Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Biol Chem ; 402(3): 253-269, 2021 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-33108336

RESUMO

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.


Assuntos
Proteínas de Membrana/metabolismo , Compostos de Sulfidrila/metabolismo , Animais , Humanos , Oxirredução , Transdução de Sinais
2.
Biochem Biophys Res Commun ; 526(2): 355-360, 2020 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-32222277

RESUMO

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.


Assuntos
Proteína ADAM17/metabolismo , Receptores de Interleucina-6/metabolismo , Proteína ADAM17/química , Sequência de Aminoácidos , Domínio Catalítico , Células HEK293 , Humanos , Mutação , Fosfatidilserinas/metabolismo , Proteólise , Receptores de Interleucina-6/química , Receptores de Interleucina-6/genética
3.
PLoS Biol ; 15(1): e2000080, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28060820

RESUMO

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.


Assuntos
Proteólise , Receptores de Interleucina-6/metabolismo , Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Processamento Alternativo/genética , Sequência de Aminoácidos , Secretases da Proteína Precursora do Amiloide/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Glicosilação , Humanos , Espaço Intracelular/metabolismo , Espectrometria de Massas , Proteínas de Membrana/metabolismo , Mutação/genética , Polissacarídeos/metabolismo , Prolina/metabolismo , Domínios Proteicos , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Interleucina-6/sangue , Receptores de Interleucina-6/química , Receptores de Interleucina-6/genética , Transdução de Sinais , Solubilidade , Valina/metabolismo
4.
Cell Physiol Biochem ; 52(4): 850-868, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30958660

RESUMO

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.


Assuntos
Proteína ADAM17/metabolismo , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Receptores de Peptídeos/metabolismo , Proteína ADAM17/genética , Membrana Celular/genética , Retículo Endoplasmático/genética , Células HEK293 , Humanos , Isomerases de Dissulfetos de Proteínas/genética , Transporte Proteico/fisiologia , Receptores de Peptídeos/genética
5.
Biochim Biophys Acta Mol Cell Res ; 1864(11 Pt B): 2088-2095, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28571693

RESUMO

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.


Assuntos
Proteína ADAM17/genética , Membrana Celular/genética , Proteínas de Membrana/genética , Proteólise , Membrana Celular/metabolismo , Regulação da Expressão Gênica/genética , Humanos , Proteínas de Membrana/metabolismo
6.
Biochim Biophys Acta ; 1863(11): 2795-2808, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27599715

RESUMO

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.


Assuntos
Proteína ADAM17/metabolismo , Exossomos/enzimologia , Células A549 , Proteína ADAM10/metabolismo , Proteína ADAM17/genética , Anfirregulina/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Sinalização do Cálcio , Proteínas de Transporte/metabolismo , Células Endoteliais/enzimologia , Ativação Enzimática , Exossomos/efeitos dos fármacos , Exossomos/metabolismo , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Lipopolissacarídeos/farmacologia , Microdomínios da Membrana/enzimologia , Proteínas de Membrana/metabolismo , Monócitos/enzimologia , Transporte Proteico , Interferência de RNA , Especificidade por Substrato , Acetato de Tetradecanoilforbol/farmacologia , Transfecção , Fator de Crescimento Transformador alfa/metabolismo
7.
Biochem J ; 468(3): 507-18, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25846075

RESUMO

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.


Assuntos
Proteínas ADAM/metabolismo , Membrana Celular/metabolismo , Condrócitos/metabolismo , Osteoartrite do Quadril/metabolismo , Proteínas/metabolismo , Receptores de Interleucina-6/metabolismo , Regulação para Cima , Proteínas ADAM/química , Proteínas ADAM/genética , Proteína ADAM17 , Substituição de Aminoácidos , Linhagem Celular Tumoral , Regulação para Baixo , Predisposição Genética para Doença , Células HEK293 , Humanos , Mutagênese Sítio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Osteoartrite do Quadril/genética , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Domínios e Motivos de Interação entre Proteínas , Proteínas/química , Proteínas/genética , Receptores de Interleucina-6/química , Receptores de Interleucina-6/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
8.
Biochemistry ; 54(38): 5791-801, 2015 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-26348730

RESUMO

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.


Assuntos
Proteínas ADAM/metabolismo , Membrana Celular/metabolismo , Bicamadas Lipídicas/metabolismo , Proteínas ADAM/análise , Proteínas ADAM/genética , Proteína ADAM17 , Sequência de Aminoácidos , Animais , Linhagem Celular , Células Hep G2 , Humanos , Interações Hidrofóbicas e Hidrofílicas , Camundongos , Dados de Sequência Molecular , Mutação , Domínios e Motivos de Interação entre Proteínas , Especificidade por Substrato
9.
J Biol Chem ; 289(23): 16336-48, 2014 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-24790088

RESUMO

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.


Assuntos
Proteínas ADAM/metabolismo , Receptores de Interleucina-6/metabolismo , Proteínas ADAM/química , Proteína ADAM17 , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Dicroísmo Circular , Sequência Conservada , Primers do DNA , Células HEK293 , Humanos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Homologia de Sequência de Aminoácidos
10.
Nat Chem Biol ; 9(1): 37-42, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23143413

RESUMO

Human pathogens often produce soluble protein toxins that generate pores inside membranes, resulting in the death of target cells and tissue damage. In pathogenic amoebae, this has been exemplified with amoebapores of the enteric protozoan parasite Entamoeba histolytica. Here we characterize acanthaporin, to our knowledge the first pore-forming toxin to be described from acanthamoebae, which are free-living, bacteria-feeding, unicellular organisms that are opportunistic pathogens of increasing importance and cause severe and often fatal diseases. We isolated acanthaporin from extracts of virulent Acanthamoeba culbertsoni by tracking its pore-forming activity, molecularly cloned the gene of its precursor and recombinantly expressed the mature protein in bacteria. Acanthaporin was cytotoxic for human neuronal cells and exerted antimicrobial activity against a variety of bacterial strains by permeabilizing their membranes. The tertiary structures of acanthaporin's active monomeric form and inactive dimeric form, both solved by NMR spectroscopy, revealed a currently unknown protein fold and a pH-dependent trigger mechanism of activation.


Assuntos
Acanthamoeba/química , Proteínas de Protozoários/química , Proteínas de Protozoários/fisiologia , Acanthamoeba/patogenicidade , Sequência de Aminoácidos , Dimerização , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Virulência
11.
RNA Biol ; 12(9): 1043-53, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26383776

RESUMO

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.


Assuntos
Aptâmeros de Nucleotídeos/metabolismo , Receptores de Interleucina-6/metabolismo , Animais , Aptâmeros de Nucleotídeos/química , Sítios de Ligação , Linhagem Celular , Humanos , Interleucina-6/metabolismo , Camundongos , Modelos Moleculares , Conformação Molecular , Conformação de Ácido Nucleico , Motivos de Nucleotídeos , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Receptores de Interleucina-6/química , Técnica de Seleção de Aptâmeros
12.
J Biol Chem ; 288(6): 4346-54, 2013 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-23209286

RESUMO

IL-27 consists of the cytokine subunit p28 and the non-signaling α-receptor EBI3. p28 was shown to additionally act via the non-signaling membrane-bound IL-6 α-receptor (IL-6R) as an agonistic cytokine but also as a gp130 ß-receptor antagonist, leading to inhibition of IL-6 signaling. Here, we developed a strategy for bacterial expression, purification, and refolding of murine p28. We show that p28 did not interfere with IL-6- or IL-27-induced signaling, indicating that p28 has no antagonistic properties. Moreover, we demonstrate that murine p28 acts as an agonistic cytokine via the murine and human IL-6R, indicating that p28 exhibits no species specificity. p28 was able to induce p28-trans-signaling via the soluble IL-6R (sIL-6R), a characteristic property that was initially described for trans-signaling of IL-6 via the sIL-6R. Of notice, p28/sIL-6R trans-signaling was inhibited by the IL-6 trans-signaling antagonist, soluble gp130. At higher concentrations, p28 but not IL-6 was able to induce signaling even in the absence of IL-6R or EBI3. Although IL-27 signals via a heterodimer of the ß-receptor chains gp130 and Wsx-1, p28/IL-6R specifically recruits two gp130 ß-receptor chains for signal transduction. The binding of p28 to a gp130/Wsx-1 heterodimer or a gp130 homodimer is highly selective and controlled by a novel molecular switch induced by EBI3 or IL-6R, respectively.


Assuntos
Receptor gp130 de Citocina/metabolismo , Interleucinas/metabolismo , Multimerização Proteica/fisiologia , Transdução de Sinais/fisiologia , Animais , Células COS , Chlorocebus aethiops , Receptor gp130 de Citocina/genética , Humanos , Interleucinas/genética , Camundongos , Células NIH 3T3 , Receptores de Citocinas/genética , Receptores de Citocinas/metabolismo , Receptores de Interleucina/genética , Receptores de Interleucina/metabolismo
13.
RNA Biol ; 11(1): 57-65, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24440854

RESUMO

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.


Assuntos
Aptâmeros de Nucleotídeos/metabolismo , RNA/química , RNA/genética , Receptores de Interleucina-6/metabolismo , Aptâmeros de Nucleotídeos/química , Fluoresceínas , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Ligação Proteica , Estabilidade de RNA
14.
J Am Chem Soc ; 135(15): 5776-81, 2013 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-23521534

RESUMO

A disintegrin and metalloprotease-17 (ADAM17) is a major sheddase responsible for the regulation of a wide range of biological processes, like cellular differentiation, regeneration, or cancer progression. Hitherto, the mechanism regulating the enzymatic activity of ADAM17 is poorly understood. Recently, protein-disulfide isomerase (PDI) was shown to interact with ADAM17 and to down-regulate its enzymatic activity. Here we demonstrate by NMR spectroscopy and tandem-mass spectrometry that PDI directly interacts with the membrane-proximal domain (MPD), a domain of ADAM17 involved in its dimerization and substrate recognition. PDI catalyzes an isomerization of disulfide bridges within the thioredoxin motif C600XXC603 of the MPD and results in a drastic structural change between an active open state and an inactive closed conformation. This conformational change of the MPD putatively acts as a molecular switch, facilitating a global reorientation of the extracellular domains in ADAM17 and regulating its shedding activity.


Assuntos
Proteínas ADAM/química , Proteínas ADAM/metabolismo , Membrana Celular/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Proteína ADAM17 , Cisteína/química , Humanos , Isomerismo , Modelos Moleculares , Estrutura Terciária de Proteína
15.
Biochem Biophys Res Commun ; 436(1): 66-72, 2013 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-23707937

RESUMO

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.


Assuntos
Citocinas/química , Proteínas de Drosophila/química , Drosophila melanogaster/química , Sequência de Aminoácidos , Animais , Dimerização , Dissulfetos/química , Escherichia coli/metabolismo , Humanos , Interleucina-6/metabolismo , Dados de Sequência Molecular , Sinais Direcionadores de Proteínas , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Transdução de Sinais
16.
Cancer Immunol Immunother ; 62(3): 411-21, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22940887

RESUMO

A disintegrin and metalloproteinase 17 (ADAM17) is significantly upregulated not only in malignant cells but also in the pro-inflammatory microenvironment of breast cancer. There, ADAM17 is critically involved in the processing of tumor-promoting proteins. Therefore, ADAM17 appears to be an attractive therapeutic target to address not only tumor cells but also the tumor-promoting environment. In a previous study, we generated a monoclonal anti-ADAM17 antibody (A300E). Although showing no complement-dependent cytotoxicity or antibody-dependent cellular cytotoxicity, the antibody was rapidly internalized by ADAM17-expressing cells and was able to transport a conjugated toxin into target cells. As a result, doxorubicin-coupled A300E or Pseudomonas exotoxin A-loaded A300E was able to kill ADAM17-expressing cells. This effect was strictly dependent on the presence of ADAM17 on the surface of target cells. As a proof of principle, both immunotoxins killed MDA-MB-231 breast cancer cells in an ADAM17-dependent manner. These data suggest that the use of anti-ADAM17 monoclonal antibodies as a carrier might be a promising new strategy for selective anti-cancer drug delivery.


Assuntos
Proteínas ADAM/imunologia , Antibióticos Antineoplásicos/administração & dosagem , Neoplasias da Mama/terapia , Doxorrubicina/administração & dosagem , Imunotoxinas/uso terapêutico , Proteínas ADAM/metabolismo , Proteína ADAM17 , Anticorpos Monoclonais/uso terapêutico , Citotoxicidade Celular Dependente de Anticorpos , Neoplasias da Mama/metabolismo , Membrana Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Proteínas do Sistema Complemento/imunologia , Feminino , Humanos , Terapia de Alvo Molecular
17.
RNA Biol ; 10(2): 216-27, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23235494

RESUMO

Aptamers are oligonucleotides that bind targets with high specificity and affinity. They have become important tools for biosensing, target detection, drug delivery and therapy. We selected the quadruplex-forming 16-mer DNA aptamer AID-1 [d(GGGT) 4] with affinity for the interleukin-6 receptor (IL-6R) and identified single nucleotide variants that showed no significant loss of binding ability. The RNA counterpart of AID-1 [r(GGGU) 4] also bound IL-6R as quadruplex structure. AID-1 is identical to the well-known HIV inhibitor T30923, which inhibits both HIV infection and HIV-1 integrase. We also demonstrated that IL-6R specific RNA aptamers not only bind HIV-1 integrase and inhibit its 3' processing activity in vitro, but also are capable of preventing HIV de novo infection with the same efficacy as the established inhibitor T30175. All these aptamer target interactions are highly dependent on formation of quadruplex structure.


Assuntos
Aptâmeros de Nucleotídeos/farmacologia , Inibidores de Integrase de HIV/farmacologia , HIV-1/efeitos dos fármacos , Receptores de Interleucina-6/metabolismo , Dicroísmo Circular , Avaliação Pré-Clínica de Medicamentos , Quadruplex G/efeitos dos fármacos , Proteína gp120 do Envelope de HIV/genética , Proteína gp120 do Envelope de HIV/metabolismo , Infecções por HIV/patologia , Infecções por HIV/virologia , Integrase de HIV/genética , Integrase de HIV/metabolismo , HIV-1/enzimologia , HIV-1/patogenicidade , Células HeLa , Humanos , Oligonucleotídeos/farmacologia , Ligação Viral/efeitos dos fármacos
18.
Biochem J ; 445(1): 135-44, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22509934

RESUMO

ADAM17 (A disintegrin and metalloproteinase 17) is a membrane-bound protease that cleaves various cell surface proteins, including cytokines and cytokine receptors. Recently it was shown that ADAM17 is highly expressed on the surface of many cancer cells, whereas normal cells express low levels of ADAM17, implying that ADAM17 is a potential immunotherapeutic target. We have generated a monoclonal antibody against human ADAM17, which recognized the membrane proximal cysteine-rich extension of the ADAM17 protein. Unlike normal cells, tumour cell lines, such as a prostate cancer cell line, pancreatic cancer cell lines, a breast cancer cell line and a non-small lung cancer cell line, expressed ADAM17 on the cell surface. Using the sequence of the antibody we generated an ADAM17-specific scFv (single-chain variable fragment) and fused this to a CD3-specific scFv to generate a bispecific T-cell engager antibody [A300E-BiTE (bispecific T-cell engager antibody)]. Specificity was demonstrated on cells in which ADAM17 was knocked down with a specific shRNA (short hairpin RNA). A300E-BiTE recognized ADAM17 and CD3 on the cell surface of tumour cells and T-cells respectively. In the presence of primary human peripheral blood mononuclear cells or human T-cells the addition of A300E-BiTE led to ADAM17-specific killing of prostate tumour cells indicating a novel strategy for the treatment of cancer.


Assuntos
Proteínas ADAM/metabolismo , Anticorpos Biespecíficos/farmacologia , Complexo CD3/metabolismo , Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia , Anticorpos de Cadeia Única/imunologia , Linfócitos T/metabolismo , Proteínas ADAM/imunologia , Proteína ADAM17 , Anticorpos Biespecíficos/imunologia , Apoptose , Western Blotting , Complexo CD3/imunologia , Proliferação de Células , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Imunoprecipitação , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Masculino , Neoplasias da Próstata/imunologia , RNA Mensageiro/genética , Linfócitos T/imunologia , Células Tumorais Cultivadas
19.
J Biol Chem ; 286(50): 42959-70, 2011 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-21990364

RESUMO

IL-6 trans-signaling via the soluble IL-6 receptor (sIL-6R) plays a critical role in chronic inflammation and cancer. Soluble gp130 (sgp130) specifically inhibits IL-6 trans-signaling but was described to not interfere with classic signaling via the membrane-bound IL-6R. Physiological and most pathophysiological conditions are characterized by a molar excess of serum sIL-6R over IL-6 characterized by free IL-6 and IL-6 found in IL-6·sIL-6R complexes allowing both classic and trans-signaling. Surprisingly, under these conditions, sgp130 was able to trap all free IL-6 molecules in IL-6·sIL-6R·sgp130 complexes, resulting in inhibition of classic signaling. Because a significant fraction of IL-6 molecules did not form complexes with sIL-6R, our results demonstrate that compared with the anti-IL-6R antibody tocilizumab or the anti-trans-signaling monoclonal antibody 25F10, much lower concentrations of the dimeric sgp130Fc were sufficient to block trans-signaling. In vivo, sgp130Fc blocked IL-6 signaling in the colon but not in liver and lung, indicating that the colon is a prominent target of IL-6 trans-signaling. Our results point to a so far unanticipated role of sgp130 in the blockade of classic signaling and indicate that in vivo only low therapeutic concentrations of sgp130Fc guarantee blockade of IL-6 trans-signaling without affecting IL-6 classic signaling.


Assuntos
Receptor gp130 de Citocina/metabolismo , Interleucina-6/metabolismo , Receptores de Interleucina-6/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados/farmacologia , Western Blotting , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Receptor gp130 de Citocina/antagonistas & inibidores , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Interleucina-6/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Interleucina-6/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos
20.
RNA Biol ; 9(1): 67-80, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22258147

RESUMO

Aptamers represent an emerging strategy to deliver cargo molecules, including dyes, drugs, proteins or even genes, into specific target cells. Upon binding to specific cell surface receptors aptamers can be internalized, for example by macropinocytosis or receptor mediated endocytosis. Here we report the in vitro selection and characterization of RNA aptamers with high affinity (Kd = 20 nM) and specificity for the human IL-6 receptor (IL-6R). Importantly, these aptamers trigger uptake without compromising the interaction of IL-6R with its natural ligands the cytokine IL-6 and glycoprotein 130 (gp130). We further optimized the aptamers to obtain a shortened, only 19-nt RNA oligonucleotide retaining all necessary characteristics for high affinity and selective recognition of IL-6R on cell surfaces. Upon incubation with IL-6R presenting cells this aptamer was rapidly internalized. Importantly, we could use our aptamer, to deliver bulky cargos, exemplified by fluorescently labeled streptavidin, into IL-6R presenting cells, thereby setting the stage for an aptamer-mediated escort of drug molecules to diseased cell populations or tissues.


Assuntos
Aptâmeros de Nucleotídeos/metabolismo , Interleucina-6/metabolismo , Receptores de Interleucina-6/metabolismo , Animais , Aptâmeros de Nucleotídeos/normas , Sequência de Bases , Linhagem Celular , Receptor gp130 de Citocina/metabolismo , DNA Complementar/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Camundongos , Dados de Sequência Molecular , Dobramento de RNA , Estabilidade de RNA , Estreptavidina/química , Especificidade por Substrato , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA