Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 9(1): 16893, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31729440

RESUMO

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.


Assuntos
Heme/metabolismo , Interleucina-1/metabolismo , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Células Cultivadas , Citocinas/agonistas , Citocinas/química , Citocinas/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Mediadores da Inflamação/agonistas , Mediadores da Inflamação/química , Mediadores da Inflamação/metabolismo , Interleucina-1/agonistas , Interleucina-1/química , Modelos Moleculares , Simulação de Dinâmica Molecular , Ligação Proteica , Conformação Proteica , Psoríase/metabolismo , Psoríase/patologia , Relação Estrutura-Atividade , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia
3.
Metallomics ; 9(11): 1576-1584, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-29043344

RESUMO

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.


Assuntos
Cátions Bivalentes/farmacologia , Ensaios Enzimáticos/métodos , Metais/farmacologia , Proteínas Quinases/metabolismo , Biocatálise/efeitos dos fármacos , Cádmio/farmacologia , Cálcio/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Humanos , Magnésio/farmacologia , Manganês/farmacologia , Nucleotídeos/metabolismo , Ligação Proteica/efeitos dos fármacos , Especificidade por Substrato , Ressonância de Plasmônio de Superfície , Zinco/farmacologia
4.
Nat Commun ; 7: 11310, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-27090491

RESUMO

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.


Assuntos
Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Heterocromatina/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Sequência de Aminoácidos , Western Blotting , Linhagem Celular Tumoral , Cromatina/genética , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/química , Proteínas Cromossômicas não Histona/genética , Cristalografia por Raios X , Heterocromatina/genética , Histonas/química , Humanos , Cinética , Lisina/química , Metilação , Microscopia de Fluorescência , Modelos Moleculares , Dados de Sequência Molecular , Nucleossomos/química , Nucleossomos/metabolismo , Ligação Proteica , Multimerização Proteica , Homologia de Sequência de Aminoácidos , Eletricidade Estática
5.
ACS Chem Biol ; 10(10): 2303-15, 2015 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-26200257

RESUMO

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.


Assuntos
Cálcio/farmacologia , Cátions Bivalentes/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Magnésio/farmacologia , Sequência de Aminoácidos , Sítios de Ligação , Cálcio/química , Cátions Bivalentes/química , Proteínas Quinases Dependentes de AMP Cíclico/química , Ativação Enzimática/efeitos dos fármacos , Íons , Magnésio/química , Modelos Biológicos , Dados de Sequência Molecular , Alinhamento de Sequência
6.
Hum Vaccin Immunother ; 10(4): 926-37, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24495867

RESUMO

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.


Assuntos
Anticorpos Antibacterianos/imunologia , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais/imunologia , Antígenos de Bactérias/imunologia , Imunoglobulina G/imunologia , Infecções Estafilocócicas/terapia , Staphylococcus aureus/imunologia , Animais , Atividade Bactericida do Sangue , Humanos , Camundongos , Viabilidade Microbiana/efeitos dos fármacos , Ligação Proteica
7.
Biochem J ; 459(1): 59-69, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24438162

RESUMO

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.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Multimerização Proteica/fisiologia , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/fisiologia , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/fisiologia , Animais , Apoproteínas/química , Apoproteínas/fisiologia , Camelus , Domínio Catalítico/fisiologia , Cristalização/métodos , Ativação Enzimática/fisiologia , Humanos , Conformação Proteica
8.
Chem Biol ; 20(6): 847-56, 2013 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-23790495

RESUMO

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.


Assuntos
Proteína Quinase 1 Ativada por Mitógeno/análise , Animais , Técnicas Biossensoriais , Butadienos/farmacologia , Ativação Enzimática/efeitos dos fármacos , Células HEK293 , Humanos , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Musculares/química , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutagênese Sítio-Dirigida , Células NIH 3T3 , Nitrilas/farmacologia , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilação , Ligação Proteica , Pirimidinonas/química , Especificidade por Substrato
9.
Proc Natl Acad Sci U S A ; 110(16): E1533-42, 2013 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-23509299

RESUMO

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.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Proto-Oncogênicas c-raf/metabolismo , 3',5'-AMP Cíclico Fosfodiesterases/genética , Animais , Western Blotting , Primers do DNA/genética , Drosophila melanogaster , Deleção de Genes , Células HEK293 , Células HeLa , Humanos , Imunoprecipitação , Sistema de Sinalização das MAP Quinases/genética , Espectrometria de Massas , Camundongos , Camundongos Knockout , Mutagênese Sítio-Dirigida , Fosforilação , Ressonância de Plasmônio de Superfície
10.
J Biol Chem ; 287(40): 33756-65, 2012 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-22815475

RESUMO

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.


Assuntos
Proteínas Cromossômicas não Histona/química , Histonas/química , Lisina/química , Nucleossomos/química , Biotinilação , Calorimetria/métodos , Cromatina/química , Cromatina/metabolismo , Homólogo 5 da Proteína Cromobox , DNA/química , Epigênese Genética , Histonas/metabolismo , Humanos , Espectroscopia de Ressonância Magnética/métodos , Metilação , Nucleossomos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Schizosaccharomyces/metabolismo
11.
Proc Natl Acad Sci U S A ; 109(34): E2248-57, 2012 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-22843676

RESUMO

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.


Assuntos
Repetição de Anquirina , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Animais , Células COS , Biologia Computacional/métodos , Cristalização , Cristalografia por Raios X/métodos , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Células HEK293 , Humanos , Fosforilação , Ligação Proteica , Conformação Proteica , Proteínas Recombinantes/química , Ribossomos/química
12.
J Microbiol Methods ; 88(2): 229-36, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22178430

RESUMO

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.


Assuntos
Proteínas de Bactérias/análise , Proteínas de Transporte/análise , GMP Cíclico/análogos & derivados , Proteômica/métodos , Pseudomonas aeruginosa/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , GMP Cíclico/química , GMP Cíclico/metabolismo , Pseudomonas aeruginosa/química , Transdução de Sinais , Ressonância de Plasmônio de Superfície
13.
PLoS One ; 6(3): e18184, 2011 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-21445368

RESUMO

The characterization of factors contributing to the formation and development of surface-associated bacterial communities known as biofilms has become an area of intense interest since biofilms have a major impact on human health, the environment and industry. Various studies have demonstrated that motility, including swimming, swarming and twitching, seems to play an important role in the surface colonization and establishment of structured biofilms. Thereby, the impact of chemotaxis on biofilm formation has been less intensively studied. Pseudomonas aeruginosa has a very complex chemosensory system with two Che systems implicated in flagella-mediated motility. In this study, we demonstrate that the chemotaxis protein CheR1 is a methyltransferase that binds S-adenosylmethionine and transfers a methyl group from this methyl donor to the chemoreceptor PctA, an activity which can be stimulated by the attractant serine but not by glutamine. We furthermore demonstrate that CheR1 does not only play a role in flagella-mediated chemotaxis but that its activity is essential for the formation and maintenance of bacterial biofilm structures. We propose a model in which motility and chemotaxis impact on initial attachment processes, dispersion and reattachment and increase the efficiency and frequency of surface sampling in P. aeruginosa.


Assuntos
Quimiotaxia , Metiltransferases/metabolismo , Pseudomonas aeruginosa/enzimologia , Biofilmes , Metiltransferases/genética , Mutação , Pseudomonas aeruginosa/isolamento & purificação , Pseudomonas aeruginosa/fisiologia
14.
J Biol Chem ; 286(11): 9079-96, 2011 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-21177871

RESUMO

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.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Miócitos Cardíacos/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Sistemas do Segundo Mensageiro/fisiologia , Proteínas de Ancoragem à Quinase A/genética , Regulação Alostérica/efeitos dos fármacos , Regulação Alostérica/genética , Animais , Doença Crônica , AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/genética , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/metabolismo , Masculino , Contração Miocárdica/efeitos dos fármacos , Ratos , Ratos Endogâmicos WKY , Sistemas do Segundo Mensageiro/efeitos dos fármacos
15.
Antimicrob Agents Chemother ; 55(1): 165-73, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20956605

RESUMO

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.


Assuntos
Anticorpos Antibacterianos/imunologia , Anticorpos Antibacterianos/uso terapêutico , Antígenos de Bactérias/imunologia , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Animais , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Masculino , Camundongos , Sepse/tratamento farmacológico , Sepse/microbiologia , Infecções Estafilocócicas/microbiologia
16.
Genes Dev ; 24(23): 2621-6, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21123649

RESUMO

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.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Fator Proteico 1 do Hospedeiro/metabolismo , RNA Bacteriano/metabolismo , Ligação Proteica
17.
Biochem J ; 413(1): 93-101, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18373497

RESUMO

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.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Magnésio/farmacologia , Ligação Competitiva/efeitos dos fármacos , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/química , Inibidores Enzimáticos/metabolismo , Modelos Moleculares , Nucleotídeos/metabolismo , Peptídeos/metabolismo , Ligação Proteica/efeitos dos fármacos , Conformação Proteica , Subunidades Proteicas , Ressonância de Plasmônio de Superfície
18.
Proteomics ; 8(6): 1212-20, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18338824

RESUMO

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.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Software , Biologia Computacional/métodos , Ligação Proteica , Proteômica/instrumentação , Proteômica/métodos , Ressonância de Plasmônio de Superfície
19.
Artif Organs ; 31(6): 466-71, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17537059

RESUMO

Selective adhesion of plasma proteins to immobilized heparin is considered to be beneficial regarding hemocompatibility of foreign materials in contact with blood. Prothrombin, thrombin, antithrombin III (AT3), and fibrinogen were selected for analysis in an experimental model. Biomolecular interaction analysis employing surface plasmon resonance was utilized to record and analyze their binding properties in real time. Biotinylated heparin, heparin-albumin conjugate, and albumin, respectively, were immobilized onto streptavidin-coated sensors as ligands. Prothrombin did not bind to any of the ligand surfaces and no specific binding of any of the plasma proteins to albumin was observed. Binding kinetics of thrombin to heparin and to heparin-albumin conjugate were calculated using two different methods. For heparin, identical K(D)(equilibrium dissociation constant) values of 61 x 10(-9) M were obtained with both methods. For the conjugate, only slightly different K(D) values of 111 x 10(-9) and 104 x 10(-9) M, respectively, were calculated. The affinity of thrombin toward the heparin-coated surface proved to be higher than its affinity toward the heparin conjugate. The binding pattern of AT3 to both heparin and heparin-albumin conjugate, although specific, was biphasic, possibly due to a conformational change during the binding process. Steady-state kinetic analysis revealed a K(D) value of 281 +/- 24 x 10(-9) M for the heparin surface. For the conjugate surface, a K(D) of 53 +/- 5 x 10(-9) M was calculated, indicating a higher affinity toward heparin-albumin conjugate. A high-affinity binding of fibrinogen to high-density surfaces of both heparin and the conjugate was observed. However, as binding to low-density surfaces was considerably reduced, specificity remained uncertain.


Assuntos
Antitrombina III/metabolismo , Heparina/metabolismo , Protrombina/metabolismo , Albumina Sérica/metabolismo , Trombina/metabolismo , Adsorção , Materiais Biocompatíveis , Humanos , Ligação Proteica , Ressonância de Plasmônio de Superfície , Propriedades de Superfície
20.
Proteomics ; 6(19): 5132-9, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16912968

RESUMO

A variety of different in vivo and in vitro technologies provide comprehensive insights in protein-protein interaction networks. Here we demonstrate a novel approach to analyze, verify and quantify putative interactions between two members of the S100 protein family and 80 recombinant proteins derived from a proteome-wide protein expression library. Surface plasmon resonance (SPR) using Biacore technology and functional protein microarrays were used as two independent methods to study protein-protein interactions. With this combined approach we were able to detect nine calcium-dependent interactions between Arg-Gly-Ser-(RGS)-His6 tagged proteins derived from the library and GST-tagged S100B and S100A6, respectively. For the protein microarray affinity-purified proteins from the expression library were spotted onto modified glass slides and probed with the S100 proteins. SPR experiments were performed in the same setup and in a vice-versa approach reversing analytes and ligands to determine distinct association and dissociation patterns of each positive interaction. Besides already known interaction partners, several novel binders were found independently with both detection methods, albeit analogous immobilization strategies had to be applied in both assays.


Assuntos
Análise Serial de Proteínas , Ressonância de Plasmônio de Superfície , Técnicas Biossensoriais , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Humanos , Fatores de Crescimento Neural/química , Fatores de Crescimento Neural/metabolismo , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Ligação Proteica , Mapeamento de Interação de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteína A6 Ligante de Cálcio S100 , Subunidade beta da Proteína Ligante de Cálcio S100 , Proteínas S100/química , Proteínas S100/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...