Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Int J Mol Sci ; 23(6)2022 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-35328453

RESUMEN

During general anesthesia, alterations in neuronal metabolism may induce neurotoxicity and/or neuroprotection depending on the dose and type of the applied anesthetic. In this study, we investigate the effects of clinically relevant concentrations of sevoflurane (2% and 4%, i.e., 1 and 2 MAC) on different activity states in hippocampal slices of young Wistar rats. We combine electrophysiological recordings, partial tissue oxygen (ptiO2) measurements, and flavin adenine dinucleotide (FAD) imaging with computational modeling. Sevoflurane minimally decreased the cerebral metabolic rate of oxygen (CMRO2) while decreasing synaptic transmission in naive slices. During pharmacologically induced gamma oscillations, sevoflurane impaired network activity, thereby decreasing CMRO2. During stimulus-induced neuronal activation, sevoflurane decreased CMRO2 and excitability while basal metabolism remained constant. In this line, stimulus-induced FAD transients decreased without changes in basal mitochondrial redox state. Integration of experimental data and computer modeling revealed no evidence for a direct effect of sevoflurane on key enzymes of the citric acid cycle or oxidative phosphorylation. Clinically relevant concentrations of sevoflurane generated a decent decrease in energy metabolism, which was proportional to the present neuronal activity. Mitochondrial function remained intact under sevoflurane, suggesting a better metabolic profile than isoflurane or propofol.


Asunto(s)
Anestésicos por Inhalación , Isoflurano , Anestésicos por Inhalación/farmacología , Animales , Metabolismo Energético , Flavina-Adenina Dinucleótido/metabolismo , Isoflurano/farmacología , Mitocondrias/metabolismo , Oxígeno/metabolismo , Ratas , Ratas Wistar , Sevoflurano/farmacología
2.
J Neurogenet ; 34(1): 106-114, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31980003

RESUMEN

The cellular analysis of mushroom body (MB)-dependent memory forming processes is far advanced, whereas, the molecular and physiological understanding of their synaptic basis lags behind. Recent analysis of the Drosophila olfactory system showed that Unc13A, a member of the M(Unc13) release factor family, promotes a phasic, high release probability component, while Unc13B supports a slower tonic release component, reflecting their different nanoscopic positioning within individual active zones. We here use STED super-resolution microscopy of MB lobe synapses to show that Unc13A clusters closer to the active zone centre than Unc13B. Unc13A specifically supported phasic transmission and short-term plasticity of Kenyon cell:output neuron synapses, measured by combining electrophysiological recordings of output neurons with optogenetic stimulation. Knockdown of unc13A within Kenyon cells provoked drastic deficits of olfactory aversive short-term and anaesthesia-sensitive middle-term memory. Knockdown of unc13B provoked milder memory deficits. Thus, a low frequency domain transmission component is probably crucial for the proper representation of memory-associated activity patterns, consistent with sparse Kenyon cell activation during memory acquisition and retrieval. Notably, Unc13A/B ratios appeared highly diversified across MB lobes, leaving room for an interplay of activity components in memory encoding and retrieval.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas de la Membrana/metabolismo , Memoria/fisiología , Cuerpos Pedunculados/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Plasticidad Neuronal/fisiología , Percepción Olfatoria/fisiología , Animales , Drosophila , Femenino , Isoformas de Proteínas , Sinapsis/metabolismo
3.
Eur J Hum Genet ; 31(9): 1066-1072, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-36732662

RESUMEN

Incidental research findings pose a considerable challenge to hospital-based research biobanks since they are acting as intermediaries between healthcare and research. In a joint action the centralized biobank ibdw (Interdisciplinary Bank of Biomaterials and Data Wuerzburg) together with local authorities drafted a coherent concept to manage incidental research findings in full compliance with relevant ethical and data privacy regulations. The concept was developed and elaborated in close collaboration with the German Biobank Alliance (GBA). Comprehensive documentation of all steps guarantees the traceability of the process. By a mandatory assessment of the findings prior to re-identification of the individual concerned, unnecessary measures can be avoided. The individual's "right not to know" is respected according to the stipulations of the informed consent. As a general principle any communication with the individual occurs exclusively through the hospital and by competent physicians with appropriate knowledge and communication skills. We propose this scheme as a blueprint for reporting workflows for incidental research findings at hospital-based biobanks.


Asunto(s)
Bancos de Muestras Biológicas , Investigación Biomédica , Humanos , Consenso , Retroalimentación , Hallazgos Incidentales , Flujo de Trabajo , Hospitales
4.
Clin Chem Lab Med ; 50(7): 1253-62, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22149738

RESUMEN

BACKGROUND: Determinations of platelet receptor functions are indispensable diagnostic indicators of cardiovascular and hemostatic diseases including hereditary and acquired receptor defects and receptor responses to drugs. However, presently available techniques for assessing platelet function have some disadvantages, such as low sensitivity and the requirement of large sample sizes and unphysiologically high agonist concentrations. Our goal was to develop and initially characterize a new technique designed to quantitatively analyze platelet receptor activation and platelet function on the basis of measuring changes in low angle light scattering. METHODS: We developed a novel technique based on low angle light scattering registering changes in light scattering at a range of different angles in platelet suspensions during activation. RESULTS: The method proved to be highly sensitive for simultaneous real time detection of changes in size and shape of platelets during activation. Unlike commonly-used methods, the light scattering method could detect platelet shape change and aggregation in response to nanomolar concentrations of extracellular nucleotides. Furthermore, our results demonstrate that the advantages of the light scattering method make it a choice method for platelet receptor monitoring and for investigation of both murine and human platelets in disease models. CONCLUSIONS: Our data demonstrate the suitability and superiority of this new low angle light scattering method for comprehensive analyses of platelet receptors and functions. This highly sensitive, quantitative, and online detection of essential physiological, pathophysiological and pharmacological-response properties of human and mouse platelets is a significant improvement over conventional techniques.


Asunto(s)
Plaquetas/química , Interpretación de Imagen Asistida por Computador/métodos , Pruebas de Función Plaquetaria/métodos , Receptores Purinérgicos P2/sangre , Adenosina Difosfato/sangre , Adenosina Trifosfato/sangre , Animales , Plaquetas/metabolismo , Colágeno/farmacología , Humanos , Luz , Ratones , Persona de Mediana Edad , Agregación Plaquetaria/efectos de los fármacos , Plasma Rico en Plaquetas/química , Plasma Rico en Plaquetas/metabolismo , Dispersión de Radiación , Tromboxano A2/sangre
5.
J Biol Chem ; 285(24): 18352-63, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20356841

RESUMEN

Protein kinase A (PKA) activation by cAMP phosphorylates multiple target proteins in numerous platelet inhibitory pathways that have a very important role in maintaining circulating platelets in a resting state. Here we show that in thrombin- and collagen-stimulated platelets, PKA is activated by cAMP-independent mechanisms involving dissociation of the catalytic subunit of PKA (PKAc) from an NFkappaB-IkappaBalpha-PKAc complex. We demonstrate mRNA and protein expression for most of the NFkappaB family members in platelets. From resting platelets, PKAc was co-immunoprecipitated with IkappaBalpha, and conversely, IkappaBalpha was also co-immunoprecipitated with PKAc. This interaction was significantly reduced in thrombin- and collagen-stimulated platelets. Stimulation of platelets with thrombin- or collagen-activated IKK, at least partly by PI3 kinase-dependent pathways, leading to phosphorylation of IkappaBalpha, disruption of an IkappaBalpha-PKAc complex, and release of free, active PKAc, which phosphorylated VASP and other PKA substrates. IKK inhibitor inhibited thrombin-stimulated IkBalpha phosphorylation, PKA-IkBalpha dissociation, and VASP phosphorylation, and potentiated integrin alphaIIbbeta3 activation and the early phase of platelet aggregation. We conclude that thrombin and collagen not only cause platelet activation but also appear to fine-tune this response by initiating downstream NFkappaB-dependent PKAc activation, as a novel feedback inhibitory signaling mechanism for preventing undesired platelet activation.


Asunto(s)
Plaquetas/metabolismo , Colágeno/química , Proteínas Quinasas Dependientes de AMP Cíclico/química , Quinasa I-kappa B/metabolismo , FN-kappa B/metabolismo , Trombina/química , Dominio Catalítico , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Células HL-60 , Humanos , Proteínas I-kappa B/metabolismo , Inhibidor NF-kappaB alfa , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Agregación Plaquetaria
6.
Thromb Res ; 171: 22-30, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30240944

RESUMEN

INTRODUCTION: Arachidonic acid induced aggregation is a generally accepted test for aspirin resistance. However, doubts have been raised that arachidonic acid stimulated aggregation can be regarded as reliable testing for aspirin resistance. Arachidonic acid, in addition to platelet activation, can induce phosphatidylserine translocation on the outer surface of platelet membrane which could be mediated by apoptosis pathways or transformation of platelets to the procoagulant state. MATERIALS AND METHODS: We explored effects of arachidonic acid over a vast range of concentrations and a wide range of read-outs for human platelet activation, procoagulant activity, and platelet viability. Additionally we tested whether cAMP- or cGMP-dependent protein kinase activation can inhibit procoagulant activity or platelet viability. RESULTS: Arachidonic acid-induced washed platelet activation was detected at low micromolar concentrations during the first 2 min of stimulation. After longer incubation and/or at higher concentrations arachidonic acid triggered platelet procoagulant activity and reduced platelet viability. At the same time, arachidonic acid stimulated adenylate cyclase mediated protein phosphorylation which correlated with reduced platelet activation. Moreover, additional stimulation of cAMP- or cGMP-dependent protein kinase inhibited only platelet activation, but did not prevent pro-coagulant activity and platelet death. CONCLUSIONS: While arachidonic acid induces platelet activation at low concentrations and during short incubation time, higher concentrations and lasting incubation evokes adenylate cyclase activation and subsequent protein phosphorylation corresponding to reduced platelet activation, but also enhanced pro-coagulant activity and reduced viability. Our observations provide further proof for the complex fine tuning of platelet responses in a time and agonist concentration dependent manner.


Asunto(s)
Ácido Araquidónico/farmacología , Plaquetas/efectos de los fármacos , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Activación Plaquetaria/efectos de los fármacos , Aspirina/farmacología , Plaquetas/citología , Plaquetas/metabolismo , Supervivencia Celular/efectos de los fármacos , Resistencia a Medicamentos , Humanos , Fosforilación/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Pruebas de Función Plaquetaria/métodos , Proteínas Quinasas/metabolismo
7.
Cell Rep ; 23(5): 1259-1274, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29719243

RESUMEN

High-throughput electron microscopy has started to reveal synaptic connectivity maps of single circuits and whole brain regions, for example, in the Drosophila olfactory system. However, efficacy, timing, and frequency tuning of synaptic vesicle release are also highly diversified across brain synapses. These features critically depend on the nanometer-scale coupling distance between voltage-gated Ca2+ channels (VGCCs) and the synaptic vesicle release machinery. Combining light super resolution microscopy with in vivo electrophysiology, we show here that two orthogonal scaffold proteins (ELKS family Bruchpilot, BRP, and Syd-1) cluster-specific (M)Unc13 release factor isoforms either close (BRP/Unc13A) or further away (Syd-1/Unc13B) from VGCCs across synapses of the Drosophila olfactory system, resulting in different synapse-characteristic forms of short-term plasticity. Moreover, BRP/Unc13A versus Syd-1/Unc13B ratios were different between synapse types. Thus, variation in tightly versus loosely coupled scaffold protein/(M)Unc13 modules can tune synapse-type-specific release features, and "nanoscopic molecular fingerprints" might identify synapses with specific temporal features.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Proteínas de la Membrana/metabolismo , Cuerpos Pedunculados , Proteínas del Tejido Nervioso/metabolismo , Vesículas Sinápticas , Animales , Drosophila melanogaster , Cuerpos Pedunculados/metabolismo , Cuerpos Pedunculados/ultraestructura , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestructura
8.
Stroke ; 34(3): 764-9, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12624305

RESUMEN

BACKGROUND AND PURPOSE: Dipyridamole and in particular dipyridamole in combination with low-dose aspirin are very effective in preventing recurrent stroke. However, the mechanism(s) underlying this dipyridamole effect have not been elucidated. Since dipyridamole inhibits the cGMP-specific phosphodiesterase type V in vitro, we hypothesized and tested whether therapeutically relevant dipyridamole concentrations enhance NO/cGMP-mediated effects in intact human platelets studied ex vivo. METHODS: Phosphorylation of vasodilator-stimulated phosphoprotein (VASP), an established marker of NO/cGMP effects in human platelets, was quantified by phosphorylation-specific antibodies and Western blots. Serotonin secretion and thromboxane synthase activity were determined by fluorometric quantification of derivatized serotonin and synthase products, respectively. RESULTS: Endothelium-derived factors such as NO and prostaglandin I2 are known to elevate both cGMP and cAMP levels with concomitant platelet inhibition and VASP phosphorylation. In our in vitro experiments, therapeutically relevant concentrations (3.5 micromol/L) of dipyridamole amplified only cGMP-mediated VASP phosphorylation due to the NO donor sodium nitroprusside, but not cAMP-mediated effects. Furthermore, thromboxane synthase activity and serotonin secretion, events important for initial platelet activation, were inhibited by sodium nitroprusside, an effect also enhanced by dipyridamole, demonstrating the functional relevance of these observations. Finally, the ex vivo enhancement of NO/cGMP effects was also observed with platelets obtained from healthy volunteers treated with extended-release dipyridamole. CONCLUSIONS: Under therapeutically relevant conditions, dipyridamole enhances platelet inhibition by amplifying the signaling of the NO donor sodium nitroprusside. These data support the concept that enhancement of endothelium-dependent NO/cGMP-mediated signaling may be an important in vivo component of dipyridamole action.


Asunto(s)
Plaquetas/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , GMP Cíclico/metabolismo , Dipiridamol/farmacología , Óxido Nítrico/metabolismo , Fosfoproteínas/metabolismo , Vasodilatadores/farmacología , Adulto , Alprostadil/farmacología , Plaquetas/metabolismo , Preparaciones de Acción Retardada/farmacología , Femenino , Humanos , Técnicas In Vitro , Masculino , Proteínas de Microfilamentos , Persona de Mediana Edad , Donantes de Óxido Nítrico/farmacología , Nitroprusiato/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Fosforilación/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Valores de Referencia , Serotonina/biosíntesis , Serotonina/metabolismo , Transducción de Señal/efectos de los fármacos
9.
Eur J Pharmacol ; 740: 15-27, 2014 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-25003953

RESUMEN

Platelets are permanently exposed to a variety of prostanoids formed by blood cells or the vessel wall. The two major prostanoids, prostacyclin and thromboxane act through well established pathways mediated by their respective G-protein coupled receptors inhibiting or promoting platelet aggregation accordingly. Yet the role of other prostanoids and prostanoid receptors for platelet function regulation has not been thoroughly investigated. We aimed at a comprehensive analysis of prostanoid effects on platelets, the receptors and pathways involved and functional consequences. We analyzed cAMP formation and phosphorylation of proteins pivotal to platelet function as well as functional platelet responses such as secretion, aggregation and phosphorylation. The types of prostanoid receptors contributing and their individual share in signaling pathways were analyzed and indicated a major role for prostanoid IP1 and DP1 receptors followed by prostanoid EP4 and EP3 receptors while prostanoid EP2 receptors appear less relevant. We could show for the first time the reciprocal action of the endogenous prostaglandin PGE2 on platelets by functional responses and phosphorylation events. PGE2 evokes stimulatory as well as inhibitory effects in a concentration dependent manner in platelets via prostanoid EP3 or EP4 and prostanoid DP1 receptors. A mathematical model integrating the pathway components was established which successfully reproduces the observed platelet responses. Additionally we could show that human platelets themselves produce sufficient PGE2 to act in an autocrine or paracrine fashion. These mechanisms may provide a fine tuning of platelet responses in the circulating blood by either promoting or limiting endogenous platelet activation.


Asunto(s)
Plaquetas/efectos de los fármacos , Prostaglandinas/metabolismo , Receptores de Prostaglandina/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Plaquetas/metabolismo , Plaquetas/fisiología , Calcio/metabolismo , Moléculas de Adhesión Celular/metabolismo , AMP Cíclico/metabolismo , Humanos , Proteínas de Microfilamentos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Selectina-P/metabolismo , Fosfoproteínas/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Serotonina/metabolismo , Proteínas de Unión al GTP rap1/metabolismo
10.
Methods Mol Biol ; 893: 101-13, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22665297

RESUMEN

Quantitative proteomics has become a routinely used technique to globally compare protein content and expression profiles of biological samples, for instance after differential stimulation. In this context, chemical stable isotope-based labeling techniques, such as ICAT and iTRAQ, have been successfully applied in a large variety of studies. Since iTRAQ labels are isobaric, quantitation is conducted on the MS/MS level. Consequently, up to eight samples can be multiplexed and quantified in a single experiment without increasing sample complexity. Here, we present a robust workflow to conduct iTRAQ quantification of biological samples such as human platelets, which comprises (a) an adequate sample preparation procedure, (b) an optimized tryptic digestion protocol, (c) SPE desalting and subsequent peptide labeling using a 4-plex iTRAQ labeling kit, and (d) fractionation of the obtained peptide mixture by strong cation exchange chromatography.


Asunto(s)
Fragmentos de Péptidos/química , Proteoma/química , Plaquetas/química , Extractos Celulares/química , Extractos Celulares/aislamiento & purificación , Cromatografía por Intercambio Iónico , Electroforesis en Gel de Poliacrilamida , Humanos , Marcaje Isotópico , Metilación , Fragmentos de Péptidos/aislamiento & purificación , Proteolisis , Proteoma/aislamiento & purificación , Espectrometría de Masas en Tándem , Tripsina/química
11.
Br J Pharmacol ; 167(4): 826-38, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22612416

RESUMEN

BACKGROUND AND PURPOSE: cGMP is involved in the regulation of many cellular processes including cardiac and smooth muscle contractility, aldosterone synthesis and inhibition of platelet activation. Intracellular effects cGMP are mediated by cGMP-dependent PKs, cGMP-regulated PDEs and cGMP-gated ion channels. PKG inhibitors are widely used to discriminate PKG-specific effects. They can be divided into cyclic nucleotide-binding site inhibitors such as Rp-phosphorothioate analogues (Rp-cGMPS), ATP-binding site inhibitors such as KT5823, and substrate binding site inhibitors represented by the recently described DT-oligopeptides. As it has been shown that Rp-cGMPS and KT5823 have numerous non-specific effects, we analysed the pharmacological properties of the oligopeptide (D)-DT-2 described as a highly specific, membrane-permeable, PKG inhibitor. EXPERIMENTAL APPROACH: Specificity and potency of (D)-DT-2 to inhibit PKG activity was evaluated using biochemical assays in vitro and by substrate phosphorylation analysis in various cell types including human platelets, rat mesangial cells and rat neonatal cardiomyocytes. KEY RESULTS: Despite potent inhibition of PKGI in vitro, (D)-DT-2 lost specificity for PKG in cell homogenates and particularly in living cells, as demonstrated by phosphorylation of different substrates. Instead, (D)-DT-2 modulated activity of other kinases including ERK, p38, PKB and PKC, thereby inducing unpredicted and often opposing functional effects. CONCLUSIONS AND IMPLICATIONS: We conclude that DT-oligopeptides, as other inhibitors, cannot be used to specifically inhibit PKG in intact cells. Therefore, no specific pharmacological PKG inhibitors are available, and reliable studies of PKG signalling can only be made by using RNA knockdown or genetic deletion methods.


Asunto(s)
Proteína Quinasa Dependiente de GMP Cíclico Tipo I/antagonistas & inhibidores , Oligopéptidos/farmacología , Animales , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células HEK293 , Humanos , Células Mesangiales/efectos de los fármacos , Células Mesangiales/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA