Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Biol Chem ; 402(3): 253-269, 2021 02 23.
Article in English | MEDLINE | ID: mdl-33108336

ABSTRACT

Redox-mediated signal transduction depends on the enzymatic production of second messengers such as hydrogen peroxide, nitric oxide and hydrogen sulfite, as well as specific, reversible redox modifications of cysteine-residues in proteins. So-called thiol switches induce for instance conformational changes in specific proteins that regulate cellular pathways e.g., cell metabolism, proliferation, migration, gene expression and inflammation. Reduction, oxidation and disulfide isomerization are controlled by oxidoreductases of the thioredoxin family, including thioredoxins, glutaredoxins, peroxiredoxins and protein dsisulfide isomerases. These proteins are located in different cellular compartments, interact with substrates and catalyze specific reactions. Interestingly, some of these proteins are released by cells. Their extracellular functions and generally extracellular redox control have been widely underestimated. Here, we give an insight into extracellular redox signaling, extracellular thiol switches and their regulation by secreted oxidoreductases and thiol-isomerases, a topic whose importance has been scarcely studied so far, likely due to methodological limitations. We focus on the secreted redox proteins and characterized thiol switches in the ectodomains of membrane proteins, such as integrins and the metalloprotease ADAM17, which are among the best-characterized proteins and discuss their underlying mechanisms and biological implications.


Subject(s)
Membrane Proteins/metabolism , Sulfhydryl Compounds/metabolism , Animals , Humans , Oxidation-Reduction , Signal Transduction
2.
Biochem Biophys Res Commun ; 526(2): 355-360, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32222277

ABSTRACT

Proteolytic processing of membrane proteins by A disintegrin and metalloprotease-17 (ADAM17) is a key regulatory step in many physiological and pathophysiological processes. This so-called shedding is essential for development, regeneration and immune defense. An uncontrolled ADAM17 activity promotes cancer development, chronic inflammation and autoimmune diseases. Consequently, the ADAM17 activity is tightly regulated. As a final trigger for the shedding event a phosphatidylserine (PS) flip to the outer leaflet of the cell membrane was recently described. PS interacts with the extracellular part of ADAM17, which results in the shedding event by shifting the catalytic domain towards the membrane close to the cleavage sites within ADAM17 substrates. Our data indicate that the intrinsic proteolytic activity of the catalytic domain is prerequisite for the shedding activity and constantly present. However, the accessibility for substrate cleavage sites is controlled on several levels. In this report, we demonstrate that the positioning of the catalytic domain towards the cleavage sites is a crucial part of the shedding process. This finding contributes to the understanding of the complex and multilayered regulation of ADAM17 at the cell surface.


Subject(s)
ADAM17 Protein/metabolism , Receptors, Interleukin-6/metabolism , ADAM17 Protein/chemistry , Amino Acid Sequence , Catalytic Domain , HEK293 Cells , Humans , Mutation , Phosphatidylserines/metabolism , Proteolysis , Receptors, Interleukin-6/chemistry , Receptors, Interleukin-6/genetics
3.
Cell Physiol Biochem ; 52(4): 850-868, 2019.
Article in English | MEDLINE | ID: mdl-30958660

ABSTRACT

BACKGROUND/AIMS: Endoplasmic reticulum (ER)-resident proteins with a C-terminal KDEL ERretention sequence are captured in the Golgi apparatus by KDEL receptors (KDELRs). The binding of such proteins to these receptors induces their retrograde transport. Nevertheless, some KDEL proteins, such as Protein Disulfide Isomerases (PDIs), are found at the cell surface. PDIs target disulfide bridges in the extracellular domains of proteins, such as integrins or A Disintegrin And Metalloprotease 17 (ADAM17) leading to changes in the structure and function of these molecules. Integrins become activated and ADAM17 inactivated upon disulfide isomerization. The way that PDIs escape from retrograde transport and reach the plasma membrane remains far from clear. Various mechanisms might exist, depending on whether a local cell surface association or a more global secretion is required. METHODS: To get a more detailed insight in the transport of PDIs to the cell surface, methods such as cell surface biotinylation, flow cytometric analysis, immunoprecipitation, fluorescence microscopy as well as labeling of cells with fluorescence labled recombinant PDIA6 was performed. RESULTS: Here, we show that the C-terminal KDEL ER retention sequence is sufficient to prevent secretion of PDIA6 into the extracellular space but is mandatory for its association with the cell surface. The cell surface trafficking of PDIA1, PDIA3, and PDIA6 is dependent on KDELR1, which travels in a dynamic manner to the cell surface. This transport is assumed to result in PDI cell surface association, which differs from PDI inducible secretion into the extracellular space. Distinct PDIs differ in their trafficking properties. Endogenous KDELR1, detectable at the cell surface, might be involved not only in the transport of cell-surface-associated PDIs, but also in their retrieval and internalization from the extracellular space. CONCLUSION: Beside their ER retention motive PDIs travel to the cell surface. Here they target different proteins to render their function. To escape the ER PDIs travel via various pathways. One of them depends on the KDELR1, which can transport its target to the cell surface, where it is to be expected to release its cargo in close vicinity to its target molecules. Hence, the KDEL sequence is needed for cell surface association of PDIs, such as PDIA6.


Subject(s)
ADAM17 Protein/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Protein Disulfide-Isomerases/metabolism , Receptors, Peptide/metabolism , ADAM17 Protein/genetics , Cell Membrane/genetics , Endoplasmic Reticulum/genetics , HEK293 Cells , Humans , Protein Disulfide-Isomerases/genetics , Protein Transport/physiology , Receptors, Peptide/genetics
4.
Front Immunol ; 9: 2474, 2018.
Article in English | MEDLINE | ID: mdl-30455686

ABSTRACT

Integrin α5ß1 is a crucial adhesion molecule that mediates the adherence of many cell types to the extracellular matrix through recognition of its classic ligand fibronectin as well as to other cells through binding to an alternative counter-receptor, the metalloproteinase ADAM17/TACE. Interactions between integrin α5ß1 and ADAM17 may take place both in trans (between molecules expressed on different cells) or in cis (between molecules expressed on the same cell) configurations. It has been recently reported that the cis association between α5ß1 and ADAM17 keeps both molecules inactive, whereas their dissociation results in activation of their adhesive and metalloproteinase activities. Here we show that the tetraspanin CD9 negatively regulates integrin α5ß1-mediated cell adhesion by enhancing the cis interaction of this integrin with ADAM17 on the cell surface. Additionally we show that, similarly to CD9, the monoclonal antibody 2A10 directed to the disintegrin domain of ADAM17 specifically inhibits integrin α5ß1-mediated cell adhesion to its ligands fibronectin and ADAM17.


Subject(s)
ADAM17 Protein/metabolism , Leukocytes/immunology , Neoplastic Cells, Circulating/immunology , Tetraspanin 29/metabolism , ADAM17 Protein/genetics , ADAM17 Protein/immunology , Antibodies, Monoclonal/metabolism , CRISPR-Cas Systems , Cell Adhesion , Fibronectins/metabolism , Gene Knockdown Techniques , Humans , Integrin alpha5beta1/metabolism , K562 Cells , Protein Binding
5.
Sci Rep ; 8(1): 1103, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29348576

ABSTRACT

A Disintegrin and Metalloprotease 17 (ADAM17) can cause the fast release of growth factors and inflammatory mediators from the cell surface. Its activity has to be turned on which occurs by various stimuli. The active form can be inactivated by a structural change in its ectodomain, related to the pattern of the formed disulphide bridges. The switch-off is executed by protein disulfide isomerases (PDIs) that catalyze an isomerization of two disulfide bridges and thereby cause a disulfide switch. We demonstrate that the integrity of the CGHC-motif within the active site of PDIs is indispensable. In particular, no major variation is apparent in the activities of the two catalytic domains of PDIA6. The affinities between PDIA1, PDIA3, PDIA6 and the targeted domain of ADAM17 are all in the nanomolar range and display no significant differences. The redundancy between PDIs and their disulfide switch activity in ectodomains of transmembrane proteins found in vitro appears to be a basic characteristic. However, different PDIs might be required in vivo for disulfide switches in different tissues and under different cellular and physiological situations.


Subject(s)
ADAM17 Protein/metabolism , Disintegrins/metabolism , Disulfides/metabolism , Protein Disulfide-Isomerases/metabolism , Catalysis , Humans , Isomerism , Protein Disulfide-Isomerases/chemistry , Substrate Specificity
6.
Oxid Med Cell Longev ; 2017: 8459402, 2017.
Article in English | MEDLINE | ID: mdl-29118897

ABSTRACT

Redox regulation depends on the enzymatically controlled production and decay of redox active molecules. NADPH oxidases, superoxide dismutases, nitric oxide synthases, and others produce the redox active molecules superoxide, hydrogen peroxide, nitric oxide, and hydrogen sulfide. These react with target proteins inducing spatiotemporal modifications of cysteine residues within different signaling cascades. Thioredoxin family proteins are key regulators of the redox state of proteins. They regulate the formation and removal of oxidative modifications by specific thiol reduction and oxidation. All of these redox enzymes affect inflammatory processes and the innate and adaptive immune response. Interestingly, this regulation involves different mechanisms in different biological compartments and specialized cell types. The localization and activity of distinct proteins including, for instance, the transcription factor NFκB and the immune mediator HMGB1 are redox-regulated. The transmembrane protein ADAM17 releases proinflammatory mediators, such as TNFα, and is itself regulated by a thiol switch. Moreover, extracellular redox enzymes were shown to modulate the activity and migration behavior of various types of immune cells by acting as cytokines and/or chemokines. Within this review article, we will address the concept of redox signaling and the functions of both redox enzymes and redox active molecules in innate and adaptive immune responses.


Subject(s)
NADPH Oxidases/metabolism , Nitric Oxide Synthase/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Animals , Humans , Inflammation/enzymology , Inflammation/pathology , Inflammation Mediators/metabolism , Oxidation-Reduction
7.
FEBS Lett ; 591(21): 3567-3587, 2017 11.
Article in English | MEDLINE | ID: mdl-28949004

ABSTRACT

The shedding of ectodomains is a crucial mechanism in many physiological and pathological events. A disintegrin and metalloprotease-17 (ADAM17) is a key sheddase involved in essential processes, such as development, regeneration, and immune defense. ADAM17 exists in two conformations which differ in their disulfide connection in the membrane-proximal domain (MPD). Protein-disulfide isomerases (PDIs) on the cell surface convert the open MPD into a rigid closed form, which corresponds to inactive ADAM17. ADAM17 is expressed in its open activatable form in the endoplasmic reticulum (ER) and consequently must be protected against ER-resident PDI activity. Here, we show that the chaperone 78-kDa glucose-regulated protein (GRP78) protects the MPD against PDI-dependent disulfide-bond isomerization by binding to this domain and, thereby, preventing ADAM17 inhibition.


Subject(s)
ADAM17 Protein/metabolism , Endoplasmic Reticulum/metabolism , Heat-Shock Proteins/metabolism , Protein Disulfide-Isomerases/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum Chaperone BiP , Enzyme Activation , HEK293 Cells , Heat-Shock Proteins/genetics , Humans , Protein Disulfide-Isomerases/genetics , Protein Domains
8.
Biochim Biophys Acta Mol Cell Res ; 1864(11 Pt B): 2088-2095, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28571693

ABSTRACT

In contrast to many other signalling mechanisms shedding of membrane-anchored proteins is an irreversible process. A Disintegrin And Metalloproteinase (ADAM) 17 is one of the major sheddases involved in a variety of physiological and pathophysiological processes including regeneration, differentiation, and cancer progression. Due to its central role in signalling the shedding activity of ADAM17 is tightly regulated, especially on the cell surface, where shedding events take place. The activity of ADAM17 can be subdivided into a catalytic activity and the actual shedding activity. Whereas the catalytic activity is constitutively present, the shedding activity has to be induced and is tightly controlled to prevent pathological situations induced by the release of its substrates. The regulation of the shedding activity of ADAM17 is multilayered and different regions of the protease are involved. Intriguingly, its extracellular domains play crucial roles in different regulatory mechanisms. We will discuss the role of these domains in the control of ADAM17 activity. This article is part of a Special Issue entitled: Proteolysis as a Regulatory Event in Pathophysiology edited by Stefan Rose-John.


Subject(s)
ADAM17 Protein/genetics , Cell Membrane/genetics , Membrane Proteins/genetics , Proteolysis , Cell Membrane/metabolism , Gene Expression Regulation/genetics , Humans , Membrane Proteins/metabolism
9.
PLoS Biol ; 15(1): e2000080, 2017 01.
Article in English | MEDLINE | ID: mdl-28060820

ABSTRACT

Signaling of the cytokine interleukin-6 (IL-6) via its soluble IL-6 receptor (sIL-6R) is responsible for the proinflammatory properties of IL-6 and constitutes an attractive therapeutic target, but how the sIL-6R is generated in vivo remains largely unclear. Here, we use liquid chromatography-mass spectrometry to identify an sIL-6R form in human serum that originates from proteolytic cleavage, map its cleavage site between Pro-355 and Val-356, and determine the occupancy of all O- and N-glycosylation sites of the human sIL-6R. The metalloprotease a disintegrin and metalloproteinase 17 (ADAM17) uses this cleavage site in vitro, and mutation of Val-356 is sufficient to completely abrogate IL-6R proteolysis. N- and O-glycosylation were dispensable for signaling of the IL-6R, but proteolysis was orchestrated by an N- and O-glycosylated sequon near the cleavage site and an N-glycan exosite in domain D1. Proteolysis of an IL-6R completely devoid of glycans is significantly impaired. Thus, glycosylation is an important regulator for sIL-6R generation.


Subject(s)
Proteolysis , Receptors, Interleukin-6/metabolism , ADAM10 Protein/metabolism , ADAM17 Protein/metabolism , Alternative Splicing/genetics , Amino Acid Sequence , Amyloid Precursor Protein Secretases/metabolism , Cell Line , Cell Membrane/metabolism , Glycosylation , Humans , Intracellular Space/metabolism , Mass Spectrometry , Membrane Proteins/metabolism , Mutation/genetics , Polysaccharides/metabolism , Proline/metabolism , Protein Domains , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Interleukin-6/blood , Receptors, Interleukin-6/chemistry , Receptors, Interleukin-6/genetics , Signal Transduction , Solubility , Valine/metabolism
11.
Sci Rep ; 6: 35067, 2016 10 12.
Article in English | MEDLINE | ID: mdl-27731361

ABSTRACT

An important, irreversible step in many signalling pathways is the shedding of membrane-anchored proteins. A Disintegrin And Metalloproteinase (ADAM) 17 is one of the major sheddases involved in a variety of physiological and pathophysiological processes including regeneration, differentiation, and cancer progression. This central role in signalling implies that ADAM17 activity has to be tightly regulated, including at the level of localisation. Most mature ADAM17 is localised intracellularly, with only a small amount at the cell surface. We found that ADAM17 is constitutively internalised by clathrin-coated pits and that physiological stimulators such as GPCR ligands induce ADAM17-mediated shedding, but do not alter the cell-surface abundance of the protease. In contrast, the PKC-activating phorbol ester PMA, often used as a strong inducer of ADAM17, causes not only proteolysis by ADAM17 but also a rapid increase of the mature protease at the cell surface. This is followed by internalisation and subsequent degradation of the protease. Eventually, this leads to a substantial downregulation of mature ADAM17. Our results therefore imply that physiological activation of ADAM17 does not rely on its relocalisation, but that PMA-induced PKC activity drastically dysregulates the localisation of ADAM17.


Subject(s)
ADAM17 Protein/metabolism , Cell Membrane/metabolism , Clathrin-Coated Vesicles/metabolism , Phorbol Esters/pharmacology , Down-Regulation , Enzyme Activation/drug effects , HEK293 Cells , HeLa Cells , Humans , Protein Kinase C/metabolism , Proteolysis/drug effects , Signal Transduction
12.
Biochim Biophys Acta ; 1863(11): 2795-2808, 2016 11.
Article in English | MEDLINE | ID: mdl-27599715

ABSTRACT

By mediating proteolytic shedding on the cell surface the disintegrin and metalloproteinases ADAM10 and ADAM17 function as critical regulators of growth factors, cytokines and adhesion molecules. We here report that stimulation of lung epithelial A549 tumor cells with phorbol-12-myristate-13-acetate (PMA) leads to the downregulation of the surface expressed mature form of ADAM17 without affecting ADAM10 expression. This reduction could not be sufficiently explained by metalloproteinase-mediated degradation, dynamin-mediated internalization or microdomain redistribution of ADAM17. Instead, surface downregulation of ADAM17 was correlated with the presence of its mature form in exosomes. Exosomal ADAM17 release was also observed in monocytic and primary endothelial cells where it could be induced by stimulation with lipopolysaccharide. Antibody-mediated surface labelling of ADAM17 revealed that at least part of exosomal ADAM17 was oriented with the metalloproteinase domain outside and had been expressed on the cell surface. Suppression of iRHOM2-mediated ADAM17 maturation prevented surface expression and exosomal release of ADAM17. Further, deletion of the protease's C-terminus or cell treatment with a calcium chelator diminished exosomal release as well as surface downregulation of ADAM17, underlining that both processes are closely associated. Co-incubation of ADAM17 containing exosomes with cells expressing the ADAM17 substrates TGFα or amphiregulin lead to increased shedding of both substrates. This was prevented when exosomes were prepared from cells with shRNA-mediated ADAM17 knockdown. These data indicate that cell stimulation can downregulate expression of mature ADAM17 from the cell surface and induce release of exosomal ADAM17, which can then distribute and contribute to substrate shedding on more distant cells.


Subject(s)
ADAM17 Protein/metabolism , Exosomes/enzymology , A549 Cells , ADAM10 Protein/metabolism , ADAM17 Protein/genetics , Amphiregulin/metabolism , Amyloid Precursor Protein Secretases/metabolism , Calcium Signaling , Carrier Proteins/metabolism , Endothelial Cells/enzymology , Enzyme Activation , Exosomes/drug effects , Exosomes/metabolism , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Lipopolysaccharides/pharmacology , Membrane Microdomains/enzymology , Membrane Proteins/metabolism , Monocytes/enzymology , Protein Transport , RNA Interference , Substrate Specificity , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Transforming Growth Factor alpha/metabolism
13.
Nat Commun ; 7: 11523, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27161080

ABSTRACT

ADAM17, a prominent member of the 'Disintegrin and Metalloproteinase' (ADAM) family, controls vital cellular functions through cleavage of transmembrane substrates. Here we present evidence that surface exposure of phosphatidylserine (PS) is pivotal for ADAM17 to exert sheddase activity. PS exposure is tightly coupled to substrate shedding provoked by diverse ADAM17 activators. PS dependency is demonstrated in the following: (a) in Raji cells undergoing apoptosis; (b) in mutant PSA-3 cells with manipulatable PS content; and (c) in Scott syndrome lymphocytes genetically defunct in their capacity to externalize PS in response to intracellular Ca(2+) elevation. Soluble phosphorylserine but not phosphorylcholine inhibits substrate cleavage. The isolated membrane proximal domain (MPD) of ADAM17 binds to PS but not to phosphatidylcholine liposomes. A cationic PS-binding motif is identified in this domain, replacement of which abrogates liposome-binding and renders the protease incapable of cleaving its substrates in cells. We speculate that surface-exposed PS directs the protease to its targets where it then executes its shedding function.


Subject(s)
ADAM17 Protein/metabolism , Phosphatidylserines/metabolism , ADAM17 Protein/chemistry , ADAM17 Protein/deficiency , ADAM17 Protein/genetics , Amino Acid Sequence , Animals , Apoptosis/physiology , Blood Coagulation Disorders/blood , Blood Coagulation Disorders/genetics , Cell Line , Enzyme Activation , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Melitten/pharmacology , Mice , Mice, Knockout , Models, Biological , Protein Domains , Substrate Specificity
14.
Biochemistry ; 54(38): 5791-801, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26348730

ABSTRACT

A wide variety of biological processes including differentiation, regeneration, and cancer progression are regulated by shedding of membrane-anchored proteins. One of the major sheddases is A Disintegrin And Metalloprotease-17 (ADAM17) whose extracellular region consists of a pro-, a catalytic, a disintegrin-, and a membrane-proximal domain (MPD) as well as a short juxtamembrane segment of 17 amino acid residues that has been named "Conserved ADAM-seventeeN Dynamic Interaction Sequence" (CANDIS). This segment is involved in substrate recognition. Key mediators of inflammation including interleukin-6 receptor (IL-6R) and tumor necrosis factor (TNF-α) are substrates of ADAM17. The shedding activity of ADAM17 is regulated by the conformation of the membrane-proximal domain preceding the CANDIS segment. Here, we show that CANDIS, besides being involved in substrate recognition, is able to interact with lipid bilayers in vitro and that this property could be involved in regulating ADAM17 shedding activity.


Subject(s)
ADAM Proteins/metabolism , Cell Membrane/metabolism , Lipid Bilayers/metabolism , ADAM Proteins/analysis , ADAM Proteins/genetics , ADAM17 Protein , Amino Acid Sequence , Animals , Cell Line , Hep G2 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Mice , Molecular Sequence Data , Mutation , Protein Interaction Domains and Motifs , Substrate Specificity
15.
RNA Biol ; 12(9): 1043-53, 2015.
Article in English | MEDLINE | ID: mdl-26383776

ABSTRACT

Aptamers are an emerging class of highly specific targeting ligands. They can be selected in vitro for a large variety of targets, ranging from small molecules to whole cells. Most aptamers selected are nucleic acid-based, allowing chemical synthesis and easy modification. Although their properties make them interesting drug candidates for a broad spectrum of applications and an interesting alternative to antibodies or fusion proteins, they are not yet broadly used. One major drawback of aptamers is their susceptibility to abundant serum nucleases, resulting in their fast degradation in biological fluids. Using modified nucleic acids has become a common strategy to overcome these disadvantages, greatly increasing their half-life under cell culture conditions or even in vivo. Whereas pre-selective modifications of the initial library for aptamer selection are relatively easy to obtain, post-selective modifications of already selected aptamers are still generally very labor-intensive and often compromise the aptamers ability to bind its target molecule. Here we report the selection, characterization and post-selective modification of a 34 nucleotide (nt) RNA aptamer for a non-dominant, novel target site (domain 3) of the interleukin-6 receptor (IL-6R). We performed structural analyses and investigated the affinity of the aptamer to the membrane-bound and soluble forms (sIL-6R) of the IL-6R. Further, we performed structural analyses of the aptamer in solution using small-angle X-ray scattering and determined its overall shape and oligomeric state. Post-selective exchange of all pyrimidines against their 2'-fluoro analogs increased the aptamers stability significantly without compromising its affinity for the target protein. The resulting modified aptamer could be shortened to its minimal binding motif without loss of affinity.


Subject(s)
Aptamers, Nucleotide/metabolism , Receptors, Interleukin-6/metabolism , Animals , Aptamers, Nucleotide/chemistry , Binding Sites , Cell Line , Humans , Interleukin-6/metabolism , Mice , Models, Molecular , Molecular Conformation , Nucleic Acid Conformation , Nucleotide Motifs , Protein Binding , Protein Interaction Domains and Motifs , Receptors, Interleukin-6/chemistry , SELEX Aptamer Technique
16.
Biochem J ; 468(3): 507-18, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25846075

ABSTRACT

To avoid malformation and disease, tissue development and homoeostasis are co-ordinated precisely in time and space. Secreted Frizzled-related protein 3 (sFRP3), encoded by the Frizzled-related protein gene (FRZB), acts as an antagonist of Wnt signalling in bone development by delaying the maturation of proliferative chondrocytes into hypertrophic chondrocytes. A disintegrin and metalloprotease 17 (ADAM17) is a transmembrane protease that is essential for developmental processes and promotes cartilage maturation into bone. sFRP3 is chondroprotective and is expressed in chondrocytes of healthy articular cartilage. Upon damage to cartilage, sFRP3 is down-regulated. Rare variants of sFRP3 are associated with osteoarthritis. The present study demonstrates a novel function of sFRP3 in suppression of the enzymatic activity of ADAM17 which results in the inhibition of ADAM17-meditated interleukin-6 receptor (IL-6R) shedding. By contrast, the rare double variant of sFRP3 failed to suppress ADAM17. The shed soluble IL-6R (sIL-6R) is linked to inflammation, cartilage degeneration and osteolysis. Accordingly, enhanced activity of ADAM17 in cartilage, caused by the expression of the rare double sFRP3 variant, provides an explanation for the genetic effect of sFRP3 variants in joint disease. The finding that sFRP3 interacts with the ADAM17 substrate IL-6R also suggests a new regulatory mechanism by which the substrate is protected against shedding.


Subject(s)
ADAM Proteins/metabolism , Cell Membrane/metabolism , Chondrocytes/metabolism , Osteoarthritis, Hip/metabolism , Proteins/metabolism , Receptors, Interleukin-6/metabolism , Up-Regulation , ADAM Proteins/chemistry , ADAM Proteins/genetics , ADAM17 Protein , Amino Acid Substitution , Cell Line, Tumor , Down-Regulation , Genetic Predisposition to Disease , HEK293 Cells , Humans , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Osteoarthritis, Hip/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Proteins/chemistry , Proteins/genetics , Receptors, Interleukin-6/chemistry , Receptors, Interleukin-6/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
17.
J Biol Chem ; 289(23): 16336-48, 2014 Jun 06.
Article in English | MEDLINE | ID: mdl-24790088

ABSTRACT

A disintegrin and metalloprotease 17 (ADAM17) is a major sheddase involved in the regulation of a wide range of biological processes. Key substrates of ADAM17 are the IL-6 receptor (IL-6R) and TNF-α. The extracellular region of ADAM17 consists of a prodomain, a catalytic domain, a disintegrin domain, and a membrane-proximal domain as well as a small stalk region. This study demonstrates that this juxtamembrane segment is highly conserved, α-helical, and involved in IL-6R binding. This process is regulated by the structure of the preceding membrane-proximal domain, which acts as molecular switch of ADAM17 activity operated by a protein-disulfide isomerase. Hence, we have termed the conserved stalk region "Conserved ADAM seventeen dynamic interaction sequence" (CANDIS). Finally, we identified the region in IL-6R that binds to CANDIS. In contrast to the type I transmembrane proteins, the IL-6R, and IL-1RII, CANDIS does not bind the type II transmembrane protein TNF-α, demonstrating fundamental differences in the respective shedding by ADAM17.


Subject(s)
ADAM Proteins/metabolism , Receptors, Interleukin-6/metabolism , ADAM Proteins/chemistry , ADAM17 Protein , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Circular Dichroism , Conserved Sequence , DNA Primers , HEK293 Cells , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Homology, Amino Acid
18.
Neurobiol Aging ; 35(8): 1913-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24629672

ABSTRACT

Pathologic aggregation of α-synuclein is a central process in the pathogenesis of Parkinson's disease. The α-synuclein gene (SNCA) encodes at least 4 different α-synuclein isoforms through alternative splicing (SNCA140, SNCA126, SNCA112, SNCA98). Differential expression of α-synuclein isoforms has been shown in Lewy body diseases. In contrast to the canonical α-synuclein isoform of 140 amino acid residues (SNCA140), which has been investigated in detail, little is known about the properties of the 3 alternative isoforms. We have investigated the aggregation properties of all 4 isoforms in cultured cells and analyzed fibril-formation of 3 isoforms (SNCA140, SNCA126, and SNCA98) in vitro by electron microscopy. Each of the 3 alternative isoforms aggregates significantly less than the canonical isoform SNCA140. Electron microscopy showed that SNCA140 formed the well-known relatively straight fibrils while SNCA126 formed shorter fibrils, which were arranged in parallel fibril bundles and SNCA98 formed annular structures. Expression analysis of α-synuclein isoforms in different human brain regions demonstrated low expression levels of the alternative isoforms in comparison to the canonical SNCA140 isoform. These findings demonstrate that α-synuclein isoforms differ qualitatively and quantitatively in their aggregation properties. The biological consequences of these findings remain to be explored in vitro and in vivo.


Subject(s)
Parkinson Disease/genetics , Protein Aggregates , Protein Aggregation, Pathological/metabolism , alpha-Synuclein/metabolism , Amino Acid Sequence , Brain/metabolism , HEK293 Cells , Humans , Molecular Sequence Data , Protein Isoforms/metabolism , alpha-Synuclein/chemistry
19.
RNA Biol ; 11(1): 57-65, 2014.
Article in English | MEDLINE | ID: mdl-24440854

ABSTRACT

Interleukin-6 (IL-6) is a multifunctional cytokine that is involved in the progression of various inflammatory diseases, such as rheumatoid arthritis and certain cancers; for example, multiple myeloma or hepatocellular carcinoma. To interfere with IL-6-dependent diseases, targeting IL-6 receptor (IL-6R)-presenting tumor cells using aptamers might be a valuable strategy to broaden established IL-6- or IL-6R-directed treatment regimens. Recently, we reported on the in vitro selection of RNA aptamers binding to the human IL-6 receptor (IL-6R) with nanomolar affinity. One aptamer, namely AIR-3A, was 19 nt in size and able to deliver bulky cargos into IL-6R-presenting cells. As AIR-3A is a natural RNA molecule, its use for in vivo applications might be limited due to its susceptibility to ubiquitous ribonucleases. Aiming at more robust RNA aptamers targeting IL-6R, we now report on the generation of stabilized RNA aptamers for potential in vivo applications. The new 2'-F-modified RNA aptamers bind to IL-6R via its extracellular portion with low nanomolar affinity comparable to the previously identified unmodified counterpart. Aptamers do not interfere with the IL-6 receptor complex formation. The work described here represents one further step to potentially apply stabilized IL-6R-binding RNA aptamers in IL-6R-connected diseases, like multiple myeloma and hepatocellular carcinoma.


Subject(s)
Aptamers, Nucleotide/metabolism , RNA/chemistry , RNA/genetics , Receptors, Interleukin-6/metabolism , Aptamers, Nucleotide/chemistry , Fluoresceins , Humans , Neoplasms/genetics , Neoplasms/metabolism , Protein Binding , RNA Stability
20.
Biochem Biophys Res Commun ; 436(1): 66-72, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23707937

ABSTRACT

The unpaired-like protein 3 (Upd3) is one of the three cytokines of Drosophila melanogaster supposed to activate the JAK/STAT signaling pathway (Janus tyrosine kinases/signal transducer and activator of transcription). This activation occurs via the type-I cytokine receptor domeless, an orthologue of gp130, the common signal transducer of all four-helix bundle interleukin-6 (IL-6) type cytokines. Both receptors are known to exist as preformed dimers in the plasma membrane and initiate the acute-phase response. These facts indicate an evolutionary relation between vertebrate IL-6 and the Drosophila protein Upd3. Here we presented data which strengthen this notion. Upd3 was recombinantly expressed, a renaturation and purification protocol was established which allows to obtain high amounts of biological active protein. This protein is, like human IL-6, a monomeric-α helical cytokine, implicating that Upd3 is an "ancestor" of the four-helix bundle cytokines.


Subject(s)
Cytokines/chemistry , Drosophila Proteins/chemistry , Drosophila melanogaster/chemistry , Amino Acid Sequence , Animals , Dimerization , Disulfides/chemistry , Escherichia coli/metabolism , Humans , Interleukin-6/metabolism , Molecular Sequence Data , Protein Sorting Signals , Protein Structure, Secondary , Recombinant Proteins/chemistry , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...