Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Eur J Appl Physiol ; 110(2): 395-404, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20512503

RESUMO

Emerging evidence suggests that nitric oxide (NO) plays a pivotal role in the mechanism of vascular hyporesponsiveness contributing to microgravity-induced orthostatic intolerance. The cellular and enzymatic source of the NO, however, remains controversial. In addition, the time course of the endothelial-dependent contribution remains unstudied. We tested the hypotheses that the change in vasoresponsiveness seen in acute (3-day) hindlimb unweighted (HLU) animals is due to an endothelium-dependent mechanism and that endothelial-dependent attenuation in vasoreactivity is due to endothelial nitric oxide synthase (NOS-3) dependent activation. Vasoreactivity was investigated in rat aortic rings following acute HLU treatment. Dose responsiveness to norepinepherine (NE) was depressed after 3-day HLU [1,338 +/- 54 vs. 2,325 +/- 58 mg at max (NE), HLU vs. C, P < 0.001]. However, removal of the endothelium restored the vascular contractility to that of C. In addition, 1H-oxadiazole quinoxalin-1-one (ODQ), a soluble guanylyl cyclase inhibitor, restored the reduced vasoconstrictor responses to phenylephrine (PE) seen in 3-day HLU rings (1.30 +/- 0.10 vs. 0.53 +/- 0.07 g, HLU + ODQ vs. HLU, P = 0.0001). Ca(+) dependent nitric oxide synthase (NOS) activity was increased, as was vascular NO products as a result of HLU. While NOS-3 expression was not increased in HLU rats, phosphorylation of NOS-3 at serine-1177 (an activator of NOS-3) was increased while phosphorylation of serine-495 (an inactivator of NOS-3) was decreased. These findings demonstrate that changes in vasoresponsiveness in the acute HLU model of microgravity are due to an upregulation of the endothelial-dependent NO/cGMP pathway through NOS phosphorylation.


Assuntos
Aorta/metabolismo , GMP Cíclico/metabolismo , Endotélio Vascular/metabolismo , Óxido Nítrico/metabolismo , Sistemas do Segundo Mensageiro , Vasoconstrição , Vasodilatação , Simulação de Ausência de Peso , Animais , Aorta/efeitos dos fármacos , Caveolina 1/metabolismo , Relação Dose-Resposta a Droga , Endotélio Vascular/efeitos dos fármacos , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Guanilato Ciclase/antagonistas & inibidores , Guanilato Ciclase/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Elevação dos Membros Posteriores , Masculino , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação , Ratos , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/metabolismo , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Serina , Guanilil Ciclase Solúvel , Fatores de Tempo , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
2.
Circ Res ; 107(1): 117-25, 2010 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-20489165

RESUMO

RATIONALE: Although an age-related decrease in NO bioavailability contributes to vascular stiffness, the underlying molecular mechanisms remain incompletely understood. We hypothesize that NO constrains the activity of the matrix crosslinking enzyme tissue transglutaminase (TG2) via S-nitrosylation in young vessels, a process that is reversed in aging. OBJECTIVE: We sought to determine whether endothelium-dependent NO regulates TG2 activity by S-nitrosylation and whether this contributes to age-related vascular stiffness. METHODS AND RESULTS: We first demonstrate that NO suppresses activity and increases S-nitrosylation of TG2 in cellular models. Next, we show that nitric oxide synthase (NOS) inhibition leads to increased surface and extracellular matrix-associated TG2. We then demonstrate that endothelium-derived bioactive NO primarily mediates its effects through TG2, using TG2(-/-) mice chronically treated with the NOS inhibitor l-N(G)-nitroarginine methyl ester (L-NAME). We confirm that TG2 activity is modulated by endothelium-derived bioactive NO in young rat aorta. In aging rat aorta, although TG2 expression remains unaltered, its activity increases and S-nitrosylation decreases. Furthermore, TG2 inhibition decreases vascular stiffness in aging rats. Finally, TG2 activity and matrix crosslinks are augmented with age in human aorta, whereas abundance remains unchanged. CONCLUSIONS: Decreased S-nitrosylation of TG2 and increased TG activity lead to enhanced matrix crosslinking and contribute to vascular stiffening in aging. TG2 appears to be the member of the transglutaminase family primarily contributing to this phenotype. Inhibition of TG2 could thus represent a therapeutic target for age-associated vascular stiffness and isolated systolic hypertension.


Assuntos
Envelhecimento/metabolismo , Endotélio Vascular/enzimologia , Proteínas de Ligação ao GTP/metabolismo , Óxido Nítrico/fisiologia , Transglutaminases/metabolismo , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/patologia , Animais , Células Cultivadas , Endotélio Vascular/patologia , Proteínas de Ligação ao GTP/antagonistas & inibidores , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Células NIH 3T3 , Óxido Nítrico/antagonistas & inibidores , Proteína 2 Glutamina gama-Glutamiltransferase , Ratos , Ratos Endogâmicos F344 , Transglutaminases/antagonistas & inibidores
3.
J Appl Physiol (1985) ; 106(6): 2002-8, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19299573

RESUMO

We have previously shown that microgravity and simulated microgravity induce an increase in human and rat aortic stiffness. We attempted to elucidate the mechanism(s) responsible for this increase in stiffness. We hypothesize that an alteration in vessel wall collagen or elastin content or in extracellular matrix (ECM) cross-linking either individually or in a combination is responsible for the increased vessel stiffness. Rats underwent hindlimb unweighting (HLU) for a period of 7 days to simulate microgravity. The contribution of ECM cross-linking to the vessel wall stiffness was evaluated by measuring aortic pulse wave velocity following inhibition of the cross-linking enzymes lysyl oxidase (LOX) and transglutaminase (tTG) and the nonenzymatic advanced glycation end product cross-linking pathway during HLU. Aortic collagen and elastin content was quantified using established colorimetric assays. Collagen subtype composition was determined via immunofluorescent staining. The increase in aortic pulse wave velocity after HLU was significantly attenuated in the LOX and tTG inhibition groups compared with saline (1.13 +/- 0.11 vs. 3.00 +/- 0.15 m/s, LOX vs. saline, P < 0.001; 1.16 +/- 0.25 vs. 3.00 +/- 0.15 m/s, tTG vs. saline, P < 0.001). Hydroxyproline content, a measure of collagen content, was increased in all groups after HLU (2.01 +/- 0.62 vs. 3.69 +/- 0.68% dry weight, non-HLU vs. HLU, P = 0.009). Collagen subtype composition and aortic elastin content were not altered by HLU. Together, these data indicate that HLU-induced increases in aortic stiffness are due to both increased aortic collagen content and enzyme cross-linking activity.


Assuntos
Aorta Torácica/patologia , Matriz Extracelular/metabolismo , Elevação dos Membros Posteriores/fisiologia , Simulação de Ausência de Peso/efeitos adversos , Animais , Aorta Torácica/metabolismo , Aorta Torácica/fisiopatologia , Velocidade do Fluxo Sanguíneo , Pressão Sanguínea , Colágeno/metabolismo , Elastina/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Hidroxiprolina/metabolismo , Masculino , Maleabilidade , Proteína-Lisina 6-Oxidase/antagonistas & inibidores , Fluxo Pulsátil , Ratos , Ratos Wistar , Transglutaminases/metabolismo
5.
J Appl Physiol (1985) ; 102(3): 853-8, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17082368

RESUMO

Microgravity (microG)-induced orthostatic intolerance (OI) in astronauts is characterized by a marked decrease in cardiac output (CO) in response to an orthostatic stress. Since CO is highly dependent on venous return, alterations in the resistance to venous return (RVR) may be important in contributing to OI. The RVR is directly dependent on arterial compliance (C(a)), where aortic compliance (C(ao)) contributes up to 60% of C(a). We tested the hypothesis that microG-induced changes in C(a) may represent a protective mechanism against OI. A retrospective analysis on hemodynamic data collected from astronauts after 5- to 18-day spaceflight missions revealed that orthostatically tolerant (OT) astronauts showed a significant decrease in C(a) after spaceflight, while OI astronauts showed a slight increase in C(a). A ground-based animal model simulating microG, hindlimb-unweighted rats, was used to explore this phenomenon. Two independent assessments of C(ao), in vivo pulse wave velocity (PWV) of the thoracic aorta and in vitro pressure-diameter squared relationship (PDSR) measurements of the excised thoracic aorta, were determined. PWV showed a significant increase in aortic stiffness compared with control, despite unchanged blood pressures. This increase in aortic stiffness was confirmed by the PDSR analysis. Thus both actual microG in humans and simulated microG in rats induces changes in C(ao). The difference in C(a) in OT and OI astronaut suggests that the microG-induced decrease in C(a) is a protective adaptation to spaceflight that reduces the RVR and allows for the maintenance of adequate CO in response to an orthostatic stress.


Assuntos
Adaptação Fisiológica , Aorta/fisiologia , Astronautas , Postura/fisiologia , Ausência de Peso/efeitos adversos , Animais , Artérias/fisiologia , Elevação dos Membros Posteriores , Humanos , Masculino , Pressão , Pulso Arterial , Ratos , Ratos Wistar
6.
BMC Neurosci ; 7: 58, 2006 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-16863593

RESUMO

BACKGROUND: Earlier observations in our lab had indicated that large, time-varying magnetic fields could elicit action potentials that travel in only one direction in at least some of the myelinated axons in peripheral nerves. The objective of this study was to collect quantitative evidence for magnetically induced unidirectional action potentials in peripheral nerves of human subjects. A magnetic coil was maneuvered to a location on the upper arm where physical effects consistent with the creation of unidirectional action potentials were observed. Electromyographic (EMG) and somatosensory evoked potential (SEP) recordings were then made from a total of 20 subjects during stimulation with the magnetic coil. RESULTS: The relative amplitudes of the EMG and SEP signals changed oppositely when the current direction in the magnetic coil was reversed. This effect was consistent with current direction in the coil relative to the arm for all subjects. CONCLUSION: A differential evocation of motor and sensory fibers was demonstrated and indicates that it may be possible to induce unidirectional action potentials in myelinated peripheral nerve fibers with magnetic stimulation.


Assuntos
Magnetismo/instrumentação , Neurônios Motores/fisiologia , Fibras Nervosas Mielinizadas/fisiologia , Vias Neurais/fisiologia , Neurônios Aferentes/fisiologia , Nervos Periféricos/fisiologia , Potenciais de Ação/fisiologia , Vias Aferentes/fisiologia , Vias Eferentes/fisiologia , Estimulação Elétrica/instrumentação , Estimulação Elétrica/métodos , Eletromiografia , Potenciais Somatossensoriais Evocados/fisiologia , Humanos , Mecanorreceptores/fisiologia , Córtex Motor/fisiologia , Contração Muscular/fisiologia , Músculo Esquelético/inervação , Músculo Esquelético/fisiologia , Condução Nervosa/fisiologia , Inibição Neural/fisiologia , Córtex Somatossensorial/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA