Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
Am J Physiol Heart Circ Physiol ; 306(7): H1011-7, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24531813

RESUMO

Removal of plasma proteins from perfusates increases vascular permeability. The common interpretation of the action of albumin is that it forms part of the permeability barrier by electrostatic binding to the endothelial glycocalyx. We tested the alternate hypothesis that removal of perfusate albumin in rat venular microvessels decreased the availability of sphingosine-1-phosphate (S1P), which is normally carried in plasma bound to albumin and lipoproteins and is required to maintain stable baseline endothelial barriers (Am J Physiol Heart Circ Physiol 303: H825-H834, 2012). Red blood cells (RBCs) are a primary source of S1P in the normal circulation. We compared apparent albumin permeability coefficients [solute permeability (Ps)] measured using perfusates containing albumin (10 mg/ml, control) and conditioned by 20-min exposure to rat RBCs with Ps when test perfusates were in RBC-conditioned protein-free Ringer solution. The control perfusate S1P concentration (439 ± 46 nM) was near the normal plasma value at 37 °C and established a stable baseline Ps (0.9 ± 0.4 × 10(-6) cm/s). Ringer solution perfusate contained 52 ± 8 nM S1P and increased Ps more than 10-fold (16.1 ± 3.9 × 10(-6) cm/s). Consistent with albumin-dependent transport of S1P from RBCs, S1P concentrations in RBC-conditioned solutions decreased as albumin concentration, hematocrit, and temperature decreased. Protein-free Ringer solution perfusates that used liposomes instead of RBCs as flow markers failed to maintain normal permeability, reproducing the "albumin effect" in these mammalian microvessels. We conclude that the albumin effect depends on the action of albumin to facilitate the release and transport of S1P from RBCs that normally provide a significant amount of S1P to the endothelium.


Assuntos
Permeabilidade Capilar , Eritrócitos/metabolismo , Lisofosfolipídeos/sangue , Microcirculação , Perfusão , Albumina Sérica/metabolismo , Esfingosina/análogos & derivados , Vênulas/fisiologia , Animais , Transporte Biológico , Hematócrito , Soluções Isotônicas/metabolismo , Lipossomos , Masculino , Ratos , Ratos Sprague-Dawley , Solução de Ringer , Esfingosina/sangue , Fatores de Tempo
2.
Am J Physiol Heart Circ Physiol ; 304(8): H1077-84, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23417864

RESUMO

Endothelial cells in a cultured monolayer change from a "cobblestone" configuration when grown under static conditions to a more elongated shape, aligned with the direction of flow, after exposure to sustained uniform shear stress. Sustained blood flow acts to protect regions of large arteries from injury. We tested the hypothesis that the stable permeability state of individually perfused microvessels is also characteristic of flow conditioning. In individually perfused rat mesenteric venular microvessels, microvascular permeability, measured as hydraulic conductivity (Lp), was stable [mean 1.0 × 10(-7) cm/(s × cmH2O)] and independent of shear stress (3-14 dyn/cm(2)) for up to 3 h. Vessels perfused opposite to the direction of normal blood flow exhibited a delayed Lp increase [ΔLp was 7.6 × 10(-7) cm/(s × cmH2O)], but the increase was independent of wall shear stress. Addition of chondroitin sulfate and hyaluronic acid to perfusates increased the shear stress range, but did not modify the asymmetry in response to flow direction. Increased Lp in reverse-perfused vessels was associated with numerous discontinuities of VE-cadherin and occludin, while both proteins were continuous around the periphery of forward-perfused vessels. The results are not consistent with a general mechanism for graded shear-dependent permeability increase, but they are consistent with the idea that a stable Lp under normal flow contributes to prevention of edema formation and also enables physiological regulation of shear-dependent small solute permeabilities (e.g., glucose). The responses during reverse flow are consistent with reports that disturbed flows result in a less stable endothelial barrier in venular microvessels.


Assuntos
Permeabilidade Capilar/fisiologia , Células Endoteliais/fisiologia , Hemorreologia/fisiologia , Microcirculação/fisiologia , Vênulas/fisiologia , Água/metabolismo , Animais , Antígenos CD/metabolismo , Caderinas/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Adesão Celular , Sulfatos de Condroitina/farmacologia , Células Endoteliais/efeitos dos fármacos , Glicocálix/efeitos dos fármacos , Glicocálix/fisiologia , Ácido Hialurônico/farmacologia , Masculino , Veias Mesentéricas/efeitos dos fármacos , Veias Mesentéricas/fisiologia , Microcirculação/efeitos dos fármacos , Ocludina/metabolismo , Ratos , Ratos Sprague-Dawley , Vênulas/efeitos dos fármacos , Viscossuplementos/farmacologia
3.
Am J Physiol Heart Circ Physiol ; 303(7): H825-34, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22865384

RESUMO

Exogenous sphingosine-1-phosphate (S1P), a lipid mediator in blood, attenuates acute microvascular permeability increases via receptor S1P1 to stabilize the endothelium. To evaluate the contribution of erythrocytes as an endogenous source of S1P to the regulation of basal permeability, we measured permeability coefficients in intact individually perfused venular microvessels of rat mesentery. This strategy also enabled the contributions of other endogenous S1P sources to be evaluated. Apparent permeability coefficients (P(S)) to albumin and α-lactalbumin and the hydraulic conductivity of mesenteric microvessels were measured in the presence or absence of rat erythrocytes or rat erythrocyte-conditioned perfusate. Rat erythrocytes added to the perfusate were the principal source of S1P in these microvessels. Basal P(S) to albumin was stable and typical of blood-perfused microvessels (mean 0.5 × 10(-6) cm/s) when erythrocytes or erythrocyte-conditioned perfusates were present. When they were absent, P(S) to albumin or α-lactalbumin increased up to 40-fold (over 10 min). When exogenous S1P was added to perfusates, permeability returned to levels comparable with those seen in the presence of erythrocytes. Addition of SEW 2871, an agonist specific for S1P1, in the absence of red blood cells reduced P(S)(BSA) (40-fold reduction) toward basal. The specific S1P1 receptor antagonist (W-146) reversed the stabilizing action of erythrocytes and increased permeability (27-fold increase) in a manner similar to that seen in the absence of erythrocytes. Erythrocytes are a primary source of S1P that maintains normal venular microvessel permeability. Absence of erythrocytes or conditioned perfusate in in vivo and in vitro models of endothelial barriers elevates basal permeability.


Assuntos
Permeabilidade Capilar , Endotélio Vascular/metabolismo , Eritrócitos/metabolismo , Lisofosfolipídeos/metabolismo , Mesentério/irrigação sanguínea , Comunicação Parácrina , Esfingosina/análogos & derivados , Albuminas/metabolismo , Animais , Permeabilidade Capilar/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Eritrócitos/efeitos dos fármacos , Lactalbumina/metabolismo , Masculino , Oxidiazóis/farmacologia , Comunicação Parácrina/efeitos dos fármacos , Pressão , Ratos , Ratos Sprague-Dawley , Receptores de Lisoesfingolipídeo/agonistas , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/metabolismo , Tiofenos/farmacologia , Fatores de Tempo , Vênulas/metabolismo
4.
Am J Physiol Heart Circ Physiol ; 302(10): H1929-35, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22427519

RESUMO

To evaluate the hypothesis that sphingosine-1-phosphate (S1P) and cAMP attenuate increased permeability of individually perfused mesenteric microvessels through a common Rac1-dependent pathway, we measured the attenuation of the peak hydraulic conductivity (L(p)) in response to the inflammatory agent bradykinin (BK) by either S1P or cAMP. We varied the extent of exposure to each agent (test) and measured the ratio L(p)(test)/L(p)(BK alone) for each vessel (anesthetized rats). S1P (1 µM) added at the same time as BK (concurrent, no pretreatment) was as effective to attenuate the response to BK (L(p) ratio: 0.14 ± 0.05; n = 5) as concurrent plus pretreatment with S1P for 30 min (L(p) ratio: 0.26 ± 0.06; n = 11). The same pretreatment with S1P, but with no concurrent S1P, caused no inhibition of the BK response (L(p) ratio 1.07 ± 0.11; n = 8). The rapid on and off action of S1P demonstrated by these results was in contrast to cAMP-dependent changes induced by rolipram and forskolin (RF), which developed more slowly, lasted longer, and resulted in partial inhibition when given either as pretreatment or concurrent with BK. In cultured endothelium, there was no Rac activation or peripheral cortactin localization at 1 min with RF, but cortactin localization and Rac activation were maximal at 1 min with S1P. When S1P was removed, Rac activation returned to control within 2 min. Because of such differing time courses, S1P and cAMP are unlikely to act through fully common effector mechanisms.


Assuntos
Bradicinina/farmacologia , Permeabilidade Capilar/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Microvasos/efeitos dos fármacos , Esfingosina/análogos & derivados , Vasodilatadores/farmacologia , Animais , Bradicinina/efeitos dos fármacos , Permeabilidade Capilar/fisiologia , Colforsina/farmacologia , AMP Cíclico/farmacologia , Masculino , Microvasos/fisiologia , Modelos Animais , Ratos , Rolipram/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Esfingosina/farmacologia , Fatores de Tempo , Proteínas rac1 de Ligação ao GTP/metabolismo
5.
Ann Biomed Eng ; 40(4): 828-39, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22009311

RESUMO

Endothelial cells are covered with a polysaccharide rich layer more than 400 nm thick, mechanical properties of which limit access of circulating plasma components to endothelial cell membranes. The barrier properties of this endothelial surface layer are deduced from the rate of tracer penetration into the layer and the mechanics of red and white cell movement through capillary microvessels. This review compares the mechanosensor and permeability properties of an inner layer (100-150 nm, close to the endothelial membrane) characterized as a quasi-periodic structure which accounts for key aspects of transvascular exchange and vascular permeability with those of the whole endothelial surface layers. We conclude that many of the barrier properties of the whole surface layer are not representative of the primary fiber matrix forming the molecular filter determining transvascular exchange. The differences between the properties of the whole layer and the inner glycocalyx structures likely reflect dynamic aspects of the endothelial surface layer including tracer binding to specific components, synthesis and degradation of key components, activation of signaling pathways in the endothelial cells when components of the surface layer are lost or degraded, and the spatial distribution of adhesion proteins in microdomains of the endothelial cell membrane.


Assuntos
Capilares/metabolismo , Moléculas de Adesão Celular/metabolismo , Células Endoteliais/metabolismo , Glicocálix/metabolismo , Microdomínios da Membrana/metabolismo , Transdução de Sinais/fisiologia , Animais , Células Endoteliais/citologia , Humanos , Permeabilidade
6.
Proc Natl Acad Sci U S A ; 105(4): 1374-9, 2008 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-18216252

RESUMO

A phenomenon that has defied explanation for two decades is the time scale for transient reabsorption in the classic experiments of Michel and Phillips on individually perfused frog mesentery microvessels. One finds that transient reabsorption lasts <2 min before a new steady state of low filtration is established when the lumen pressure is abruptly dropped from a high to a low value. Our experiments in frog and rat venular microvessels under a variety of conditions revealed the same time trend for new steady states to be established as in Michel and Phillips' experiments. In contrast, one theoretically predicts herein that the time required for the tissue albumin concentration to increase to values for a new steady state to be achieved through reabsorption is in the order of several hours. In this paper we propose a new hypothesis and theoretical model for this rapid regulation, namely that pericytes covering the interendothelial cleft exits create small trapped microdomains outside the cleft exits which regulate this transient behavior. Our electron microscopy studies on rat mesenteric venular microvessels reveal an average pericyte coverage of approximately 85%. The theoretical model based on this ultrastructural study predicts an equilibration time on the order of 1 min when the lumen pressure is abruptly lowered. The basic concept of a trapped microdomain can also be extended to microdomains in the interstitial space surrounding skeletal muscle capillaries.


Assuntos
Endotélio Vascular/metabolismo , Mesentério/metabolismo , Pericitos/metabolismo , Albumina Sérica/química , Albumina Sérica/metabolismo , Vênulas/metabolismo , Animais , Aorta , Endotélio Vascular/fisiologia , Endotélio Vascular/ultraestrutura , Masculino , Mesentério/irrigação sanguínea , Mesentério/ultraestrutura , Microcirculação/metabolismo , Microcirculação/ultraestrutura , Pericitos/ultraestrutura , Estrutura Terciária de Proteína/fisiologia , Transporte Proteico/fisiologia , Rana pipiens , Rana temporaria , Ratos , Ratos Sprague-Dawley , Albumina Sérica/fisiologia , Vênulas/fisiologia , Vênulas/ultraestrutura
7.
Am J Physiol Heart Circ Physiol ; 294(3): H1188-96, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18178724

RESUMO

Experiments in cultured endothelial cell monolayers demonstrate that increased intracellular cAMP strongly inhibits the acute permeability responses by both protein kinase A (PKA)-dependent and -independent pathways. The contribution of the PKA-independent pathways to the anti-inflammatory mechanisms of cAMP in intact mammalian microvessels has not been systematically investigated. We evaluated the role of the cAMP-dependent activation of the exchange protein activated by cAMP (Epac), a guanine nucleotide exchange factor for the small GTPase Rap1, in rat venular microvessels exposed to the platelet-activating factor (PAF). The cAMP analog 8-pCPT-2'-O-methyl-cAMP (O-Me-cAMP), which stimulates the Epac/Rap1 pathway but has no effect on PKA, significantly attenuated the PAF increase in microvessel permeability as measured by hydraulic conductivity (Lp). We also demonstrated that PAF induced a rearrangement of vascular endothelial (VE)-cadherin seen as numerous lateral spikes and frequent short breaks in the otherwise continuous peripheral immunofluorescent label. Pretreatment with O-Me-cAMP completely prevented the PAF-induced rearrangement of VE-cadherin. We conclude that the action of the Epac/Rap1 pathway to stabilize cell-cell adhesion is a significant component of the activity of cAMP to attenuate an acute increase in vascular permeability. Our results indicate that increased permeability in intact microvessels by acute inflammatory agents such as PAF is the result of the decreased effectiveness of the Epac/Rap1 pathway modulation of cell-cell adhesion.


Assuntos
Permeabilidade Capilar/fisiologia , Fator de Ativação de Plaquetas/fisiologia , Transdução de Sinais/fisiologia , Circulação Esplâncnica/fisiologia , Proteínas rap1 de Ligação ao GTP/fisiologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Caderinas/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Colforsina/farmacologia , AMP Cíclico/análogos & derivados , AMP Cíclico/metabolismo , AMP Cíclico/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Indicadores e Reagentes , Isoproterenol/farmacologia , Masculino , Microscopia Confocal , Inibidores de Fosfodiesterase/farmacologia , Fator de Ativação de Plaquetas/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Rolipram/farmacologia , Transdução de Sinais/efeitos dos fármacos , Circulação Esplâncnica/efeitos dos fármacos , Proteínas rap1 de Ligação ao GTP/genética
8.
Histochem Cell Biol ; 129(2): 179-91, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18030489

RESUMO

From studies using macrovascular endothelium, it was concluded that Rho A activation generally leads to endothelial barrier breakdown. Here, we characterized the role of Rho GTPases in endothelial barrier regulation in four different cell lines, both microvascular and macrovascular. Rho A activation by cytotoxic necrotizing factor y (CNFy) induced stress fiber formation in all cell lines. This was paralleled by gap formation and barrier breakdown in microvascular mesenteric endothelial cells (MesEnd), human dermal microvascular endothelial cells (HDMEC) as well as in macrovascular pulmonary artery endothelial cells (PAEC) but not in microvascular myocardial endothelial cells (MyEnd). In MyEnd cells, activation of Rac 1 and Cdc42 by CNF-1 strengthened barrier properties whereas in MesEnd, HDMEC and PAEC all three GTPases were activated which increased permeability in PAEC but not in MesEnd and HDMEC. In PAEC, CNF-1-induced decrease of barrier properties was blocked by the Rho kinase inhibitor Y27632 indicating that co-activation of Rho A dominated the barrier response. Inactivation of Rac 1 by toxin B or by lethal toxin (LT) compromised barrier properties in all cell lines. Taken together, Rac 1 requirement for endothelial barrier maintenance but not the destabilizing role of Rho A seems to be ubiquitous.


Assuntos
Permeabilidade Capilar/fisiologia , Células Endoteliais/fisiologia , Proteínas rho de Ligação ao GTP/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia , Amidas/farmacologia , Animais , Toxinas Bacterianas/farmacologia , Permeabilidade Capilar/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Proteínas de Escherichia coli/farmacologia , Humanos , Camundongos , Piridinas/farmacologia , Suínos , Proteína cdc42 de Ligação ao GTP/fisiologia , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/fisiologia , Proteínas rho de Ligação ao GTP/efeitos dos fármacos , Proteína rhoA de Ligação ao GTP/antagonistas & inibidores
10.
Infect Immun ; 73(8): 4879-87, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16041001

RESUMO

Epsilon-toxin, the primary virulence factor of Clostridium perfringens type D, causes mortality in livestock, particularly sheep and goats, in which it induces an often-fatal enterotoxemia. It is believed to compromise the intestinal barrier and then enter the gut vasculature, from which it is carried systemically, causing widespread vascular endothelial damage and edema. Here we used single perfused venular microvessels in rat mesentery, which enabled direct observation of permeability properties of the in situ vascular wall during exposure to toxin. We determined the hydraulic conductivity (L(p)) of microvessels as a measure of the response to epsilon-toxin. We found that microvessels were highly sensitive to toxin. At 10 microg ml(-1) the L(p) increased irreversibly to more than 15 times the control value by 10 min. At 0.3 microg ml(-1) no increase in L(p) was observed for up to 90 min. The toxin-induced increase in L(p) was consistent with changes in ultrastructure of microvessels exposed to the toxin. Those microvessels exhibited gaps either between or through endothelial cells where perfusate had direct access to the basement membrane. Many endothelial cells appeared necrotic, highly attenuated, and with dense cytoplasm. We showed that epsilon-toxin, in a time- and dose-dependent manner, rapidly and irreversibly compromised the barrier function of venular microvessel endothelium. The results conformed to the hypothesis that epsilon-toxin interacts with vascular endothelial cells and increases the vessel wall permeability by direct damage of the endothelium.


Assuntos
Toxinas Bacterianas/farmacologia , Clostridium perfringens , Veias Mesentéricas/efeitos dos fármacos , Animais , Anticorpos Monoclonais , Toxinas Bacterianas/imunologia , Veias Mesentéricas/imunologia , Veias Mesentéricas/patologia , Veias Mesentéricas/ultraestrutura , Microscopia Eletrônica de Transmissão , Permeabilidade/efeitos dos fármacos , Ratos , Fatores de Tempo
11.
J Physiol ; 564(Pt 3): 817-27, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15718259

RESUMO

Vascular permeability is assumed to be regulated by the cytosolic Ca(2+) concentration ([Ca(2+)](c)) of the endothelial cells. When permeability is increased, however, the maximum [Ca(2+)](c) appears to occur after the maximum permeability increase, suggesting that Ca(2+)-dependent mechanisms other than the absolute Ca(2+) concentration may regulate permeability. Here we investigate whether the rate of increase of the [Ca(2+)](c) (d[Ca(2+)](c)/dt) may more closely approximate the time course of the permeability increase. Hydraulic conductivity (L(p)) and endothelial [Ca(2+)](c) were measured in single perfused frog mesenteric microvessels in vivo. The relationships between the time courses of the increased L(p), [Ca(2+)](c) and d[Ca(2+)](c)/dt were examined. L(p) peaked significantly earlier than [Ca(2+)](c) in all drug treatments examined (Ca(2+) store release, store-mediated Ca(2+) influx, and store-independent Ca(2+) influx). When L(p) was increased in a store-dependent manner the time taken for L(p) to peak (3.6 +/- 0.9 min during store release, 1.2 +/- 0.3 min during store-mediated Ca(2+) influx) was significantly less than the time taken for [Ca(2+)](c) to peak (9.2 +/- 2.8 min during store release, 2.1 +/- 0.7 min during store-mediated influx), but very similar to that for the peak d[Ca(2+)](c)/dt to occur (4.3 +/- 2.0 min during store release, 1.1 +/- 0.5 min during Ca(2+) influx). Additionally, when the increase was independent of intracellular Ca(2+) stores, L(p) (0.38 +/- 0.03 min) and d[Ca(2+)](c)/dt (0.30 +/- 0.1 min) both peaked significantly before the [Ca(2+)](c) (1.05 +/- 0.31 min). These data suggest that the regulation of vascular permeability by endothelial cell Ca(2+) may be regulated by the rate of change of the [Ca(2+)](c) rather than the global [Ca(2+)].


Assuntos
Cálcio/metabolismo , Permeabilidade Capilar/fisiologia , Citosol/metabolismo , Endotélio Vascular/fisiologia , Rana temporaria/fisiologia , Animais , Cinética , Masculino , Taxa de Depuração Metabólica
12.
Am J Physiol Heart Circ Physiol ; 288(3): H1296-305, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15528228

RESUMO

We tested the hypothesis that the equilibrium between F- and G-actin in endothelial cells modulates the integrity of the actin cytoskeleton and is important for the maintenance of endothelial barrier functions in vivo and in vitro. We used the actin-depolymerizing agent cytochalasin D and jasplakinolide, an actin filament (F-actin) stabilizing and promoting substance, to modulate the actin cytoskeleton. Low doses of jasplakinolide (0.1 microM), which we have previously shown to reduce the permeability-increasing effect of cytochalasin D, had no influence on resting permeability of single-perfused mesenteric microvessels in vivo as well as on monolayer integrity. The F-actin content of cultured endothelial cells remained unchanged. In contrast, higher doses (10 microM) of jasplakinolide increased permeability (hydraulic conductivity) to the same extent as cytochalasin D and induced formation of intercellular gaps in cultured myocardial endothelial (MyEnd) cell monolayers. This was accompanied by a 34% increase of F-actin and pronounced disorganization of the actin cytoskeleton in MyEnd cells. Furthermore, we tested whether an increase of cAMP by forskolin and rolipram would prevent the cytochalasin D-induced barrier breakdown. Conditions that increase intracellular cAMP failed to block the cytochalasin D-induced permeability increase in vivo and the reduction of vascular endothelial cadherin-mediated adhesion in vitro. Taken together, these data support the hypothesis that the state of polymerization of the actin cytoskeleton is critical for maintenance of endothelial barrier functions and that both depolymerization by cytochalasin D and hyperpolymerization of actin by jasplakinolide resulted in an increase of microvessel permeability in vivo. However, cAMP, which is known to support endothelial barrier functions, seems to work by mechanisms other than stabilizing F-actin.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Endotélio Vascular/metabolismo , Citoesqueleto de Actina/efeitos dos fármacos , Animais , Antígenos CD , Antineoplásicos/farmacologia , Caderinas/metabolismo , Linhagem Celular Transformada , AMP Cíclico/metabolismo , Citocalasina D/farmacologia , Depsipeptídeos/farmacologia , Relação Dose-Resposta a Droga , Junções Comunicantes/efeitos dos fármacos , Masculino , Camundongos , Microcirculação/fisiologia , Inibidores da Síntese de Ácido Nucleico/farmacologia , Ratos , Ratos Sprague-Dawley
13.
Am J Physiol Heart Circ Physiol ; 287(6): H2427-33, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15271660

RESUMO

cAMP enhances endothelial barrier properties and is protective against various inflammatory mediators both in vivo and in vitro. However, the mechanisms whereby cAMP stabilizes the endothelial barrier are largely unknown. Recently we demonstrated that the Rho family GTPase Rac-1 is required for maintenance of endothelial barrier functions in vivo and in vitro. Therefore, in the present study we investigated the effect of forskolin (5 microM)- and rolipram (10 microM)-induced cAMP increase on reduction of barrier functions in response to Rac-1 inhibition by Clostridium sordellii lethal toxin (LT). Forskolin and rolipram treatment blocked LT (200 ng/ml)-induced hydraulic conductivity (Lp) increase in mesenteric microvessels in vivo. Likewise, LT-induced intercellular gap formation in monolayers of cultured microvascular myocardial endothelial (MyEnd) cells and LT-induced loss of adhesion of vascular endothelial cadherin-coated microbeads were abolished. Inhibition of PKA by myristoylated inhibitor peptide (14-22) of PKA (100 microM) reduced the protective effect of cAMP on LT-induced Lp increase in vivo and gap formation in vitro, indicating that the effect of cAMP on Rac-1 inhibition was PKA dependent. Glucosylation assays demonstrated that cAMP prevents inhibitory Rac-1 glucosylation by LT, indicating that one way that cAMP enhances endothelial barrier functions may be by regulating Rac-1 signaling. Our study suggests that cAMP may provide its well-established protective effects at least in part by regulation of Rho proteins.


Assuntos
Permeabilidade Capilar/fisiologia , AMP Cíclico/metabolismo , Endotélio Vascular/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Antígenos CD , Toxinas Bacterianas/farmacologia , Caderinas/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Linhagem Celular Transformada , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Junções Comunicantes/efeitos dos fármacos , Junções Comunicantes/metabolismo , Técnicas In Vitro , Veias Mesentéricas/efeitos dos fármacos , Veias Mesentéricas/metabolismo , Camundongos , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores , Proteínas rho de Ligação ao GTP/metabolismo
14.
J Physiol ; 557(Pt 3): 889-907, 2004 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15073281

RESUMO

We hypothesized that ultrafiltrate crossing the luminal endothelial glycocalyx through infrequent discontinuities (gaps) in the tight junction (TJ) strand of endothelial clefts reduces albumin diffusive flux from tissue into the 'protected region' of the cleft on the luminal side of the TJ. Thus, the effective oncotic pressure difference (sigma black triangle down pi) opposing filtration is greater than that measured between lumen and interstitial fluid. To test this we measured sigma black triangle down pi across rat mesenteric microvessels perfused with albumin (50 mg ml(-1)) with and without interstitial albumin at the same concentration within a few micrometres of the endothelium as demonstrated by confocal microscopy. We found sigma black triangle down pi was near 70% of luminal oncotic pressure when the tissue concentration equalled that in the lumen. We determined size and frequency of TJ strand gaps in endothelial clefts using serial section electron microscopy. We found nine gaps in the reconstructed clefts having mean spacing of 3.59 microm and mean length of 315 nm. The mean depth of the TJ strand near gaps was 67 nm and the mean cleft path length from lumen to interstitium was 411 nm. With these parameters our three-dimensional hydrodynamic model confirmed that fluid velocity was high at gaps in the TJ strand so that even at relatively low hydraulic pressures the albumin concentration on the tissue side of the glycocalyx was significantly lower than in the interstitium. The results conform to the hypothesis that colloid osmotic forces opposing filtration across non-fenestrated continuous capillaries are developed across the endothelial glycocalyx and that the oncotic pressure of interstitial fluid does not directly determine fluid balance across microvascular endothelium.


Assuntos
Capilares/fisiologia , Pressão Hidrostática , Pressão Osmótica , Equilíbrio Hidroeletrolítico/fisiologia , Albuminas/química , Animais , Capilares/ultraestrutura , Endotélio Vascular/metabolismo , Endotélio Vascular/ultraestrutura , Eritrócitos/metabolismo , Eritrócitos/ultraestrutura , Glicocálix/metabolismo , Glicocálix/ultraestrutura , Técnicas In Vitro , Masculino , Microscopia Confocal , Microscopia Eletrônica , Modelos Estatísticos , Ratos , Ratos Sprague-Dawley , Soluções , Junções Íntimas/metabolismo , Junções Íntimas/ultraestrutura
15.
Am J Physiol Heart Circ Physiol ; 287(2): H704-11, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15044196

RESUMO

We demonstrated previously that inhibition of the small GTPase Rac-1 by Clostridium sordellii lethal toxin (LT) increased the hydraulic conductivity (L(p)) of rat venular microvessels and induced gap formation in cultured myocardial endothelial cells (MyEnd). In MyEnd cells, we also demonstrated that both LT and cytochalasin D reduced cellular adhesion of vascular endothelial (VE)-cadherin-coated beads. Here we further evaluate the contribution of actin depolymerization, myosin-based contraction, and VE-cadherin linkage to the actin cytoskeleton to LT-induced permeability. The actin-depolymerizing agent cytochalasin D increased L(p) in single rat mesenteric microvessels to the same extent as LT over 80 min. However, whereas the actin-stabilizing agent jasplakinolide blunted the L(p) increase due to cytochalasin D by 78%, it had no effect on the LT response. This conforms to the hypothesis that the predominant mechanism whereby Rac-1 stabilizes the endothelial barrier in intact microvessels is separate from actin polymerization and likely at the level of the VE-cadherin linkage to the actin cytoskeleton. In intact vessels, neither inhibition of contraction (butanedione monoxime, an inhibitor of myosin ATPase) nor inhibition of Rho kinase (Y-27632) modified the response to LT, even though both inhibitors lowered resting L(p). In contrast butanedione monoxime and inhibition of myosin light chain kinase completely inhibited LT-induced intercellular gap formation and largely reduced the LT-induced permeability increase in MyEnd monolayers. These results support the hypothesis that the contractile mechanisms that contribute to the formation of large gaps between cultured endothelial cells exposed to inflammatory conditions do not significantly contribute to increased permeability in intact microvessels.


Assuntos
Permeabilidade Capilar/fisiologia , Depsipeptídeos , Diacetil/análogos & derivados , Endotélio Vascular/metabolismo , Circulação Esplâncnica/fisiologia , Vasoconstrição/fisiologia , Proteínas rac1 de Ligação ao GTP/fisiologia , Actinas/metabolismo , Amidas/farmacologia , Animais , Antígenos CD , Azepinas/farmacologia , Toxinas Bacterianas/farmacologia , Caderinas/fisiologia , Permeabilidade Capilar/efeitos dos fármacos , Adesão Celular/fisiologia , Linhagem Celular Transformada , Citocalasina D/farmacologia , Citoesqueleto/efeitos dos fármacos , Diacetil/farmacologia , Endotélio Vascular/citologia , Inibidores Enzimáticos/farmacologia , Espaço Extracelular/efeitos dos fármacos , Camundongos , Microcirculação/fisiologia , Miosinas/fisiologia , Naftalenos/farmacologia , Inibidores da Síntese de Ácido Nucleico/farmacologia , Peptídeos Cíclicos/farmacologia , Piridinas/farmacologia , Ratos
16.
Am J Physiol Heart Circ Physiol ; 286(1): H394-401, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14512275

RESUMO

Our previous experiments indicated that GTPases, other than RhoA, are important for the maintenance of endothelial barrier integrity in both intact microvessels of rats and mice and cultured mouse myocardial endothelial (MyEnd) cell monolayers. In the present study, we inhibited the endothelial GTPase Rac by Clostridium sordellii lethal toxin (LT) and investigated the relation between the degree of inhibition of Rac by glucosylation and increased endothelial barrier permeability. In rat venular microvessels, LT (200 ng/ml) increased hydraulic conductivity from a control value of 2.5 +/- 0.6 to 100.8 +/- 18.7 x 10-7 cm x s(-1) x cm H2O(-1) after 80 min. In cultured MyEnd cells exposed to LT (200 ng/ml), up to 60% of cellular Rac was glucosylated after 90 min, resulting in depolymerization of F-actin and interruptions of junctional distribution of vascular endothelial cadherin (VE-cadherin) and beta-catenin as well as the formation of intercellular gaps. To understand the mechanism by which inhibition of Rac caused disassembly of adherens junctions, we used laser tweezers to quantify VE-cadherin-mediated adhesion. LT and cytochalasin D, an actin depolymerizing agent, both reduced adhesion of VE-cadherin-coated microbeads to the endothelial cell surface, whereas the inhibitor of Rho kinase Y-27632 did not. Stabilization of actin filaments by jasplakinolide completely blocked the effect of cytochalasin D but not of LT on bead adhesion. We conclude that Rac regulates endothelial barrier properties in vivo and in vitro by 1) modulation of actin filament polymerization and 2) acting directly on the tether between VE-cadherin and the cytoskeleton.


Assuntos
Capilares/metabolismo , Permeabilidade Capilar/fisiologia , Endotélio Vascular/metabolismo , Proteínas rac de Ligação ao GTP/fisiologia , Actinas/metabolismo , Amidas/farmacologia , Animais , Toxinas Bacterianas/farmacologia , Caderinas/metabolismo , Linhagem Celular Transformada , Citocalasina D/farmacologia , Endotélio Vascular/citologia , Inibidores Enzimáticos/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular , Lasers , Masculino , Camundongos , Microcirculação/efeitos dos fármacos , Microesferas , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Piridinas/farmacologia , Ratos , Ratos Sprague-Dawley , Circulação Esplâncnica/efeitos dos fármacos , Quinases Associadas a rho
17.
Am J Physiol Heart Circ Physiol ; 285(6): H2446-53, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12893636

RESUMO

Thrombin is widely used to stimulate a variety of responses in cultured endothelial cell monolayers as a model of acute vascular endothelial response to inflammatory mediators. However, preliminary results indicated that rat mesenteric venules did not respond acutely to thrombin. We tested the hypothesis that rat venules would respond to thrombin 24 h after prior injury by microperfusion. Vessel responsiveness was measured as hydraulic conductivity (Lp). When venules were exposed to rat thrombin (10 U/ml) within 2 h of initial perfusion with vehicle control, there was no increase in Lp of any vessel from a mean baseline of 1.2 +/- 0.2 x 10(-7) cm.s-1.cmH2O-1. In contrast, when perfused with thrombin at 25-27 h after initial perfusion, every venule responded to thrombin with a transient increase in Lp. The mean peak Lp on day 2 in response to thrombin was 24 +/- 4.2 x 10(-7) cm.s-1.cmH2O-1. Our results suggest that prior endothelial injury modifies the endothelial cell phenotype and alters the response of endothelial cells to thrombin after 24 h. Phenotypic plasticity of endothelial cells may play a key role in the regulation of permeability of some endothelial cells in culture and in intact venules, where localized leaky sites may form where there had been a previous inflammatory response.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/imunologia , Hemostáticos/farmacologia , Trombina/farmacologia , Animais , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/imunologia , Masculino , Fator de Ativação de Plaquetas/farmacologia , Ratos , Ratos Sprague-Dawley , Vasculite/fisiopatologia , Vênulas/efeitos dos fármacos , Vênulas/imunologia , Vênulas/metabolismo
18.
Am J Physiol Heart Circ Physiol ; 285(1): H406-17, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12649070

RESUMO

We tested the hypothesis that acutely induced hyperpermeability is dependent on actin-myosin contractility by using individually perfused mesentery venules of pentobarbital-anesthetized rats. Venule hydraulic conductivity (Lp) was measured to monitor hyperpermeability response to the platelet-activating factor (PAF) 1-O-hexadecyl-2-acetyl-sn-glycero-3-phosphocholine or bradykinin. Perfusion with PAF (10 nM) induced a robust transient high Lp [24.3 +/- 1.7 x 10-7 cm/(s.cmH2O)] that peaked in 8.9 +/- 0.5 min and then returned toward control Lp [1.6 +/- 0.1 x 10-7 cm/(s.cmH2O)]. Reconstruction of venular segments with the use of transmission electron microscopy of serial sections confirmed that PAF induces paracellular inflammatory gaps. Specific inhibition of myosin light chain kinase (MLCK) with 1-10 microM 1-(5-iodonaphthalene-1-sulfonyl)-1H-hexahydro-1,4-diazepine hydrochloride (ML-7) failed to block the PAF Lp response or change the time-to-peak Lp. ML-7 reduced baseline Lp 50% at 40 min of pretreatment. ML-7 also increased the rate of recovery from PAF hyperpermeability measured as the decrease of half-time of recovery from 4.8 +/- 0.7 to 3.2 +/- 0.3 min. Inhibition of myosin ATPase with 5-20 mM 2,3-butanedione 2-monoxime also failed to alter the hyperpermeability response to PAF. Similar results were found using ML-7 to modulate responses. These experiments indicate that an actin-myosin contractile mechanism modulated by MLCK does not contribute significantly to the robust initial increase in permeability of rat venular microvessels exposed to two common inflammatory mediators. The results are consistent with paracellular gap formation by local release of endothelial-endothelial cell adhesion structures in the absence of contraction by the actin-myosin network.


Assuntos
Actinas/fisiologia , Bradicinina/farmacologia , Permeabilidade Capilar/efeitos dos fármacos , Músculo Liso Vascular/fisiologia , Miosinas/fisiologia , Fator de Ativação de Plaquetas/farmacologia , Actinas/antagonistas & inibidores , Algoritmos , Animais , Azepinas/farmacologia , Adesão Celular/efeitos dos fármacos , Creatina Quinase/antagonistas & inibidores , AMP Cíclico/metabolismo , Inibidores Enzimáticos/farmacologia , Junções Comunicantes/efeitos dos fármacos , Hemostáticos/farmacologia , Técnicas In Vitro , Masculino , Microscopia Eletrônica , Músculo Liso Vascular/efeitos dos fármacos , Miosinas/antagonistas & inibidores , Naftalenos/farmacologia , Ratos , Ratos Sprague-Dawley , Trombina/farmacologia , Vênulas/efeitos dos fármacos , Vênulas/ultraestrutura
19.
Microcirculation ; 9(6): 537-50, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12483550

RESUMO

OBJECTIVE: Localized inflammatory leaky sites form at regions of the microvessel wall with the largest increase in endothelial cell cytoplasmic calcium concentration, [Ca(2+)](i). We investigated the mechanisms that modulate localized increases in [Ca(2+)](i) in individual endothelial cells of microvessels after exposure to ATP. METHODS: [Ca(2+)](i) was measured by using digital fluorescence microscopy and fura-2 in the endothelial cells forming the walls of individually perfused frog mesenteric microvessels. The spread of [Ca(2+)](i) from a localized mechanical stimulus was also measured. RESULTS: The peak [Ca(2+)](i) after ATP showed marked heterogeneity, ranging from 227 to 1469 nM from resting values of 69 +/- 5 nM. After depolarization with high-potassium solutions, the endothelial cells with the largest peak increase in [Ca(2+)](i) had the largest fractional reduction. Localized increases in [Ca(2+)](i) due to mechanical stimulus did not spread. CONCLUSION: The key mechanism regulating the heterogeneity in initial peak increase in [Ca(2+)](i) is a calcium-dependent process regulated by the calcium influx itself. One such mechanism, the calcium-dependent opening of additional potassium channels leading to membrane hyperpolarization and increased driving force for calcium entry through passive conductance pathways, accounts for a significant amount of the heterogeneity of [Ca(2+)](i) in our experiments. Further investigations of both localized calcium influx and membrane potentials in the endothelial cells of intact microvessels in both frog and mammals using the imaging methods developed for these investigations are needed to understand the formation of localized leaky sites in inflamed microvessels.


Assuntos
Cálcio/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Endotélio Vascular/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Transporte Biológico , Comunicação Celular , Endotélio Vascular/citologia , Fura-2 , Microscopia de Fluorescência , Rana pipiens , Circulação Esplâncnica , Vênulas/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA