Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 186
Filter
1.
Am J Physiol Heart Circ Physiol ; 306(7): H1011-7, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24531813

ABSTRACT

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.


Subject(s)
Capillary Permeability , Erythrocytes/metabolism , Lysophospholipids/blood , Microcirculation , Perfusion , Serum Albumin/metabolism , Sphingosine/analogs & derivatives , Venules/physiology , Animals , Biological Transport , Hematocrit , Isotonic Solutions/metabolism , Liposomes , Male , Rats , Rats, Sprague-Dawley , Ringer's Solution , Sphingosine/blood , Time Factors
2.
Am J Physiol Heart Circ Physiol ; 304(8): H1077-84, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23417864

ABSTRACT

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.


Subject(s)
Capillary Permeability/physiology , Endothelial Cells/physiology , Hemorheology/physiology , Microcirculation/physiology , Venules/physiology , Water/metabolism , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Capillary Permeability/drug effects , Cell Adhesion , Chondroitin Sulfates/pharmacology , Endothelial Cells/drug effects , Glycocalyx/drug effects , Glycocalyx/physiology , Hyaluronic Acid/pharmacology , Male , Mesenteric Veins/drug effects , Mesenteric Veins/physiology , Microcirculation/drug effects , Occludin/metabolism , Rats , Rats, Sprague-Dawley , Venules/drug effects , Viscosupplements/pharmacology
3.
Acta Physiol (Oxf) ; 207(4): 628-49, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23374222

ABSTRACT

Our major theme is that the layered structure of the endothelial barrier requires continuous activation of signalling pathways regulated by sphingosine-1-phosphate (S1P) and intracellular cAMP. These pathways modulate the adherens junction, continuity of tight junction strands, and the balance of synthesis and degradation of glycocalyx components. We evaluate recent evidence that baseline permeability is maintained by constant activity of mechanisms involving the small GTPases Rap1 and Rac1. In the basal state, the barrier is compromised when activities of the small GTPases are reduced by low S1P supply or delivery. With inflammatory stimulus, increased permeability can be understood in part as the action of signalling to reduce Rap1 and Rac1 activation. With the hypothesis that microvessel permeability and selectivity under both normal and inflammatory conditions are regulated by mechanisms that are continuously active, it follows that when S1P or intracellular cAMP are elevated at the time of inflammatory stimulus, they can buffer changes induced by inflammatory agents and maintain normal barrier stability. When endothelium is exposed to inflammatory conditions and subsequently exposed to elevated S1P or intracellular cAMP, the same processes restore the functional barrier by first re-establishing the adherens junction, then modulating tight junctions and glycocalyx. In more extreme inflammatory conditions, loss of the inhibitory actions of Rac1-dependent mechanisms may promote expression of more inflammatory endothelial phenotypes by contributing to the up-regulation of RhoA-dependent contractile mechanisms and the sustained loss of surface glycocalyx allowing access of inflammatory cells to the endothelium.


Subject(s)
Blood Vessels/physiology , Capillary Permeability/physiology , Signal Transduction/physiology , Animals , Blood Vessels/cytology , Cell Membrane Permeability/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Humans , Mice , Microvessels/cytology , Microvessels/physiology , Models, Animal
4.
Am J Physiol Heart Circ Physiol ; 303(7): H825-34, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22865384

ABSTRACT

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.


Subject(s)
Capillary Permeability , Endothelium, Vascular/metabolism , Erythrocytes/metabolism , Lysophospholipids/metabolism , Mesentery/blood supply , Paracrine Communication , Sphingosine/analogs & derivatives , Albumins/metabolism , Animals , Capillary Permeability/drug effects , Endothelium, Vascular/drug effects , Erythrocytes/drug effects , Lactalbumin/metabolism , Male , Oxadiazoles/pharmacology , Paracrine Communication/drug effects , Pressure , Rats , Rats, Sprague-Dawley , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/metabolism , Sphingosine/metabolism , Thiophenes/pharmacology , Time Factors , Venules/metabolism
5.
Am J Physiol Heart Circ Physiol ; 302(10): H1929-35, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22427519

ABSTRACT

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.


Subject(s)
Bradykinin/pharmacology , Capillary Permeability/drug effects , Lysophospholipids/pharmacology , Microvessels/drug effects , Sphingosine/analogs & derivatives , Vasodilator Agents/pharmacology , Animals , Bradykinin/drug effects , Capillary Permeability/physiology , Colforsin/pharmacology , Cyclic AMP/pharmacology , Male , Microvessels/physiology , Models, Animal , Rats , Rolipram/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Sphingosine/pharmacology , Time Factors , rac1 GTP-Binding Protein/metabolism
6.
Ann Biomed Eng ; 40(4): 828-39, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22009311

ABSTRACT

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.


Subject(s)
Capillaries/metabolism , Cell Adhesion Molecules/metabolism , Endothelial Cells/metabolism , Glycocalyx/metabolism , Membrane Microdomains/metabolism , Signal Transduction/physiology , Animals , Endothelial Cells/cytology , Humans , Permeability
7.
Proc Natl Acad Sci U S A ; 105(4): 1374-9, 2008 Jan 29.
Article in English | MEDLINE | ID: mdl-18216252

ABSTRACT

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.


Subject(s)
Endothelium, Vascular/metabolism , Mesentery/metabolism , Pericytes/metabolism , Serum Albumin/chemistry , Serum Albumin/metabolism , Venules/metabolism , Animals , Aorta , Endothelium, Vascular/physiology , Endothelium, Vascular/ultrastructure , Male , Mesentery/blood supply , Mesentery/ultrastructure , Microcirculation/metabolism , Microcirculation/ultrastructure , Pericytes/ultrastructure , Protein Structure, Tertiary/physiology , Protein Transport/physiology , Rana pipiens , Rana temporaria , Rats , Rats, Sprague-Dawley , Serum Albumin/physiology , Venules/physiology , Venules/ultrastructure
8.
Am J Physiol Heart Circ Physiol ; 294(3): H1188-96, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18178724

ABSTRACT

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.


Subject(s)
Capillary Permeability/physiology , Platelet Activating Factor/physiology , Signal Transduction/physiology , Splanchnic Circulation/physiology , rap1 GTP-Binding Proteins/physiology , Adrenergic beta-Agonists/pharmacology , Animals , Cadherins/metabolism , Capillary Permeability/drug effects , Colforsin/pharmacology , Cyclic AMP/analogs & derivatives , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/physiology , Indicators and Reagents , Isoproterenol/pharmacology , Male , Microscopy, Confocal , Phosphodiesterase Inhibitors/pharmacology , Platelet Activating Factor/drug effects , Rats , Rats, Sprague-Dawley , Rolipram/pharmacology , Signal Transduction/drug effects , Splanchnic Circulation/drug effects , rap1 GTP-Binding Proteins/genetics
10.
Infect Immun ; 73(8): 4879-87, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16041001

ABSTRACT

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.


Subject(s)
Bacterial Toxins/pharmacology , Clostridium perfringens , Mesenteric Veins/drug effects , Animals , Antibodies, Monoclonal , Bacterial Toxins/immunology , Mesenteric Veins/immunology , Mesenteric Veins/pathology , Mesenteric Veins/ultrastructure , Microscopy, Electron, Transmission , Permeability/drug effects , Rats , Time Factors
11.
Am J Physiol Heart Circ Physiol ; 288(3): H1296-305, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15528228

ABSTRACT

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.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Endothelium, Vascular/metabolism , Actin Cytoskeleton/drug effects , Animals , Antigens, CD , Antineoplastic Agents/pharmacology , Cadherins/metabolism , Cell Line, Transformed , Cyclic AMP/metabolism , Cytochalasin D/pharmacology , Depsipeptides/pharmacology , Dose-Response Relationship, Drug , Gap Junctions/drug effects , Male , Mice , Microcirculation/physiology , Nucleic Acid Synthesis Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley
12.
Am J Physiol Heart Circ Physiol ; 287(6): H2427-33, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15271660

ABSTRACT

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.


Subject(s)
Capillary Permeability/physiology , Cyclic AMP/metabolism , Endothelium, Vascular/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Antigens, CD , Bacterial Toxins/pharmacology , Cadherins/metabolism , Capillary Permeability/drug effects , Cell Line, Transformed , Cyclic AMP-Dependent Protein Kinases/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Gap Junctions/drug effects , Gap Junctions/metabolism , In Vitro Techniques , Mesenteric Veins/drug effects , Mesenteric Veins/metabolism , Mice , rac1 GTP-Binding Protein/antagonists & inhibitors , rho GTP-Binding Proteins/metabolism
13.
J Physiol ; 557(Pt 3): 889-907, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15073281

ABSTRACT

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.


Subject(s)
Capillaries/physiology , Hydrostatic Pressure , Osmotic Pressure , Water-Electrolyte Balance/physiology , Albumins/chemistry , Animals , Capillaries/ultrastructure , Endothelium, Vascular/metabolism , Endothelium, Vascular/ultrastructure , Erythrocytes/metabolism , Erythrocytes/ultrastructure , Glycocalyx/metabolism , Glycocalyx/ultrastructure , In Vitro Techniques , Male , Microscopy, Confocal , Microscopy, Electron , Models, Statistical , Rats , Rats, Sprague-Dawley , Solutions , Tight Junctions/metabolism , Tight Junctions/ultrastructure
14.
Am J Physiol Heart Circ Physiol ; 287(2): H704-11, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15044196

ABSTRACT

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.


Subject(s)
Capillary Permeability/physiology , Depsipeptides , Diacetyl/analogs & derivatives , Endothelium, Vascular/metabolism , Splanchnic Circulation/physiology , Vasoconstriction/physiology , rac1 GTP-Binding Protein/physiology , Actins/metabolism , Amides/pharmacology , Animals , Antigens, CD , Azepines/pharmacology , Bacterial Toxins/pharmacology , Cadherins/physiology , Capillary Permeability/drug effects , Cell Adhesion/physiology , Cell Line, Transformed , Cytochalasin D/pharmacology , Cytoskeleton/drug effects , Diacetyl/pharmacology , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , Extracellular Space/drug effects , Mice , Microcirculation/physiology , Myosins/physiology , Naphthalenes/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology , Peptides, Cyclic/pharmacology , Pyridines/pharmacology , Rats
15.
Am J Physiol Heart Circ Physiol ; 286(1): H394-401, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14512275

ABSTRACT

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.


Subject(s)
Capillaries/metabolism , Capillary Permeability/physiology , Endothelium, Vascular/metabolism , rac GTP-Binding Proteins/physiology , Actins/metabolism , Amides/pharmacology , Animals , Bacterial Toxins/pharmacology , Cadherins/metabolism , Cell Line, Transformed , Cytochalasin D/pharmacology , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , Intracellular Signaling Peptides and Proteins , Lasers , Male , Mice , Microcirculation/drug effects , Microspheres , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Splanchnic Circulation/drug effects , rho-Associated Kinases
16.
Am J Physiol Heart Circ Physiol ; 285(6): H2446-53, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12893636

ABSTRACT

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.


Subject(s)
Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Hemostatics/pharmacology , Thrombin/pharmacology , Animals , Capillary Permeability/drug effects , Capillary Permeability/immunology , Male , Platelet Activating Factor/pharmacology , Rats , Rats, Sprague-Dawley , Vasculitis/physiopathology , Venules/drug effects , Venules/immunology , Venules/metabolism
17.
Am J Physiol Heart Circ Physiol ; 285(1): H406-17, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12649070

ABSTRACT

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.


Subject(s)
Actins/physiology , Bradykinin/pharmacology , Capillary Permeability/drug effects , Muscle, Smooth, Vascular/physiology , Myosins/physiology , Platelet Activating Factor/pharmacology , Actins/antagonists & inhibitors , Algorithms , Animals , Azepines/pharmacology , Cell Adhesion/drug effects , Creatine Kinase/antagonists & inhibitors , Cyclic AMP/metabolism , Enzyme Inhibitors/pharmacology , Gap Junctions/drug effects , Hemostatics/pharmacology , In Vitro Techniques , Male , Microscopy, Electron , Muscle, Smooth, Vascular/drug effects , Myosins/antagonists & inhibitors , Naphthalenes/pharmacology , Rats , Rats, Sprague-Dawley , Thrombin/pharmacology , Venules/drug effects , Venules/ultrastructure
18.
J Physiol ; 539(Pt 1): 295-308, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11850521

ABSTRACT

Previous experiments using cultured endothelial monolayers indicate that Rho-family small GTPases are involved in modulation of endothelial monolayer permeability by regulating assembly of the cellular actin filament scaffold, activity of myosin-based contractility and junctional distribution of the Ca2+-dependent endothelial cell adhesion molecule, VE-cadherin. We investigated these mechanisms using both cultured endothelial cells (from porcine pulmonary artery and mouse heart) and vascular endothelium in situ (mouse aorta, and individually perfused venular microvessels of mouse and rat mesentery). Exposure to Clostridium difficile toxin B (100 ng x ml(-1)) inactivated 50-90% of all endothelial Rho proteins within 60-90 min. This was accompanied by considerable reduction of actin filament stress fibres and junctional F-actin in cultured endothelial monolayers and in mouse aortic endothelium in situ. Also, VE-cadherin became discontinuous along endothelial junctions. Inhibition of Rho kinase with Y-27632 (30 microM) for 90-120 min induced F-actin reduction both in vitro and in situ but did not cause redistribution or reduction of VE-cadherin staining. Perfusion of microvessels with toxin B increased basal hydraulic permeability (L(p)) but did not attenuate the transient increase in L(p) of microvessels exposed to bradykinin. Perfusion of microvessels with Y-27632 (30 microM) for up to 100 min reduced basal L(p) but did not attenuate the permeability increase induced by platelet activating factor (PAF) or bradykinin. These results show that toxin B-mediated reduction of endothelial barrier properties is due to inactivation of small GTPases other than RhoA. Rho proteins as well as RhoA-mediated contractile mechanisms are not involved in bradykinin- or PAF-induced hyperpermeability of intact microvessels.


Subject(s)
Acute-Phase Proteins/physiology , Bacterial Proteins , Capillary Permeability/physiology , Endothelium, Vascular/metabolism , Protein Serine-Threonine Kinases/physiology , Amides/pharmacology , Animals , Aorta/drug effects , Bacterial Toxins/pharmacology , Bradykinin/pharmacology , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Glycosylation , Inflammation Mediators/pharmacology , Intracellular Signaling Peptides and Proteins , Mice , Microcirculation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/pharmacology , Rats , Swine , Venules/drug effects , rho-Associated Kinases
20.
Am J Physiol Heart Circ Physiol ; 279(4): H1724-36, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11009460

ABSTRACT

We tested the hypothesis that the effective oncotic force that opposes fluid filtration across the microvessel wall is the local oncotic pressure difference across the endothelial surface glycocalyx and not the global difference between the plasma and tissue. In single frog mesenteric microvessels perfused and superfused with solutions containing 50 mg/ml albumin, the effective oncotic pressure exerted across the microvessel wall was not significantly different from that measured when the perfusate alone contained albumin at 50 mg/ml. Measurements were made during transient and steady-state filtration at capillary pressures between 10 and 35 cmH(2)O. A cellular-level model of coupled water and solute flows in the interendothelial cleft showed water flux through small breaks in the junctional strand limited back diffusion of albumin into the protected space on the tissue side of the glycocalyx. Thus oncotic forces opposing filtration are larger than those estimated from blood-to-tissue protein concentration differences, and transcapillary fluid flux is smaller than estimated from global differences in oncotic and hydrostatic pressures.


Subject(s)
Blood Proteins/metabolism , Models, Cardiovascular , Animals , Anura , Capillary Permeability , Glycocalyx/metabolism , Microcirculation/physiology , Osmotic Pressure , Serum Albumin/metabolism , Splanchnic Circulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...