Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Am Heart Assoc ; 7(15)2018 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-30021807

RESUMO

BACKGROUND: The proximal aorta normally functions as a critical shock absorber that protects small downstream vessels from damage by pressure and flow pulsatility generated by the heart during systole. This shock absorber function is impaired with age because of aortic stiffening. METHODS AND RESULTS: We examined the contribution of common genetic variation to aortic stiffness in humans by interrogating results from the AortaGen Consortium genome-wide association study of carotid-femoral pulse wave velocity. Common genetic variation in the N-WASP (WASL) locus is associated with carotid-femoral pulse wave velocity (rs600420, P=0.0051). Thus, we tested the hypothesis that decoy proteins designed to disrupt the interaction of cytoskeletal proteins such as N-WASP with its binding partners in the vascular smooth muscle cytoskeleton could decrease ex vivo stiffness of aortas from a mouse model of aging. A synthetic decoy peptide construct of N-WASP significantly reduced activated stiffness in ex vivo aortas of aged mice. Two other cytoskeletal constructs targeted to VASP and talin-vinculin interfaces similarly decreased aging-induced ex vivo active stiffness by on-target specific actions. Furthermore, packaging these decoy peptides into microbubbles enables the peptides to be ultrasound-targeted to the wall of the proximal aorta to attenuate ex vivo active stiffness. CONCLUSIONS: We conclude that decoy peptides targeted to vascular smooth muscle cytoskeletal protein-protein interfaces and microbubble packaged can decrease aortic stiffness ex vivo. Our results provide proof of concept at the ex vivo level that decoy peptides targeted to cytoskeletal protein-protein interfaces may lead to substantive dynamic modulation of aortic stiffness.


Assuntos
Envelhecimento , Aorta Torácica/fisiopatologia , Proteínas do Citoesqueleto/genética , Hipertensão/fisiopatologia , Músculo Liso Vascular/fisiopatologia , Polimorfismo de Nucleotídeo Único , Rigidez Vascular/fisiologia , Animais , Aorta Torácica/metabolismo , Aorta Torácica/patologia , Pressão Sanguínea , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , DNA/genética , Estudo de Associação Genômica Ampla/métodos , Humanos , Hipertensão/genética , Hipertensão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/patologia , Análise de Onda de Pulso
2.
Pharmacol Rev ; 68(2): 476-532, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27037223

RESUMO

The smooth muscle cell directly drives the contraction of the vascular wall and hence regulates the size of the blood vessel lumen. We review here the current understanding of the molecular mechanisms by which agonists, therapeutics, and diseases regulate contractility of the vascular smooth muscle cell and we place this within the context of whole body function. We also discuss the implications for personalized medicine and highlight specific potential target molecules that may provide opportunities for the future development of new therapeutics to regulate vascular function.


Assuntos
Contração Muscular/fisiologia , Músculo Liso Vascular/fisiologia , Doenças Vasculares/tratamento farmacológico , Animais , Pressão Sanguínea/fisiologia , Cálcio/fisiologia , Citoesqueleto/fisiologia , Epigenômica , Humanos , Músculo Liso Vascular/metabolismo , Miosinas/metabolismo , Doenças Vasculares/genética , Doenças Vasculares/metabolismo , Doenças Vasculares/fisiopatologia
3.
J Am Heart Assoc ; 4(9): e002384, 2015 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-26376991

RESUMO

BACKGROUND: Sirtuin-1 (SirT1), a nicotinamide adenine dinucleotide(+)-dependent deacetylase, is a key enzyme in the cellular response to metabolic, inflammatory, and oxidative stresses; however, the role of endogenous SirT1 in the vasculature has not been fully elucidated. Our goal was to evaluate the role of vascular smooth muscle SirT1 in the physiological response of the aortic wall to angiotensin II, a potent hypertrophic, oxidant, and inflammatory stimulus. METHODS AND RESULTS: Mice lacking SirT1 in vascular smooth muscle (ie, smooth muscle SirT1 knockout) had drastically high mortality (70%) caused by aortic dissection after angiotensin II infusion (1 mg/kg per day) but not after an equipotent dose of norepinephrine, despite comparable blood pressure increases. Smooth muscle SirT1 knockout mice did not show any abnormal aortic morphology or blood pressure compared with wild-type littermates. Nonetheless, in response to angiotensin II, aortas from smooth muscle SirT1 knockout mice had severely disorganized elastic lamellae with frequent elastin breaks, increased oxidant production, and aortic stiffness compared with angiotensin II-treated wild-type mice. Matrix metalloproteinase expression and activity were increased in the aortas of angiotensin II-treated smooth muscle SirT1 knockout mice and were prevented in mice overexpressing SirT1 in vascular smooth muscle or with use of the oxidant scavenger tempol. CONCLUSIONS: Endogenous SirT1 in aortic smooth muscle is required to maintain the structural integrity of the aortic wall in response to oxidant and inflammatory stimuli, at least in part, by suppressing oxidant-induced matrix metalloproteinase activity. SirT1 activators could potentially be a novel therapeutic approach to prevent aortic dissection and rupture in patients at risk, such as those with hypertension or genetic disorders, such as Marfan's syndrome.


Assuntos
Angiotensina II , Aneurisma Aórtico/prevenção & controle , Dissecção Aórtica/prevenção & controle , Hipertensão/enzimologia , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Sirtuína 1/metabolismo , Dissecção Aórtica/induzido quimicamente , Dissecção Aórtica/enzimologia , Dissecção Aórtica/genética , Dissecção Aórtica/patologia , Animais , Aorta Torácica/enzimologia , Aorta Torácica/patologia , Aneurisma Aórtico/induzido quimicamente , Aneurisma Aórtico/enzimologia , Aneurisma Aórtico/genética , Aneurisma Aórtico/patologia , Células Cultivadas , Óxidos N-Cíclicos/farmacologia , Modelos Animais de Doenças , Tecido Elástico/metabolismo , Tecido Elástico/patologia , Elastina/metabolismo , Sequestradores de Radicais Livres/farmacologia , Hipertensão/induzido quimicamente , Hipertensão/genética , Metaloproteinases da Matriz/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Sirtuína 1/deficiência , Sirtuína 1/genética , Marcadores de Spin , Fatores de Tempo
4.
J Physiol ; 593(17): 3929-41, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26096914

RESUMO

Most cardiovascular research focuses on arterial mechanisms of disease, largely ignoring venous mechanisms. Here we examine ex vivo venous stiffness, spanning tissue to molecular levels, using biomechanics and magnetic microneedle technology, and show for the first time that venous stiffness is regulated by a molecular actin switch within the vascular smooth muscle cell in the wall of the vein. This switch connects the contractile apparatus within the cell to adhesion structures and facilitates stiffening of the vessel wall, regulating blood flow return to the heart. These studies also demonstrate that passive stiffness, the component of total stiffness not attributable to vascular smooth muscle activation, is severalfold lower in venous tissue than in arterial tissue. We show here that the activity of the smooth muscle cells plays a dominant role in determining total venous stiffness and regulating venous return. The literature on arterial mechanics is extensive, but far less is known about mechanisms controlling mechanical properties of veins. We use here a multi-scale approach to identify subcellular sources of venous stiffness. Portal vein tissue displays a severalfold decrease in passive stiffness compared to aortic tissues. The α-adrenergic agonist phenylephrine (PE) increased tissue stress and stiffness, both attenuated by cytochalasin D (CytoD) and PP2, inhibitors of actin polymerization and Src activity, respectively. We quantify, for the first time, cortical cellular stiffness in freshly isolated contractile vascular smooth muscle cells using magnetic microneedle technology. Cortical stiffness is significantly increased by PE and CytoD inhibits this increase but, surprisingly, PP2 does not. No detectable change in focal adhesion size, measured by immunofluorescence of FAK and zyxin, accompanies the PE-induced changes in cortical stiffness. Probing with phospho-specific antibodies confirmed activation of FAK/Src and ERK pathways and caldesmon phosphorylation. Thus, venous tissue stiffness is regulated both at the level of the smooth muscle cell cortex, via cortical actin polymerization, and by downstream smooth muscle effectors of Src/ERK signalling pathways. These findings identify novel potential molecular targets for the modulation of venous capacitance and venous return in health and disease.


Assuntos
Actinas/fisiologia , Adesões Focais/fisiologia , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/fisiologia , Veia Porta/fisiologia , Animais , Fenômenos Biomecânicos , Furões , Técnicas In Vitro , Masculino , Contração Muscular/fisiologia , Músculo Liso Vascular/citologia , Quinases da Família src/fisiologia
5.
Am J Physiol Heart Circ Physiol ; 307(8): H1252-61, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25128168

RESUMO

Increased aortic stiffness is an early and independent biomarker of cardiovascular disease. Here we tested the hypothesis that vascular smooth muscle cells (VSMCs) contribute significantly to aortic stiffness and investigated the mechanisms involved. The relative contributions of VSMCs, focal adhesions (FAs), and matrix to stiffness in mouse aorta preparations at optimal length and with confirmed VSMC viability were separated by the use of small-molecule inhibitors and activators. Using biomechanical methods designed for minimal perturbation of cellular function, we directly quantified changes with aging in aortic material stiffness. An alpha adrenoceptor agonist, in the presence of N(G)-nitro-l-arginine methyl ester (l-NAME) to remove interference of endothelial nitric oxide, increases stiffness by 90-200% from baseline in both young and old mice. Interestingly, increases are robustly suppressed by the Src kinase inhibitor PP2 in young but not old mice. Phosphotyrosine screening revealed, with aging, a biochemical signature of markedly impaired agonist-induced FA remodeling previously associated with Src signaling. Protein expression measurement confirmed a decrease in Src expression with aging. Thus we report here an additive model for the in vitro biomechanical components of the mouse aortic wall in which 1) VSMCs are a surprisingly large component of aortic stiffness at physiological lengths and 2) regulation of the VSMC component through FA signaling and hence plasticity is impaired with aging, diminishing the aorta's normal shock absorption function in response to stressors.


Assuntos
Envelhecimento , Aorta/fisiologia , Adesões Focais/metabolismo , Miócitos de Músculo Liso/fisiologia , Estresse Mecânico , Rigidez Vascular , Agonistas Adrenérgicos/farmacologia , Animais , Aorta/citologia , Aorta/crescimento & desenvolvimento , Aorta/metabolismo , Inibidores Enzimáticos/farmacologia , Hemodinâmica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , Fenilefrina/farmacologia , Pirimidinas/farmacologia , Quinases da Família src/metabolismo
6.
PLoS One ; 8(4): e62461, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23626821

RESUMO

Increased aortic stiffness is an acknowledged predictor and cause of cardiovascular disease. The sources and mechanisms of vascular stiffness are not well understood, although the extracellular matrix (ECM) has been assumed to be a major component. We tested here the hypothesis that the focal adhesions (FAs) connecting the cortical cytoskeleton of vascular smooth muscle cells (VSMCs) to the matrix in the aortic wall are a component of aortic stiffness and that this component is dynamically regulated. First, we examined a model system in which magnetic tweezers could be used to monitor cellular cortical stiffness, serum-starved A7r5 aortic smooth muscle cells. Lysophosphatidic acid (LPA), an activator of myosin that increases cell contractility, increased cortical stiffness. A small molecule inhibitor of Src-dependent FA recycling, PP2, was found to significantly inhibit LPA-induced increases in cortical stiffness, as well as tension-induced increases in FA size. To directly test the applicability of these results to force and stiffness development at the level of vascular tissue, we monitored mouse aorta ring stiffness with small sinusoidal length oscillations during agonist-induced contraction. The alpha-agonist phenylephrine, which also increases myosin activation and contractility, increased tissue stress and stiffness in a PP2- and FAK inhibitor 14-attenuated manner. Subsequent phosphotyrosine screening and follow-up with phosphosite-specific antibodies confirmed that the effects of PP2 and FAK inhibitor 14 in vascular tissue involve FA proteins, including FAK, CAS, and paxillin. Thus, in the present study we identify, for the first time, the FA of the VSMC, in particular the FAK-Src signaling complex, as a significant subcellular regulator of aortic stiffness and stress.


Assuntos
Aorta/metabolismo , Matriz Extracelular/metabolismo , Adesões Focais , Músculo Liso Vascular/metabolismo , Rigidez Vascular , Animais , Aorta/efeitos dos fármacos , Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Adesões Focais/efeitos dos fármacos , Técnicas In Vitro , Lisofosfolipídeos/farmacologia , Contração Muscular , Músculo Liso Vascular/efeitos dos fármacos , Quinase de Cadeia Leve de Miosina/antagonistas & inibidores , Quinase de Cadeia Leve de Miosina/metabolismo , Fosforilação , Pirimidinas/farmacologia , Ratos , Rigidez Vascular/efeitos dos fármacos , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
7.
J Comp Neurol ; 520(13): 2888-902, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22315181

RESUMO

The general organization of neocortical connectivity in rhesus monkey is relatively well understood. However, mounting evidence points to an organizing principle that involves clustered synapses at the level of individual dendrites. Several synaptic plasticity studies have reported cooperative interaction between neighboring synapses on a given dendritic branch, which may potentially induce synapse clusters. Additionally, theoretical models have predicted that such cooperativity is advantageous, in that it greatly enhances a neuron's computational repertoire. However, largely because of the lack of sufficient morphologic data, the existence of clustered synapses in neurons on a global scale has never been established. The majority of excitatory synapses are found within dendritic spines. In this study, we demonstrate that spine clusters do exist on pyramidal neurons by analyzing the three-dimensional locations of ∼40,000 spines on 280 apical dendritic branches in layer III of the rhesus monkey prefrontal cortex. By using clustering algorithms and Monte Carlo simulations, we quantify the probability that the observed extent of clustering does not occur randomly. This provides a measure that tests for spine clustering on a global scale, whenever high-resolution morphologic data are available. Here we demonstrate that spine clusters occur significantly more frequently than expected by pure chance and that spine clustering is concentrated in apical terminal branches. These findings indicate that spine clustering is driven by systematic biological processes. We also found that mushroom-shaped and stubby spines are predominant in clusters on dendritic segments that display prolific clustering, independently supporting a causal link between spine morphology and synaptic clustering.


Assuntos
Espinhas Dendríticas/ultraestrutura , Neocórtex/ultraestrutura , Células Piramidais/ultraestrutura , Algoritmos , Animais , Análise por Conglomerados , Espinhas Dendríticas/fisiologia , Processamento de Imagem Assistida por Computador , Imageamento Tridimensional/métodos , Macaca mulatta , Masculino , Microscopia Confocal , Neocórtex/fisiologia , Células Piramidais/fisiologia
8.
J Surg Res ; 136(1): 70-7, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17007881

RESUMO

BACKGROUND: The management of severe hepatic trauma frequently involves exposing the liver to varying periods of warm ischemia. The ischemic tolerance of the liver, in the setting of hemorrhagic shock (HS) and trauma, is presently unknown. We tested the hypothesis that warm ischemic tolerance of the porcine liver will be decreased following resuscitation from HS. MATERIALS AND METHODS: Twenty-three Yorkshire pigs were divided into three groups: 1) hepatic ischemia alone (HI, n = 9); 2) hemorrhagic shock alone (HS, n = 3); and 3) hemorrhagic shock plus hepatic ischemia combined (HSHI, n = 11). Following reperfusion, a liver biopsy was obtained and serial blood chemistries were sampled. RESULTS: Post-operative day 7 mortality was increased in the HSHI group (7/11) compared to the HI (0/9) group, P = 0.038. Notably, deaths did not result from acute liver failure, but rather from intra-operative hemodynamic collapse shortly following hepatic reperfusion. In addition, the HSHI group experienced significantly elevated lactic acid, serum creatinine and liver enzyme levels. Analysis of the liver biopsy samples is consistent with a more severe liver injury in the HSHI group. CONCLUSIONS: The warm ischemic tolerance of the liver following resuscitation from HS is significantly decreased in this porcine model compared to HS or HI alone. Mortality was associated with acute intra-operative hemodynamic collapse occurring shortly after hepatic reperfusion.


Assuntos
Isquemia/patologia , Fígado/patologia , Choque Hemorrágico/patologia , Doença Aguda , Reação de Fase Aguda/fisiopatologia , Animais , Aspartato Aminotransferases/metabolismo , Biópsia , Creatinina/sangue , Temperatura Alta , Isquemia/mortalidade , Isquemia/fisiopatologia , Rim/fisiologia , Ácido Láctico/sangue , Fígado/fisiopatologia , Circulação Hepática , Mecânica Respiratória , Ressuscitação , Choque Hemorrágico/mortalidade , Choque Hemorrágico/fisiopatologia , Sus scrofa , Equilíbrio Hidroeletrolítico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...