Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Endocrinol Metab ; 309(4): E320-33, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26081283

RESUMO

Feeding is regulated by perception in the hypothalamus, particularly the first-order arcuate nucleus (ARC) neurons, of the body's energy state. However, the cellular device for converting energy states to the activity of critical neurons in ARC is less defined. We here show that Na(+),K(+)-ATPase (NKA) in ARC senses energy states to regulate feeding. Fasting-induced systemic ghrelin rise and glucose lowering reduced ATP-hydrolyzing activity of NKA and its substrate ATP level, respectively, preferentially in ARC. Lowering glucose concentration (LG), which mimics fasting, decreased intracellular NAD(P)H and increased Na(+) concentration in single ARC neurons that subsequently exhibited [Ca(2+)]i responses to LG, showing that they were glucose-inhibited (GI) neurons. Third ventricular injection of the NKA inhibitor ouabain induced c-Fos expression in agouti-related protein (AgRP) neurons in ARC and evoked neuropeptide Y (NPY)-dependent feeding. When injected focally into ARC, ouabain stimulated feeding and mRNA expressions for NPY and AgRP. Ouabain increased [Ca(2+)]i in single NPY/AgRP neurons with greater amplitude than in proopiomelanocortin neurons in ARC. Conversely, the specific NKA activator SSA412 suppressed fasting-induced feeding and LG-induced [Ca(2+)]i increases in ARC GI neurons. NPY/AgRP neurons highly expressed NKAα3, whose knockdown impaired feeding behavior. These results demonstrate that fasting, via ghrelin rise and LG, suppresses NKA enzyme/pump activity in ARC and thereby promotes the activation of GI neurons and NPY/AgRP-dependent feeding. This study identifies ARC NKA as a hypothalamic sensor and converter of metabolic states to key neuronal activity and feeding behaviour, providing a new target to treat hyperphagic obesity and diabetes.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Metabolismo Energético/genética , Comportamento Alimentar/fisiologia , Glucose/farmacologia , Neurônios/efeitos dos fármacos , ATPase Trocadora de Sódio-Potássio/fisiologia , Trifosfato de Adenosina/metabolismo , Proteína Relacionada com Agouti/metabolismo , Animais , Comportamento Animal/fisiologia , Masculino , Neurônios/metabolismo , Neuropeptídeo Y/metabolismo , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , ATPase Trocadora de Sódio-Potássio/genética
2.
MAbs ; 6(3): 577-618, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24589717

RESUMO

The 24th Antibody Engineering & Therapeutics meeting brought together a broad range of participants who were updated on the latest advances in antibody research and development. Organized by IBC Life Sciences, the gathering is the annual meeting of The Antibody Society, which serves as the scientific sponsor. Preconference workshops on 3D modeling and delineation of clonal lineages were featured, and the conference included sessions on a wide variety of topics relevant to researchers, including systems biology; antibody deep sequencing and repertoires; the effects of antibody gene variation and usage on antibody response; directed evolution; knowledge-based design; antibodies in a complex environment; polyreactive antibodies and polyspecificity; the interface between antibody therapy and cellular immunity in cancer; antibodies in cardiometabolic medicine; antibody pharmacokinetics, distribution and off-target toxicity; optimizing antibody formats for immunotherapy; polyclonals, oligoclonals and bispecifics; antibody discovery platforms; and antibody-drug conjugates.


Assuntos
Anticorpos/química , Anticorpos/uso terapêutico , Animais , Anticorpos/genética , Evolução Molecular Direcionada , Desenho de Fármacos , Humanos , Imunoterapia , Neoplasias/terapia , Engenharia de Proteínas , Sociedades Científicas , Biologia de Sistemas
3.
J Diabetes Complications ; 27(1): 29-33, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22906552

RESUMO

BACKGROUND: Type-1 diabetes causes serious complications. Detailed molecular pathways of type-1 diabetes-mediated organ dysfunction are not completely understood. Significantly depressed (Na(+)+K(+))-ATPase (NKA) activity has been found in erythrocytes, pancreatic ß-cells, nerve cells, and muscle tissues of type-1 diabetic patients and rodent animal models. The characteristics of NKA in the development of the type-1 diabetes-mediated complications remain obscure. Here we investigated whether alterations of NKA activity in heart, kidney, and pancreas of type-1 diabetic mice occur simultaneously and whether depressed NKA activity is a universal phenomenon in major organs in the development of type-1 diabetes-induced complications. METHODS: Female non-obese diabetic (NOD) and non-obese resistant mice were used for the study. Mice blood glucose was monitored and ouabain-sensitive NKA activity was determined. RESULTS: Experimental results reveal that reduced NKA activity correlates with the progression of elevated blood glucose along with marked altered NKA apparent Na(+) affinity in all three organs of NOD mice. No significant changes of NKA protein expression were detected while the enzyme activity was reduced in tested mice, suggesting an inactive form of NKA might present in different tissues of the NOD mice. CONCLUSION: Our study suggests that concurrent impairment of NKA function in multi-organ may serve as one of the molecular pathways participating in and contributing to the mechanism of type-1 diabetes-induced complications in NOD mice. A successful protection of NKA function from injury might offer a good intervention for controlling the progression of the disease.


Assuntos
Diabetes Mellitus Tipo 1/enzimologia , Insuficiência de Múltiplos Órgãos/enzimologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Glicemia/análise , Glicemia/metabolismo , Diabetes Mellitus Tipo 1/complicações , Feminino , Rim/enzimologia , Camundongos , Camundongos Endogâmicos NOD , Insuficiência de Múltiplos Órgãos/etiologia , Miocárdio/enzimologia , Pâncreas/enzimologia , ATPase Trocadora de Sódio-Potássio/genética
4.
Biochem Biophys Res Commun ; 415(3): 479-84, 2011 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-22056556

RESUMO

(Na(+)+K(+))-ATPase (NKA) comprises two basic α and ß subunits: The larger α subunit catalyzes the hydrolysis of ATP for active transport of Na(+) and K(+) ions across the plasma membrane; the smaller ß subunit does not take part in the catalytic process of the enzyme. Little is known about allosteric regulation of the NKA ß subunit. Here, we report a surprising finding that extracellular stimuli on the native ß(1) subunit can generate a significant impact on the catalytic function of NKA. By using a ß(1) subunit-specific monoclonal antibody JY2948, we found that the JY2948-ß(1) subunit interaction markedly enhances the catalytic activity of the enzyme and increases the apparent affinity of Na(+) and K(+) ions for both ouabain-resistant rat NKA and ouabain-sensitive dog NKA. This study provides the first evidence to identify an allosteric binding site residing on the NKA ß(1) subunit and uncovers the latent allosteric property of the ß(1) subunit, which remotely controls the NKA catalytic function.


Assuntos
ATPase Trocadora de Sódio-Potássio/metabolismo , Regulação Alostérica , Animais , Cães , Humanos , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Ratos , Ratos Sprague-Dawley , ATPase Trocadora de Sódio-Potássio/química
5.
Biochem Biophys Res Commun ; 406(2): 200-3, 2011 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-21303662

RESUMO

(Na(+)+K(+))-ATPase (NKA) mediates positive inotropy in the heart. Extensive studies have demonstrated that the reverse-mode Na(+)/Ca(2+)-exchanger (NCX) plays a critical role in increasing intracellular Ca(2+) concentration through the inhibition of NKA-induced positive inotropy by cardiac glycosides. Little is known about the nature of the NCX functional mode in the activation of NKA-induced positive inotropy. Here, we examined the effect of an NKA activator SSA412 antibody on (45)Ca influx in isolated rat myocytes and found that KB-R7943, a NCX reverse-mode inhibitor, fails to inhibit the activation of NKA-induced (45)Ca influx, suggesting that the Ca(2+) influx via the reverse-mode NCX does not mediate this process. Nifedipine, an L-type Ca(2+) channel (LTCC) inhibitor, completely blocks the activation of NKA-induced (45)Ca influx, suggesting that the LTCC is responsible for the moderate increase in intracellular Ca(2+). In contrast, the inhibition of NKA by ouabain induces 4.7-fold (45)Ca influx compared with the condition of activation of NKA. Moreover, approximately 70% of ouabain-induced (45)Ca influx was obstructed by KB-R7943 and only 30% was impeded by nifedipine, indicating that both the LTCC and the NCX contribute to the rise in intracellular Ca(2+) and that the NCX reverse-mode is the major source for the (45)Ca influx induced by the inhibition of NKA. This study provides direct evidence to demonstrate that the activation of NKA-induced Ca(2+) increase is independent of the reverse-mode NCX and pinpoints a mechanistic distinction between the activation and inhibition of the NKA-mediated Ca(2+) influx path ways in cardiomyocytes.


Assuntos
Cálcio/metabolismo , Contração Miocárdica , Miócitos Cardíacos/enzimologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Células Cultivadas , Transporte de Íons , Miócitos Cardíacos/efeitos dos fármacos , Ratos , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Tioureia/análogos & derivados , Tioureia/farmacologia
6.
Biochem Biophys Res Commun ; 402(2): 319-23, 2010 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-20937253

RESUMO

Activation of (Na++K+)-ATPase (NKA) regulates cardiac L-type Ca2+ channel (LTCC) function through molecular crosstalk. The mechanism underlying NKA-LTCC crosstalk remains poorly understood. We have previously shown that activation of NKA leads to phosphorylation of LTCC α1 Ser1928. Here we investigated whether LTCC ß2 subunit is modulated by NKA activation and found that LTCC ß2 Ser496 is phosphorylated in response to activation of NKA. Src inhibitor PP1 and Erk1/2 inhibitor PD98059 abolish LTCC ß2 Ser496 phosphorylation, suggesting that NKA-mediated ß2 Ser496 phosphorylation is dependent of Src/Erk1/2 signaling pathway. Protein kinase G (PKG) inhibitor KT5823 failed to inhibit the phosphorylation of ß2 Ser496, indicating that the NKA-LTCC crosstalk is independent of PKG activity. The results of nifedipine sensitive 45Ca influx experiments suggest that phosphorylation of ß2 Ser496 may play a key down-regulation role in attenuating the accelerated activity of α1 subunit of the channel. Ouabain does not cause a phosphorylation on ß2 Ser496, indicating a fundamental difference between activation and inhibition of NKA-mediated biological processes. This study provides the first evidence to demonstrate that LTCC ß2 subunit is coupled with the movement of signals in the mechanism of activation of NKA-mediated crosstalk with LTCC.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Serina/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Canais de Cálcio Tipo L/genética , Carbazóis/farmacologia , Células Cultivadas , Ativação Enzimática , Humanos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Ratos , Serina/genética , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
7.
Mol Pharmacol ; 75(4): 774-81, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19122004

RESUMO

Cellular Ca(2+) signaling underlies diverse vital biological processes, including muscle contractility, memory encoding, fertilization, cell survival, and cell death. Despite extensive studies, the fundamental control mechanisms that regulate intracellular Ca(2+) movement remain enigmatic. We have found recently that activation of the (Na(+)+K(+))-ATPase markedly potentiates intracellular Ca(2+) transients and contractility of rat heart cells. Little is known about the pathway responsible for the activation of the (Na(+)+K(+))-ATPase-initiated Ca(2+) signaling. Here, we demonstrate a novel mechanism in which activation of the (Na(+)+K(+))-ATPase is coupled to increased L-type Ca(2+) channel function through a signaling cascade involving Src and ERK1/2 but not well established regulators of the channel, such as adrenergic receptor system or activation of PKA or CaMKII. We have also identified Ser(1928), a phosphorylation site for the alpha1 subunit of the L-type Ca(2+) channel that may participate in the activation of the (Na(+)+K(+))-ATPase-mediated Ca(2+) signaling. The findings reported here uncover a novel molecular cross-talk between activation of the (Na(+)+K(+))-ATPase and L-type Ca(2+) channel and provide new insights into Ca(2+) signaling mechanisms for deeper understanding of the nature of cellular Ca(2+) handling in heart.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Miócitos Cardíacos/enzimologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Sequência de Aminoácidos , Animais , Sinalização do Cálcio/fisiologia , Ativação Enzimática/fisiologia , Masculino , Dados de Sequência Molecular , Ratos , Ratos Sprague-Dawley , ATPase Trocadora de Sódio-Potássio/fisiologia
8.
Biochem Biophys Res Commun ; 377(2): 469-473, 2008 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-18848919

RESUMO

(Na(+)+K(+))-ATPase is a target receptor of digitalis (cardiac glycoside) drugs. It has been demonstrated that the H1-H2 domain of the alpha-subunit of the (Na(+)+K(+))-ATPase is one of the digitalis drug interaction sites of the enzyme. Despite the extensive studies of the inhibitory effect of digitalis on the (Na(+)+K(+))-ATPase, the functional property of the H1-H2 domain of the enzyme and its role in regulating enzyme activity is not completely understood. Here we report a surprise finding: instead of inhibiting the enzyme, binding of a specific monoclonal antibody SSA78 to the H1-H2 domain of the (Na(+)+K(+))-ATPase elevates the catalytic activity of the enzyme. In the presence of low concentration of ouabain, monoclonal antibody SSA78 significantly protects enzyme function against ouabain-induced inhibition. However, higher concentration of ouabain completely inactivates the (Na(+)+K(+))-ATPase even in the presence of SSA78. These results suggest that the H1-H2 domain of the (Na(+)+K(+))-ATPase is capable of regulating enzyme function in two distinct ways for both ouabain-sensitive and -resistant forms of the enzyme: it increases the activity of the (Na(+)+K(+))-ATPase during its interaction with an activator; it also participates in the mechanism of digitalis or ouabain-induced inhibition of the enzyme. Understanding the dual activity of the H1-H2 domain will help better understand the structure-function relationships of the (Na(+)+K(+))-ATPase and the biological processes mediated by the enzyme.


Assuntos
ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Antígenos/genética , Antígenos/imunologia , Antígenos/metabolismo , Cães , Inibidores Enzimáticos/farmacologia , Ouabaína/farmacologia , Estrutura Terciária de Proteína , Ratos , ATPase Trocadora de Sódio-Potássio/genética
9.
Biochem Biophys Res Commun ; 349(2): 582-7, 2006 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-16949050

RESUMO

OBJECTIVE: We have recently identified an activation site on (Na+ + K+)-ATPase and found that binding of antibody SSA412 to this specific site of the enzyme markedly augments (Na+ + K+)-ATPase catalytic activity. Demonstration of whether activation of (Na+ + K+)-ATPase affects heart function in animal in vivo was the object of this investigation. METHODS: Male wild-type CD-1 mouse and specific antibody SSA412 were used for the study. A pressure-volume micromanometer-conductance catheter in anesthetized mouse assessed in vivo cardiac functions. RESULTS: Specific antibody SSA412 infusion in mouse shifted pressure-volume loop leftward with increased stroke volume and enhanced end-systolic elastance. Global systolic parameters such as ejection fraction and cardiac output, and load independent contractile parameters including dP/dtmax/IP, PMX/EDV, Ees, and PRSW, were all increased without any effect on relaxation following administration of SSA412. Cardiac preload indexed by EDV and afterload by ESP did not alter, suggesting that SSA412-enhanced myocardial performance is a direct cardiac effect caused by the activation of (Na+ + K+)-ATPase. CONCLUSION: Our study provides the first in vivo physiological evidence to demonstrate that activation of (Na+ + K+)-ATPase induces significant positive inotropic effect in intact animal heart. The finding may lead to new therapeutic strategies for the treatment of heart failure.


Assuntos
Miocárdio/enzimologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Sequência de Aminoácidos , Animais , Ativação Enzimática , Masculino , Camundongos , Dados de Sequência Molecular , Células Musculares/patologia , Contração Miocárdica/efeitos dos fármacos , Miocárdio/patologia , Ratos , Ratos Sprague-Dawley , Homologia de Sequência de Aminoácidos , ATPase Trocadora de Sódio-Potássio/fisiologia
10.
Biochem Biophys Res Commun ; 338(4): 1669-77, 2005 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-16263081

RESUMO

Enzymes catalyze essential chemical reactions needed for living processes. (Na+ +K+)-ATPase (NKA) is one of the key enzymes that control intracellular ion homeostasis and regulate cardiac function. Little is known about activation of NKA and its biological impact. Here we show that native activity of NKA is markedly elevated when protein-protein interaction occurs at the extracellular DVEDSYGQQWTYEQR (D-R) region in the alpha-subunit of the enzyme. The apparent catalytic turnover of NKA is approximately twice as fast as the controls for both ouabain-resistant and ouabain-sensitive enzymes. Activation of NKA not only markedly protects enzyme function against denaturing, but also directly affects cellular activities by regulating intracellular Ca2+ transients and inducing a positive inotropic effect in isolated rat cardiac myocytes. Immunofluorescent labeling indicates that the D-R region of NKA is not a conventional digitalis-binding site. Our findings uncover a novel activation site of NKA that is capable of promoting the catalytic function of the enzyme and establish a new concept that activating of NKA mediates cardiac contraction.


Assuntos
ATPase Trocadora de Sódio-Potássio/metabolismo , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação de Anticorpos , Cães , Ativação Enzimática , Cinética , Dados de Sequência Molecular , Contração Miocárdica/fisiologia , Miócitos Cardíacos/fisiologia , Ouabaína/metabolismo , Coelhos , Ratos , Ratos Sprague-Dawley , Alinhamento de Sequência , ATPase Trocadora de Sódio-Potássio/imunologia
11.
Biochem Biophys Res Commun ; 336(4): 1190-3, 2005 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-16169521

RESUMO

Copper-zinc superoxide dismutase (CuZnSOD) specifically catalyzes the removal of superoxide radicals to protect cellular function against the generation of superoxide-dependent hydroxyl radicals ((.)OH). However, an unexpected observation reveals that denatured CuZnSOD (dCuZnSOD) itself induces (.)OH formation. This dCuZnSOD-dependent (.)OH generation was not inhibited by active CuZnSOD, suggesting that it is a superoxide-independent process. Sodium cyanide, histidine, and N,N'-diethyldithiocarbamate abolished (.)OH generation, implying that Cu may be responsible for dCuZnSOD-induced (.)OH formation. Catalase eliminated ()OH generation, suggesting that hydrogen peroxide may be involved in the mechanism of dCuZnSOD-mediated (.)OH production. Furthermore, nitric oxide ((.)NO) completely inhibited dCuZnSOD-induced (.)OH radical generation, indicating that (.)NO is an important (.)OH radical scavenger. Our results shed new light on the effect of dysfunctional CuZnSOD and suggest that structural disorder of the enzyme may be one of the endogenous pathways of toxic (.)OH formation in biological systems.


Assuntos
Cobre/química , Radical Hidroxila/química , Superóxido Dismutase/química , Zinco/química , Catalase/química , Ditiocarb/química , Espectroscopia de Ressonância de Spin Eletrônica , Histidina/química , Peróxido de Hidrogênio/química , Cianeto de Sódio/química
12.
FASEB J ; 19(1): 53-61, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15629895

RESUMO

(Na++K+)-ATPase (NKA) plays an important role in ion homeostasis and regulates cardiac contraction. To understand the molecular basis of its cardiac regulatory functions, we investigated whether the primary structure of the H1-H2 domain in alpha-1 (alpha1) subunit of the enzyme plays a role in myocardial contractile regulation. Here we show that site-specific binding to this 1 H1-H2 domain with a targeted antibody (SSA78) markedly augments intracellular Ca2+ transients and contraction of rat ventricular cardiomyocytes without inactivating NKA. In vivo SSA78 infusion in mice results in a positive inotropic effect with enhanced contractile function yet no change in relaxation, indicating a direct cardiac effect linked to the H1-H2 domain. Competitive immunofluorescent staining and flow cytometry reveal that SSA78 binding is antagonized by ouabain, supporting the interaction of SSA78 at one of the glycoside-effecter sites. These new findings suggest that the H1-H2 domain of 1 subunit of NKA is a critical determinant of enzyme biologic activity, which couples to enhanced myocyte calcium transient and inotropic action.


Assuntos
Contração Miocárdica/fisiologia , Peptídeos/fisiologia , ATPase Trocadora de Sódio-Potássio/fisiologia , Animais , Anticorpos/metabolismo , Anticorpos/farmacologia , Anticorpos/fisiologia , Especificidade de Anticorpos/fisiologia , Cálcio/metabolismo , Cardiotônicos/farmacologia , Cães , Coração/efeitos dos fármacos , Camundongos , Miócitos Cardíacos/química , Miócitos Cardíacos/metabolismo , Peptídeos/síntese química , Peptídeos/imunologia , Estrutura Terciária de Proteína/fisiologia , Coelhos , Ratos , ATPase Trocadora de Sódio-Potássio/química , ATPase Trocadora de Sódio-Potássio/imunologia
13.
J Biol Chem ; 278(43): 41798-803, 2003 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-12904295

RESUMO

Nitric oxide (NO.) generated from nitric oxide synthase (NOS) isoforms bound to cellular membranes may serve to modulate oxidative stresses in cardiac muscle and thereby regulate the function of key membrane-associated enzymes. Ischemia is known to inhibit the function of sarcolemmal enzymes, including the (Na+ + K+)-ATPase, but it is unknown whether concomitant injury to sarcolemma (SL)-associated NOS isoforms may contribute to this process by reducing the availability of locally generated NO. Here we report that nNOS, as well as eNOS (SL NOSs), are tightly associated with cardiac SL membranes in several different species. In isolated perfused rat hearts, global ischemia caused a time-dependent irreversible injury to cardiac SL NOSs and a disruption of SL NO. generation. Pretreatment with low concentrations of the NO. donor 1-hydroxy-2-oxo-3-(N-3-methyl-aminopropyl)-3-methyl-1-triazene (NOC-7) markedly protected both SL NOS and (Na+ + K+)-ATPase functions against ischemia-induced inactivation. Moreover, ischemia impaired SL Na+/K+ binding, and NOC-7 significantly prevented ischemic injury to the ion binding sites on (Na+ + K+)-ATPase. These novel findings indicate that NO. can protect cardiac SL NOSs and (Na+ + K+)-ATPase against ischemia-induced inactivation and suggest that locally generated NO. may serve to regulate SL Na+/K+ ion active transport in the heart.


Assuntos
Transporte de Íons , Isquemia/enzimologia , Miocárdio/ultraestrutura , Óxido Nítrico/farmacologia , Sarcolema/enzimologia , Animais , Transporte Biológico Ativo , Ativação Enzimática , Feminino , Técnicas In Vitro , Isquemia/metabolismo , Proteínas de Membrana/metabolismo , Miocárdio/enzimologia , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo I , Óxido Nítrico Sintase Tipo III , Substâncias Protetoras/farmacologia , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Sarcolema/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo
14.
Biochem Biophys Res Commun ; 294(5): 1030-5, 2002 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-12074580

RESUMO

Nitric oxide (NO*) is produced endogenously from NOS isoforms bound to sarcolemmal (SL) and sarcoplasmic reticulum (SR) membranes. To investigate whether locally generated NO* directly affects the activity of enzymes mediating ion active transport, we studied whether knockout of selected NOS isoforms would affect the functions of cardiac SL (Na+ + K+)-ATPase and SR Ca2+-ATPase. Cardiac SL and SR vesicles containing either SL (Na+ + K+)-ATPase or SR Ca2+-ATPase were isolated from mice lacking either nNOS or eNOS, or both, and tested for enzyme activities. Western blot analysis revealed that absence of single or double NOS isoforms did not interrupt the protein expression of SL (Na+ + K+)-ATPase and SR Ca2+-ATPase in cardiac muscle cells. However, lack of NOS isoforms in cardiac muscle significantly altered both (Na+ + K+)-ATPase activity and SR Ca2+-ATPase function. Our experimental results suggest that disrupted endogenous NO* production may change local redox conditions and lead to an unbalanced free radical homeostasis in cardiac muscle cells which, in turn, may affect key enzyme activities and membrane ion active transport systems in the heart.


Assuntos
Miocárdio/enzimologia , Miocárdio/metabolismo , Óxido Nítrico Sintase/fisiologia , Animais , Cálcio/metabolismo , ATPases Transportadoras de Cálcio/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica , Membranas Intracelulares/enzimologia , Transporte de Íons , Camundongos , Camundongos Knockout , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo I , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Ratos , Sarcolema/enzimologia , Retículo Sarcoplasmático/enzimologia , ATPase Trocadora de Sódio-Potássio/metabolismo
15.
Biochem Biophys Res Commun ; 291(1): 111-5, 2002 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11829469

RESUMO

Structural localization of a peptide region, KRQPRNPKTDKLVNE, in the catalytic subunit of (Na(+) + K(+))-ATPase was investigated using a specific antibody directed against this peptide in cultured African green monkey kidney CV-1 cells. Immunofluorescence staining of frozen cell sections shows that an anti-KRQPRNPKTDKLVNE antibody (SSA95) interacts with its antigenic site and binds to the extracellular side of the cell membrane. Indirect immunofluorescence and flow cytometric analyses confirmed the presence of this epitope on intact cell surfaces. These results suggest that the KRQPRNPKTDKLVNE region of the (Na(+) + K(+))-ATPase is expressed on the cellular membrane surface.


Assuntos
Domínio Catalítico/imunologia , Membrana Celular/imunologia , Epitopos/imunologia , Fragmentos de Peptídeos/imunologia , ATPase Trocadora de Sódio-Potássio/imunologia , Sequência de Aminoácidos , Animais , Anticorpos/isolamento & purificação , Anticorpos/metabolismo , Especificidade de Anticorpos/imunologia , Linhagem Celular , Membrana Celular/enzimologia , Chlorocebus aethiops , Citometria de Fluxo , Técnica Indireta de Fluorescência para Anticorpo , Rim/citologia , Rim/enzimologia , Dados de Sequência Molecular , Subunidades Proteicas , ATPase Trocadora de Sódio-Potássio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...