crit Rev Biochem Mol Biol ; 50(5): 427-39, 2015.
Article in English | MEDLINE | ID: mdl-26288337

ABSTRACT

Periostin (PN) and TGF-ß-induced protein (ßig-h3) are paralogs that contain a single emilin and four fasciclin-1 modules and are secreted from cells. PN receives attention because of its up-regulation in cancer and degenerative and allergic diseases. ßig-h3 is highly enriched in cornea and best known for harboring mutations in humans associated with corneal dystrophies. Both proteins are expressed widely, and many functions, some over-lapping, have been attributed to PN and ßig-h3 based on biochemical, cell culture, and whole animal experiments. We attempt to organize this knowledge so as to facilitate research on these interesting and incompletely understood proteins. We focus particularly on whether PN and ßig-h3 are modified by vitamin K-dependent γ-glutamyl carboxylation, a question of considerable importance given the profound effects of γ-carboxylation on structure and function of other proteins. We consider the roles of PN and ßig-h3 in formation of extracellular matrix and as ligands for integrin receptors. We attempt to reconcile the contradictory results that have arisen concerning the role of PN, which has emerged as a marker of TH2 immunity, in murine models of allergic asthma. Finally, when possible we compare and contrast the structures and functions of the two proteins.


Subject(s)
Cell Adhesion Molecules/metabolism , Extracellular Matrix Proteins/metabolism , Extracellular Matrix/metabolism , Immunity, Cellular , Integrins/agonists , Models, Molecular , Transforming Growth Factor beta/metabolism , Amino Acid Sequence , Animals , Cell Adhesion , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Conserved Sequence , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/genetics , Humans , Integrins/metabolism , Ligands , Phylogeny , Protein Conformation , Protein Processing, Post-Translational , Protein Structure, Tertiary , Sequence Alignment , Sequence Homology, Amino Acid , Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/genetics
8.
Circ Res ; 117(2): 129-41, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-25940549

ABSTRACT

RATIONALE: Histological examination of abdominal aortic aneurysm (AAA) tissues demonstrates extracellular matrix destruction and infiltration of inflammatory cells. Previous work with mouse models of AAA has shown that anti-inflammatory strategies can effectively attenuate aneurysm formation. Thrombospondin-1 is a matricellular protein involved in the maintenance of vascular structure and homeostasis through the regulation of biological functions, such as cell proliferation, apoptosis, and adhesion. Expression levels of thrombospondin-1 correlate with vascular disease conditions. OBJECTIVE: To use thrombospondin-1-deficient (Thbs1(-/-)) mice to test the hypothesis that thrombospondin-1 contributes to pathogenesis of AAAs. METHODS AND RESULTS: Mouse experimental AAA was induced through perivascular treatment with calcium phosphate, intraluminal perfusion with porcine elastase, or systemic administration of angiotensin II. Induction of AAA increased thrombospondin-1 expression in aortas of C57BL/6 or apoE-/- mice. Compared with Thbs1(+/+) mice, Thbs1(-/-) mice developed significantly smaller aortic expansion when subjected to AAA inductions, which was associated with diminished infiltration of macrophages. Thbs1(-/-) monocytic cells had reduced adhesion and migratory capacity in vitro compared with wild-type counterparts. Adoptive transfer of Thbs1(+/+) monocytic cells or bone marrow reconstitution rescued aneurysm development in Thbs1(-/-) mice. CONCLUSIONS: Thrombospondin-1 expression plays a significant role in regulation of migration and adhesion of mononuclear cells, contributing to vascular inflammation during AAA development.


Subject(s)
Aortic Aneurysm, Abdominal/physiopathology , Macrophages/physiology , Thrombospondin 1/physiology , Adoptive Transfer , Angiotensin II/toxicity , Animals , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/etiology , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/prevention & control , Apolipoproteins E/deficiency , Bone Marrow Transplantation , Calcium Phosphates/toxicity , Cell Line , Cell Movement , Disease Models, Animal , Inflammation , Macrophages/transplantation , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/physiology , Monocytes/transplantation , Pancreatic Elastase/toxicity , Radiation Chimera , Recombinant Proteins/therapeutic use , Thrombospondin 1/biosynthesis , Thrombospondin 1/deficiency , Thrombospondin 1/therapeutic use , Up-Regulation
9.
Biochem J ; 456(2): 283-95, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24070235

ABSTRACT

Fibrillins constitute the backbone of extracellular multifunctional assemblies present in elastic and non-elastic matrices, termed microfibrils. Assembly of fibrillins into microfibrils and their homoeostasis is poorly understood and is often compromised in connective tissue disorders such as Marfan syndrome and other fibrillinopathies. Using interaction mapping studies, we demonstrate that fibrillins require the complete gelatin-binding region of fibronectin for interaction, which comprises domains FNI6-FNI9. However, the interaction of fibrillin-1 with the gelatin-binding domain of fibronectin is not involved in fibrillin-1 network assembly mediated by human skin fibroblasts. We show further that the fibronectin network is essential for microfibril homoeostasis in early stages. Fibronectin is present in extracted mature microfibrils from tissue and cells as well as in some in situ microfibrils observed at the ultrastructural level, indicating an extended mechanism for the involvement of fibronectin in microfibril assembly and maturation.


Subject(s)
Fibronectins/metabolism , Microfibrils/metabolism , Microfilament Proteins/metabolism , Adhesins, Bacterial/chemistry , Adolescent , Binding Sites , Binding, Competitive , Cells, Cultured , Child , Child, Preschool , Fibrillin-1 , Fibrillins , Fibronectins/chemistry , Homeostasis , Humans , Infant , Microfilament Proteins/chemistry , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Multimerization , Protein Transport
10.
Am J Respir Cell Mol Biol ; 48(4): 503-10, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23306834

ABSTRACT

Periostin is an extracellular matrix protein that is up-regulated by T helper cell type 2 cytokines in the asthmatic airway and implicated in mouse studies as promoting eosinophil recruitment. We asked whether periostin modulates eosinophil adhesion and motility in vitro. Periostin adsorbed to polystyrene supported adhesion of purified human blood eosinophils stimulated by IL-5, IL-3, or granulocyte/macrophage colony-stimulating factor, but did not support adhesion of eosinophils treated with IL-4 or IL-13. The degree of adhesion depended on the concentrations of periostin during coating and activating cytokine during the adhesion assay. Both full-length periostin and alternatively spliced periostin, lacking C-terminal exons 17, 18, 19, and 21, supported adhesion. Adhesion was inhibited by monoclonal antibody to α(M) or ß(2) integrin subunits, but not by antibodies to other eosinophil integrin subunits. Adsorbed periostin also supported α(M)ß(2)-dependent random motility of IL-5-stimulated eosinophils with optimal movement at an intermediate coating concentration. In the presence of IL-5, eosinophils adherent on periostin formed punctate structures positive for filamentous actin, gelsolin, and phosphotyrosine. These structures fit the criteria for podosomes, highly dynamic adhesive contacts that are distinct from classical focal adhesions. The results establish α(M)ß(2) (CD11b/CD18, Mac-1) as an adhesive and promigratory periostin receptor on cytokine-stimulated eosinophils, and suggest that periostin may function as a haptotactic stimulus able to guide eosinophils to areas of high periostin density in the asthmatic airway.


Subject(s)
CD11b Antigen/metabolism , CD18 Antigens/metabolism , Cell Adhesion Molecules/metabolism , Cell Movement/drug effects , Eosinophils/metabolism , Interleukin-5/pharmacology , Animals , Asthma/metabolism , Asthma/pathology , Cell Adhesion/drug effects , Eosinophils/pathology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Female , Humans , Male , Mice
11.
J Proteome Res ; 12(7): 3393-404, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23750785

ABSTRACT

Fibronectin (Fn) is a large glycoprotein present in plasma and extracellular matrix and is important for many processes. Within Fn the 70 kDa N-terminal region (70k-Fn) is involved in cell-mediated Fn assembly, a process that contributes to embryogenesis, development, and platelet thrombus formation. In addition, major human pathogens including Staphlycoccus aureus and Streptococcus pyogenes bind the 70k-Fn region by a novel form of protein-protein interaction called ß-zipper formation, facilitating bacterial spread and colonization. Knowledge of blood plasma and platelet proteins that interact with 70k-Fn by ß-zipper formation is incomplete. In the current study, we aimed to characterize these proteins through affinity purification. For this affinity purification, we used a novel purification technique termed immiscible filtration assisted by surface tension (IFAST). The foundation of this technology is immiscible phase filtration, using a magnet to draw paramagnetic particle (PMP)-bound analyte through an immiscible barrier (oil or organic solvent) that separates an aqueous sample from an aqueous eluting buffer. The immiscible barrier functions to remove unbound proteins via exclusion rather than dilutive washing used in traditional isolation methods. We identified 31 interactors from plasma, of which only seven were previously known to interact with Fn. Furthermore, five proteins were identified to interact with 70k-Fn from platelet lysate, of which one was previously known. These results demonstrate that IFAST offers advantages for proteomic studies of interacting molecules in that the technique requires small sample volumes, can be done with high enough throughput to sample multiple interaction conditions, and is amenable to exploratory mass spectrometric and confirmatory immuno-blotting read-outs.


Subject(s)
Bacterial Proteins/isolation & purification , Blood Proteins/isolation & purification , Fibronectins/blood , Protein Binding , Bacterial Proteins/metabolism , Blood Platelets/metabolism , Blood Proteins/metabolism , Fibronectins/chemistry , Humans , Mass Spectrometry , Proteomics , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Streptococcus pyogenes/metabolism , Streptococcus pyogenes/pathogenicity
12.
J Biol Chem ; 287(20): 16759-67, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22447929

ABSTRACT

The Yersinia pestis adhesin molecule Ail interacts with the extracellular matrix protein fibronectin (Fn) on host cells to facilitate efficient delivery of cytotoxic Yop proteins, a process essential for plague virulence. A number of bacterial pathogens are known to bind to the N-terminal region of Fn, comprising type I Fn (FNI) repeats. Using proteolytically generated Fn fragments and purified recombinant Fn fragments, we demonstrated that Ail binds the centrally located 120-kDa fragment containing type III Fn (FNIII) repeats. A panel of monoclonal antibodies (mAbs) that recognize specific epitopes within the 120-kDa fragment demonstrated that mAb binding to (9)FNIII blocks Ail-mediated bacterial binding to Fn. Epitopes of three mAbs that blocked Ail binding to Fn were mapped to a similar face of (9)FNIII. Antibodies directed against (9)FNIII also inhibited Ail-dependent cell binding activity, thus demonstrating the biological relevance of this Ail binding region on Fn. Bacteria expressing Ail on their surface could also bind a minimal fragment of Fn containing repeats (9-10)FNIII, and this binding was blocked by a mAb specific for (9)FNIII. These data demonstrate that Ail binds to (9)FNIII of Fn and presents Fn to host cells to facilitate cell binding and delivery of Yops (cytotoxins of Y. pestis), a novel interaction, distinct from other bacterial Fn-binding proteins.


Subject(s)
Bacterial Adhesion/physiology , Bacterial Outer Membrane Proteins/metabolism , Fibronectins/metabolism , Virulence Factors/metabolism , Yersinia pestis/metabolism , Antibodies, Bacterial/chemistry , Antibodies, Monoclonal, Murine-Derived/chemistry , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Epitopes/chemistry , Epitopes/genetics , Epitopes/metabolism , Fibronectins/chemistry , Fibronectins/genetics , Peptide Mapping/methods , Protein Structure, Tertiary , Repetitive Sequences, Amino Acid , Virulence Factors/chemistry , Virulence Factors/genetics , Yersinia pestis/chemistry , Yersinia pestis/genetics
13.
J Biol Chem ; 287(16): 13303-12, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22351755

ABSTRACT

How fibronectin (FN) converts from a compact plasma protein to a fibrillar component of extracellular matrix is not understood. "Functional upstream domain" (FUD), a polypeptide based on F1 adhesin of Streptococcus pyogenes, binds by anti-parallel ß-strand addition to discontinuous sets of N-terminal FN type I modules, (2-5)FNI of the fibrin-binding domain and (8-9)FNI of the gelatin-binding domain. Such binding blocks assembly of FN. To learn whether ligation of (2-5)FNI, (8-9)FNI, or the two sets in combination is important for inhibition, we tested "high affinity downstream domain" (HADD), which binds by ß-strand addition to the continuous set of FNI modules, (1-5)FNI, comprising the fibrin-binding domain. HADD and FUD were similarly active in blocking fibronectin assembly. Binding of HADD or FUD to soluble plasma FN exposed the epitope to monoclonal antibody mAbIII-10 in the tenth FN type III module ((10)FNIII) and caused expansion of FN as assessed by dynamic light scattering. Soluble N-terminal constructs truncated after (9)FNI or (3)FNIII competed better than soluble FN for binding of FUD or HADD to adsorbed FN, indicating that interactions involving type III modules more C-terminal than (3)FNIII limit ß-strand addition to (1-5)FNI within intact soluble FN. Preincubation of FN with mAbIII-10 or heparin modestly increased binding to HADD or FUD. Thus, ligation of FNIII modules involved in binding of integrins and glycosaminoglycans, (10)FNIII and (12-14)FNIII, increases accessibility of (1-5)FNI. Allosteric loss of constraining interactions among (1-5)FNI, (10)FNIII, and (12-14)FNIII likely enables assembly of FN into extracellular fibrils.


Subject(s)
Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Fibronectins/chemistry , Fibronectins/metabolism , Adhesins, Bacterial/metabolism , Allosteric Regulation/physiology , Animals , Antibodies, Monoclonal/metabolism , Binding, Competitive/physiology , Epitope Mapping , Fibrin/metabolism , Fibroblasts/metabolism , Heparin/metabolism , Humans , Ligands , Mice , Protein Structure, Tertiary , Solubility , Streptococcus pyogenes/metabolism
14.
J Biol Chem ; 286(37): 32220-30, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21757696

ABSTRACT

Transglutaminase 2 (TG2) is secreted by a non-classical pathway into the extracellular space, where it has several activities pertinent to fibronectin (FN), including binding to the gelatin-binding domain of FN and acting as an integrin co-receptor. Glutamines in the N-terminal tail of FN are known to be susceptible to transamidation by both TG2 and activated blood coagulation factor XIII (FXIIIa). We used immunoblotting, limited proteolysis, and mass spectrometry to localize glutamines within FN that are subject to TG2-catalyzed incorporation of dansylcadaverine in comparison to residues modified by FXIIIa. Such analysis of plasma FN indicated that Gln-3, Gln-7, and Gln-9 in the N-terminal tail and Gln-246 of the linker between fifth and sixth type I modules ((5)F1 and (6)F1) are transamidated by both enzymes. Only minor incorporation of dansylcadaverine was detected elsewhere. Labeling of C-terminally truncated FN constructs revealed efficient TG2- or FXIIIa-catalyzed dansylcadaverine incorporation into the N-terminal residues of constructs as small as the 29-kDa fragment that includes (1-5)F1 and lacks modules from the adjacent gelatin-binding domain. However, when only (1-3)F1 were present, dansylcadaverine incorporation into the N-terminal residues of FN was lost and instead was in the enzymes, near the active site of TG2 and terminal domains of FXIIIa. Thus, these results demonstrate that FXIIIa and TG2 act similarly on glutamines at either end of (1-5)F1 and transamidation specificity of both enzymes is achieved through interactions with the intact 29K fragment.


Subject(s)
Factor XIIIa/metabolism , Fibronectins/metabolism , Glutamine/metabolism , Protein Processing, Post-Translational/physiology , Transglutaminases/metabolism , Catalytic Domain , Factor XIIIa/chemistry , Factor XIIIa/genetics , Fibronectins/chemistry , Fibronectins/genetics , GTP-Binding Proteins , Glutamine/chemistry , Glutamine/genetics , Humans , Protein Glutamine gamma Glutamyltransferase 2 , Protein Structure, Tertiary , Transglutaminases/chemistry , Transglutaminases/genetics
15.
Biochemistry ; 50(23): 5322-32, 2011 Jun 14.
Article in English | MEDLINE | ID: mdl-21561146

ABSTRACT

Homocystinuria is a genetic disorder resulting in elevated levels of homocysteine in plasma and tissues. Some of the skeletal and ocular symptoms such as long bone overgrowth, scoliosis, and ectopia lentis overlap with symptoms seen in Marfan syndrome. Marfan syndrome is caused by mutations in the extracellular matrix protein fibrillin-1. We previously showed that fibrillin-1 is a target for homocysteine and that the deposition of homocysteinylated fibrillin-1 in the extracellular matrix is compromised. Since the assembly of fibrillin-1 is critically dependent on fibronectin, we analyzed the consequences of fibronectin homocysteinylation and its interaction with fibrillin-1. Cellular fibronectin and proteolytic fragments were homocysteinylated and tested in various interaction assays with recombinant fibrillin-1 and heparin. Fibronectin homocysteinylation consistently compromised the fibronectin-fibrillin-1 interaction, while the interaction with heparin was not affected. Fibronectin homocysteinylation, but not cysteinylation, reduced the fibronectin dimers to monomers as shown by Western blotting. ELISA analyses of homocysteinylated fibronectin with three monoclonal antibodies demonstrated structural changes in the disulfide-containing FNI domains FNI(2), FNI(4), and FNI(9). Using fluorescently labeled fibronectin, we studied the consequence of fibronectin homocysteinylation on assembly in cell culture. Modified fibronectin showed deficiencies in denovo matrix incorporation and initial assembly. In conclusion, we define here characteristic structural changes of fibronectin upon homocysteinylation that translate into functional deficiencies in the fibronectin-fibrillin-1 interaction and in fibronectin assembly. Since fibronectin is a major organizer of various extracellular protein networks, these structural and functional alterations may contribute to the pathogenesis of homocystinuria and Marfan syndrome.


Subject(s)
Fibronectins/chemistry , Fibronectins/metabolism , Homocysteine/chemistry , Microfilament Proteins/metabolism , Fibrillin-1 , Fibrillins , Fibroblasts/metabolism , Homocysteine/metabolism , Humans , Microfilament Proteins/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship
16.
J Biol Chem ; 285(12): 8563-71, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20097751

ABSTRACT

Fibronectin (FN) without an RGD sequence (FN-RGE), and thus lacking the principal binding site for alpha5beta1 integrin, is deposited into the extracellular matrix of mouse embryos. Spontaneous conversion of (263)NGR and/or (501)NGR to iso-DGR possibly explains this enigma, i.e. ligation of iso-DGR by alphavbeta3 integrin may allow cells to assemble FN. Partial modification of (263)NGR to DGR or iso-DGR was detected in purified plasma FN by mass spectrometry. To test functions of the conversion, one or both NGR sequences were mutated to QGR in recombinant N-terminal 70-kDa construct of FN (70K), full-length FN, or FN-RGE. The mutations did not affect the binding of soluble 70K to already adherent fibroblasts or the ability of soluble 70K to compete with non-mutant FN or FN-RGE for binding to FN assembly sites. Non-mutant FN and FN-N263Q/N501Q with both NGRs mutated to QGRs were assembled equally well by adherent fibroblasts. FN-RGE and FN-RGE-N263Q/N501Q were also assembled equally well. Although substrate-bound 70K mediated cell adhesion in the presence of 1 mm Mn(2+) by a mechanism that was inhibited by cyclic RGD peptide, the peptide did not inhibit 70K binding to cell surface. Mutations of the NGR sequences had no effect on Mn(2+)-enhanced cell adhesion to adsorbed 70K but caused a decrease in cell adhesion to reduced and alkylated 70K. These results demonstrate that iso-DGR sequences spontaneously converted from NGR are cryptic and do not mediate the interaction of the 70K region of FN with the cell surface during FN assembly.


Subject(s)
Fibroblasts/metabolism , Fibronectins/chemistry , Animals , Binding Sites , Cell Adhesion , Cell Membrane/metabolism , Extracellular Matrix/metabolism , Humans , Mass Spectrometry/methods , Mice , Mutation , Oligopeptides/chemistry , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Vitronectin/chemistry
17.
J Biol Chem ; 285(52): 41087-99, 2010 Dec 24.
Article in English | MEDLINE | ID: mdl-20947497

ABSTRACT

The 49-residue functional upstream domain (FUD) of Streptococcus pyogenes F1 adhesin interacts with fibronectin (FN) in a heretofore unknown manner that prevents assembly of a FN matrix. Biotinylated FUD (b-FUD) bound to adsorbed FN or its recombinant N-terminal 70-kDa fibrin- and gelatin-binding fragment (70K). Binding was blocked by FN or 70K, but not by fibrin- or gelatin-binding subfragments of 70K. Isothermal titration calorimetry showed that FUD binds with K(d) values of 5.2 and 59 nM to soluble 70K and FN, respectively. We tested sets of FUD mutants and epitope-mapped monoclonal antibodies (mAbs) for ability to compete with b-FUD for binding to FN or to block FN assembly by cultured fibroblasts. Deletions or alanine substitutions throughout FUD caused loss of both activities. mAb 4D1 to the (2)FNI module had little effect, whereas mAb 7D5 to the (4)FNI module in the fibrin-binding region, 5C3 to the (9)FNI module in the gelatin-binding region, or L8 to the G-strand of (1)FNIII module adjacent to (9)FNI caused loss of binding of b-FUD to FN and decreased FN assembly. Conversely, FUD blocked binding of 7D5, 5C3, or L8, but not of 4D1, to FN. Circular dichroism indicated that FUD binds to 70K by ß-strand addition, a possibility supported by modeling based on crystal structures of peptides bound to (2)FNI-(5)FNI of the fibrin-binding domain and (8)FNI-(9)FNI of the gelatin-binding domain. Thus, the interaction likely involves an extensive anti-parallel ß-zipper in which FUD interacts with the E-strands of (2)FNI-(5)FNI and (8)FNI-(9)FNI.


Subject(s)
Adhesins, Bacterial/chemistry , Fibronectins/chemistry , Streptococcus pyogenes/chemistry , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Antibodies, Monoclonal/chemistry , Binding Sites , Epitopes/chemistry , Fibronectins/genetics , Fibronectins/metabolism , Humans , Protein Binding , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Streptococcus pyogenes/genetics , Streptococcus pyogenes/metabolism
18.
J Biol Chem ; 285(12): 8733-42, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20056600

ABSTRACT

Endogenous inhibitors of angiogenesis, such as thrombospondin-1 (TSP-1), are promising sources of therapeutic agents to treat angiogenesis-driven diseases, including cancer. TSP-1 regulates angiogenesis through different mechanisms, including binding and sequestration of the angiogenic factor fibroblast growth factor-2 (FGF-2), through a site located in the calcium binding type III repeats. We hypothesized that the FGF-2 binding sequence of TSP-1 might serve as a template for the development of inhibitors of angiogenesis. Using a peptide array approach followed by binding assays with synthetic peptides and recombinant proteins, we identified a FGF-2 binding sequence of TSP-1 in the 15-mer sequence DDDDDNDKIPDDRDN. Molecular dynamics simulations, taking the full flexibility of the ligand and receptor into account, and nuclear magnetic resonance identified the relevant residues and conformational determinants for the peptide-FGF interaction. This information was translated into a pharmacophore model used to screen the NCI2003 small molecule databases, leading to the identification of three small molecules that bound FGF-2 with affinity in the submicromolar range. The lead compounds inhibited FGF-2-induced endothelial cell proliferation in vitro and affected angiogenesis induced by FGF-2 in the chicken chorioallantoic membrane assay. These small molecules, therefore, represent promising leads for the development of antiangiogenic agents. Altogether, this study demonstrates that new biological insights obtained by integrated multidisciplinary approaches can be used to develop small molecule mimics of endogenous proteins as therapeutic agents.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Fibroblast Growth Factor 2/antagonists & inhibitors , Thrombospondin 1/physiology , Animals , Cell Proliferation , Chickens , Chorioallantoic Membrane/metabolism , Chorion/metabolism , Humans , Kinetics , Magnetic Resonance Spectroscopy , Peptides/chemistry , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Thrombospondin 1/chemistry
19.
J Biol Chem ; 284(38): 25879-88, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19617627

ABSTRACT

Bone morphogenetic protein-1-like proteinases play key roles in formation of the extracellular matrix (ECM) in vertebrates via biosynthetic processing of precursors into mature functional proteins involved in ECM assembly. Such processing includes proteolytic activation of the zymogen for lysyl oxidase. Fibronectin (FN) is an abundant protein component of the ECM that is capable of regulating manifold cellular functions through its interactions with various ECM and cell surface proteins. It was previously shown that proteolytic activation of lysyl oxidase is much reduced in cultures of FN-null mouse embryo fibroblasts (MEFs). Here we demonstrate that cellular fibronectin, the form produced by fibroblasts and various other tissue cell types, and plasma fibronectin bind BMP1 with dissociation constants (KD) of approximately 100 nM, consistent with a physiological role. Also consistent with such a role, cellular fibronectin FN is shown to positively regulate BMP1 processing activity against Chordin, probiglycan, and type I procollagen in vitro. Endogenous FN and BMP1 are demonstrated to co-localize in cell layers and to form complexes in culture medium. In addition, processing of endogenous BMP1 substrates Chordin, probiglycan, and procollagen is demonstrated to be strikingly reduced in cultures of FN(-/-) MEFs compared with FN(+/-) MEF cultures despite similar levels of endogenous BMP1. These data support the conclusion that FN binds BMP1-like proteinases in vivo and that FN is an important determinant of the in vivo activity levels of BMP1-like proteinases.


Subject(s)
Bone Morphogenetic Protein 1/metabolism , Fibroblasts/metabolism , Fibronectins/metabolism , Animals , Biglycan , Bone Morphogenetic Protein 1/genetics , Cell Line, Tumor , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Fibroblasts/cytology , Fibronectins/genetics , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Procollagen/genetics , Procollagen/metabolism , Protein Binding/physiology , Protein-Lysine 6-Oxidase/genetics , Protein-Lysine 6-Oxidase/metabolism , Proteoglycans/genetics , Proteoglycans/metabolism , Substrate Specificity/physiology
20.
Nat Struct Mol Biol ; 12(10): 910-4, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16186819

ABSTRACT

Thrombospondins (THBSs) are secreted glycoproteins that have key roles in interactions between cells and the extracellular matrix. Here, we describe the 2.6-A-resolution crystal structure of the glycosylated signature domain of human THBS2, which includes three epidermal growth factor-like modules, 13 aspartate-rich repeats and a lectin-like module. These elements interact extensively to form three structural regions termed the stalk, wire and globe. The THBS2 signature domain is stabilized by these interactions and by a network of 30 bound Ca(2+) ions and 18 disulfide bonds. The structure suggests how genetic alterations of THBSs result in disease.


Subject(s)
Calcium/chemistry , Thrombospondins/chemistry , Amino Acid Sequence , Bone Diseases/genetics , Calcium/metabolism , Crystallization , Humans , Joint Diseases/genetics , Molecular Sequence Data , Mutation , Protein Structure, Tertiary , Thrombospondins/genetics , Thrombospondins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL