Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Oncol Rep ; 49(2)2023 02.
Article in English | MEDLINE | ID: mdl-36524367

ABSTRACT

Carbonic anhydrase IX (CA IX) is a transmembrane enzyme participating in adaptive responses of tumors to hypoxia and acidosis. CA IX regulates pH, facilitates metabolic reprogramming, and supports migration, invasion and metastasis of cancer cells. Extracellular domain (ECD) of CA IX can be shed to medium and body fluids by a disintegrin and metalloproteinase (ADAM) 17. Here we show for the first time that CA IX ECD shedding can be also executed by ADAM10, a close relative of ADAM17, via an overlapping cleavage site in the stalk region of CA IX connecting its exofacial catalytic site with the transmembrane region. This finding is supported by biochemical evidence using recombinant human ADAM10 protein, colocalization of ADAM10 with CA IX, ectopic expression of a dominant­negative mutant of ADAM10 and RNA interference­mediated suppression of ADAM10. Induction of the CA IX ECD cleavage with ADAM17 and/or ADAM10 activators revealed their additive effect. Similarly, additive effect was observed with an ADAM17­inhibiting antibody and an ADAM10­preferential inhibitor GI254023X. These data indicated that ADAM10 is a CA IX sheddase acting on CA IX non­redundantly to ADAM17.


Subject(s)
ADAM Proteins , Carbonic Anhydrase IX , Humans , ADAM Proteins/chemistry , ADAM Proteins/metabolism , ADAM10 Protein/chemistry , ADAM10 Protein/metabolism , ADAM17 Protein/chemistry , ADAM17 Protein/metabolism , Carbonic Anhydrase IX/chemistry , Carbonic Anhydrase IX/metabolism , Membrane Proteins/metabolism , Neoplasms/metabolism
2.
Biochem Biophys Res Commun ; 593: 52-56, 2022 02 19.
Article in English | MEDLINE | ID: mdl-35063769

ABSTRACT

COVID-19, the respiratory infection caused by the novel coronavirus SARS-CoV-2, presents a clinical picture consistent with the dysregulation of many of the pathways mediated by the metalloprotease ADAM17. ADAM17 is a sheddase that plays a key role in the modulation of ACE2, the receptor which also functions as the point of attachment leading to cell entry by the virus. This work investigates the possibility that ADAM17 dysregulation and attachment of the SARS-CoV-2 virion to the ACE2 receptor are linked events, with the latter causing the former. Tetraspanins, the transmembrane proteins that function as scaffolds for the construction of viral entry platforms, are mooted as key components in this connection.


Subject(s)
ADAM17 Protein/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Receptors, Virus/metabolism , SARS-CoV-2/metabolism , Tetraspanin 29/metabolism , Virus Internalization , ADAM17 Protein/chemistry , Angiotensin-Converting Enzyme 2/chemistry , Binding Sites , COVID-19/epidemiology , COVID-19/transmission , COVID-19/virology , Humans , Models, Biological , Molecular Docking Simulation , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Pandemics , Protein Binding , Protein Domains , Receptors, Virus/chemistry , SARS-CoV-2/physiology , Tetraspanin 29/chemistry
3.
Biochem Biophys Res Commun ; 573: 158-163, 2021 10 08.
Article in English | MEDLINE | ID: mdl-34416436

ABSTRACT

The angiotensin Converting Enzyme 2 (ACE2) receptor is a key component of the renin-angiotensin-aldesterone system (RAAS) that mediates numerous effects in the cardiovascular system. It is also the cellular point of contact for the coronavirus spike protein. Cleavage of the receptor is both important to its physiological function as well as being necessary for cell entry by the virus. Shedding of ACE2 by the metalloprotease ADAM17 releases a catalytically active soluble form of ACE2, but cleavage by the serine protease TMPRSS2 is necessary for virion internalization. Complicating the issue is the observation that circulating ACE2 can also bind to the virus effectively blocking attachment to the membrane-bound receptor. This work investigates the possibility that the inflammatory response to coronavirus infection can abrogate shedding by ADAM17, thereby favoring cleavage by TMPRSS2 and thus cell entry by the virion.


Subject(s)
ADAM17 Protein/chemistry , ADAM17 Protein/metabolism , Angiotensin-Converting Enzyme 2/metabolism , HSP20 Heat-Shock Proteins/metabolism , Host-Pathogen Interactions/physiology , SARS-CoV-2/pathogenicity , Angiotensin-Converting Enzyme 2/chemistry , Binding Sites , HSP20 Heat-Shock Proteins/chemistry , Heat-Shock Response/physiology , Humans , Protein Domains , Protein Interaction Domains and Motifs , Serine Endopeptidases/metabolism , Virus Internalization
4.
Aging (Albany NY) ; 13(12): 16859-16872, 2021 06 28.
Article in English | MEDLINE | ID: mdl-34182543

ABSTRACT

The ADAMs (a disintegrin and metalloproteinase) are a family of cell surface proteins with crucial roles in the regulation of cell adhesion, cell proliferation to migration, proteolysis and cell signaling transduction pathways. Among these enzymes, the ADAM17 shows significant effects in the "ectodomain shedding" of its substrates such as cytokines (e.g., tumor necrosis factor α, TNFα), growth factors (e.g., epidermal growth factor, EGF), adhesion proteins (e.g., L-selectin), and their receptors (e.g., IL-6R and TNFα). Several studies focus on the underlying molecular mechanisms of ADAM17 in diseased conditions. Here, we took several different approaches to elucidate the function of ADAM17, the participation of ADAM17 in several human diseases, and the potential as targeted therapy reagents. As more and more studies verify the miRNA-mediated expression variation of ADAM17, the specific regulation network of miRNAs and ADAM17 was exploited in this review as well.


Subject(s)
ADAM17 Protein/metabolism , Disease , ADAM17 Protein/chemistry , ADAM17 Protein/genetics , Gene Regulatory Networks , Humans , Models, Biological , Molecular Targeted Therapy , Protein Processing, Post-Translational
5.
J Biol Chem ; 296: 100733, 2021.
Article in English | MEDLINE | ID: mdl-33957124

ABSTRACT

A disintegrin and metalloprotease 17 (ADAM17) is a cell-surface metalloprotease that serves as the principle sheddase for tumor necrosis factor α (TNFα), interleukin-6 receptor (IL-6R), and several ligands of the epidermal growth factor receptor (EGFR), regulating these crucial signaling pathways. ADAM17 activation requires its transmembrane domain, but not its cytoplasmic domain, and little is known about the role of this domain in vivo. To investigate, we used CRISPR-Cas9 to mutate the endogenous Adam17 locus in mice to produce a mutant ADAM17 lacking its cytoplasmic domain (Adam17Δcyto). Homozygous Adam17Δcyto animals were born at a Mendelian ratio and survived into adulthood with slightly wavy hair and curled whiskers, consistent with defects in ADAM17/EGFR signaling. At birth, Adam17Δcyto mice resembled Adam17-/- mice in that they had open eyes and enlarged semilunar heart valves, but they did not have bone growth plate defects. The deletion of the cytoplasmic domain resulted in strongly decreased ADAM17 protein levels in all tissues and cells examined, providing a likely cause for the hypomorphic phenotype. In functional assays, Adam17Δcyto mouse embryonic fibroblasts and bone-marrow-derived macrophages had strongly reduced ADAM17 activity, consistent with the reduced protein levels. Nevertheless, ADAM17Δcyto could be stimulated by PMA, a well-characterized posttranslational activator of ADAM17, corroborating that the cytoplasmic domain of endogenous ADAM17 is not required for its rapid response to PMA. Taken together, these results provide the first evidence that the cytoplasmic domain of ADAM17 plays a pivotal role in vivo in regulating ADAM17 levels and function.


Subject(s)
ADAM17 Protein/chemistry , ADAM17 Protein/metabolism , Cytoplasm/metabolism , ADAM17 Protein/genetics , Amino Acid Sequence , Animals , Base Sequence , CRISPR-Cas Systems , Female , Fibroblasts/metabolism , Male , Mice , Mice, Inbred C57BL , Phenotype , Protein Domains , Protein Stability , Sequence Deletion
6.
Cell Mol Life Sci ; 78(11): 5015-5040, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33950315

ABSTRACT

Membrane-tethered signalling proteins such as TNFα and many EGF receptor ligands undergo shedding by the metalloproteinase ADAM17 to get released. The pseudoproteases iRhom1 and iRhom2 are important for the transport, maturation and activity of ADAM17. Yet, the structural and functional requirements to promote the transport of the iRhom-ADAM17 complex have not yet been thoroughly investigated. Utilising in silico and in vitro methods, we here map the conserved iRhom homology domain (IRHD) and provide first insights into its structure and function. By focusing on iRhom2, we identified different structural and functional factors within the IRHD. We found that the structural integrity of the IRHD is a key factor for ADAM17 binding. In addition, we identified a highly conserved motif within an unstructured region of the IRHD, that, when mutated, restricts the transport of the iRhom-ADAM17 complex through the secretory pathway in in vitro, ex vivo and in vivo systems and also increases the half-life of iRhom2 and ADAM17. Furthermore, the disruption of this IRHD motif was also reflected by changes in the yet undescribed interaction profile of iRhom2 with proteins involved in intracellular vesicle transport. Overall, we provide the first insights into the forward trafficking of iRhoms which is critical for TNFα and EGF receptor signalling.


Subject(s)
ADAM17 Protein/metabolism , Carrier Proteins/metabolism , EGF Family of Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , ADAM17 Protein/chemistry , Amino Acid Motifs , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Line , Half-Life , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mutagenesis , Protein Binding , Protein Domains , Protein Transport , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction
7.
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
8.
FASEB J ; 34(4): 4956-4969, 2020 04.
Article in English | MEDLINE | ID: mdl-32103528

ABSTRACT

The metalloprotease ADAM17 (a disintegrin and metalloprotease 17) regulates EGF-receptor and TNFα signaling, thereby not only protecting the skin and intestinal barrier, but also contributing to autoimmunity. ADAM17 can be rapidly activated by many stimuli through its transmembrane domain (TMD), with the seven membrane-spanning inactive Rhomboids (iRhom) 1 and 2 implicated as candidate regulatory partners. However, several alternative models of ADAM17 regulation exist that do not involve the iRhoms, such as regulation through disulfide bond exchange or through interaction with charged phospholipids. Here, we report that a non-activatable mutant of ADAM17 with the TMD of betacellulin (BTC) can be rescued by restoring residues from the ADAM17 TMD, but only in Adam17-/- cells, which contain iRhoms, not in iRhom1/2-/- cells. We also provide the first evidence that the extracellular juxtamembrane domains (JMDs) of ADAM17 and iRhom2 regulate the stimulation and substrate selectivity of ADAM17. Interestingly, a point mutation in the ADAM17 JMD identified in a patient with Tetralogy of Fallot, a serious heart valve defect, affects the substrate selectivity of ADAM17 toward Heparin-binding epidermal growth factor like growth factor (HB-EGF), a crucial regulator of heart valve development in mice. These findings provide new insights into the regulation of ADAM17 through an essential interaction with the TMD1 and JMD1 of iRhom2.


Subject(s)
ADAM17 Protein/metabolism , Carrier Proteins/metabolism , Mutation , ADAM17 Protein/chemistry , ADAM17 Protein/genetics , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Protein Domains , Substrate Specificity , Tetralogy of Fallot/genetics
9.
Cancer Lett ; 467: 50-57, 2019 12 28.
Article in English | MEDLINE | ID: mdl-31593799

ABSTRACT

ADAM proteases are multi domain transmembrane metalloproteases that cleave a range of cell surface proteins and activate signaling pathways implicated in tumor progression, including those mediated by Notch, EFGR, and the Eph receptors. Consequently, they have emerged as key therapeutic targets in the efforts to inhibit tumor initiation and progression. To that end, two main approaches have been taken to develop ADAM antagonists: (i) small molecule inhibitors, and (ii) monoclonal antibodies. In this mini-review we describe the distinct features of ADAM proteases, particularly of ADAM10 and ADAM17, their domain organization, conformational rearrangements, regulation, as well as their emerging importance as therapeutic targets in cancer. Further, we highlight an anti-ADAM10 monoclonal antibody that we have recently developed, which has shown significant promise in inhibiting Notch signaling and deterring growth of solid tumors in pre-clinical settings.


Subject(s)
ADAM Proteins/chemistry , ADAM Proteins/metabolism , Neoplasms/metabolism , ADAM Proteins/antagonists & inhibitors , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/chemistry , ADAM10 Protein/metabolism , ADAM17 Protein/chemistry , ADAM17 Protein/metabolism , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Catalytic Domain , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/drug therapy , Protein Conformation , Protein Domains
10.
Life Sci Alliance ; 2(5)2019 10.
Article in English | MEDLINE | ID: mdl-31455669

ABSTRACT

ADAM17, prominent member of the "Disintegrin and Metalloproteinase" (ADAM) family, controls vital cellular functions through cleavage of transmembrane substrates. Several of these play central roles in oncogenesis and inflammation, yet despite its importance, the mechanism by which ADAM17 is activated is not fully understood. We recently presented evidence that surface exposure of phosphatidylserine (PS) is the penultimate event required for sheddase activation, which occurs upon binding of a membrane-proximal, cationic binding motif to the anionic phospholipid headgroup. Here, we show that mutagenesis of the 3 amino acids constituting the PS-binding motif leads to embryonic lethality in mice. Heterozygotes showed no abnormalities. Primary hepatocytes and fibroblasts were analysed and found to express the mutant protease on the cell surface. However, PMA-stimulated release of ADAM17 substrates was completely abolished. The results directly support the novel concept of transiently externalised PS as essential trigger of extracellular protease function in vivo.


Subject(s)
ADAM17 Protein/chemistry , ADAM17 Protein/genetics , Mutation , Phosphatidylserines/metabolism , ADAM17 Protein/metabolism , Animals , Binding Sites , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Genes, Lethal , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Mice , Primary Cell Culture , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology
11.
FASEB J ; 33(11): 11925-11940, 2019 11.
Article in English | MEDLINE | ID: mdl-31381863

ABSTRACT

Meprin ß is a membrane-bound metalloprotease involved in extracellular matrix assembly and inflammatory processes in health and disease. A disintegrin and metalloproteinase (ADAM)10 and ADAM17 are physiologic relevant sheddases of inactive promeprin ß, which influences its substrate repertoire and subsequent biologic functions. Proteomic analysis also revealed several ADAMs as putative meprin ß substrates. Here, we demonstrate specific N-terminal processing of ADAM9, 10, and 17 by meprin ß and identify cleavage sites within their prodomains. Because ADAM prodomains can act as specific inhibitors, we postulate a role for meprin ß in the regulation of ADAM activities. Indeed, prodomain cleavage by meprin ß caused increased ADAM protease activities, as observed by peptide-based cleavage assays and demonstrated by increased ectodomain shedding activity. Direct interaction of meprin ß and ADAM proteases could be shown by immunofluorescence microscopy and immunoprecipitation experiments. As demonstrated by a bacterial activator of meprin ß and additional measurement of TNF-α shedding on bone marrow-derived macrophages, meprin ß/ADAM protease interactions likely influence inflammatory conditions. Thus, we identified a novel proteolytic pathway of meprin ß with ADAM proteases to control protease activities at the cell surface as part of the protease web.-Wichert, R., Scharfenberg, F., Colmorgen, C., Koudelka, T., Schwarz, J., Wetzel, S., Potempa, B., Potempa, J., Bartsch, J. W., Sagi, I., Tholey, A., Saftig, P., Rose-John, S., Becker-Pauly, C. Meprin ß induces activities of A disintegrin and metalloproteinases 9, 10, and 17 by specific prodomain cleavage.


Subject(s)
ADAM Proteins/metabolism , ADAM10 Protein/metabolism , ADAM17 Protein/metabolism , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , ADAM Proteins/chemistry , ADAM Proteins/genetics , ADAM10 Protein/chemistry , ADAM10 Protein/genetics , ADAM17 Protein/chemistry , ADAM17 Protein/genetics , Animals , Cell Membrane/metabolism , Cells, Cultured , Extracellular Matrix/metabolism , HEK293 Cells , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Metalloendopeptidases/genetics , Mice, Inbred C57BL , Protein Domains , Proteolysis , Proteomics/methods , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
12.
Biochim Biophys Acta Mol Cell Res ; 1866(10): 1567-1583, 2019 10.
Article in English | MEDLINE | ID: mdl-31330158

ABSTRACT

Several membrane-bound proteins with a single transmembrane domain are subjected to limited proteolysis at the cell surface. This cleavage leads to the release of their biologically active ectodomains, which can trigger different signalling pathways. In many cases, this ectodomain shedding is mediated by members of the family of a disintegrins and metalloproteinases (ADAMs). ADAM17 in particular is responsible for the cleavage of several proinflammatory mediators, growth factors, receptors and adhesion molecules. Due to its direct involvement in the release of these signalling molecules, ADAM17 can be positively and negatively involved in various physiological processes as well as in inflammatory, fibrotic and malignant pathologies. This central role of ADAM17 in a variety of processes requires strict multi-level regulation, including phosphorylation, various conformational changes and endogenous inhibitors. Recent research has shown that an early, crucial control mechanism is interaction with certain adapter proteins identified as iRhom1 and iRhom2, which are pseudoproteases of the rhomboid superfamily. Thus, iRhoms have also a decisive influence on physiological and pathophysiological signalling processes regulated by ADAM17. Their characteristic gene expression profiles, the specific consequences of gene knockouts and finally the occurrence of disease-associated mutations suggest that iRhom1 and iRhom2 undergo different gene regulation in order to fulfil their function in different cell types and are therefore only partially redundant. Therefore, there is not only interest in ADAM17, but also in iRhoms as therapeutic targets. However, to exploit the therapeutic potential, the regulation of ADAM17 activity and in particular its interaction with iRhoms must be well understood.


Subject(s)
ADAM17 Protein/chemistry , ADAM17 Protein/metabolism , Membrane Proteins/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Gene Knockout Techniques , Genetic Predisposition to Disease/genetics , Humans , Inflammation , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Metalloproteases , Mice , Mutation , Phosphorylation , Protein Conformation , Signal Transduction , Transcriptome
13.
Int J Mol Sci ; 20(9)2019 May 04.
Article in English | MEDLINE | ID: mdl-31060243

ABSTRACT

Colorectal cancer is one of the most commonly diagnosed malignancies in the Western world and is associated with elevated expression and activity of epidermal growth factor receptors (EGF-R). The metalloproteinase ADAM17 is involved in EGF-R activation by processing EGF-R ligands from membrane-bound pro-ligands. Underlining the link between colon cancer and ADAM17, genetic intestinal cancer models in ADAM17-deficient mice show a reduced tumor burden. In this study, we characterize point mutations within the ADAM17 gene found in the tissue of colon cancer patients. In order to shed light on the role of ADAM17 in cancer development, as well as into the mechanisms that regulate maturation and cellular trafficking of ADAM17, we here perform overexpression studies of four ADAM17 variants located in the pro-, membrane-proximal- and cytoplasmic-domain of the ADAM17 protein in ADAM10/17-deficient HEK cells. Interestingly, we found a cancer-associated point mutation within the pro-domain of ADAM17 (R177C) to be most impaired in its proteolytic activity and trafficking to the cell membrane. By comparing this variant to an ADAM17 construct lacking the entire pro-domain, we discovered similar functional limitations and propose a crucial role of the pro-domain for ADAM17 maturation, cellular trafficking and thus proteolytic activity.


Subject(s)
ADAM17 Protein/genetics , ADAM17 Protein/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Mutation , ADAM17 Protein/chemistry , Animals , Biomarkers , Colonic Neoplasms/pathology , Disease Susceptibility , Humans , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Proteolysis , Structure-Activity Relationship
14.
Biochemistry ; 58(20): 2509-2518, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30946563

ABSTRACT

Regulated proteolysis of signaling proteins under mechanical tension enables cells to communicate with their environment in a variety of developmental and physiologic contexts. The role of force in inducing proteolytic sensitivity has been explored using magnetic tweezers at the single-molecule level with bead-tethered assays, but such efforts have been limited by challenges in ensuring that beads not be restrained by multiple tethers. Here, we describe a multiplexed assay for single-molecule proteolysis that overcomes the multiple-tether problem using a flow-extension strategy on a microscope equipped with magnetic tweezers. Particle tracking and computational sorting of flow-induced displacements allow assignment of tethered substrates to singly captured and multiply tethered bins, with the fraction of fully mobile, single-tether substrates depending inversely on the concentration of substrate loaded on the coverslip. Computational exclusion of multiple-tether beads enables robust assessment of on-target proteolysis by the highly specific tobacco etch virus protease and the more promiscuous metalloprotease ADAM17. This method should be generally applicable to a wide range of proteases and readily extensible to robust evaluation of proteolytic sensitivity as a function of applied magnetic force.


Subject(s)
ADAM17 Protein/chemistry , Endopeptidases/chemistry , Peptides/analysis , Proteolysis , Single Molecule Imaging/methods , DNA/chemistry , Humans , Magnetic Phenomena , Microfluidics/methods , Motion , Peptides/chemistry , Potyvirus/enzymology , Proof of Concept Study
15.
Analyst ; 144(5): 1825-1830, 2019 Feb 25.
Article in English | MEDLINE | ID: mdl-30675599

ABSTRACT

A disintegrin and metalloproteinase 17 (ADAM17) has become a novel biomarker and potential therapeutic target for the early detection and treatment of human cancers. In this work, by covalently attaching fluorescently labeled ADAM17 substrate peptide (Pep-FAM) molecules to carboxylated graphene oxide (cGO) and monitoring the cleavage of the peptide substrate by ADAM17, we developed a cGO-Pep-FAM fluorescence sensor for the rapid, sensitive and accurate detection of ADAM17. The sensor was highly sensitive with a detection limit of 17.5 picomolar. Furthermore, the sensor was selective: structure similar proteases such as ADAM9 and MMP-9 would not interfere with ADAM17 detection. In addition, simulated serum samples were successfully analyzed. Our developed cGO-Pep-FAM sensing strategy should find useful applications in disease diagnosis and drug screening.


Subject(s)
ADAM17 Protein/blood , Biosensing Techniques/methods , Enzyme Assays/methods , Graphite/chemistry , Oxides/chemistry , ADAM17 Protein/chemistry , Amino Acid Sequence , Fluoresceins/chemistry , Fluorescence , Fluorescent Dyes/chemistry , Humans , Limit of Detection , Peptides/chemistry , Proteolysis
16.
J Immunol ; 201(10): 3106-3118, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30355783

ABSTRACT

A disintegrin and metalloproteinase (ADAM) 17 has been implicated in many shedding processes. Major substrates of ADAM17 are TNF-α, IL-6R, and ligands of the epidermal growth factor receptor. The essential role of the protease is emphasized by the fact that ADAM17 deficiency is lethal in mice. To study ADAM17 function in vivo, we generated viable hypomorphic ADAM17 mice called ADAM17ex/ex mice. Recent studies indicated regulation of proteolytic ADAM17 activity by cellular processes such as cytoplasmic phosphorylation and removal of the prodomain by furin cleavage. Maturation and thus activation of ADAM17 is not fully understood. So far, studies of ADAM17 maturation have been mainly limited to mouse embryonic fibroblasts or transfected cell lines relying on nonphysiologic stimuli such as phorbol esters, thus making interpretation of the results difficult in a physiologic context. In this article, we present a robust cell system to study ADAM17 maturation and function in primary cells of the immune system. To this end, HoxB8 conditionally immortalized macrophage precursor cell lines were derived from bone marrow of wild-type and hypomorphic ADAM17ex/ex mice, which are devoid of measurable ADAM17 activity. ADAM17 mutants were stably expressed in macrophage precursor cells, differentiated to macrophages under different growth factor conditions (M-CSF versus GM-CSF), and analyzed for cellular localization, proteolytic activity, and podosome disassembly. Our study reveals maturation and activity of ADAM17 in a more physiological-immune cell system. We show that this cell system can be further exploited for genetic modifications of ADAM17 and for studying its function in immune cells.


Subject(s)
ADAM17 Protein/chemistry , ADAM17 Protein/metabolism , Cell Culture Techniques/methods , Dendritic Cells/enzymology , Macrophages/enzymology , Animals , Cell Line , Homeodomain Proteins , Mice
17.
Arch Cardiovasc Dis ; 110(12): 700-711, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29097110

ABSTRACT

Tumour necrosis factor alpha converting enzyme (TACE/ADAM17) is a member of the A disintegrin and metalloproteinase (ADAM) family of ectodomain shedding proteinases. It regulates many inflammatory processes by cleaving several transmembrane proteins, including tumour necrosis factor alpha (TNFα) and its receptors tumour necrosis factor alpha receptor 1 and tumour necrosis factor alpha receptor 2. There is evidence that TACE is involved in several inflammatory diseases, such as ischaemia, heart failure, arthritis, atherosclerosis, diabetes and cancer as well as neurological and immune diseases. This review summarizes the latest discoveries regarding the mechanism of action and regulation of TACE. It also focuses on the role of TACE in atherosclerosis and coronary artery disease (CAD), highlighting clinical studies that have investigated its expression and protein activity. The multitude of substrates cleaved by TACE make this enzyme an attractive target for therapy and a candidate for biomarker research and development in CAD.


Subject(s)
ADAM17 Protein/metabolism , Cardiovascular System/enzymology , Coronary Artery Disease/enzymology , ADAM17 Protein/chemistry , Animals , Biomarkers/metabolism , Cardiovascular System/physiopathology , Coronary Artery Disease/diagnosis , Coronary Artery Disease/physiopathology , Enzyme Activation , Humans , Prognosis , Protein Conformation , Signal Transduction , Structure-Activity Relationship , Substrate Specificity
18.
Int J Biochem Cell Biol ; 85: 6-14, 2017 04.
Article in English | MEDLINE | ID: mdl-28143719

ABSTRACT

Proteolytic cleavage of the membrane-bound Interleukin-6 receptor (IL-6R) by the metalloprotease ADAM17 releases an agonistic soluble form of the IL-6R (sIL-6R), which is responsible for the pro-inflammatory trans-signaling branch of the cytokine's activities. This proteolytic step, which is also called ectodomain shedding, is critically regulated by the cleavage site within the IL-6R stalk, because mutations or small deletions within this region are known to render the IL-6R irresponsive towards proteolysis. In the present study, we employed cleavage site profiling data of ADAM17 to generate an IL-6R with increased cleavage susceptibility. Using site-directed mutagenesis, we showed that the non-prime sites P3 and P2 and the prime site P1' were critical for this increase in proteolysis, whereas other positions within the cleavage site were of minor importance. Insertion of this optimized cleavage site into the stalk of the Interleukin-11 receptor (IL-11R) was not sufficient to enable ADAM17-mediated proteolysis, but transfer of different parts of the IL-6R stalk enabled shedding by ADAM17. These findings shed light on the cleavage site specificities of ADAM17 using a native substrate and reveal further differences in the proteolysis of IL-6R and IL-11R.


Subject(s)
Interleukin-6/metabolism , Receptors, Interleukin-11/metabolism , Receptors, Interleukin-6/metabolism , ADAM17 Protein/chemistry , ADAM17 Protein/genetics , Binding Sites , Blotting, Western , Catalytic Domain/genetics , Genetic Variation , HEK293 Cells , Humans , Protein Processing, Post-Translational , Proteolysis , Receptors, Interleukin-11/genetics , Receptors, Interleukin-6/genetics
19.
J Cell Sci ; 130(5): 868-878, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28104813

ABSTRACT

A disintegrin and metalloproteinase 17 (ADAM17) controls the release of the pro-inflammatory cytokine tumor necrosis factor α (TNFα, also known as TNF) and is crucial for protecting the skin and intestinal barrier by proteolytic activation of epidermal growth factor receptor (EGFR) ligands. The seven-membrane-spanning protein called inactive rhomboid 2 (Rhbdf2; also known as iRhom2) is required for ADAM17-dependent TNFα shedding and crosstalk with the EGFR, and a point mutation (known as sinecure, sin) in the first transmembrane domain (TMD) of Rhbdf2 (Rhbdf2sin) blocks TNFα shedding, yet little is known about the underlying mechanism. Here, we used a structure-function analysis informed by structural modeling to evaluate the interaction between the TMD of ADAM17 and the first TMD of Rhbdf2, and the role of this interaction in Rhbdf2-ADAM17-dependent shedding. Moreover, we show that double mutant mice that are homozygous for Rhbdf2sin/sin and lack Rhbdf1 closely resemble Rhbdf1/2-/- double knockout mice, highlighting the severe functional impact of the Rhbdf2sin/sin mutation on ADAM17 during mouse development. Taken together, these findings provide new mechanistic and conceptual insights into the critical role of the TMDs of ADAM17 and Rhbdf2 in the regulation of the ADAM17 and EGFR, and ADAM17 and TNFα signaling pathways.


Subject(s)
ADAM17 Protein/chemistry , ADAM17 Protein/metabolism , Carrier Proteins/metabolism , Models, Molecular , Proteolysis , Amino Acid Sequence , Amino Acids/metabolism , Animals , Bone Marrow Cells/cytology , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Membrane/metabolism , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Growth Plate/metabolism , Heart Valves/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Docking Simulation , Molecular Dynamics Simulation , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation/genetics , Protein Binding , Structure-Activity Relationship
20.
Sci Rep ; 6: 35598, 2016 12 16.
Article in English | MEDLINE | ID: mdl-27982031

ABSTRACT

Dysregulated activity of A Disintegrin And Metalloproteinase 17 (ADAM17)/TNFα Converting Enzyme (TACE) is associated with inflammatory disorders and cancer progression by releasing regulatory membrane-tethered proteins like TNFα, IL6R and EGFR ligands. Although specific inhibition of TACE is thought to be a viable strategy for inflammatory disorders and for malignancies treatment, the generation of effective inhibitors in vivo has been proven to be challenging. Here we report on the development of a protein inhibitor that leverages the endogenous modulator of TACE. We have generated a stable form of the auto-inhibitory TACE prodomain (TPD), which specifically inhibits in vitro and cell-surface TACE, but not the related ADAM10, and effectively modulated TNFα secretion in cells. TPD significantly attenuated TACE-mediated disease models of sepsis, rheumatoid arthritis (RA) and inflammatory bowel disease (IBD), and reduced TNFα in synovial fluids from RA patients. Our results demonstrate that intervening with endogenous ADAM sheddase modulatory mechanisms holds potential as a general strategy for the design of ADAM inhibitors.


Subject(s)
ADAM17 Protein/chemistry , Arthritis/drug therapy , Colitis/drug therapy , Enzyme Inhibitors/administration & dosage , Shock, Septic/drug therapy , ADAM10 Protein/metabolism , ADAM17 Protein/antagonists & inhibitors , Animals , Arthritis/chemically induced , Arthritis/metabolism , Cells, Cultured , Colitis/chemically induced , Colitis/metabolism , Collagen/adverse effects , Disease Models, Animal , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Lipopolysaccharides/adverse effects , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Protein Domains , Shock, Septic/chemically induced , Shock, Septic/metabolism , Trinitrobenzenesulfonic Acid/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...