Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Cells ; 13(11)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38891113

ABSTRACT

Tigilanol tiglate (TT, also known as EBC-46) is a novel, plant-derived diterpene ester possessing anticancer and wound-healing properties. Here, we show that TT-evoked PKC-dependent S985 phosphorylation of the tyrosine kinase MET leads to subsequent degradation of tyrosine phosphorylated p-Y1003 and p-Y1234/5 MET species. PKC inhibition with BIM-1 blocked S985 phosphorylation of MET and led to MET cell surface accumulation. Treatment with metalloproteinase inhibitors prevented MET-ECD release into cell culture media, which was also blocked by PKC inhibitors. Furthermore, unbiased secretome analysis, performed using TMT-technology, identified additional targets of TT-dependent release of cell surface proteins from H357 head and neck cancer cells. We confirm that the MET co-signalling receptor syndecan-1 was cleaved from the cell surface in response to TT treatment. This was accompanied by rapid cleavage of the cellular junction adhesion protein Nectin-1 and the nerve growth factor receptor NGFRp75/TNFR16. These findings, that TT is a novel negative regulator of protumorigenic c-MET and NGFRp75/TNFR16 signalling, as well as regulating Nectin-1-mediated cell adhesion, further contribute to our understanding of the mode of action and efficacy of TT in the treatment of solid tumours.


Subject(s)
Head and Neck Neoplasms , Proto-Oncogene Proteins c-met , Humans , Proto-Oncogene Proteins c-met/metabolism , Phosphorylation/drug effects , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Cell Line, Tumor , Secretome/metabolism , Diterpenes/pharmacology , Membrane Proteins/metabolism , Signal Transduction/drug effects , Syndecan-1/metabolism , Nectins/metabolism , Protein Kinase C/metabolism
2.
Int J Mol Sci ; 25(4)2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38397010

ABSTRACT

A wound healing model was developed to elucidate the role of mesenchymal-matrix-associated transglutaminase 2 (TG2) in keratinocyte re-epithelialisation. TG2 drives keratinocyte migratory responses by activation of disintegrin and metalloproteinase 17 (ADAM17). We demonstrate that epidermal growth factor (EGF) receptor ligand shedding leads to EGFR-transactivation and subsequent rapid keratinocyte migration on TG2-positive ECM. In contrast, keratinocyte migration was impaired in TG2 null conditions. We show that keratinocytes express the adhesion G-protein-coupled receptor, ADGRG1 (GPR56), which has been proposed as a TG2 receptor. Using ADAM17 activation as a readout and luciferase reporter assays, we demonstrate that TG2 activates GPR56. GPR56 activation by TG2 reached the same level as observed with an agonistic N-GPR56 antibody. The N-terminal GPR56 domain is required for TG2-regulated signalling response, as the constitutively active C-GPR56 receptor was not activated by TG2. Signalling required the C-terminal TG2 ß-barrel domains and involved RhoA-associated protein kinase (ROCK) and ADAM17 activation, which was blocked by specific inhibitors. Cell surface binding of TG2 to the N-terminal GPR56 domain is rapid and is associated with TG2 and GPR56 endocytosis. TG2 and GPR56 represent a ligand receptor pair causing RhoA and EGFR transactivation. Furthermore, we determined a binding constant for the interaction of human TG2 with N-GPR56 and show for the first time that only the calcium-enabled "open" TG2 conformation associates with N-GPR56.


Subject(s)
Protein Glutamine gamma Glutamyltransferase 2 , Receptors, G-Protein-Coupled , Humans , ADAM17 Protein/genetics , ADAM17 Protein/metabolism , ErbB Receptors/metabolism , Ligands , Protein Glutamine gamma Glutamyltransferase 2/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
3.
Alzheimers Dement ; 20(3): 2016-2033, 2024 03.
Article in English | MEDLINE | ID: mdl-38184788

ABSTRACT

INTRODUCTION: Genome-wide association studies link susceptibility to late-onset Alzheimer's disease (LOAD) with EphA1. Sequencing identified a non-synonymous substitution P460L as a LOAD risk variant. Other Ephs regulate vascular permeability and immune cell recruitment. We hypothesized that P460L dysregulates EphA1 receptor activity and impacts neuroinflammation. METHODS: EphA1/P460L receptor activity was assayed in isogenic Human Embryonic Kidney (HEK) cells. Soluble EphA1/P460L (sEphA1/sP460L) reverse signaling in brain endothelial cells was assessed by T-cell recruitment and barrier function assays. RESULTS: EphA1 and P460L were expressed in HEK cells, but membrane and soluble P460L were significantly reduced. Ligand engagement induced Y781 phosphorylation of EphA1 but not P460L. sEphA1 primed brain endothelial cells for increased T-cell recruitment; however, sP460L was less effective. sEphA1 decreased the integrity of the brain endothelial barrier, while sP460L had no effect. DISCUSSION: These findings suggest that P460L alters EphA1-dependent forward and reverse signaling, which may impact blood-brain barrier function in LOAD. HIGHLIGHTS: EphA1-dependent reverse signaling controls recruitment of T cells by brain endothelial cells. EphA1-dependent reverse signaling remodels brain endothelial cell contacts. LOAD-associated P460L variant of EphA1 shows reduced membrane expression and reduced ligand responses. LOAD-associated P460L variant of EphA1 fails to reverse signal to brain endothelial cells.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/genetics , Blood-Brain Barrier , Endothelial Cells , Genome-Wide Association Study , Ligands , Receptor, EphA1/metabolism
4.
Front Oncol ; 12: 841890, 2022.
Article in English | MEDLINE | ID: mdl-35600402

ABSTRACT

G protein-coupled receptor 56 (GPR56/ADGRG1) is an adhesion GPCR with an essential role in brain development and cancer. Elevated expression of GPR56 was observed in the clinical specimens of Glioblastoma (GBM), a highly invasive primary brain tumor. However, we found the expression to be variable across the specimens, presumably due to the intratumor heterogeneity of GBM. Therefore, we re-examined GPR56 expression in public domain spatial gene expression data and single-cell expression data for GBM, which revealed that GPR56 expression was high in cellular tumors, infiltrating tumor cells, and proliferating cells, low in microvascular proliferation and peri-necrotic areas of the tumor, especially in hypoxic mesenchymal-like cells. To gain a better understanding of the consequences of GPR56 downregulation in tumor cells and other molecular changes associated with it, we generated a sh-RNA-mediated GPR56 knockdown in the GBM cell line U373 and performed transcriptomics, proteomics, and phospho-proteomics analysis. Our analysis revealed enrichment of gene signatures, pathways, and phosphorylation of proteins potentially associated with mesenchymal (MES) transition in the tumor and concurrent increase in cell invasion and migration behavior of the GPR56 knockdown GBM cells. Interestingly, our analysis also showed elevated expression of Transglutaminase 2 (TG2) - a known interactor of GPR56, in the knockdown cells. The inverse expression of GPR56 and TG2 was also observed in intratumoral, spatial gene expression data for GBM and in GBM cell lines cultured in vitro under hypoxic conditions. Integrating all these observations, we propose a putative functional link between the inverse expression of the two proteins, the hypoxic niche and the mesenchymal status in the tumor. Hypoxia-induced downregulation of GPR56 and activation of TG2 may result in a network of molecular events that contribute to the mesenchymal transition of GBM cells, and we propose a putative model to explain this functional and regulatory relationship of the two proteins.

5.
J Thromb Haemost ; 18(4): 942-954, 2020 04.
Article in English | MEDLINE | ID: mdl-31894636

ABSTRACT

BACKGROUND: Atherosclerotic plaque rupture and subsequent thrombosis underpin thrombotic syndromes. Under inflammatory conditions in the unstable plaque, perturbed endothelial cells secrete von Willebrand Factor (VWF) which, via its interaction with GpIbα, enables platelet rolling across and adherence to the damaged endothelium. Following plaque rupture, VWF and platelets are exposed to subendothelial collagen, which supports stable platelet adhesion, activation, and aggregation. Plaque-derived matrix metalloproteinase (MMP)-13 is also released into the surrounding lumen where it may interact with VWF, collagen, and platelets. OBJECTIVES: We sought to discover whether MMP-13 can cleave VWF and whether this might regulate its interaction with both collagen and platelets. METHODS: We have used platelet adhesion assays and whole blood flow experiments to assess the effects of VWF cleavage by MMP-13 on platelet adhesion and thrombus formation. RESULTS: Unlike the shear-dependent cleavage of VWF by a disintegrin and metalloprotease with thrombospondin motif member 13 (ADAMTS13), MMP-13 is able to cleave VWF under static conditions. Following cleavage by MMP-13, immobilized VWF cannot bind to collagen but interacts more strongly with platelets, supporting slower platelet rolling in whole blood under shear. Compared with intact VWF, the interaction of cleaved VWF with platelets results in greater GpIbα upregulation and P-selectin expression, and the thrombi formed on cleaved VWF-collagen co-coatings are larger and more contractile than platelet aggregates on intact VWF-collagen co-coatings or on collagen alone. CONCLUSIONS: Our data suggest a VWF-mediated role for MMP-13 in the recruitment of platelets to the site of vascular injury and may provide new insights into the association of MMP-13 in atherothrombotic and stroke pathologies.


Subject(s)
Blood Platelets , Collagen , Matrix Metalloproteinase 13 , von Willebrand Factor , Endothelial Cells , Humans , Platelet Adhesiveness
6.
Sci Rep ; 9(1): 12540, 2019 08 29.
Article in English | MEDLINE | ID: mdl-31467400

ABSTRACT

A Disintegrin and Metalloproteinase-15 (ADAM15) is a transmembrane protein involved in protein ectodomain shedding, cell adhesion and signalling. We previously cloned and characterised alternatively spliced variants of ADAM15 that differ in their intracellular domains and demonstrated correlation of the expression of specific variants with breast cancer prognosis. In this study we have created isogenic cell panels (MDA-MB-231 and MCF-7) expressing five ADAM15 variants including wild-type and catalytically inactive forms. The expression of ADAM15 isoforms in MDA-MB-231 cells led to cell clustering to varying degree, without changes in EMT markers vimentin, slug and E-cadherin. Analysis of tight junction molecules revealed ADAM15 isoform specific, catalytic function dependent upregulation of Claudin-1. The expression of ADAM15A, and to a lesser degree of C and E isoforms led to an increase in Claudin-1 expression in MDA-MB-231 cells, while ADAM15B had no effect. In MCF-7 cells, ADAM15E was the principal variant inducing Claudin-1 expression. Sh-RNA mediated down-regulation of ADAM15 in ADAM15 over-expressing cells reduced Claudin-1 levels. Additionally, downregulation of endogenous ADAM15 expression in T47D cells by shRNA reduced endogenous Claudin-1 expression confirming a role for ADAM15 in regulating Claudin-1 expression. The PI3K/Akt/mTOR pathway was involved in regulating Claudin-1 expression downstream of ADAM15. Immunofluorescence analysis of MDA-MB-231 ADAM15A expressing cells showed Claudin-1 at cell-cell junctions, in the cytoplasm and nuclei. ADAM15 co-localised with Claudin-1 and ZO1 at cell-cell junctions. Immunoprecipitation analysis demonstrated complex formation between ADAM15 and ZO1/ZO2. These findings highlight the importance of ADAM15 Intra Cellular Domain-mediated interactions in regulating substrate selection and breast cancer cell phenotype.


Subject(s)
ADAM Proteins/metabolism , Breast Neoplasms/genetics , Claudin-1/genetics , Membrane Proteins/metabolism , ADAM Proteins/genetics , Breast Neoplasms/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Claudin-1/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Membrane Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction , Transcriptional Activation , Up-Regulation
7.
Sci Rep ; 9(1): 5487, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30940840

ABSTRACT

L-selectin on T-cells is best known as an adhesion molecule that supports recruitment of blood-borne naïve and central memory cells into lymph nodes. Proteolytic shedding of the ectodomain is thought to redirect activated T-cells from lymph nodes to sites of infection. However, we have shown that activated T-cells re-express L-selectin before lymph node egress and use L-selectin to locate to virus-infected tissues. Therefore, we considered other roles for L-selectin proteolysis during T cell activation. In this study, we used T cells expressing cleavable or non-cleavable L-selectin and determined the impact of L-selectin proteolysis on T cell activation in virus-infected mice. We confirm an essential and non-redundant role for ADAM17 in TCR-induced proteolysis of L-selectin in mouse and human T cells and show that L-selectin cleavage does not regulate T cell activation measured by CD69 or TCR internalisation. Following virus infection of mice, L-selectin proteolysis promoted early clonal expansion of cytotoxic T cells resulting in an 8-fold increase over T cells unable to cleave L-selectin. T cells unable to cleave L-selectin showed delayed proliferation in vitro which correlated with lower CD25 expression. Based on these results, we propose that ADAM17-dependent proteolysis of L-selectin should be considered a regulator of T-cell activation at sites of immune activity.


Subject(s)
ADAM17 Protein/metabolism , Clone Cells/immunology , L-Selectin/metabolism , T-Lymphocytes, Cytotoxic/immunology , Virus Diseases/metabolism , ADAM17 Protein/genetics , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Cell Movement , Cells, Cultured , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , L-Selectin/genetics , Lectins, C-Type/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mutation , Proteolysis , Virus Diseases/immunology
8.
Res Pract Thromb Haemost ; 2(2): 370-379, 2018 Apr.
Article in English | MEDLINE | ID: mdl-30046741

ABSTRACT

BACKGROUND: Acute thrombotic syndromes lead to atherosclerotic plaque rupture with subsequent thrombus formation, myocardial infarction and stroke. Following rupture, flowing blood is exposed to plaque components, including collagen, which triggers platelet activation and aggregation. However, plaque rupture releases other components into the surrounding vessel which have the potential to influence platelet function and thrombus formation. OBJECTIVES: Here we sought to elucidate whether matrix metalloproteinase-13 (MMP-13), a collagenolytic metalloproteinase up-regulated in atherothrombotic and inflammatory conditions, affects platelet aggregation and thrombus formation. RESULTS: We demonstrate that MMP-13 is able to bind to platelet receptors alphaIIbbeta3 (αIIbß3) and platelet glycoprotein (GP)VI. The interactions between MMP-13, GPVI and αIIbß3 are sufficient to significantly inhibit washed platelet aggregation and decrease thrombus formation on fibrillar collagen. CONCLUSIONS: Our data demonstrate a role for MMP-13 in the inhibition of both platelet aggregation and thrombus formation in whole flowing blood, and may provide new avenues of research into the mechanisms underlying the subtle role of MMP-13 in atherothrombotic pathologies.

9.
Cell Biol Int ; 42(3): 273-280, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28762604

ABSTRACT

TMEFF2 is a type I transmembrane protein with two follistatin (FS) and one EGF-like domain over-expressed in prostate cancer; however its biological role in prostate cancer development and progression remains unclear, which may, at least in part, be explained by its proteolytic processing. The extracellular part of TMEFF2 (TMEFF2-ECD) is cleaved by ADAM17 and the membrane-retained fragment is further processed by the gamma-secretase complex. TMEFF2 shedding is increased with cell crowding, a condition associated with the tumour microenvironment, which was mediated by oxidative stress signalling, requiring jun-kinase (JNK) activation. Moreover, we have identified that TMEFF2 is also a novel substrate for other proteases implicated in prostate cancer, including two ADAMs (ADAM9 and ADAM12) and the type II transmembrane serine proteinases (TTSPs) matriptase-1 and hepsin. Whereas cleavage by ADAM9 and ADAM12 generates previously identified TMEFF2-ECD, proteolytic processing by matriptase-1 and hepsin produced TMEFF2 fragments, composed of TMEFF2-ECD or FS and/or EGF-like domains as well as novel membrane retained fragments. Differential TMEFF2 processing from a single transmembrane protein may be a general mechanism to modulate transmembrane protein levels and domains, dependent on the repertoire of ADAMs or TTSPs expressed by the target cell.


Subject(s)
ADAM Proteins/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Oxidative Stress/physiology , Prostatic Neoplasms/metabolism , ADAM12 Protein/metabolism , ADAM17 Protein/metabolism , Amino Acid Sequence , Cell Membrane/metabolism , HEK293 Cells , Humans , Male , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Reactive Oxygen Species/metabolism , Serine Endopeptidases/metabolism
10.
Amino Acids ; 49(3): 453-460, 2017 03.
Article in English | MEDLINE | ID: mdl-27562793

ABSTRACT

Transglutaminases have important roles in stabilizing extracellular protein assemblies in tissue repair processes but some reaction products can stimulate immune activation, leading to chronic inflammatory conditions or autoimmunity. Exacerbated disease in models of inflammatory arthritis has been ascribed to sustained extracellular enzyme activity alongside formation of select protein modifications. Here, we review the evidence, with a focus on the link between P2X7R signaling and TG2 export, a pathway that we have recently discovered which ties extracellular protein modifications into the danger signal-mediated innate immune response. These recent insights offer new opportunities for therapeutic intervention.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Bone and Bones/immunology , Cartilage/immunology , GTP-Binding Proteins/immunology , Receptors, Purinergic P2X7/immunology , Transglutaminases/immunology , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Autoimmunity , Bone and Bones/pathology , Cartilage/pathology , Cytokines/genetics , Cytokines/immunology , GTP-Binding Proteins/genetics , Gene Expression Regulation , Humans , Immunity, Innate , Inflammasomes/genetics , Inflammasomes/immunology , Inflammation , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Protein Glutamine gamma Glutamyltransferase 2 , Protein Transport/immunology , Receptors, Purinergic P2X7/genetics , Signal Transduction , Transglutaminases/genetics
11.
Cell Rep ; 15(8): 1757-70, 2016 05 24.
Article in English | MEDLINE | ID: mdl-27184850

ABSTRACT

Natural killer (NK) cells possess potent cytotoxic mechanisms that need to be tightly controlled. Here, we explored the regulation and function of GPR56/ADGRG1, an adhesion G protein-coupled receptor implicated in developmental processes and expressed distinctively in mature NK cells. Expression of GPR56 was triggered by Hobit (a homolog of Blimp-1 in T cells) and declined upon cell activation. Through studying NK cells from polymicrogyria patients with disease-causing mutations in ADGRG1, encoding GPR56, and NK-92 cells ectopically expressing the receptor, we found that GPR56 negatively regulates immediate effector functions, including production of inflammatory cytokines and cytolytic proteins, degranulation, and target cell killing. GPR56 pursues this activity by associating with the tetraspanin CD81. We conclude that GPR56 inhibits natural cytotoxicity of human NK cells.


Subject(s)
Killer Cells, Natural/metabolism , Receptors, G-Protein-Coupled/metabolism , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line , Cells, Cultured , Cytokines/pharmacology , Cytotoxicity, Immunologic/drug effects , Down-Regulation/drug effects , Humans , Immunological Synapses/drug effects , Inflammation Mediators/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Malformations of Cortical Development/pathology , Receptors, G-Protein-Coupled/deficiency , Tetraspanin 28/metabolism , Transcription Factors/metabolism
12.
J Cell Sci ; 128(24): 4615-28, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26542019

ABSTRACT

Transglutaminases (denoted TG or TGM) are externalized from cells via an unknown unconventional secretory pathway. Here, we show for the first time that purinergic signaling regulates active secretion of TG2 (also known as TGM2), an enzyme with a pivotal role in stabilizing extracellular matrices and modulating cell-matrix interactions in tissue repair. Extracellular ATP promotes TG2 secretion by macrophages, and this can be blocked by a selective antagonist against the purinergic receptor P2X7 (P2X7R, also known as P2RX7). Introduction of functional P2X7R into HEK293 cells is sufficient to confer rapid, regulated TG2 export. By employing pharmacological agents, TG2 release could be separated from P2X7R-mediated microvesicle shedding. Neither Ca(2+) signaling alone nor membrane depolarization triggered TG2 secretion, which occurred only upon receptor membrane pore formation and without pannexin channel involvement. A gain-of-function mutation in P2X7R associated with autoimmune disease caused enhanced TG2 externalization from cells, and this correlated with increased pore activity. These results provide a mechanistic explanation for a link between active TG2 secretion and inflammatory responses, and aberrant enhanced TG2 activity in certain autoimmune conditions.


Subject(s)
Adenosine Triphosphate/metabolism , Calcium Signaling , GTP-Binding Proteins/metabolism , Membrane Potentials , Receptors, Purinergic P2X7/metabolism , Transglutaminases/metabolism , Autoimmune Diseases/genetics , Autoimmune Diseases/metabolism , Cell Line, Tumor , Female , GTP-Binding Proteins/genetics , HEK293 Cells , Humans , Male , Mutation , Protein Glutamine gamma Glutamyltransferase 2 , Receptors, Purinergic P2X7/genetics , Transglutaminases/genetics
13.
Cell Signal ; 27(7): 1325-35, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25817572

ABSTRACT

TROP2, a cancer cell surface protein with both pro-oncogenic and anti-oncogenic properties is cleaved by ADAM17. ADAM17 dependent cleavage requires novel PKC activity which is blocked by the ADAM10/ADAM17 inhibitor GW64 as well as by the PKC inhibitor Bim-1. Full length TROP2 release is induced by classical PKC activation and blocked by Gö6979, without affecting ADAM17 dependent TROP2 cleavage. Full length TROP2 is released in ectosomes, as inhibition of endocytosis did not prevent release. Inhibition of the atypical PKC isoform PKCζ stimulated metalloproteinase dependent N-terminal alternative TROP2 cleavage. The resulting alternative TROP2 cleavage product remains membrane associated via a disulphide bond, but is released in microvesicles with an average size of 107nm. Inhibition of endocytosis following PKCζ inhibition prevented alternative cleavage and release of TROP2, suggesting that these events require endocytic uptake and exosomal release of the corresponding microvesicles. The alternative TROP2 cleavage product was also found in PC3 cell lysates following deglycosylation, and may represent a novel biomarker in prostate cancer.


Subject(s)
ADAM Proteins/metabolism , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Protein Kinase C/metabolism , ADAM17 Protein , Antigens, Neoplasm/genetics , Cell Adhesion Molecules/genetics , Endocytosis/drug effects , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , RNA Interference , RNA, Small Interfering/metabolism , Tetradecanoylphorbol Acetate/pharmacology
14.
BMC Res Notes ; 7: 913, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25516095

ABSTRACT

BACKGROUND: Recently, the orphan G-protein coupled receptor 83 (GPR83) was identified as a new participant in body weight regulation. This receptor is highly expressed in the hypothalamic arcuate nucleus and is regulated in response to nutrient availability. Gpr83 knock-out mice are protected from diet-induced obesity. Moreover, in a previous study, we designed and characterized several artificial constitutively activating mutations (CAMs) in GPR83. A particular CAM was located in the extracellular N-terminal domain (eNDo) that is highly conserved among GPR83 orthologs. This suggests the contribution of this receptor part into regulation of signaling, which needed a more detailed investigation. FINDINGS: In this present study, therefore, we further explored the role of the eNDo in regulating GPR83-signaling and demonstrate a proof-of-principle approach in that deletion mutants are characterized by a strong increase in basal Gq/11-mediated signaling, whilst none of the additionally characterized signaling pathways (Gs, Gi, G12/13) were activated by the N-terminal deletion variants. Of note, we detected basal GPR83 MAPK-activity of the wild type receptor, which was not increased in the deletion variants. CONCLUSIONS: Finally, the extracellular portion of GPR83 has a strong regulatory function on this receptor. A suppressive - inverse agonistic - effect of the eNDo on GPR83 signaling activity is demonstrated here, which also suggests a putative link between extracellular receptor activation and proteolytic cleavage. These new insights highlight important aspects of GPR83-regulation and might open options in the development of tools to modulate GPR83-signaling.


Subject(s)
Protein Structure, Tertiary , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Binding Sites/genetics , COS Cells , Cell Membrane/metabolism , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Mice , Mitogen-Activated Protein Kinases/metabolism , Models, Molecular , Molecular Sequence Data , Mutation , Receptors, G-Protein-Coupled/genetics , Sequence Homology, Amino Acid
15.
J Biol Chem ; 289(35): 24091-101, 2014 Aug 29.
Article in English | MEDLINE | ID: mdl-25008319

ABSTRACT

Remodeling of collagen by matrix metalloproteinases (MMPs) is crucial to tissue homeostasis and repair. MMP-13 is a collagenase with a substrate preference for collagen II over collagens I and III. It recognizes a specific, well-known site in the tropocollagen molecule where its binding locally perturbs the triple helix, allowing the catalytic domain of the active enzyme to cleave the collagen α chains sequentially, at Gly(775)-Leu(776) in collagen II. However, the specific residues upon which collagen recognition depends within and surrounding this locus have not been systematically mapped. Using our triple-helical peptide Collagen Toolkit libraries in solid-phase binding assays, we found that MMP-13 shows little affinity for Collagen Toolkit III, but binds selectively to two triple-helical peptides of Toolkit II. We have identified the residues required for the adhesion of both proMMP-13 and MMP-13 to one of these, Toolkit peptide II-44, which contains the canonical collagenase cleavage site. MMP-13 was unable to bind to a linear peptide of the same sequence as II-44. We also discovered a second binding site near the N terminus of collagen II (starting at helix residue 127) in Toolkit peptide II-8. The pattern of binding of the free hemopexin domain of MMP-13 was similar to that of the full-length enzyme, but the free catalytic subunit bound none of our peptides. The susceptibility of Toolkit peptides to proteolysis in solution was independent of the very specific recognition of immobilized peptides by MMP-13; the enzyme proved able to cleave a range of dissolved collagen peptides.


Subject(s)
Collagen Type II/metabolism , Matrix Metalloproteinase 13/metabolism , Peptides/metabolism , Amino Acid Sequence , Base Sequence , Collagen Type II/chemistry , DNA Primers , Matrix Metalloproteinase 13/chemistry , Molecular Sequence Data , Proteolysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Substrate Specificity
16.
PLoS One ; 8(3): e55667, 2013.
Article in English | MEDLINE | ID: mdl-23469166

ABSTRACT

Tissue inhibitors of metalloproteinases (TIMPs) while originally characterized as inhibitors of matrix metalloproteinases (MMPs) have recently been shown to have a wide range of functions that are independent of their MMP inhibitory properties. Tissue inhibitor of metalloproteinases-3 (TIMP-3) is a potent inhibitor of VEGF-mediated angiogenesis and neovascularization through its ability to block the binding of VEGF to its receptor VEGFR-2. To identify and characterize the anti-angiogenic domain of TIMP-3, structure function analyses and synthetic peptide studies were performed using VEGF-mediated receptor binding, signaling, migration and proliferation. In addition, the ability of TIMP-3 peptides to inhibit CNV in a mouse model was evaluated. We demonstrate that the anti-angiogenic property resides in the COOH-terminal domain of TIMP-3 protein which can block the binding of VEGF specifically to its receptor VEGFR-2, but not to VEGFR-1 similar to the full-length wild-type protein. Synthetic peptides corresponding to putative loop 6 and tail region of TIMP-3 have anti-angiogenic properties as determined by inhibition of VEGF binding to VEGFR-2, VEGF-induced phosphorylation of VEGFR-2 and downstream signaling pathways as well as endothelial cell proliferation and migration in response to VEGF. In addition, we show that intravitreal administration of TIMP-3 peptide could inhibit the size of laser-induced choroidal neovascularization lesions in mice. Thus, we have identified TIMP-3 peptides to be efficient inhibitors of angiogenesis and have a potential to be used therapeutically in diseases with increased neovascularization.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Bruch Membrane/blood supply , Bruch Membrane/drug effects , Choroidal Neovascularization/prevention & control , Peptides/pharmacology , Tissue Inhibitor of Metalloproteinase-3/antagonists & inhibitors , Tissue Inhibitor of Metalloproteinase-3/chemistry , Angiogenesis Inhibitors/chemical synthesis , Animals , Bruch Membrane/injuries , Cells, Cultured , Choroidal Neovascularization/etiology , Choroidal Neovascularization/genetics , Choroidal Neovascularization/metabolism , Endothelium, Vascular , Gene Expression Regulation/drug effects , Humans , Laser Coagulation/adverse effects , Mice , Mice, Inbred C57BL , Peptides/chemical synthesis , Phosphorylation , Signal Transduction/drug effects , Solid-Phase Synthesis Techniques , Tissue Inhibitor of Metalloproteinase-3/genetics , Tissue Inhibitor of Metalloproteinase-3/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
17.
Methods Mol Biol ; 622: 233-43, 2010.
Article in English | MEDLINE | ID: mdl-20135286

ABSTRACT

The degradation of the extracellular matrix during development and in disease is thought to result from the combined action of several proteolytic enzyme systems, including the matrix metalloproteinases (MMPs), serine proteinases, and cysteine proteinases. The majority of the soluble MMPs are synthesized as proenzymes which require extracellular activation in order to gain proteolytic activity and the analysis of their activation mechanism is a prerequisite for understanding MMP-mediated proteolysis.The emphasis of this chapter is the provision of the experimental tools to study MMP activation in vitro and in cellular model systems. Hence, we use the activation of procollagenase-3 (proMMP-13) and progelatinase A (proMMP-2) as examples of the methods used.


Subject(s)
Matrix Metalloproteinases/metabolism , Molecular Biology/methods , Cell Line, Tumor , Collagenases/metabolism , Enzyme Activation/drug effects , Enzyme Precursors/metabolism , Gelatinases/metabolism , Humans , Models, Biological , Phenylmercuric Acetate/analogs & derivatives , Phenylmercuric Acetate/pharmacology
18.
J Biol Chem ; 282(52): 37378-88, 2007 Dec 28.
Article in English | MEDLINE | ID: mdl-17942404

ABSTRACT

The transmembrane protein with epidermal growth factor and two follistatin motifs 2 (TMEFF2) is expressed in prostate and brain and shed from the cell surface in a metalloproteinase-dependent fashion. Neither the sheddase(s) responsible for TMEFF2 shedding nor the physiological significance or activity of the soluble TMEFF2 ectodomain (TMEFF2-ECD) has been identified. In the present study we present new evidence that a disintegrin and metalloproteinase-17 (ADAM17) is responsible for phorbol 12-myristate 13-acetate-induced release of TMEFF2-ECD using small interfering RNA to ablate ADAM17 expression or by inhibiting enzymatic activity. A single well shedding assay monitoring the release of alkaline phosphatase-tagged TMEFF2-ECD into medium and the generation of 22- and 14-kDa C-terminal fragments in lysates were dependent on ADAM17 activity. A gamma-secretase inhibitor prevented the formation of a 10-kDa fragment in cell lysates, thus establishing TMEFF2 as a novel substrate for regulated intramembrane proteolysis. We assigned proliferation-inducing activity to TMEFF2. Inhibition of TMEFF2 shedding using synthetic metalloproteinase inhibitors or small interfering RNA targeting TMEFF2 expression yielded a statistically significant reduction of cell proliferation in the lymph node-derived prostate cancer cells (LNCaPs) and a human embryonic kidney (HEK293) cell line overexpressing TMEFF2. The TMEFF2-ECD was able to induce ERK1/2 phosphorylation in an epidermal growth factor receptor (or ErbB1)-dependent manner in HEK293 cells. Our data suggest that TMEFF2 contributes to cell proliferation in an ADAM17-dependent autocrine fashion in cells expressing this protein.


Subject(s)
ADAM Proteins/chemistry , Cell Membrane/metabolism , Epidermal Growth Factor/metabolism , Follistatin/chemistry , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Phorbol Esters/pharmacology , Prostatic Neoplasms/metabolism , ADAM17 Protein , Amino Acid Motifs , Cell Line , Cell Line, Tumor , Cell Proliferation , Humans , Male , Models, Biological , Protein Conformation , Protein Structure, Tertiary , RNA, Small Interfering/metabolism
19.
J Biol Chem ; 281(33): 23386-94, 2006 Aug 18.
Article in English | MEDLINE | ID: mdl-16777853

ABSTRACT

Tissue inhibitor of metalloproteinases-2 (TIMP-2) is unique as it is the only member of the TIMP family that is involved in the cellular activation of promatrix metalloproteinase-2 (pro-MMP-2) by virtue of forming a trimolecular complex with membrane type 1 matrix metalloproteinase (MT1-MMP) on the cell surface. TIMP-4 is similar in structure to TIMP-2 but is unable to support the activation of the proenzyme. Several reports have highlighted the importance of the TIMP-2 C-terminal domain in the pro-MMP-2 activation complex; however, very little is known about the role of the extended AB loop of TIMP-2 in this mechanism even though it has been shown to interact with MT1-MMP. In this study we show by mutagenesis and kinetic analysis that it is possible to transfer the MT1-MMP binding affinity of the TIMP-2 AB loop to TIMP-4 but that its transplantation into TIMP-4 does not endow the inhibitor with pro-MMP-2 activating activity. However, transfer of both the AB loop and C-terminal domain of TIMP-2 to TIMP-4 generates a mutant that can activate pro-MMP-2 and so demonstrates that both these regions of TIMP-2 are important for the activation process.


Subject(s)
Enzyme Precursors/metabolism , Matrix Metalloproteinases/metabolism , Tissue Inhibitor of Metalloproteinase-2/chemistry , Tissue Inhibitor of Metalloproteinase-2/physiology , Amino Acid Sequence , Animals , Enzyme Activation , Enzyme Precursors/antagonists & inhibitors , Humans , Matrix Metalloproteinase 14 , Matrix Metalloproteinase Inhibitors , Matrix Metalloproteinases, Membrane-Associated , Mice , Molecular Sequence Data , Mutant Chimeric Proteins/chemical synthesis , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/physiology , Protein Structure, Tertiary/genetics , Proteins/chemistry , Proteins/genetics , Recombinant Fusion Proteins/chemical synthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , Tissue Inhibitor of Metalloproteinase-2/deficiency , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinases , Tissue Inhibitor of Metalloproteinase-4
20.
Cardiovasc Res ; 67(1): 39-49, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15949468

ABSTRACT

OBJECTIVES: Vascular cell adhesion molecule-1 (VCAM-1) is a cell surface adhesion molecule involved in the recruitment of leukocytes to endothelial cells on arterial walls during the pathogenesis of atherosclerosis. The soluble ectodomain of VCAM-1 (sVCAM-1) is proteolytically released from the cell surface into the circulation, a process which is up-regulated in patients with cardiovascular or inflammatory disease. Here we investigate mechanisms involved in sVCAM-1 generation in response to cytokine stimulation. METHODS: VCAM-1 ectodomain release into the conditioned media of MCEC-1 murine endothelial cells and cells grown from primary aortic explants from timp3-/- mice and wild-type littermates was measured by sandwich ELISA and Western blot after stimulation with tumor necrosis factor alpha (TNFalpha), interleukin-1beta (IL-1beta), or the phorbol ester PMA. Protease expression was inhibited (knocked down) with siRNA and validated using real-time PCR. RESULTS: Proinflammatory cytokines IL-1beta and TNFalpha up-regulated VCAM-1 ectodomain release from the MCEC-1 cells, and this was dependant on p38 and mitogen-activated protein kinases (MAP kinases) and inhibited by the matrix metalloproteinase (MMP) inhibitor BB94 and tissue inhibitor of metalloproteinase (TIMP)-3, but not TIMP-1 or TIMP-2. Timp-3-/- cells exhibited greater VCAM-1 ectodomain release following cytokine stimulation than TIMP-3-expressing cells. Additionally, cytokine stimulation of MCEC-1 cells was shown to cause down-regulation of TIMP-3 expression. Knockdown of the metalloproteinase ADAM17, but not ADAM10 or ADAM12, gene expression reduced cytokine-stimulated VCAM-1 shedding. CONCLUSIONS: TIMP-3 regulates the release of sVCAM-1 from cytokine-stimulated endothelial cells, which is mediated by ADAM17.


Subject(s)
Cytokines/pharmacology , Endothelial Cells/metabolism , Tissue Inhibitor of Metalloproteinase-3/physiology , Vascular Cell Adhesion Molecule-1/physiology , Animals , Aorta , Blotting, Northern , Blotting, Western/methods , Cell Line , Cells, Cultured , Culture Media, Conditioned , Endothelial Cells/drug effects , Enzyme-Linked Immunosorbent Assay/methods , Extracellular Signal-Regulated MAP Kinases , Humans , Interleukin-1 , Mice , Mice, Knockout , PPAR alpha , Stimulation, Chemical , Tissue Inhibitor of Metalloproteinase-3/genetics , Tumor Necrosis Factor-alpha , Umbilical Veins , Vascular Cell Adhesion Molecule-1/analysis , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL