Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Circ Res ; 132(7): 828-848, 2023 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-36883446

RESUMO

BACKGROUND: Signaling by cAMP is organized in multiple distinct subcellular nanodomains regulated by cAMP-hydrolyzing PDEs (phosphodiesterases). Cardiac ß-adrenergic signaling has served as the prototypical system to elucidate cAMP compartmentalization. Although studies in cardiac myocytes have provided an understanding of the location and properties of a handful of cAMP subcellular compartments, an overall view of the cellular landscape of cAMP nanodomains is missing. METHODS: Here, we combined an integrated phosphoproteomics approach that takes advantage of the unique role that individual PDEs play in the control of local cAMP, with network analysis to identify previously unrecognized cAMP nanodomains associated with ß-adrenergic stimulation. We then validated the composition and function of one of these nanodomains using biochemical, pharmacological, and genetic approaches and cardiac myocytes from both rodents and humans. RESULTS: We demonstrate the validity of the integrated phosphoproteomic strategy to pinpoint the location and provide critical cues to determine the function of previously unknown cAMP nanodomains. We characterize in detail one such compartment and demonstrate that the PDE3A2 isoform operates in a nuclear nanodomain that involves SMAD4 (SMAD family member 4) and HDAC-1 (histone deacetylase 1). Inhibition of PDE3 results in increased HDAC-1 phosphorylation, leading to inhibition of its deacetylase activity, derepression of gene transcription, and cardiac myocyte hypertrophic growth. CONCLUSIONS: We developed a strategy for detailed mapping of subcellular PDE-specific cAMP nanodomains. Our findings reveal a mechanism that explains the negative long-term clinical outcome observed in patients with heart failure treated with PDE3 inhibitors.


Assuntos
AMP Cíclico , Miócitos Cardíacos , Humanos , Proteômica , Diester Fosfórico Hidrolases , Hipertrofia , Adrenérgicos
2.
Blood ; 129(26): 3476-3485, 2017 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-28515092

RESUMO

During acute inflammation, 3 neutrophil subsets are found in the blood: neutrophils with a conventional segmented nucleus, neutrophils with a banded nucleus, and T-cell-suppressing CD62Ldim neutrophils with a high number of nuclear lobes. In this study, we compared the in vivo kinetics and proteomes of banded, mature, and hypersegmented neutrophils to determine whether these cell types represent truly different neutrophil subsets or reflect changes induced by lipopolysaccharide (LPS) activation. Using in vivo pulse-chase labeling of neutrophil DNA with 6,6-2H2-glucose, we found that 2H-labeled banded neutrophils appeared much earlier in blood than labeled CD62Ldim and segmented neutrophils, which shared similar label kinetics. Comparison of the proteomes by cluster analysis revealed that CD62Ldim neutrophils were clearly separate from conventional segmented neutrophils despite having similar kinetics in peripheral blood. Interestingly, the conventional segmented cells were more related at a proteome level to banded cells despite a 2-day difference in maturation time. The differences between CD62Ldim and mature neutrophils are unlikely to have been a direct result of LPS-induced activation, because of the extremely low transcriptional capacity of CD62Ldim neutrophils and the fact that neutrophils do not directly respond to the low dose of LPS used in the study (2 ng/kg body weight). Therefore, we propose CD62Ldim neutrophils are a truly separate neutrophil subset that is recruited to the bloodstream in response to acute inflammation. This trial was registered at www.clinicaltrials.gov as #NCT01766414.


Assuntos
Selectina L/análise , Neutrófilos/citologia , Análise por Conglomerados , Deutério/administração & dosagem , Glucose/administração & dosagem , Voluntários Saudáveis , Humanos , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/farmacologia , Proteoma , Coloração e Rotulagem
3.
Circ Res ; 120(1): 110-119, 2017 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-27729468

RESUMO

RATIONALE: Junctional membrane complexes (JMCs) in myocytes are critical microdomains, in which excitation-contraction coupling occurs. Structural and functional disruption of JMCs underlies contractile dysfunction in failing hearts. However, the role of newly identified JMC protein SPEG (striated muscle preferentially expressed protein kinase) remains unclear. OBJECTIVE: To determine the role of SPEG in healthy and failing adult hearts. METHODS AND RESULTS: Proteomic analysis of immunoprecipitated JMC proteins ryanodine receptor type 2 and junctophilin-2 (JPH2) followed by mass spectrometry identified the serine-threonine kinase SPEG as the only novel binding partner for both proteins. Real-time polymerase chain reaction revealed the downregulation of SPEG mRNA levels in failing human hearts. A novel cardiac myocyte-specific Speg conditional knockout (MCM-Spegfl/fl) model revealed that adult-onset SPEG deficiency results in heart failure (HF). Calcium (Ca2+) and transverse-tubule imaging of ventricular myocytes from MCM-Spegfl/fl mice post HF revealed both increased sarcoplasmic reticulum Ca2+ spark frequency and disrupted JMC integrity. Additional studies revealed that transverse-tubule disruption precedes the development of HF development in MCM-Spegfl/fl mice. Although total JPH2 levels were unaltered, JPH2 phosphorylation levels were found to be reduced in MCM-Spegfl/fl mice, suggesting that loss of SPEG phosphorylation of JPH2 led to transverse-tubule disruption, a precursor of HF development in SPEG-deficient mice. CONCLUSIONS: The novel JMC protein SPEG is downregulated in human failing hearts. Acute loss of SPEG in mouse hearts causes JPH2 dephosphorylation and transverse-tubule loss associated with downstream Ca2+ mishandling leading to HF. Our study suggests that SPEG could be a novel target for the treatment of HF.


Assuntos
Insuficiência Cardíaca/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Musculares/biossíntese , Proteínas Musculares/metabolismo , Miócitos Cardíacos/metabolismo , Quinase de Cadeia Leve de Miosina/biossíntese , Proteômica/métodos , Adulto , Idoso , Animais , Feminino , Células HEK293 , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Humanos , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Proteínas Musculares/genética , Quinase de Cadeia Leve de Miosina/genética
4.
J Proteome Res ; 16(2): 571-582, 2017 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-28152593

RESUMO

Sphingolipids (SLs) are essential components of cell membranes and are broad-range bioactive signaling molecules. SL levels must be tightly regulated as imbalances affect cellular function and contribute to pathologies ranging from neurodegenerative and metabolic disorders to cancer and aging. Deciphering how SL homeostasis is maintained and uncovering new regulators is required for understanding lipid biology and for identifying new targets for therapeutic interventions. Here we combine omics technologies to identify the changes of the transcriptome, proteome, and phosphoproteome in the yeast Saccharomyces cerevisiae upon SL depletion induced by myriocin. Surprisingly, while SL depletion triggers important changes in the expression of regulatory proteins involved in SL homeostasis, the most dramatic regulation occurs at the level of the phosphoproteome, suggesting that maintaining SL homeostasis demands rapid responses. To discover which of the phosphoproteomic changes are required for the cell's first-line response to SL depletion, we overlaid our omics results with systematic growth screens for genes required during growth in myriocin. By following the rate of SL biosynthesis in those candidates that are both affecting growth and are phosphorylated in response to the drug, we uncovered Atg9, Stp4, and Gvp36 as putative new regulators of SL homeostasis.


Assuntos
Ácido Aspártico Endopeptidases/genética , Proteínas Relacionadas à Autofagia/genética , Regulação Fúngica da Expressão Gênica , Proteínas de Membrana/genética , Proteínas de Transporte de Monossacarídeos/genética , Fosfoproteínas/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Antifúngicos/farmacologia , Ácido Aspártico Endopeptidases/metabolismo , Proteínas Relacionadas à Autofagia/metabolismo , Ácidos Graxos Monoinsaturados/farmacologia , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Homeostase/efeitos dos fármacos , Homeostase/genética , Proteínas de Membrana/metabolismo , Proteínas de Transporte de Monossacarídeos/metabolismo , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Proteômica/métodos , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Transdução de Sinais , Esfingolipídeos/antagonistas & inibidores , Esfingolipídeos/biossíntese
5.
J Biol Chem ; 290(12): 7887-96, 2015 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-25653285

RESUMO

Protein-protein interactions are important in providing compartmentalization and specificity in cellular signal transduction. Many studies have hallmarked the well designed compartmentalization of the cAMP-dependent protein kinase (PKA) through its anchoring proteins. Much less data are available on the compartmentalization of its closest homolog, cGMP-dependent protein kinase (PKG), via its own PKG anchoring proteins (GKAPs). For the enrichment, screening, and discovery of (novel) PKA anchoring proteins, a plethora of methodologies is available, including our previously described chemical proteomics approach based on immobilized cAMP or cGMP. Although this method was demonstrated to be effective, each immobilized cyclic nucleotide did not discriminate in the enrichment for either PKA or PKG and their secondary interactors. Hence, with PKG signaling components being less abundant in most tissues, it turned out to be challenging to enrich and identify GKAPs. Here we extend this cAMP-based chemical proteomics approach using competitive concentrations of free cyclic nucleotides to isolate each kinase and its secondary interactors. Using this approach, we identified Huntingtin-associated protein 1 (HAP1) as a putative novel GKAP. Through sequence alignment with known GKAPs and secondary structure prediction analysis, we defined a small sequence domain mediating the interaction with PKG Iß but not PKG Iα. In vitro binding studies and site-directed mutagenesis further confirmed the specificity and affinity of HAP1 binding to the PKG Iß N terminus. These data fully support that HAP1 is a GKAP, anchoring specifically to the cGMP-dependent protein kinase isoform Iß, and provide further evidence that also PKG spatiotemporal signaling is largely controlled by anchoring proteins.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Isoenzimas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Células HEK293 , Humanos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/química , Proteômica , Homologia de Sequência de Aminoácidos
6.
Mol Cell Proteomics ; 13(8): 2004-16, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24925903

RESUMO

The cyclic nucleotide cyclic guanosine monophosphate (cGMP) plays an important role in learning and memory, but its signaling mechanisms in the mammalian brain are not fully understood. Using mass-spectrometry-based proteomics, we evaluated how the cerebellum adapts its (phospho)proteome in a knockout mouse model of cGMP-dependent protein kinase type I (cGKI). Our data reveal that a small subset of proteins in the cerebellum (∼3% of the quantified proteins) became substantially differentially expressed in the absence of cGKI. More changes were observed at the phosphoproteome level, with hundreds of sites being differentially phosphorylated between wild-type and knockout cerebellum. Most of these phosphorylated sites do not represent known cGKI substrates. An integrative computational network analysis of the data indicated that the differentially expressed proteins and proteins harboring differentially phosphorylated sites largely belong to a tight network in the Purkinje cells of the cerebellum involving important cGMP/cAMP signaling nodes (e.g. PDE5 and PKARIIß) and Ca(2+) signaling (e.g. SERCA3). In this way, removal of cGKI could be linked to impaired cerebellar long-term depression at Purkinje cell synapses. In addition, we were able to identify a set of novel putative (phospho)proteins to be considered in this network. Overall, our data improve our understanding of cerebellar cGKI signaling and suggest novel players in cGKI-regulated synaptic plasticity.


Assuntos
Cerebelo/metabolismo , Proteína Quinase Dependente de GMP Cíclico Tipo I/genética , Fosfoproteínas/isolamento & purificação , Animais , GMP Cíclico , Proteína Quinase Dependente de GMP Cíclico Tipo I/metabolismo , Camundongos , Camundongos Knockout , Fosforilação , Proteômica/métodos , Transdução de Sinais , Sinapses/metabolismo
7.
Biochemistry ; 54(1): 11-21, 2015 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-25097019

RESUMO

Protein kinase A (PKA) in vertebrates is localized to specific locations in the cell via A-kinase anchoring proteins (AKAPs). The regulatory subunits of the four PKA isoforms (RIα, RIß, RIIα, and RIIß) each form a homodimer, and their dimerization domain interacts with a small helical region present in each of the more than 40 AKAPs reported so far. This allows for tight anchoring of PKA and efficient communication with other signaling proteins that interact with the AKAP scaffold in a spatial and temporal manner. The hydrophobic interaction surfaces of the PKA-R dimer and several AKAP helices have been investigated in great detail. Despite this knowledge, not every suggested AKAP has had its binding motif specified. Here we created an efficient bioinformatic tool, termed THAHIT, to accurately map the PKA binding motif and/or additional motifs of all previously reported AKAPs. Moreover, THAHIT predicts its specificity toward PKA-RIα and/or PKA-RIIα binding. To verify the validity of these newly predicted anchoring sites and their putative specificities, we used computational modeling approaches (HADDOCK), biochemical affinity studies (fluorescence anisotropy), and cellular colocalization studies. We further demonstrate the potential of THAHIT to identify novel AKAPs in cAMP-based chemical proteomics discovery data sets, and the human proteome. We retrieved numerous novel AKAP candidates, including a never reported 330 kDa AKAP observed in heart tissue, which we further characterized biochemically as a PKA-RIIα binder. Altogether, THAHIT provides a comprehensive overview of known and novel PKA-AKAP interaction domains and their PKA-R specificities.


Assuntos
Proteínas de Ancoragem à Quinase A/química , Proteínas de Ancoragem à Quinase A/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/química , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Domínios e Motivos de Interação entre Proteínas/fisiologia , Proteínas de Ancoragem à Quinase A/genética , Sequência de Aminoácidos , Proteínas Quinases Dependentes de AMP Cíclico/genética , Células HEK293 , Humanos , Dados de Sequência Molecular , Ligação Proteica/fisiologia , Estrutura Secundária de Proteína
8.
J Proteome Res ; 14(7): 2976-87, 2015 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-26066639

RESUMO

In phosphorylation-directed signaling, spatial and temporal control is organized by complex interaction networks that diligently direct kinases toward distinct substrates to fine-tune specificity. How these protein networks originate and evolve into complex regulatory machineries are among the most fascinating research questions in biology. Here, spatiotemporal signaling is investigated by tracing the evolutionary dynamics of each functional domain of cAMP-dependent protein kinase (PKA) and its diverse set of A-kinase anchoring proteins (AKAPs). Homologues of the catalytic (PKA-C) and regulatory (PKA-R) domains of the (PKA-R)2-(PKA-C)2 holoenzyme were found throughout evolution. Most variation was observed in the RIIa of PKA-R, crucial for dimerization and docking to AKAPs. The RIIa domain was not observed in all PKA-R homologues. In the fungi and distinct protist lineages, the RIIa domain emerges within PKA-R, but it displays large sequence variation. These organisms do not harbor homologues of AKAPs, suggesting that efficient docking to direct spatiotemporal PKA activity evolved in multicellular eukaryotes. To test this in silico hypothesis, we experimentally screened organisms with increasing complexity by cAMP-based chemical proteomics to reveal that the occurrence of PKA-AKAP interactions indeed coincided and expanded within vertebrates, suggesting a crucial role for AKAPs in the advent of metazoan multicellularity.


Assuntos
Evolução Biológica , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Animais , Domínio Catalítico , Sequência Conservada , Dados de Sequência Molecular , Filogenia , Proteômica , Homologia de Sequência de Aminoácidos , Frações Subcelulares/enzimologia
9.
Mol Cell Proteomics ; 12(11): 3350-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23882029

RESUMO

In the past decade, mass-spectrometry-based methods have emerged for the quantitative profiling of dynamic changes in protein phosphorylation, allowing the behavior of thousands of phosphorylation sites to be monitored in a single experiment. However, when one is interested in specific signaling pathways, such shotgun methodologies are not ideal because they lack selectivity and are not cost and time efficient with respect to instrument and data analysis time. Here we evaluate and explore a peptide-centric antibody generated to selectively enrich peptides containing the cAMP-dependent protein kinase (PKA) consensus motif. This targeted phosphoproteomic strategy is used to profile temporal quantitative changes of potential PKA substrates in Jurkat T lymphocytes upon prostaglandin E2 (PGE2) stimulation, which increases intracellular cAMP, activating PKA. Our method combines ultra-high-specificity motif-based immunoaffinity purification with cost-efficient stable isotope dimethyl labeling. We identified 655 phosphopeptides, of which 642 (i.e. 98%) contained the consensus motif [R/K][R/K/X]X[pS/pT]. When our data were compared with a large-scale Jurkat T-lymphocyte phosphoproteomics dataset containing more than 10,500 phosphosites, a minimal overlap of 0.2% was observed. This stresses the need for such targeted analyses when the interest is in a particular kinase. Our data provide a resource of likely substrates of PKA, and potentially some substrates of closely related kinases. Network analysis revealed that about half of the observed substrates have been implicated in cAMP-induced signaling. Still, the other half of the here-identified substrates have been less well characterized, representing a valuable resource for future research.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Fosfoproteínas/metabolismo , Proteômica/métodos , Motivos de Aminoácidos , Cromatografia Líquida/métodos , Dinoprostona/metabolismo , Dinoprostona/farmacologia , Humanos , Imunoprecipitação/métodos , Células Jurkat , Fosfoproteínas/química , Fosfoproteínas/genética , Mapas de Interação de Proteínas , Transdução de Sinais/efeitos dos fármacos , Especificidade por Substrato , Espectrometria de Massas em Tandem/métodos
10.
J Proteome Res ; 13(1): 249-59, 2014 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-24087892

RESUMO

Post-translational modifications (PTMs) play an important role in the regulation of protein function. Mass spectrometry based proteomics experiments nowadays identify tens of thousands of PTMs in a single experiment. A wealth of data has therefore become publically available. Evidently the biological function of each PTM is the key question to be addressed; however, such analyses focus primarily on single PTM events. This ignores the fact that PTMs may act in concert in the regulation of protein function, a process termed PTM crosstalk. Relatively little is known on the frequency and functional relevance of crosstalk between PTM sites. In a bioinformatics approach, we extracted PTMs occurring in proximity in the protein sequence from publically available databases. These PTMs and their flanking sequences were subjected to stringent motif searches, including a scoring for evolutionary conservation. Our unprejudiced approach was able to detect a respectable set of motifs, of which about half were described previously. Among these we could add many new proteins harboring these motifs. We extracted also several novel motifs, which through their widespread appearance and high conservation may pinpoint at previously nonannotated concerted PTM actions. By employing network analyses on these proteins, we propose putative functional roles for these novel motifs with two PTM sites in close proximity.


Assuntos
Proteínas/fisiologia , Receptor Cross-Talk , Sequência de Aminoácidos , Animais , Humanos , Espectrometria de Massas , Dados de Sequência Molecular , Processamento de Proteína Pós-Traducional , Proteínas/química , Homologia de Sequência de Aminoácidos
11.
J Cell Mol Med ; 18(11): 2135-46, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25216213

RESUMO

The cyclic adenosine monophosphate (cAMP)-dependent protein kinase (PKA) is an elementary molecule involved in both acute and chronic modulation of cardiac function. Substantial research in recent years has highlighted the importance of A-kinase anchoring proteins (AKAP) therein as they act as the backbones of major macromolecular signalling complexes of the ß-adrenergic/cAMP/PKA pathway. This review discusses the role of AKAP-associated protein complexes in acute and chronic cardiac modulation by dissecting their role in altering the activity of different ion channels, which underlie cardiac action potential (AP) generation. In addition, we review the involvement of different AKAP complexes in mechanisms of cardiac remodelling and arrhythmias.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Eletrofisiologia , Coração/fisiopatologia , Miocárdio/enzimologia , Potenciais de Ação , AMP Cíclico/metabolismo , Humanos , Canais Iônicos/metabolismo , Miocárdio/metabolismo , Transdução de Sinais , Quinases de Receptores Adrenérgicos beta/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 33(7): 1635-8, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23640497

RESUMO

OBJECTIVE: Platelet activation and subsequent protein release play an important role in healthy hemostasis and inflammatory responses, yet the identity and quantity of proteins in the platelet releasate are still debated. Here, we present a reversed releasate proteomics approach to determine unambiguously and quantitatively proteins released from activated platelets. APPROACH AND RESULTS: Isolated platelets were mock and fully stimulated after which the released proteins in the supernatant were removed. Using high-end proteomics technology (2D chromatography, stable isotope labeling, electron transfer dissociation, and high collision dissociation fragmentation) allowed us to quantitatively discriminate the released proteins from uncontrolled lysis products. Monitoring the copy numbers of ≈ 4500 platelet proteins, we observed that after stimulation via thrombin and collagen, only 124 (<3%) proteins were significantly released (P<0.05). The released proteins span a concentration range of ≥ 5 orders, as confirmed by ELISA. The released proteins were highly enriched in secretion tags and contained all known factors at high concentrations (>100 ng/mL, eg, thrombospondin, von Willebrand factor, and platelet factor 4). Interestingly, in the lower concentration range of the releasate many novel factors were identified. CONCLUSIONS: Our reversed releasate dataset forms the first unambiguous, in depth repository for molecular factors released by platelets.


Assuntos
Plaquetas/metabolismo , Ativação Plaquetária , Proteínas/metabolismo , Proteômica/métodos , Cromatografia por Troca Iônica , Cromatografia Líquida , Colágeno/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Marcação por Isótopo , Espectrometria de Massas em Tandem , Trombina/metabolismo
13.
Arterioscler Thromb Vasc Biol ; 33(7): 1538-43, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23619296

RESUMO

OBJECTIVE: Platelet adhesion to subendothelial collagen is dependent on the integrin α2ß1 and glycoprotein VI (GPVI) receptors. The major signaling routes in collagen-dependent platelet activation are outlined; however, crucial detailed knowledge of the actual phosphorylation events mediating them is still limited. Here, we explore phosphotyrosine signaling events downstream of GPVI with site-specific detail. APPROACH AND RESULTS: Immunoprecipitations of phosphotyrosine-modified peptides from protein digests of GPVI-activated and resting human platelets were compared by stable isotope-based quantitative mass spectrometry. We surveyed 214 unique phosphotyrosine sites over 2 time points, of which 28 showed a significant increase in phosphorylation on GPVI activation. Among these was Tyr370 of oligophrenin-1 (OPHN1), a Rho GTPase-activating protein. To elucidate the function of OPHN1 in platelets, we performed an array of functional platelet analyses within a small cohort of patients with rare oligophrenia. Because of germline mutations in the OPHN1 gene locus, these patients lack OPHN1 expression entirely and are in essence a human knockout model. Our studies revealed that among other unaltered properties, patients with oligophrenia show normal P-selectin exposure and αIIbß3 activation in response to GPVI, as well as normal aggregate formation on collagen under shear conditions. Finally, the major difference in OPHN1-deficient platelets turned out to be a significantly reduced collagen-induced filopodia formation. CONCLUSIONS: In-depth phosphotyrosine screening revealed many novel signaling recipients downstream of GPVI activation uncovering a new level of detail within this important pathway. To illustrate the strength of such data, functional follow-up of OPHN1 in human platelets deficient in this protein showed reduced filopodia formation on collagen, an important parameter of platelet hemostatic function.


Assuntos
Plaquetas/metabolismo , Proteínas do Citoesqueleto/sangue , Proteínas Ativadoras de GTPase/sangue , Erros Inatos do Metabolismo/sangue , Proteínas Nucleares/sangue , Glicoproteínas da Membrana de Plaquetas/metabolismo , Pseudópodes/metabolismo , Transdução de Sinais , Estudos de Casos e Controles , Criança , Colágeno/metabolismo , Proteínas do Citoesqueleto/deficiência , Proteínas do Citoesqueleto/genética , Proteínas Ativadoras de GTPase/deficiência , Proteínas Ativadoras de GTPase/genética , Hemostasia , Humanos , Imunoprecipitação , Masculino , Espectrometria de Massas , Erros Inatos do Metabolismo/genética , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Selectina-P/sangue , Fosforilação , Adesividade Plaquetária , Testes de Função Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Proteômica/métodos , Estresse Mecânico , Fatores de Tempo , Tirosina
14.
Angew Chem Int Ed Engl ; 53(36): 9660-4, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25044833

RESUMO

Protein phosphorylation is a widespread process forming the mechanistic basis of cellular signaling. Up to now, different aspects, for example, site-specificity, kinetics, role of co-factors, and structure-function relationships have been typically investigated by multiple techniques that are incompatible with one another. The approach introduced here maximizes the amount of information gained on protein (complex) phosphorylation while minimizing sample handling. Using high-resolution native mass spectrometry on intact protein (assemblies) up to 150 kDa we track the sequential incorporation of phosphate groups and map their localization by peptide LC-MS/MS. On two model systems, the protein kinase G and the interplay between Aurora kinase A and Bora, we demonstrate the simultaneous monitoring of various aspects of the phosphorylation process, namely the effect of different cofactors on PKG autophosphorylation and the interaction of AurA and Bora as both an enzyme-substrate pair and physical binding partners.


Assuntos
Fosforilação/fisiologia , Proteínas/metabolismo , Animais , Aurora Quinase A/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Humanos , Cinética , Espectrometria de Massas , Especificidade por Substrato
15.
J Biol Chem ; 287(52): 43789-97, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23115245

RESUMO

Protein kinase A-anchoring proteins (AKAPs) provide spatio-temporal specificity for the omnipotent cAMP-dependent protein kinase (PKA) via high affinity interactions with PKA regulatory subunits (PKA-RI, RII). Many PKA-RII-AKAP complexes are heavily tethered to cellular substructures, whereas PKA-RI-AKAP complexes have remained largely undiscovered. Here, using a cAMP affinity-based chemical proteomics strategy in human heart and platelets, we uncovered a novel, ubiquitously expressed AKAP, termed small membrane (sm)AKAP due to its specific localization at the plasma membrane via potential myristoylation/palmitoylation anchors. In vitro binding studies revealed specificity of smAKAP for PKA-RI (K(d) = 7 nM) over PKA-RII (K(d) = 53 nM) subunits, co-expression of smAKAP with the four PKA R subunits revealed an even more exclusive specificity of smAKAP for PKA-RIα/ß in the cellular context. Applying the singlet oxygen-generating electron microscopy probe miniSOG indicated that smAKAP is tethered to the plasma membrane and is particularly dense at cell-cell junctions and within filopodia. Our preliminary functional characterization of smAKAP provides evidence that, like PKA-RII, PKA-RI can be tightly tethered by a novel repertoire of AKAPs, providing a new perspective on spatio-temporal control of cAMP signaling.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Membrana Celular/metabolismo , Proteína Quinase Tipo I Dependente de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Lipoilação/fisiologia , Sistemas do Segundo Mensageiro/fisiologia , Proteínas de Ancoragem à Quinase A/genética , Animais , Membrana Celular/genética , AMP Cíclico/genética , Proteína Quinase Tipo I Dependente de AMP Cíclico/genética , Feminino , Humanos , Junções Intercelulares/genética , Junções Intercelulares/metabolismo , Masculino , Camundongos , Ligação Proteica , Pseudópodes/genética , Pseudópodes/metabolismo
16.
J Am Chem Soc ; 135(8): 2867-70, 2013 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-23387960

RESUMO

Active-site directed probes are powerful in studies of enzymatic function. We report an active-site directed probe based on a warhead so far considered unreactive. By replacing the C-terminal carboxylate of ubiquitin (Ub) with an alkyne functionality, a selective reaction with the active-site cysteine residue of de-ubiquitinating enzymes was observed. The resulting product was shown to be a quaternary vinyl thioether, as determined by X-ray crystallography. Proteomic analysis of proteins bound to an immobilized Ub alkyne probe confirmed the selectivity toward de-ubiquitinating enzymes. The observed reactivity is not just restricted to propargylated Ub, as highlighted by the selective reaction between caspase-1 (interleukin converting enzyme) and a propargylated peptide derived from IL-1ß, a caspase-1 substrate.


Assuntos
Alcinos/química , Cisteína/química , Peptídeo Hidrolases/metabolismo , Domínio Catalítico
17.
Mol Cell Proteomics ; 10(10): O111.008474, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21705516

RESUMO

In quantitative proteomics stable isotope labeling has progressed from cultured cells toward the total incorporation of labeled atoms or amino acids into whole multicellular organisms. For instance, the recently introduced (13)C(6)-lysine labeled SILAC mouse allows accurate comparison of protein expression directly in tissue. In this model, only lysine, but not arginine, residues are isotope labeled, as the latter may cause complications to the quantification by in vivo conversion of arginine to proline. The sole labeling of lysines discourages the use of trypsin, as not all peptides will be quantifiable. Therefore, in the initial work Lys-C was used for digestion. Here, we demonstrate that the lysine-directed protease metalloendopeptidase Lys-N is an excellent alternative. As lysine directed peptides generally yield longer and higher charged peptides, alongside the more traditional collision induced dissociation we also implemented electron transfer dissociation in a quantitative stable isotope labeling with amino acid in cell culture workflow for the first time. The utility of these two complementary approaches is highlighted by investigating the differences in protein expression between the left and right ventricle of a mouse heart. Using Lys-N and electron transfer dissociation yielded coverage to a depth of 3749 proteins, which is similar as earlier investigations into the murine heart proteome. In addition, this strategy yields quantitative information on ∼ 2000 proteins with a median coverage of four peptides per protein in a single strong cation exchange-liquid chromatography-MS experiment, revealing that the left and right ventricle proteomes are very similar qualitatively as well as quantitatively.


Assuntos
Ventrículos do Coração/metabolismo , Metaloendopeptidases/química , Proteoma/metabolismo , Proteômica/métodos , Animais , Isótopos de Carbono/química , Transporte de Elétrons , Feminino , Regulação da Expressão Gênica , Ventrículos do Coração/química , Marcação por Isótopo , Lisina/química , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/química , Miócitos Cardíacos/metabolismo , Peptídeos/análise , Peptídeos/química , Peptídeos/isolamento & purificação , Proteoma/análise
18.
Proteomics ; 12(6): 749-51, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22539426

RESUMO

A newly reorganized HUPO Cardiovascular Initiative was announced at the HUPO 2011 Cardiovascular Initiative Workshop at Geneva. The new initiative is now part of the biology- and disease-driven component of the HUPO Human Proteome Project (B/D-HPP). Here we report the recent achievements and future directions of the initiative, and offer a perspective on the present challenges of cardiovascular proteomics and its integration with the cardiovascular biology community at large.


Assuntos
Doenças Cardiovasculares/metabolismo , Sistema Cardiovascular/metabolismo , Proteômica/métodos , Animais , Sistema Cardiovascular/química , Humanos , Cooperação Internacional , Proteoma/análise , Proteoma/metabolismo , Suíça
19.
J Mol Cell Cardiol ; 52(2): 511-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21712045

RESUMO

Here we reveal that the characterization of large-scale re-arrangements of signaling scaffolds induced by heart failure can serve as a novel concept to identify more specific therapeutic targets. In the mammalian heart, the cAMP pathway, with the cAMP-dependent protein kinase (PKA) in a central role, acts directly downstream of adrenergic receptors to mediate cardiac contractility and rhythm. Heart failure, characterized by severe alterations in adrenergic stimulation is, amongst other interventions, often treated with ß-blockers. Contrasting results, however, have shown both beneficial and detrimental effects of decreased cAMP levels in failing hearts. We hypothesize that the origin of this behavior lies in the complex spatiotemporal organization of the regulatory subunit of PKA (PKA-R), which associates tightly with various A-kinase anchoring proteins (AKAPs) to specifically localize PKA's activity. Using chemical proteomics directly applied to human patient and control heart tissue we demonstrate that the association profile of PKA-R with several AKAPs is severely altered in the failing heart, for instance effecting the interaction between PKA and the novel AKAP SPHKAP was 6-fold upregulated upon failing heart conditions. Also a significant increase in captured cGMP-dependent protein kinase (PKG) and phosphodiesterase 2 (PDE2) was observed. The observed altered profiles can already explain many aspects of the aberrant cAMP-response in the failing human heart, validating that this dataset may provide a resource for several novel, more specific, treatment options. This article is part of a Special Issue entitled "Local Signaling in Myocytes".


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Insuficiência Cardíaca/metabolismo , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Miocárdio/metabolismo , Miofibrilas/metabolismo , Ligação Proteica , Mapeamento de Interação de Proteínas , Proteoma/metabolismo , Transdução de Sinais , Adulto Jovem
20.
J Proteome Res ; 11(11): 5235-44, 2012 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-23020738

RESUMO

Coronary atherosclerosis represents the major cause of death in Western societies. As atherosclerosis typically progresses over years without giving rise to clinical symptoms, biomarkers are urgently needed to identify patients at risk. Over the past decade, evidence has accumulated suggesting cross-talk between the diseased vasculature and cells of the innate immune system. We therefore employed proteomics to search for biomarkers associated with severe atherosclerotic coronary lumen stenosis in circulating leukocytes. In a two-phase approach, we first performed in-depth quantitative profiling of the granulocyte proteome on a small pooled cohort of patients suffering from chronic (sub)total coronary occlusion and matched control patients using stable isotope peptide labeling, two-dimensional LC-MS/MS and data-dependent decision tree fragmentation. Over 3000 proteins were quantified, among which 57 candidate biomarker proteins remained after stringent filtering. The most promising biomarker candidates were subsequently verified in the individual samples of the discovery cohort using label-free, single-run LC-MS/MS analysis, as well as in an independent verification cohort of 25 patients with total coronary occlusion (CTO) and 19 matched controls. Our data reveal bactericidal/permeability-increasing protein (BPI) as a promising biomarker for severe atherosclerotic coronary stenosis, being down-regulated in circulating granulocytes of CTO patients.


Assuntos
Peptídeos Catiônicos Antimicrobianos/sangue , Biomarcadores/sangue , Doença da Artéria Coronariana/sangue , Estenose Coronária/sangue , Granulócitos/metabolismo , Proteoma , Proteínas Sanguíneas , Separação Celular , Estudos de Coortes , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA