Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Genes Dev ; 24(23): 2621-6, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21123649

RESUMEN

Hfq, a protein required for small RNA (sRNA)-mediated regulation in bacteria, binds RNA with low-nanomolar K(d) values and long half-lives of complexes (>100 min). This cannot be reconciled with the 1- 2-min response time of regulation in vivo. We show that RNAs displace each other on Hfq on a short time scale by RNA concentration-driven (active) cycling. Already at submicromolar concentrations of competitor RNA, half-lives of RNA-Hfq complexes are ≈1 min. We propose that competitor RNA associates transiently with RNA-Hfq complexes, RNAs exchange binding sites, and one of the RNAs eventually dissociates. This solves the "strong binding-high turnover" paradox and permits efficient use of the Hfq pool.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Proteína de Factor 1 del Huésped/metabolismo , ARN Bacteriano/metabolismo , Unión Proteica
2.
Proc Natl Acad Sci U S A ; 110(16): E1533-42, 2013 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-23509299

RESUMEN

V-raf-1 murine leukemia viral oncogene homolog 1 (Raf-1) is a key activator of the ERK pathway and is a target for cross-regulation of this pathway by the cAMP signaling system. The cAMP-activated protein kinase, PKA, inhibits Raf-1 by phosphorylation on S259. Here, we show that the cAMP-degrading phosphodiesterase-8A (PDE8A) associates with Raf-1 to protect it from inhibitory phosphorylation by PKA, thereby enhancing Raf-1's ability to stimulate ERK signaling. PDE8A binds to Raf-1 with high (picomolar) affinity. Mapping of the interaction domain on PDE8A using peptide array technology identified amino acids 454-465 as the main binding site, which could be disrupted by mutation. A cell-permeable peptide corresponding to this region disrupted the PDE8A/Raf-1 interaction in cells, thereby reducing ERK activation and the cellular response to EGF. Overexpression of a catalytically inactive PDE8A in cells displayed a dominant negative phenotype on ERK activation. These effects were recapitulated at the organism level in genetically modified (PDE8A(-/-)) mice. Similarly, PDE8 deletion in Drosophila melanogaster reduced basal ERK activation and sensitized flies to stress-induced death. We propose that PDE8A is a physiological regulator of Raf-1 signaling in some cells.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Proto-Oncogénicas c-raf/metabolismo , 3',5'-AMP Cíclico Fosfodiesterasas/genética , Animales , Western Blotting , Cartilla de ADN/genética , Drosophila melanogaster , Eliminación de Gen , Células HEK293 , Células HeLa , Humanos , Inmunoprecipitación , Sistema de Señalización de MAP Quinasas/genética , Espectrometría de Masas , Ratones , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Fosforilación , Resonancia por Plasmón de Superficie
3.
Biochem J ; 459(1): 59-69, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24438162

RESUMEN

GAK (cyclin G-associated kinase) is a key regulator of clathrin-coated vesicle trafficking and plays a central role during development. Additionally, due to the unusually high plasticity of its catalytic domain, it is a frequent 'off-target' of clinical kinase inhibitors associated with respiratory side effects of these drugs. In the present paper, we determined the crystal structure of the GAK catalytic domain alone and in complex with specific single-chain antibodies (nanobodies). GAK is constitutively active and weakly associates in solution. The GAK apo structure revealed a dimeric inactive state of the catalytic domain mediated by an unusual activation segment interaction. Co-crystallization with the nanobody NbGAK_4 trapped GAK in a dimeric arrangement similar to the one observed in the apo structure, whereas NbGAK_1 captured the activation segment of monomeric GAK in a well-ordered conformation, representing features of the active kinase. The presented structural and biochemical data provide insight into the domain plasticity of GAK and demonstrate the utility of nanobodies to gain insight into conformational changes of dynamic molecules. In addition, we present structural data on the binding mode of ATP mimetic inhibitors and enzyme kinetic data, which will support rational inhibitor design of inhibitors to reduce the off-target effect on GAK.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/fisiología , Multimerización de Proteína/fisiología , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/fisiología , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/fisiología , Animales , Apoproteínas/química , Apoproteínas/fisiología , Camelus , Dominio Catalítico/fisiología , Cristalización/métodos , Activación Enzimática/fisiología , Humanos , Conformación Proteica
4.
Proc Natl Acad Sci U S A ; 109(34): E2248-57, 2012 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-22843676

RESUMEN

We have selected designed ankyrin repeat proteins (DARPins) from a synthetic library by using ribosome display that selectively bind to the mitogen-activated protein kinase ERK2 (extracellular signal-regulated kinase 2) in either its nonphosphorylated (inactive) or doubly phosphorylated (active) form. They do not bind to other kinases tested. Crystal structures of complexes with two DARPins, each specific for one of the kinase forms, were obtained. The two DARPins bind to essentially the same region of the kinase, but recognize the conformational change within the activation loop and an adjacent area, which is the key structural difference that occurs upon activation. Whereas the rigid phosphorylated activation loop remains in the same form when bound by the DARPin, the more mobile unphosphorylated loop is pushed to a new position. The DARPins can be used to selectively precipitate the cognate form of the kinases from cell lysates. They can also specifically recognize the modification status of the kinase inside the cell. By fusing the kinase with Renilla luciferase and the DARPin to GFP, an energy transfer from luciferase to GFP can be observed in COS-7 cells upon intracellular complex formation. Phosphorylated ERK2 is seen to increase by incubation of the COS-7 cells with FBS and to decrease upon adding the ERK pathway inhibitor PD98509. Furthermore, the anti-ERK2 DARPin is seen to inhibit ERK phosphorylation as it blocks the target inside the cell. This strategy of creating activation-state-specific sensors and kinase-specific inhibitors may add to the repertoire to investigate intracellular signaling in real time.


Asunto(s)
Repetición de Anquirina , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Animales , Células COS , Biología Computacional/métodos , Cristalización , Cristalografía por Rayos X/métodos , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Células HEK293 , Humanos , Fosforilación , Unión Proteica , Conformación Proteica , Proteínas Recombinantes/química , Ribosomas/química
6.
J Biol Chem ; 287(40): 33756-65, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22815475

RESUMEN

Binding of heterochromatin protein 1 (HP1) to the histone H3 lysine 9 trimethylation (H3K9me3) mark is a hallmark of establishment and maintenance of heterochromatin. Although genetic and cell biological aspects have been elucidated, the molecular details of HP1 binding to H3K9me3 nucleosomes are unknown. Using a combination of NMR spectroscopy and biophysical measurements on fully defined recombinant experimental systems, we demonstrate that H3K9me3 works as an on/off switch regulating distinct binding modes of hHP1ß to the nucleosome. The methyl-mark determines a highly flexible and very dynamic interaction of the chromodomain of hHP1ß with the H3-tail. There are no other constraints of interaction or additional multimerization interfaces. In contrast, in the absence of methylation, the hinge region and the N-terminal tail form weak nucleosome contacts mainly with DNA. In agreement with the high flexibility within the hHP1ß-H3K9me3 nucleosome complex, the chromoshadow domain does not provide a direct binding interface. Our results report the first detailed structural analysis of a dynamic protein-nucleosome complex directed by a histone modification and provide a conceptual framework for understanding similar interactions in the context of chromatin.


Asunto(s)
Proteínas Cromosómicas no Histona/química , Histonas/química , Lisina/química , Nucleosomas/química , Biotinilación , Calorimetría/métodos , Cromatina/química , Cromatina/metabolismo , Homólogo de la Proteína Chromobox 5 , ADN/química , Epigénesis Genética , Histonas/metabolismo , Humanos , Espectroscopía de Resonancia Magnética/métodos , Metilación , Nucleosomas/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Schizosaccharomyces/metabolismo
7.
J Biol Chem ; 286(11): 9079-96, 2011 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-21177871

RESUMEN

A-kinase anchoring proteins (AKAPs) tether protein kinase A (PKA) and other signaling proteins to defined intracellular sites, thereby establishing compartmentalized cAMP signaling. AKAP-PKA interactions play key roles in various cellular processes, including the regulation of cardiac myocyte contractility. We discovered small molecules, 3,3'-diamino-4,4'-dihydroxydiphenylmethane (FMP-API-1) and its derivatives, which inhibit AKAP-PKA interactions in vitro and in cultured cardiac myocytes. The molecules bind to an allosteric site of regulatory subunits of PKA identifying a hitherto unrecognized region that controls AKAP-PKA interactions. FMP-API-1 also activates PKA. The net effect of FMP-API-1 is a selective interference with compartmentalized cAMP signaling. In cardiac myocytes, FMP-API-1 reveals a novel mechanism involved in terminating ß-adrenoreceptor-induced cAMP synthesis. In addition, FMP-API-1 leads to an increase in contractility of cultured rat cardiac myocytes and intact hearts. Thus, FMP-API-1 represents not only a novel means to study compartmentalized cAMP/PKA signaling but, due to its effects on cardiac myocytes and intact hearts, provides the basis for a new concept in the treatment of chronic heart failure.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Miocitos Cardíacos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Sistemas de Mensajero Secundario/fisiología , Proteínas de Anclaje a la Quinasa A/genética , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/genética , Animales , Enfermedad Crónica , AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/metabolismo , Masculino , Contracción Miocárdica/efectos de los fármacos , Ratas , Ratas Endogámicas WKY , Sistemas de Mensajero Secundario/efectos de los fármacos
8.
Antimicrob Agents Chemother ; 55(1): 165-73, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20956605

RESUMEN

Staphylococcus aureus is the most common cause of nosocomial infections. Multiple antibiotic resistance and severe clinical outcomes provide a strong rationale for development of immunoglobulin-based strategies. Traditionally, novel immunological approaches against bacterial pathogens involve antibodies directed against cell surface-exposed virulence-associated epitopes or toxins. In this study, we generated a monoclonal antibody targeting the housekeeping protein IsaA, a suggested soluble lytic transglycosylase of S. aureus, and tested its therapeutic efficacy in two experimental mouse infection models. A murine anti-IsaA antibody of the IgG1 subclass (UK-66P) showed the highest binding affinity in Biacore analysis. This antibody recognized all S. aureus strains tested, including hospital-acquired and community-acquired methicillin-resistant S. aureus strains. Therapeutic efficacy in vivo in mice was analyzed using a central venous catheter-related infection model and a sepsis survival model. In both models, anti-IsaA IgG1 conferred protection against staphylococcal infection. Ex vivo, UK-66P activates professional phagocytes and induces highly microbicidal reactive oxygen metabolites in a dose-dependent manner, resulting in bacterial killing. The study provides proof of concept that monoclonal IgG1 antibodies with high affinity to the ubiquitously expressed, single-epitope-targeting IsaA are effective in the treatment of staphylococcal infection in different mouse models. Anti-IsaA antibodies might be a useful component in an antibody-based therapeutic for prophylaxis or adjunctive treatment of human cases of S. aureus infections.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Anticuerpos Antibacterianos/uso terapéutico , Antígenos Bacterianos/inmunología , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Animales , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Masculino , Ratones , Sepsis/tratamiento farmacológico , Sepsis/microbiología , Infecciones Estafilocócicas/microbiología
9.
Biochem J ; 413(1): 93-101, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18373497

RESUMEN

Conformational control of protein kinases is an important way of modulating catalytic activity. Crystal structures of the C (catalytic) subunit of PKA (protein kinase A) in complex with physiological inhibitors and/or nucleotides suggest a highly dynamic process switching between open and more closed conformations. To investigate the underlying molecular mechanisms, SPR (surface plasmon resonance) was used for detailed binding analyses of two physiological PKA inhibitors, PKI (heat-stable protein kinase inhibitor) and a truncated form of the R (regulatory) subunit (RIalpha 92-260), in the presence of various concentrations of metals and nucleotides. Interestingly, it could be demonstrated that high-affinity binding of each pseudosubstrate inhibitor was dependent only on the concentration of divalent metal ions. At low micromolar concentrations of Mg2+ with PKI, transient interaction kinetics with fast on- and off-rates were observed, whereas at high Mg2+ concentrations the off-rate was slowed down by a factor of 200. This effect could be attributed to the second, low-affinity metal-binding site in the C subunit. In contrast, when investigating the interaction of RIalpha 92-260 with the C subunit under the same conditions, it was shown that the association rate rather than the dissociation rate was influenced by the presence of high concentrations of Mg2+. A model is presented, where the high-affinity interaction of the C subunit with pseudosubstrate inhibitors (RIalpha and PKI) is dependent on the closed, catalytically inactive conformation induced by the binding of a nucleotide complex where both of the metal-binding sites are occupied.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Magnesio/farmacología , Unión Competitiva/efectos de los fármacos , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/química , Inhibidores Enzimáticos/metabolismo , Modelos Moleculares , Nucleótidos/metabolismo , Péptidos/metabolismo , Unión Proteica/efectos de los fármacos , Conformación Proteica , Subunidades de Proteína , Resonancia por Plasmón de Superficie
10.
Sci Rep ; 9(1): 16893, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31729440

RESUMEN

Cytokines of the interleukin (IL)-1 family regulate immune and inflammatory responses. The recently discovered IL-36 family members are involved in psoriasis, rheumatoid arthritis, and pulmonary diseases. Here, we show that IL-36α interacts with heme thereby contributing to its regulation. Based on in-depth spectroscopic analyses, we describe two heme-binding sites in IL-36α that associate with heme in a pentacoordinated fashion. Solution NMR analysis reveals structural features of IL-36α and its complex with heme. Structural investigation of a truncated IL-36α supports the notion that the N-terminus is necessary for association with its cognate receptor. Consistent with our structural studies, IL-36-mediated signal transduction was negatively regulated by heme in synovial fibroblast-like synoviocytes from rheumatoid arthritis patients. Taken together, our results provide a structural framework for heme-binding proteins and add IL-1 cytokines to the group of potentially heme-regulated proteins.


Asunto(s)
Hemo/metabolismo , Interleucina-1/metabolismo , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Células Cultivadas , Citocinas/agonistas , Citocinas/química , Citocinas/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Mediadores de Inflamación/agonistas , Mediadores de Inflamación/química , Mediadores de Inflamación/metabolismo , Interleucina-1/agonistas , Interleucina-1/química , Modelos Moleculares , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica , Psoriasis/metabolismo , Psoriasis/patología , Relación Estructura-Actividad , Membrana Sinovial/metabolismo , Membrana Sinovial/patología
11.
Proteomics ; 8(6): 1212-20, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18338824

RESUMEN

Functional proteomics aims to describe cellular protein networks in depth based on the quantification of molecular interactions. In order to study the interaction of adenosine-3',5'-cyclic monophosphate (cAMP), a general second messenger involved in several intracellular signalling networks, with one of its respective target proteins, the regulatory (R) subunit of cAMP dependent protein kinase (PKA), a number of different methods was employed. These include fluorescence polarisation (FP), isothermal titration calorimetry (ITC), surface plasmon resonance (SPR), amplified luminescence proximity homogeneous assay (ALPHA-screen), radioligand binding or activity-based assays. Kinetic, thermodynamic and equilibrium binding data of a variety of cAMP derivatives to several cAMP binding domains were integrated in a single database system, we called KinetXBase, allowing for very distinct data formats. KinetXBase is a practical data handling system for molecular interaction data of any kind, providing a synopsis of data derived from different technologies. This supports ongoing efforts in the bioinformatics community to devise formal concepts for a unified representation of interaction data, in order to enable their exchange and easy comparison. KinetXBase was applied here to analyse complex cAMP binding data and highly site-specific cAMP analogues could be identified. The software package is free for download by academic users.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Programas Informáticos , Biología Computacional/métodos , Unión Proteica , Proteómica/instrumentación , Proteómica/métodos , Resonancia por Plasmón de Superficie
12.
Biochem J ; 396(2): 297-306, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16483255

RESUMEN

PKA (protein kinase A) is tethered to subcellular compartments by direct interaction of its regulatory subunits (RI or RII) with AKAPs (A kinase-anchoring proteins). AKAPs preferentially bind RII subunits via their RII-binding domains. RII-binding domains form structurally conserved amphipathic helices with unrelated sequences. Their binding affinities for RII subunits differ greatly within the AKAP family. Amongst the AKAPs that bind RIIalpha subunits with high affinity is AKAP7delta [AKAP18delta; K(d) (equilibrium dissociation constant) value of 31 nM]. An N-terminally truncated AKAP7delta mutant binds RIIalpha subunits with higher affinity than the full-length protein presumably due to loss of an inhibitory region [Henn, Edemir, Stefan, Wiesner, Lorenz, Theilig, Schmidtt, Vossebein, Tamma, Beyermann et al. (2004) J. Biol. Chem. 279, 26654-26665]. In the present study, we demonstrate that peptides (25 amino acid residues) derived from the RII-binding domain of AKAP7delta bind RIIalpha subunits with higher affinity (K(d)=0.4+/-0.3 nM) than either full-length or N-terminally truncated AKAP7delta, or peptides derived from other RII binding domains. The AKAP7delta-derived peptides and stearate-coupled membrane-permeable mutants effectively disrupt AKAP-RII subunit interactions in vitro and in cell-based assays. Thus they are valuable novel tools for studying anchored PKA signalling. Molecular modelling indicated that the high affinity binding of the amphipathic helix, which forms the RII-binding domain of AKAP7delta, with RII subunits involves both the hydrophobic and the hydrophilic faces of the helix. Alanine scanning (25 amino acid peptides, SPOT technology, combined with RII overlay assays) of the RII binding domain revealed that hydrophobic amino acid residues form the backbone of the interaction and that hydrogen bond- and salt-bridge-forming amino acid residues increase the affinity of the interaction.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas de la Membrana/metabolismo , Péptidos/química , Unión Proteica , Proteínas de Anclaje a la Quinasa A , Proteínas Adaptadoras Transductoras de Señales/química , Secuencia de Aminoácidos , Aminoácidos/química , Aminoácidos/metabolismo , Animales , Sitios de Unión , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/química , Electrofisiología , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Proteínas de la Membrana/química , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/metabolismo , Péptidos/farmacología , Estructura Terciaria de Proteína , Subunidades de Proteína , Ratas , Alineación de Secuencia , Factores de Tiempo
13.
Metallomics ; 9(11): 1576-1584, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29043344

RESUMEN

Protein kinases are key enzymes in the regulation of eukaryotic signal transduction. As metalloenzymes they employ divalent cations for catalysis and regulation. We used the catalytic (C) subunit of cAMP-dependent protein kinase (PKA) as a model protein to investigate the role of a variety of physiologically or pathophysiologically relevant divalent metal ions in distinct steps within the catalytic cycle. It is established that divalent metal ions play a crucial role in co-binding of nucleotides and also assist in catalysis. Our studies reveal that besides the physiologically highly relevant magnesium, metals like zinc and manganese can assist in phosphoryl transfer, however, only a few support efficient substrate turnover (turnover catalysis). Those trace metals allow for substrate binding and phosphotransfer but hamper product release. We further established the unique role of magnesium as the common biologically relevant divalent metal ideally enabling (co-) substrate binding and orientation. Magnesium allows stable substrate binding and, on the other hand accelerates product release, thus being extremely efficient in turnover catalysis. We extended our studies to non-catalytic functions of protein kinases looking at pseudokinases, a subfamily of protein kinases inherently lacking critical residues for catalysis. Recently, pseudokinases have been linked to human diseases. Some pseudokinases are still capable of binding metal ions, yet have lost the ability to transfer the phosphoryl group from ATP to a given substrate. Here metal ions stabilize an active like, though catalytically unproductive conformation and are therefore crucial to maintain non-catalytic function. Finally, we demonstrate for the canonical kinase PKA that the trace metal manganese alone can stabilize protein kinases in an active like conformation allowing them to bind substrates even in the absence of nucleotides.


Asunto(s)
Cationes Bivalentes/farmacología , Pruebas de Enzimas/métodos , Metales/farmacología , Proteínas Quinasas/metabolismo , Biocatálisis/efectos de los fármacos , Cadmio/farmacología , Calcio/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , Magnesio/farmacología , Manganeso/farmacología , Nucleótidos/metabolismo , Unión Proteica/efectos de los fármacos , Especificidad por Sustrato , Resonancia por Plasmón de Superficie , Zinc/farmacología
14.
Eur J Cell Biol ; 85(7): 663-72, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16529845

RESUMEN

cAMP-dependent protein kinase (PKA) plays a key role in intracellular signalling. cAMP antagonists, acting as suppressors of PKA activity by preventing PKA-holoenzyme dissociation, have received increasing attention because of their potential use in diagnostics as well as for therapeutic purposes. A large number of cAMP analogs have been described over the last three decades and methodology has been established to monitor cAMP agonists action by either following enzymatic activity or holoenzyme dissociation. This is not the case for cAMP antagonists, where only a few substances have been demonstrated to exhibit effects in the low micromolar range, for example, Rp-8-Br-cAMPS. A main drawback in the development of new compounds is the lack of technologies to assess antagonist action in an in vitro situation as well as in living cells. Here we quantify the effect of several cAMP analogs applying three different biochemical/biophysical assay setups and one in-cell assay. This includes two methods monitoring subunit dissociation in a test tube, namely AlphaScreen, a bead-based proximity assay, and surface plasmon resonance, determining the association and dissociation patterns of the two PKA subunits in real time in response to antagonists. BRET(2), performed in living cells in a 96-well format, allows testing for the efficacy of membrane-permeable cAMP analogs based on a genetically engineered cAMP sensor. Using novel and established experimental strategies side by side, the action of cAMP and cAMP analogs was tested on type Ialpha PKA holoenzyme, thus generating methodology to screen drug libraries for potential cAMP antagonists with high accuracy, reproducibility as well as potential for automation.


Asunto(s)
AMP Cíclico/antagonistas & inhibidores , Animales , Células COS , Chlorocebus aethiops , AMP Cíclico/agonistas , Evaluación Preclínica de Medicamentos/métodos , Activación Enzimática/efectos de los fármacos , Transferencia Resonante de Energía de Fluorescencia/métodos , Humanos , Ratones , Espectrofotometría , Resonancia por Plasmón de Superficie
15.
Nat Commun ; 7: 11310, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27090491

RESUMEN

Histone H3 trimethylation of lysine 9 (H3K9me3) and proteins of the heterochromatin protein 1 (HP1) family are hallmarks of heterochromatin, a state of compacted DNA essential for genome stability and long-term transcriptional silencing. The mechanisms by which H3K9me3 and HP1 contribute to chromatin condensation have been speculative and controversial. Here we demonstrate that human HP1ß is a prototypic HP1 protein exemplifying most basal chromatin binding and effects. These are caused by dimeric and dynamic interaction with highly enriched H3K9me3 and are modulated by various electrostatic interfaces. HP1ß bridges condensed chromatin, which we postulate stabilizes the compacted state. In agreement, HP1ß genome-wide localization follows H3K9me3-enrichment and artificial bridging of chromatin fibres is sufficient for maintaining cellular heterochromatic conformation. Overall, our findings define a fundamental mechanism for chromatin higher order structural changes caused by HP1 proteins, which might contribute to the plastic nature of condensed chromatin.


Asunto(s)
Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Heterocromatina/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Secuencia de Aminoácidos , Western Blotting , Línea Celular Tumoral , Cromatina/genética , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/genética , Cristalografía por Rayos X , Heterocromatina/genética , Histonas/química , Humanos , Cinética , Lisina/química , Metilación , Microscopía Fluorescente , Modelos Moleculares , Datos de Secuencia Molecular , Nucleosomas/química , Nucleosomas/metabolismo , Unión Proteica , Multimerización de Proteína , Homología de Secuencia de Aminoácido , Electricidad Estática
16.
Methods Mol Biol ; 305: 17-46, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15939992

RESUMEN

Direct optical detection provides an excellent means to investigate interactions of molecules in biological systems. The dynamic equilibria inherent to these systems can be described in greater detail by recording the kinetics of a biomolecular interaction. Optical biosensors allow direct detection of interaction patterns without the need for labeling. An overview covering several commercially available biosensors is given, with a focus on instruments based on surface plasmon resonance (SPR) and reflectometric interference spectroscopy (RIFS). Potential assay formats and experimental design, appropriate controls, and calibration procedures, especially when handling low molecular weight substances, are discussed. The single steps of an interaction analysis combined with practical tips for evaluation, data processing, and interpretation of kinetic data are described in detail. In a practical example, a step-by-step procedure for the analysis of a low molecular weight compound interaction with serum protein, determined on a commercial SPR sensor, is presented.


Asunto(s)
Técnicas Biosensibles/métodos , Unión Proteica , Unión Competitiva , Técnicas Biosensibles/instrumentación , Técnicas Biosensibles/estadística & datos numéricos , Interpretación Estadística de Datos , Humanos , Técnicas In Vitro , Cinética , Ligandos , Peso Molecular , Óptica y Fotónica/instrumentación , Albúmina Sérica/metabolismo , Análisis Espectral/instrumentación , Análisis Espectral/métodos , Resonancia por Plasmón de Superficie/instrumentación , Resonancia por Plasmón de Superficie/métodos , Warfarina/metabolismo
17.
ACS Chem Biol ; 10(10): 2303-15, 2015 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-26200257

RESUMEN

cAMP-dependent protein kinase (PKA) is regulated primarily in response to physiological signals while nucleotides and metals may provide fine-tuning. PKA can use different metal ions for phosphoryl transfer, yet some, like Ca(2+), do not support steady-state catalysis. Fluorescence Polarization (FP) and Surface Plasmon Resonance (SPR) were used to study inhibitor and substrate interactions with PKA. The data illustrate how metals can act differentially as a result of their inherent coordination properties. We found that Ca(2+), in contrast to Mg(2+), does not induce high-affinity binding of PKA to pseudosubstrate inhibitors. However, Ca(2+) works in a single turnover mode to allow for phosphoryl-transfer. Using a novel SPR approach, we were able to directly monitor the interaction of PKA with a substrate in the presence of Mg(2+)ATP. This allows us to depict the entire kinase reaction including complex formation as well as release of the phosphorylated substrate. In contrast to Mg(2+), Ca(2+) apparently slows down the enzymatic reaction. A focus on individual reaction steps revealed that Ca(2+) is not as efficient as Mg(2+) in stabilizing the enzyme:substrate complex. The opposite holds true for product dissociation where Mg(2+) easily releases the phospho-substrate while Ca(2+) traps both reaction products at the active site. This explains the low steady-state activity in the presence of Ca(2+). Furthermore, Ca(2+) is able to modulate kinase activity as well as inhibitor binding even in the presence of Mg(2+). We therefore hypothesize that the physiological metal ions Mg(2+) and Ca(2+) both play a role in kinase activity and regulation. Since PKA is localized close to calcium channels and may render PKA activity susceptible to Ca(2+), our data provide a possible mechanism for novel crosstalk between cAMP and calcium signaling.


Asunto(s)
Calcio/farmacología , Cationes Bivalentes/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Magnesio/farmacología , Secuencia de Aminoácidos , Sitios de Unión , Calcio/química , Cationes Bivalentes/química , Proteínas Quinasas Dependientes de AMP Cíclico/química , Activación Enzimática/efectos de los fármacos , Iones , Magnesio/química , Modelos Biológicos , Datos de Secuencia Molecular , Alineación de Secuencia
18.
Hum Vaccin Immunother ; 10(4): 926-37, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24495867

RESUMEN

Multi-antigen immunotherapy approaches against Staphylococcus aureus are expected to have the best chance of clinical success when used in combinatorial therapy, potentially incorporating opsonic killing of bacteria and toxin neutralization. We recently reported the development of a murine monoclonal antibody specific for the immunodominant staphylococcal antigen A (IsaA), which showed highly efficient staphylococcal killing in experimental infection models of S. aureus. If IsaA-specific antibodies are to be used as a component of combination therapy in humans, the binding specificity and biological activity of the humanized variant must be preserved. Here, we describe the functional characterization of a humanized monoclonal IgG1 variant designated, hUK-66. The humanized antibody showed comparable binding kinetics to those of its murine parent, and recognized the target antigen IsaA on the surface of clinically relevant S. aureus lineages. Furthermore, hUK-66 enhances the killing of S. aureus in whole blood (a physiological environment) samples from healthy subjects and patients prone to staphylococcal infections such as diabetes and dialysis patients, and patients with generalized artery occlusive disease indicating no interference with already present natural antibodies. Taken together, these data indicate that hUK-66 mediates bacterial killing even in high risk patients and thus, could play a role for immunotherapy strategies to combat severe S. aureus infections.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales/inmunología , Antígenos Bacterianos/inmunología , Inmunoglobulina G/inmunología , Infecciones Estafilocócicas/terapia , Staphylococcus aureus/inmunología , Animales , Actividad Bactericida de la Sangre , Humanos , Ratones , Viabilidad Microbiana/efectos de los fármacos , Unión Proteica
19.
Chem Biol ; 20(6): 847-56, 2013 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-23790495

RESUMEN

Investigation of protein activation in living cells is fundamental to understanding how proteins are influenced by the full complement of upstream regulators they experience. Here, we describe the generation of a biosensor based on the DARPin binding scaffold suited for intracellular applications. Combining library selection and knowledge-based design, we created an ERK activity biosensor by derivatizing a DARPin specific for phosphorylated ERK with a solvatochromatic merocyanine dye, whose fluorescence increases upon pERK binding. The biosensor specifically responded to pERK2, recognized by its conformation, but not to ERK2 or other closely related mitogen-activated kinases tested. Activated endogenous ERK was visualized in mouse embryo fibroblasts, revealing greater activation in the nucleus, perinuclear regions, and especially the nucleoli. The DARPin-based biosensor will serve as a useful tool for studying biological functions of ERK in vitro and in vivo.


Asunto(s)
Proteína Quinasa 1 Activada por Mitógenos/análisis , Animales , Técnicas Biosensibles , Butadienos/farmacología , Activación Enzimática/efectos de los fármacos , Células HEK293 , Humanos , Ratones , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Musculares/química , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutagénesis Sitio-Dirigida , Células 3T3 NIH , Nitrilos/farmacología , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilación , Unión Proteica , Pirimidinonas/química , Especificidad por Sustrato
20.
J Microbiol Methods ; 88(2): 229-36, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22178430

RESUMEN

In many bacteria, high levels of the ubiquitous second messenger c-di-GMP have been demonstrated to suppress motility and to promote the establishment of surface-adherent biofilm communities. While molecular mechanisms underlying the synthesis and degradation of c-di-GMP have been comprehensively characterized, little is known about how c-di-GMP mediates its regulatory effects. In this study, we have established a chemical proteomics approach to identify c-di-GMP interacting proteins in the opportunistic pathogen Pseudomonas aeruginosa. A functionalized c-di-GMP analog, 2'-aminohexylcarbamoyl-c-di-GMP (2'-AHC-c-di-GMP), was chemically synthesized and following its immobilization used to perform affinity pull down experiments. Enriched proteins were subsequently identified by high-resolution mass spectrometry. 2'-AHC-c-di-GMP was also employed in surface plasmon resonance studies to evaluate and quantify the interaction of c-di-GMP with its potential target molecules in vitro. The biochemical tools presented here may serve the identification of novel classes of c-di-GMP effectors and thus contribute to a better characterization and understanding of the complex c-di-GMP signaling network.


Asunto(s)
Proteínas Bacterianas/análisis , Proteínas Portadoras/análisis , GMP Cíclico/análogos & derivados , Proteómica/métodos , Pseudomonas aeruginosa/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , GMP Cíclico/química , GMP Cíclico/metabolismo , Pseudomonas aeruginosa/química , Transducción de Señal , Resonancia por Plasmón de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA