Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 182
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(28): e2210152120, 2023 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-37406102

RESUMO

Sepsis has emerged as a global health burden associated with multiple organ dysfunction and 20% mortality rate in patients. Numerous clinical studies over the past two decades have correlated the disease severity and mortality in septic patients with impaired heart rate variability (HRV), as a consequence of impaired chronotropic response of sinoatrial node (SAN) pacemaker activity to vagal/parasympathetic stimulation. However, the molecular mechanism(s) downstream to parasympathetic inputs have not been investigated yet in sepsis, particularly in the SAN. Based on electrocardiography, fluorescence Ca2+ imaging, electrophysiology, and protein assays from organ to subcellular level, we report that impaired muscarinic receptor subtype 2-G protein-activated inwardly-rectifying potassium channel (M2R-GIRK) signaling in a lipopolysaccharide-induced proxy septic mouse model plays a critical role in SAN pacemaking and HRV. The parasympathetic responses to a muscarinic agonist, namely IKACh activation in SAN cells, reduction in Ca2+ mobilization of SAN tissues, lowering of heart rate and increase in HRV, were profoundly attenuated upon lipopolysaccharide-induced sepsis. These functional alterations manifested as a direct consequence of reduced expression of key ion-channel components (GIRK1, GIRK4, and M2R) in the mouse SAN tissues and cells, which was further evident in the human right atrial appendages of septic patients and likely not mediated by the common proinflammatory cytokines elevated in sepsis.


Assuntos
Lipopolissacarídeos , Sepse , Humanos , Animais , Camundongos , Lipopolissacarídeos/toxicidade , Lipopolissacarídeos/metabolismo , Nó Sinoatrial/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Sepse/induzido quimicamente , Sepse/metabolismo
2.
Nitric Oxide ; 129: 41-52, 2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-36216240

RESUMO

Hydrogen sulfide (H2S) emerged as an essential signaling molecule exerting beneficial effects in various cardiovascular, neurodegenerative, or musculoskeletal diseases with an inflammatory component, such as osteoarthritis. These protective effects were initially attributed to protein S-sulfhydration, a posttranslational modification of reactive cysteine residues. However, recent studies suggest that polysulfides and not H2S are responsible for S-sulfhydration. To distinguish between H2S and polysulfide-mediated effects in this study, we used the slow-releasing H2S and persulfide donor P*, which can be decomposed into polysulfides. The effects of P* on IL-1ß-induced inducible nitric oxide synthase (iNOS), a pro-inflammatory mediator in osteoarthritis, were determined by nitrite measurement, qPCR, and Western blotting in the murine chondrocyte-like cell line ATDC5. Decomposed P* significantly reduced IL-1ß-induced iNOS signaling via polysulfides, independently of H2S. In line with this, the fast-releasing H2S donor NaHS was ineffective. In RAW 264.7 macrophages, similar results were obtained. P*-derived polysulfides further diminished IL-1ß-induced CCAAT/enhancer-binding protein (C/EBP) ß and δ expression in ATDC5 cells, which might play a critical role in P*-mediated iNOS decline. In conclusion, our data support the view that polysulfides are essential signaling molecules as well as potential mediators of H2S signaling. Moreover, we propose that C/EBPß/δ might be a novel target involved in H2S and polysulfide-mediated anti-inflammatory signaling.


Assuntos
Sulfeto de Hidrogênio , Osteoartrite , Camundongos , Animais , Sulfeto de Hidrogênio/farmacologia , Sulfeto de Hidrogênio/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Sulfetos/farmacologia , Sulfetos/metabolismo , Anti-Inflamatórios , Óxido Nítrico/metabolismo
3.
Sensors (Basel) ; 20(6)2020 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-32192068

RESUMO

The increasing utilization of carbon fiber reinforced plastic (CFRP) in the aeronautical industry calls for a structural health monitoring (SHM) system for adhesively bonded CFRP joints. Optical glass fiber with inscribed fiber Bragg gratings (FBGs) is a promising technology for a SHM system. This paper investigates the intrusive effect of embedding optical glass fibers carrying FBGs on adhesive bond strength and adhesive layer thickness and quality. Embedding the optical glass fibers directly in the adhesive bond has the advantage of directly monitoring the targeted structure but poses the risk of significantly reducing the bond strength. Optical glass fibers with different cladding diameters (50, 80, 125 µm) and coating types (polyimide, with a thickness of 3-8 µm, and acrylate, with a thickness of ~35 µm) are embedded in structural and repair film adhesives here. Without embedded optical glass fibers, the film adhesives have an adhesive layer thickness of ~90 µm (structural) and ~100 µm (repair) after curing. The intrusive effect of the fiber embedding on the adhesive bond strength is investigated here with quasi static and fatigue single lap joint (SLJ) tensile shear tests. Also, the influence of hydrothermal aging procedures on the quasi static tensile shear strength is investigated. It is found that optical glass fibers with a total diameter (glass fiber cladding + coating) of ~145 µm significantly reduce the quasi static tensile shear strength and increase the adhesive layer thickness and number of air inclusions (or pores) in the structural film adhesive joints. In the repair adhesive joints, no significant reduction of quasi static tensile shear strength is caused by the embedding of any of the tested fiber types and diameters. However, an increase in the adhesive layer thickness is detected. In both adhesive films, no effect on the quasi-static tensile shear strength is detected when embedding optical glass fibers with total diameters <100 µm. The applied aging regime only affects the repair film adhesive joints, and the structural film adhesive joints show no significant reduction. A polyimide-coated 80 µm optical glass fiber is selected for fatigue SLJ tensile shear tests in combination with the more sensitive structural film adhesive. No significant differences between the S-N curves and tensile shear fatigue strength of the reference samples without embedded optical fibers and the samples carrying the polyimide-coated 80 µm optical glass fibers are detected. Thus, it is concluded that the influences of embedding optical glass fibers with total diameters <100 µm on the fatigue limit of the tested film adhesive joints is negligible.

4.
J Mol Cell Cardiol ; 130: 49-58, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30910669

RESUMO

Adenosine exerts numerous protective actions in the heart, including attenuation of cardiac hypertrophy. Adenosine kinase (ADK) converts adenosine to adenosine monophosphate (AMP) and is the major route of myocardial adenosine metabolism, however, the impact of ADK activity on cardiac structure and function is unknown. To examine the role of ADK in cardiac homeostasis and adaptation to stress, conditional cardiomyocyte specific ADK knockout mice (cADK-/-) were produced using the MerCreMer-lox-P system. Within 4 weeks of ADK disruption, cADK-/- mice developed spontaneous hypertrophy and increased ß-Myosin Heavy Chain expression without observable LV dysfunction. In response to 6 weeks moderate left ventricular pressure overload (transverse aortic constriction;TAC), wild type mice (WT) exhibited ~60% increase in ventricular ADK expression and developed LV hypertrophy with preserved LV function. In contrast, cADK-/- mice exhibited significantly greater LV hypertrophy and cardiac stress marker expression (atrial natrurietic peptide and ß-Myosin Heavy Chain), LV dilation, reduced LV ejection fraction and increased pulmonary congestion. ADK disruption did not decrease protein methylation, inhibit AMPK, or worsen fibrosis, but was associated with persistently elevated mTORC1 and p44/42 ERK MAP kinase signaling and a striking increase in microtubule (MT) stabilization/detyrosination. In neonatal cardiomyocytes exposed to hypertrophic stress, 2-chloroadenosine (CADO) or adenosine treatment suppressed MT detyrosination, which was reversed by ADK inhibition with iodotubercidin or ABT-702. Conversely, adenoviral over-expression of ADK augmented CADO destabilization of MTs and potentiated CADO attenuation of cardiomyocyte hypertrophy. Together, these findings indicate a novel adenosine receptor-independent role for ADK-mediated adenosine metabolism in cardiomyocyte microtubule dynamics and protection against maladaptive hypertrophy.


Assuntos
Adenosina Quinase/metabolismo , Cardiomegalia/metabolismo , Sistema de Sinalização das MAP Quinases , Microtúbulos/metabolismo , Miócitos Cardíacos/metabolismo , Disfunção Ventricular Esquerda/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Adenosina Quinase/genética , Animais , Cardiomegalia/genética , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Camundongos , Camundongos Knockout , Microtúbulos/genética , Miócitos Cardíacos/patologia , Ratos , Ratos Sprague-Dawley , Volume Sistólico/genética , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/patologia , Disfunção Ventricular Esquerda/fisiopatologia
5.
Nitric Oxide ; 89: 14-21, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31022534

RESUMO

Nitric oxide synthase (NOS) catalyzes the transformation of l-arginine, molecular oxygen (O2), and NADPH-derived electrons to nitric oxide (NO) and l-citrulline. Under some conditions, however, NOS catalyzes the reduction of O2 to superoxide (O2-) instead, a phenomenon that is generally referred to as uncoupling. In principle, both the heme in the oxygenase domain and the flavins in the reductase domain could catalyze O2- formation. In the former case the oxyferrous (Fe(II)O2) complex that is formed as an intermediate during catalysis would dissociate to heme and O2-; in the latter case the reduced flavins would reduce O2 to O2-. The NOS cofactor tetrahydrobiopterin (BH4) is indispensable for coupled catalysis. In the case of uncoupling at the heme this is explained by the essential role of BH4 as an electron donor to the oxyferrous complex; in the case of uncoupling at the flavins it is assumed that the absence of BH4 results in NOS monomerization, with the monomers incapable to sustain NO synthesis but still able to support uncoupled catalysis. In spite of little supporting evidence, uncoupling at the reductase after NOS monomerization appears to be the predominant hypothesis at present. To set the record straight we extended prior studies by determining under which conditions uncoupling of the neuronal and endothelial isoforms (nNOS and eNOS) occurred and if a correlation exists between uncoupling and the monomer/dimer equilibrium. We determined the rates of coupled/uncoupled catalysis by measuring NADPH oxidation spectrophotometrically at 340 nm and citrulline synthesis as the formation of [3H]-citrulline from [3H]-Arg. The monomer/dimer equilibrium was determined by FPLC and, for comparison, by low-temperature polyacrylamide gel electrophoresis. Uncoupling occurred in the absence of Arg and/or BH4, but not in the absence of Ca2+ or calmodulin (CaM). Since omission of Ca2+/CaM will completely block heme reduction while still allowing substantial FMN reduction, this argues against uncoupling by the reductase domain. In the presence of heme-directed NOS inhibitors uncoupling occurred to the extent that these compound allowed heme reduction, again arguing in favor of uncoupling at the heme. The monomer/dimer equilibrium showed no correlation with uncoupling. We conclude that uncoupling by BH4 deficiency takes place exclusively at the heme, with virtually no contribution from the flavins and no role for NOS monomerization.


Assuntos
Óxido Nítrico Sintase/química , Biopterinas/análogos & derivados , Biopterinas/química , Citrulina/química , Inibidores Enzimáticos/química , Heme/química , Humanos , Imidazóis/química , NADP/química , Óxido Nítrico Sintase/antagonistas & inibidores , Nitroarginina/química , Oxigênio/química , Pichia/genética , Multimerização Proteica
6.
Mol Pharmacol ; 93(2): 73-78, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29138269

RESUMO

Belonging to the class of so-called soluble guanylate cyclase (sGC) activators, cinaciguat and BAY 60-2770 are interesting therapeutic tools for the treatment of various cardiovascular pathologies. The drugs are supposed to preferentially stimulate oxidized or heme-depleted, but not native sGC. Since this concept has been challenged by studies demonstrating complete relaxation of nondiseased vessels, this study was designed to reinvestigate the mode of action in greater detail. To this purpose, the effect of cinaciguat was studied on vessel tone of porcine coronary arteries and rat thoracic aortas. Organ bath studies showed that the compound caused time- and concentration-dependent relaxation of precontracted vessels with a maximal effect observed at 90 minutes. The dilatory response was not affected by extensive washout of the drug. Cinaciguat-induced vasodilation was associated with a time- and concentration-dependent increase of cGMP levels. Experiments with purified sGC in the presence of Tween 20 showed that cinaciguat activates the heme-free enzyme in a concentration-dependent manner with an EC50 value of ∼0.2 µM and maximal cGMP formation at 10 µM. By contrast, the effect of cinaciguat on 1H-[1,2,4]oxadiazolo-[4,3-a]quinoxalin-1-one-oxidized (ferric) sGC was moderate, reaching ∼10%-15% of maximal activity. Dilution experiments of cinaciguat/Tween 20-preincubated sGC revealed the irreversible character of the drug. Assuming a sensitive balance between heme-free, ferric, and nitric oxide-sensitive ferrous sGC in cells and tissues, we propose that cinaciguat by virtue of its irreversible mode of action is capable of shifting this equilibrium toward the heme-free apo-sGC species.


Assuntos
Benzoatos/farmacologia , Inibidores Enzimáticos/farmacologia , Mimetismo Molecular , Protoporfirinas/metabolismo , Guanilil Ciclase Solúvel/antagonistas & inibidores , Vasodilatação/efeitos dos fármacos , Animais , Aorta Torácica/fisiologia , Bovinos , Vasos Coronários/metabolismo , GMP Cíclico/metabolismo , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Ativação Enzimática , Estabilidade Enzimática , Pulmão/efeitos dos fármacos , Pulmão/enzimologia , Protoporfirinas/química , Ratos Sprague-Dawley , Guanilil Ciclase Solúvel/metabolismo , Suínos , Vasodilatadores/farmacologia
7.
Mol Pharmacol ; 93(4): 335-343, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29358221

RESUMO

According to current views, oxidation of aldehyde dehydrogenase-2 (ALDH2) during glyceryltrinitrate (GTN) biotransformation is essentially involved in vascular nitrate tolerance and explains the dependence of this reaction on added thiols. Using a novel fluorescent intracellular nitric oxide (NO) probe expressed in vascular smooth muscle cells (VSMCs), we observed ALDH2-catalyzed formation of NO from GTN in the presence of exogenously added dithiothreitol (DTT), whereas only a short burst of NO, corresponding to a single turnover of ALDH2, occurred in the absence of DTT. This short burst of NO associated with oxidation of the reactive C302 residue in the active site was followed by formation of low-nanomolar NO, even without added DTT, indicating slow recovery of ALDH2 activity by an endogenous reductant. In addition to the thiol-reversible oxidation of ALDH2, thiol-refractive inactivation was observed, particularly under high-turnover conditions. Organ bath experiments with rat aortas showed that relaxation by GTN lasted longer than that caused by the NO donor diethylamine/NONOate, in line with the long-lasting nanomolar NO generation from GTN observed in VSMCs. Our results suggest that an endogenous reductant with low efficiency allows sustained generation of GTN-derived NO in the low-nanomolar range that is sufficient for vascular relaxation. On a longer time scale, mechanism-based, thiol-refractive irreversible inactivation of ALDH2, and possibly depletion of the endogenous reductant, will render blood vessels tolerant to GTN. Accordingly, full reactivation of oxidized ALDH2 may not occur in vivo and may not be necessary to explain GTN-induced vasodilation.


Assuntos
Aldeído-Desidrogenase Mitocondrial/metabolismo , Tolerância a Medicamentos/fisiologia , Músculo Liso Vascular/metabolismo , Nitratos/metabolismo , Óxido Nítrico/metabolismo , Nitroglicerina/metabolismo , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/metabolismo , Linhagem Celular Transformada , Linhagem Celular Tumoral , Ditiotreitol/farmacologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Músculo Liso Vascular/efeitos dos fármacos , Nitratos/farmacologia , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley
8.
J Biol Chem ; 291(46): 24076-24084, 2016 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-27679490

RESUMO

Aldehyde dehydrogenase-2 (ALDH2) catalyzes vascular bioactivation of the antianginal drug nitroglycerin (GTN), resulting in activation of soluble guanylate cyclase (sGC) and cGMP-mediated vasodilation. We have previously shown that a minor reaction of ALDH2-catalyzed GTN bioconversion, accounting for about 5% of the main clearance-based turnover yielding inorganic nitrite, results in direct NO formation and concluded that this minor pathway could provide the link between vascular GTN metabolism and activation of sGC. However, lack of detectable NO at therapeutically relevant GTN concentrations (≤1 µm) in vascular tissue called into question the biological significance of NO formation by purified ALDH2. We addressed this issue and used a novel, highly sensitive genetically encoded fluorescent NO probe (geNOp) to visualize intracellular NO formation at low GTN concentrations (≤1 µm) in cultured vascular smooth muscle cells (VSMC) expressing an ALDH2 mutant that reduces GTN to NO but lacks clearance-based GTN denitration activity. NO formation was compared with GTN-induced activation of sGC. The addition of 1 µm GTN to VSMC expressing either wild-type or C301S/C303S ALDH2 resulted in pronounced intracellular NO elevation, with maximal concentrations of 7 and 17 nm, respectively. Formation of GTN-derived NO correlated well with activation of purified sGC in VSMC lysates and cGMP accumulation in intact porcine aortic endothelial cells infected with wild-type or mutant ALDH2. Formation of NO and cGMP accumulation were inhibited by ALDH inhibitors chloral hydrate and daidzin. The present study demonstrates that ALDH2-catalyzed NO formation is necessary and sufficient for GTN bioactivation in VSMC.


Assuntos
Aldeído-Desidrogenase Mitocondrial/metabolismo , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Óxido Nítrico/metabolismo , Nitroglicerina/farmacocinética , Aldeído-Desidrogenase Mitocondrial/antagonistas & inibidores , Aldeído-Desidrogenase Mitocondrial/genética , Substituição de Aminoácidos , Animais , Bovinos , Hidrato de Cloral/farmacologia , Humanos , Isoflavonas/farmacologia , Camundongos , Camundongos Knockout , Mutação de Sentido Incorreto , Nitroglicerina/farmacologia , Suínos
9.
Nitric Oxide ; 70: 59-67, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28882669

RESUMO

The members of the nitric oxide synthase (NOS) family, eNOS, nNOS and iNOS, are well-characterized enzymes. However, due to the lack of suitable direct NO sensors, little is known about the kinetic properties of cellular NO generation by the different nitric oxide synthase isoenzymes. Very recently, we developed a novel class of fluorescent protein-based NO-probes, the geNOps, which allow real-time measurement of cellular NO generation and fluctuation. By applying these genetic NO biosensors to nNOS-, eNOS- and iNOS-expressing HEK293 cells we were able to characterize the respective NO dynamics in single cells that exhibited identical Ca2+ signaling as comparable activator of nNOS and eNOS. Our data demonstrate that upon Ca2+ mobilization nNOS-derived NO signals occur instantly and strictly follow the Ca2+ elevation while NO release by eNOS occurs gradually and sustained. To detect high NO levels in cells expressing iNOS, a new ratiometric probe based on two fluorescent proteins was developed. This novel geNOp variant allows the measurement of the high NO levels in cells expressing iNOS. Moreover, we used this probe to study the L-arginine-dependency of NO generation by iNOS on the level of single cells. Our experiments highlight that the geNOps technology is suitable to detect obvious differences in the kinetics, amplitude and substrate-dependence of cellular NO signals-derived from all three nitric oxide synthase isoforms.


Assuntos
Óxido Nítrico Sintase Tipo III/análise , Óxido Nítrico Sintase Tipo II/análise , Óxido Nítrico Sintase Tipo I/análise , Óxido Nítrico/biossíntese , Arginina/metabolismo , Técnicas Biossensoriais/instrumentação , Cálcio/metabolismo , Corantes Fluorescentes/química , Células HEK293/enzimologia , Humanos , Isoenzimas , Cinética , Proteínas Luminescentes/química , Microscopia de Fluorescência , Óxido Nítrico/análise , Óxido Nítrico/química
10.
Diabetologia ; 59(12): 2503-2506, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27376542

RESUMO

Medications approved for diabetes-associated renal and cardiovascular morbidities and candidate drugs currently in development are subject to substantial variability in drug response. Heterogeneity on a molecular phenotype level is not apparent at clinical presentation, which means that inter-individual differences in drug effect at the molecular level are masked. These findings identify the need for optimising patient phenotyping via use of molecular biomarkers for a personalised therapy approach. Molecular diversity may, on the one hand, result from the effect of genetic polymorphisms on drug transport, metabolism and effective target modulation. Equally relevant, differences may be due to molecular pathologies. The presence of distinct molecular phenotypes is suggested by classifiers aimed at modelling progressive disease. Such functions for prognosis incorporate a complex set of clinical variables or a multitude of molecular markers reflecting a diverse set of molecular disease mechanisms. This information on disease pathology and the mechanism of action of the drug needs to be systematically integrated with data on molecular biomarkers to develop an experimental tool for personalising medicine. The large amount of molecular data available for characterising diabetes-associated morbidities allows for elucidation of molecular process model representations of disease pathologies. Selecting biomarker candidates on such grounds and, in turn identifying their association with progressive disease allows for the identification of molecular processes associated with disease progression. The molecular effect of a drug can also be modelled at a molecular process level, and the integration of disease pathology and drug effect molecular models reveals candidate biomarkers for assessing drug response. Such tools serve as enrichment strategies aimed at adding precision to drug development and use.


Assuntos
Diabetes Mellitus/metabolismo , Nefropatias Diabéticas/metabolismo , Biologia de Sistemas/métodos , Animais , Comorbidade , Humanos , Farmacogenética
11.
J Biol Chem ; 290(41): 24932-44, 2015 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-26296888

RESUMO

Citrulline formation by both human neuronal nitric-oxide synthase (nNOS) and mouse macrophage inducible NOS was inhibited by the hydrogen sulfide (H2S) donor Na2S with IC50 values of ∼2.4·10(-5) and ∼7.9·10(-5) m, respectively, whereas human endothelial NOS was hardly affected at all. Inhibition of nNOS was not affected by the concentrations of l-arginine (Arg), NADPH, FAD, FMN, tetrahydrobiopterin (BH4), and calmodulin, indicating that H2S does not interfere with substrate or cofactor binding. The IC50 decreased to ∼1.5·10(-5) m at pH 6.0 and increased to ∼8.3·10(-5) m at pH 8.0. Preincubation of concentrated nNOS with H2S under turnover conditions decreased activity after dilution by ∼70%, suggesting irreversible inhibition. However, when calmodulin was omitted during preincubation, activity was not affected, suggesting that irreversible inhibition requires both H2S and NO. Likewise, NADPH oxidation was inhibited with an IC50 of ∼1.9·10(-5) m in the presence of Arg and BH4 but exhibited much higher IC50 values (∼1.0-6.1·10(-4) m) when Arg and/or BH4 was omitted. Moreover, the relatively weak inhibition of nNOS by Na2S in the absence of Arg and/or BH4 was markedly potentiated by the NO donor 1-(hydroxy-NNO-azoxy)-l-proline, disodium salt (IC50 ∼ 1.3-2.0·10(-5) m). These results suggest that nNOS and inducible NOS but not endothelial NOS are irreversibly inhibited by H2S/NO at modest concentrations of H2S in a reaction that may allow feedback inhibition of NO production under conditions of excessive NO/H2S formation.


Assuntos
Inibidores Enzimáticos/farmacologia , Sulfeto de Hidrogênio/farmacologia , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Óxido Nítrico/farmacologia , Animais , Citrulina/biossíntese , Interações Medicamentosas , Escherichia coli/citologia , Escherichia coli/efeitos dos fármacos , Escherichia coli/metabolismo , Glutationa/farmacologia , Humanos , Camundongos , NADP/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Oxirredução/efeitos dos fármacos , Compostos de Sulfidrila/farmacologia
12.
J Transl Med ; 14(1): 203, 2016 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-27378474

RESUMO

BACKGROUND: Individual patients show a large variability in albuminuria response to angiotensin receptor blockers (ARB). Identifying novel biomarkers that predict ARB response may help tailor therapy. We aimed to discover and validate a serum metabolite classifier that predicts albuminuria response to ARBs in patients with diabetes mellitus and micro- or macroalbuminuria. METHODS: Liquid chromatography-tandem mass spectrometry metabolomics was performed on serum samples. Data from patients with type 2 diabetes and microalbuminuria (n = 49) treated with irbesartan 300 mg/day were used for discovery. LASSO and ridge regression were performed to develop the classifier. Improvement in albuminuria response prediction was assessed by calculating differences in R(2) between a reference model of clinical parameters and a model with clinical parameters and the classifier. The classifier was externally validated in patients with type 1 diabetes and macroalbuminuria (n = 50) treated with losartan 100 mg/day. Molecular process analysis was performed to link metabolites to molecular mechanisms contributing to ARB response. RESULTS: In discovery, median change in urinary albumin excretion (UAE) was -42 % [Q1-Q3: -69 to -8]. The classifier, consisting of 21 metabolites, was significantly associated with UAE response to irbesartan (p < 0.001) and improved prediction of UAE response on top of the clinical reference model (R(2) increase from 0.10 to 0.70; p < 0.001). In external validation, median change in UAE was -43 % [Q1-Q35: -63 to -23]. The classifier improved prediction of UAE response to losartan (R(2) increase from 0.20 to 0.59; p < 0.001). Specifically ADMA impacting eNOS activity appears to be a relevant factor in ARB response. CONCLUSIONS: A serum metabolite classifier was discovered and externally validated to significantly improve prediction of albuminuria response to ARBs in diabetes mellitus.


Assuntos
Antagonistas de Receptores de Angiotensina/uso terapêutico , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/metabolismo , Metaboloma , Adulto , Albuminúria/sangue , Albuminúria/complicações , Antagonistas de Receptores de Angiotensina/farmacologia , Compostos de Bifenilo/uso terapêutico , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/tratamento farmacológico , Feminino , Humanos , Irbesartana , Losartan/uso terapêutico , Masculino , Metaboloma/efeitos dos fármacos , Pessoa de Meia-Idade , Modelos Moleculares , Tetrazóis/uso terapêutico
13.
Eur J Clin Invest ; 46(3): 213-26, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26707063

RESUMO

BACKGROUND: MicroRNAs (miRNAs) contribute to chronic kidney disease (CKD) progression via regulating mRNAs involved in renal homeostasis. However, their association with clinical outcome remains poorly understood. MATERIALS AND METHODS: We performed miRNA and mRNA expression profiling on renal biopsy sections by qPCR (miRNA) and microarrays (mRNA) in a discovery (n = 43) and in a validation (n = 29) cohort. miRNAs differentiating stable and progressive cases were inversely correlated with putative target mRNAs, which were further characterized by pathway analysis using KEGG pathways. RESULTS: miR-30d, miR-140-3p, miR-532-3p, miR-194, miR-190, miR-204 and miR-206 were downregulated in progressive cases. These seven miRNAs correlated with upregulated 29 target mRNAs involved in inflammatory response, cell-cell interaction, apoptosis and intra-cellular signalling. In particular, miR-206 and miR-532-3p were associated with distinct biological processes via the expression of their target mRNAs: Reduced expression of miR-206 in progressive disease correlated with the upregulation of target mRNAs participating in inflammatory pathways (CCL19, CXCL1, IFNAR2, NCK2, PTK2B, PTPRC, RASGRP1 and TNFRSF25). Progressive cases also showed a lower expression of miR-532-3p and an increased expression of target transcripts involved in apoptosis pathways (MAP3K14, TNFRSF10B/TRAIL-R2, TRADD and TRAF2). In the validation cohort, we confirmed the decreased expression of miR-206 and miR-532-3p, and the inverse correlation of these miRNAs with the expression of nine of the 12 target genes. The levels of the identified miRNAs and the target mRNAs correlated with clinical parameters and histological damage indices. CONCLUSIONS: These results suggest the involvement of specific miRNAs and mRNAs in biological pathways associated with the progression of CKD.


Assuntos
Rim/metabolismo , MicroRNAs/metabolismo , RNA Mensageiro/metabolismo , Insuficiência Renal Crônica/genética , Adulto , Idoso , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/genética , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/metabolismo , Estudos de Coortes , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/metabolismo , Regulação para Baixo , Feminino , Perfilação da Expressão Gênica , Glomerulonefrite por IGA/genética , Glomerulonefrite por IGA/metabolismo , Glomerulonefrite Membranoproliferativa/genética , Glomerulonefrite Membranoproliferativa/metabolismo , Glomerulonefrite Membranosa/genética , Glomerulonefrite Membranosa/metabolismo , Glomerulosclerose Segmentar e Focal/genética , Glomerulosclerose Segmentar e Focal/metabolismo , Humanos , Nefrite Lúpica/genética , Nefrite Lúpica/metabolismo , Masculino , Pessoa de Meia-Idade , Nefroesclerose/genética , Nefroesclerose/metabolismo , Nefrose Lipoide/genética , Nefrose Lipoide/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Insuficiência Renal Crônica/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcriptoma , Regulação para Cima , Adulto Jovem
14.
Nitric Oxide ; 54: 8-14, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26805578

RESUMO

Scavenging of nitric oxide (NO) often interferes with studies on NO signaling in cell-free preparations. We observed that formation of cGMP by NO-stimulated purified soluble guanylate cyclase (sGC) was virtually abolished in the presence of cytosolic preparations of porcine coronary arteries, with the scavenging activity localized in the tunica media (smooth muscle layer). Electrochemical measurement of NO release from a donor compound and light absorbance spectroscopy showed that cytosolic preparations contained a reduced heme protein that scavenged NO. This protein, which reacted with anti-human hemoglobin antibodies, was efficiently removed from the preparations by haptoglobin affinity chromatography. The cleared cytosols showed only minor scavenging of NO according to electrochemical measurements and did not decrease cGMP formation by NO-stimulated sGC. In contrast, the column flow-through caused a nearly 2-fold increase of maximal sGC activity (from 33.1 ± 1.6 to 54.9 ± 2.2 µmol × min(-1) × mg(-1)). The proteins retained on the affinity column were identified as hemoglobin α and ß subunits. The results indicate that hemoglobin, presumably derived from vasa vasorum erythrocytes, is present and scavenges NO in preparations of porcine coronary artery smooth muscle. Selective removal of hemoglobin-mediated scavenging unmasked stimulation of maximal NO-stimulated sGC activity by a soluble factor expressed in vascular tissue.


Assuntos
Vasos Coronários/metabolismo , Hemoglobinas/metabolismo , Óxido Nítrico/metabolismo , Túnica Média/metabolismo , Animais , Bovinos , GMP Cíclico/metabolismo , Citoglobina , Globinas/metabolismo , Haptoglobinas/metabolismo , Humanos , Técnicas In Vitro , Guanilil Ciclase Solúvel/metabolismo , Suínos
16.
Biochim Biophys Acta ; 1841(6): 906-17, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24657704

RESUMO

Systemic knockout of adipose triglyceride lipase (ATGL), the pivotal enzyme of triglyceride lipolysis, results in a murine phenotype that is characterized by progredient cardiac steatosis and severe heart failure. Since cardiac and vascular dysfunction have been closely related in numerous studies we investigated endothelium-dependent and -independent vessel function of ATGL knockout mice. Aortic relaxation studies and Langendorff perfusion experiments of isolated hearts showed that ATGL knockout mice suffer from pronounced micro- and macrovascular endothelial dysfunction. Experiments with agonists directly targeting vascular smooth muscle cells revealed the functional integrity of the smooth muscle cell layer. Loss of vascular reactivity was restored ~50% upon treatment of ATGL knockout mice with the PPARα agonist Wy14,643, indicating that this phenomenon is partly a consequence of impaired cardiac contractility. Biochemical analysis revealed that aortic endothelial NO synthase expression and activity were significantly reduced in ATGL deficiency. Enzyme activity was fully restored in ATGL mice treated with the PPARα agonist. Biochemical analysis of perivascular adipose tissue demonstrated that ATGL knockout mice suffer from perivascular inflammatory oxidative stress which occurs independent of cardiac dysfunction and might contribute to vascular defects. Our results reveal a hitherto unrecognized link between disturbed lipid metabolism, obesity and cardiovascular disease.


Assuntos
Insuficiência Cardíaca/patologia , Lipase/genética , Metabolismo dos Lipídeos/genética , Obesidade/genética , Triglicerídeos/metabolismo , Tecido Adiposo/enzimologia , Tecido Adiposo/metabolismo , Animais , Modelos Animais de Doenças , Regulação Enzimológica da Expressão Gênica , Insuficiência Cardíaca/enzimologia , Humanos , Lipase/biossíntese , Lipase/metabolismo , Camundongos , Camundongos Knockout , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Óxido Nítrico Sintase/biossíntese , Obesidade/enzimologia , Obesidade/patologia , Técnicas de Cultura de Órgãos , Estresse Oxidativo , PPAR alfa/genética , PPAR alfa/metabolismo
17.
EMBO J ; 30(14): 2868-80, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21666598

RESUMO

Mammalian centromeric cohesin is protected from phosphorylation-dependent displacement in mitotic prophase by shugoshin-1 (Sgo1), while shugoshin-2 (Sgo2) protects cohesin from separase-dependent cleavage in meiosis I. In higher eukaryotes, progression and faithful execution of both mitosis and meiosis are controlled by the spindle assembly checkpoint, which delays anaphase onset until chromosomes have achieved proper attachment to microtubules. According to the so-called template model, Mad1-Mad2 complexes at unattached kinetochores instruct conformational change of soluble Mad2, thus catalysing Mad2 binding to its target Cdc20. Here, we show that human Sgo2, but not Sgo1, specifically interacts with Mad2 in a manner that strongly resembles the interactions of Mad2 with Mad1 or Cdc20. Sgo2 contains a Mad1/Cdc20-like Mad2-interaction motif and competes with Mad1 and Cdc20 for binding to Mad2. NMR and biochemical analyses show that shugoshin binding induces similar conformational changes in Mad2 as do Mad1 or Cdc20. Mad2 binding regulates fine-tuning of Sgo2's sub-centromeric localization. Mad2 binding is conserved in the only known Xenopus laevis shugoshin homologue and, compatible with a putative meiotic function, the interaction occurs in oocytes.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Meiose/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Proteínas de Ligação ao Cálcio/genética , Proteínas Cdc20 , Proteínas de Ciclo Celular/genética , Células Cultivadas , Centrômero/fisiologia , Feminino , Humanos , Cinetocoros/fisiologia , Proteínas Mad2 , Dados de Sequência Molecular , Proteínas Nucleares/genética , Oócitos/citologia , Oócitos/metabolismo , Conformação Proteica , Proteínas Repressoras/genética , Homologia de Sequência de Aminoácidos , Fuso Acromático , Xenopus laevis/metabolismo
18.
Nephrol Dial Transplant ; 30(6): 910-7, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25170095

RESUMO

The discovery of novel classes of non-coding RNAs (ncRNAs) has revolutionized medicine. Long thought to be a mere cellular housekeeper, surprising functions have recently been uncovered. MicroRNAs (miRNAs), are a representative of the class of short ncRNAs, play a fundamental role in the control of DNA and protein biosynthesis and activity as well as pathology. Currently, miRNAs are being investigated as diagnostic and prognostic markers and potential therapeutic targets in kidney transplantation for such indolent processes as ischaemia-reperfusion injury, humoral rejection or viral infections. It is realistic to believe that monitoring of renal allograft recipients in the future will include genome-wide miRNA profiling of biological fluids. Based on these individual profiles, an informed decision on therapeutic consequences will be possible. A first success with a specific suppression of miRNAs by antisense oligonucleotides was achieved in experimental studies of reperfusion injury and humoral rejection. Proof of this concept in men comes from studies in such indolent viral infections as Ebola and hepatitis C, where anti-miR therapy led to sustained viral clearance. In this review, we summarize the basis of the recent ncRNA revolution and its implication for kidney transplantation.


Assuntos
Biomarcadores/metabolismo , Nefropatias/genética , Transplante de Rim , MicroRNAs/genética , Humanos , Nefropatias/diagnóstico , Nefropatias/terapia
19.
Nephrol Dial Transplant ; 30 Suppl 4: iv105-112, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26209732

RESUMO

Diabetic kidney disease (DKD) is a complex, multifactorial disease and is associated with a high risk of renal and cardiovascular morbidity and mortality. Clinical practice guidelines for diabetes recommend essentially identical treatments for all patients without taking into account how the individual responds to the instituted therapy. Yet, individuals vary widely in how they respond to medications and therefore optimal therapy differs between individuals. Understanding the underlying molecular mechanisms of variability in drug response will help tailor optimal therapy. Polymorphisms in genes related to drug pharmacokinetics have been used to explore mechanisms of response variability in DKD, but with limited success. The complex interaction between genetic make-up and environmental factors on the abundance of proteins and metabolites renders pharmacogenomics alone insufficient to fully capture response variability. A complementary approach is to attribute drug response variability to individual variability in underlying molecular mechanisms involved in the progression of disease. The interplay of different processes (e.g. inflammation, fibrosis, angiogenesis, oxidative stress) appears to drive disease progression, but the individual contribution of each process varies. Drugs at the other hand address specific targets and thereby interfere in certain disease-associated processes. At this level, biomarkers may help to gain insight into which specific pathophysiological processes are involved in an individual followed by a rational assessment whether a specific drug's mode of action indeed targets the relevant process at hand. This article describes the conceptual background and data-driven workflow developed by the SysKid consortium aimed at improving characterization of the molecular mechanisms underlying DKD at the interference of the molecular impact of individual drugs in order to tailor optimal therapy to individual patients.


Assuntos
Biomarcadores/análise , Nefropatias Diabéticas/prevenção & controle , Tratamento Farmacológico/métodos , Variação Genética/genética , Farmacogenética , Medicina de Precisão , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/genética , Progressão da Doença , Genômica , Humanos
20.
Nephrol Dial Transplant ; 30 Suppl 4: iv86-95, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26209743

RESUMO

Diabetic kidney disease occurs in ∼ 25-40% of patients with type 2 diabetes. Given the high risk of progressive renal function loss and end-stage renal disease, early identification of patients with a renal risk is important. Novel biomarkers may aid in improving renal risk stratification. In this review, we first focus on the classical panel of albuminuria and estimated glomerular filtration rate as the primary clinical predictors of renal disease and then move our attention to novel biomarkers, primarily concentrating on assay-based multiple/panel biomarkers, proteomics biomarkers and metabolomics biomarkers. We focus on multiple biomarker panels since the molecular processes of renal disease progression in type 2 diabetes are heterogeneous, rendering it unlikely that a single biomarker significantly adds to clinical risk prediction. A limited number of prospective studies of multiple biomarkers address the predictive performance of novel biomarker panels in addition to the classical panel in type 2 diabetes. However, the prospective studies conducted so far have small sample sizes, are insufficiently powered and lack external validation. Adequately sized validation studies of multiple biomarker panels are thus required. There is also a paucity of studies that assess the effect of treatments on novel biomarker panels and determine whether initial treatment-induced changes in novel biomarkers predict changes in long-term renal outcomes. Such studies can not only improve our healthcare but also our understanding of the mechanisms of actions of existing and novel drugs and may yield biomarkers that can be used to monitor drug response. We conclude that this will be an area to focus research on in the future.


Assuntos
Biomarcadores/metabolismo , Diabetes Mellitus Tipo 2/diagnóstico , Insuficiência Renal Crônica/diagnóstico , Diabetes Mellitus Tipo 2/metabolismo , Progressão da Doença , Humanos , Prognóstico , Insuficiência Renal Crônica/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA