Comprehensive Physiology Wiley Online Library

Microcirculatory Exchange Function

Full Article on Wiley Online Library



Abstract

The sections in this article are:

1 Introduction
2 Physical Principles Controlling Fluid and Solute Transport
2.1 General Characteristics of Fluid and Solute Flux
2.2 Exchange Microvessels
2.3 Experimental Models for Studying Microvascular Transport
2.4 Paracellular and Transcellular Transport
3 Regulatory Mechanisms
3.1 Agonists and Receptors: Localization and Function
3.2 Signal Transduction Pathways
4 Concluding Remarks
Figure 1. Figure 1.

Microvascular hyperpermeability response to platelet activating factor (PAF: 1.0 × 10−7 M) in the hamster cheek pouch. The details of the microvascular network are observed by fluorescence intravital microscopy using a macromolecular fluorochrome (i.e. FITC‐dextran 150kDa). The branching pattern of the arterioles (a) and tributaries to venules (v) can be followed easily. The control panel (A) shows absence of FITC‐dextran 150 in the interstitium. Topical application of PAF (B) induced arteriolar constriction and permeation of the fluorescent macromolecular tracer into the interstitium across postcapillary venules (arrows). Modified from 66 with permission. (See page 3 in colour section at the back of the book)

Figure 2. Figure 2.

The Landis technique. A single microvessel is occluded with a glass micropipette and the distance between an erythrocyte trapped and in the microvessel lumen and the occluding probe is measured as a function of time. The microvessel diameter is also measured. Volume flux (J,JS) is estimated using the initial erythrocyte velocity. Hydraulic conductivity (Lp) is determined as the slope of J,JS as a function of mean occluded intraluminal pressure (P).

Figure 3. Figure 3.

The isolated venule technique. (A) Schematic of the experimental setup. A venule is dissected, excised, and mounted on two glass micropipettes and bathed with an albumin physiological saline solution at 37°C. Pressure and flow are controlled by altering the heights of the inflow and outflow reservoirs, which are mounted on manometers. (B) To assess permeability, the fluorescence intensity of a window containing the venule and the nearby extraluminal area is measured. The inflow is quickly switched from normal APSS to APSS containing a fluorochrome‐labeled tracer (e.g. FITC‐albumin), causing a step increase fluorescence intensity (ΔIfo) that is proportional to the number of solute tracer molecules that have entered the lumen. This is followed by a gradual increase in intensity (dIf/dt)o. The rate of (dIf/dt)o is proportional to solute transport across the microvascular wall. The intensity returns to the basal level when the tracer is washed out from the lumen.

Adapted from 34 and 79 with permission
Figure 4. Figure 4.

Cultured endothelial cell (EC) permeability models. An EC monolayer is grown on a porous membrane that is placed between two chambers. (A) A vertical chamber system is shown, in which the upper chamber represents the “luminal” side and the tower chamber represents the “abluminal” side of the endothelial monolayer. Note that the fluid levels of the upper and lower chambers are kept at the same height to minimize any hydrostatic differences between the two chambers. (B) A horizontal setup.

Figure 5. Figure 5.

Ultrastructure of canine cardiac postcapillary venule. The electron micrograph demonstrates the complex morphology of endothelial cells (ECs). Possible transport pathways for solutes are indicated by different arrows. Caveolae and vesicles are abundant throughout the EC. WP. Weibel—Palade body. Durán, WN and Berendsen P (unpublished). (See page 3 in colour section at the back of the book)

Figure 6. Figure 6.

Routes of fluid and solute transport across the endothelium. Two pathways, one paracellular and one transcellular, have been proposed. In the paracellular pathway, fluid and solutes pass between endothelial cells (ECs), with the major barrier being the tightness of junctions between ECs. Several molecules thought to regulate junctional integrity are shown. In the transcellular pathway, active vesicular transport, or the formation of channels of VVOs, is proposed as a route for fluid and solute passage.

Figure 7. Figure 7.

Signal transduction pathways in microvascular permeability. Receptors are activated upon binding to their respective agonists and interact with downstream signaling pathways. The diagram reflects a typical set of cascades activated by permeability agonists; VEGF is used as an example.

Figure 8. Figure 8.

Transfection of ca‐ROCK protein increases coronary venular permeability. Isolated porcine coronary venules were transfected with 3 μg/ml ca‐ROCK protein for 120min in absence (solid line, squares, n = 6) or presence (dashed line, triangles, n = 4) of the ROCK inhibitor Y‐27632 (5 × 10−6M). Y‐27632 was added to the bath 20min prior to the addition of ca‐ROCK. Transfection was initiated at 0min and continued throughout the time course. *Indicates P < 0.05 vs. baseline permeability (0‐min time point). ‡Indicates P < 0.05 vs. ca‐ROCK transfection when Y‐27632 is present. From Ref. 307 with permission.

Figure 9. Figure 9.

MLC phosphorylation as a mechanism of endothelial barrier regulation. Inflammatory stimuli cause elevated phosphorylation of MLC on Thr‐18/Ser‐19 in endothelial cells, which in turn elevates actin—myosin mediated contraction. This leads to increased cellular centrifugal force that promotes opening of endothelial cell—cell junctions, causing increased endothelial permeability. MLC phosphorylation increases when MLCK activity is elevated or MLCP activity is reduced. The Rho/ROCK pathway can inactivate MLCP through phosphorylation of the MLCP targeting subunit MYPT‐1 on Thr‐696. (See page 4 in colour section at the back of the book)

Figure 10. Figure 10.

ROCK‐mediated tension and barrier dysfunction in coronary venular endothelial cell (EC) monolayers. Transfection of ca‐ROCK (3 μg/ml) into cultured coronary venular EC (CVEC) monolayers increases isometric tension (triangles) and causes a drop in TER (solid line). A time course overlay of ca‐ROCK‐stimulated changes in TER and tension shows that these two events occur simultaneously. From Ref. 307 with permission.

Figure 11. Figure 11.

Adherens junctions in endothelial cells (EC). (A) Adhesion between ECs is achieved by homotypic binding of VE‐cadherin molecules at endothelial cell—cell junctions. Several catenins (p120, α, β, γ) are associated with VE‐cadherin and regulate its homotypic binding, as well as linkage to the cytoskeleton. (B) Under stimulated conditions, such as during VEGF treatment, VE‐cadherin, β‐catenin, and p120 catenin are phosphorylated on tyrosine residues, and a reorganization of endothelial cell—cell junctions occurs, characterized by the appearance of several VE‐cadherin rich finger‐like projections (arrows). Scale bar represents 20μm. Images from Ref. 207 with permission. (See page 4 in colour section at the back of the book)

Figure 12. Figure 12.

PAF‐induced NO production. (A) NO concentration was measured in the suffusate of the hamster cheek pouch using a Nitric Oxide Analyzer 407. (B) Perivenular NO concentration was determined using a NO‐sensitive electrode in the mouse cremaster muscle. NO concentration rose quickly upon topical administration of PAF in both tissues. “From 407 with permission.

Figure 13. Figure 13.

Obligatory role of eNOS‐mediated NO production in PAF‐induced hyperpermeability in vivo. PAF‐induced hyperpermeability is markedly reduced in eNOS−/−mice compared to wild‐type control mice. Typical intravital fluorescence microscopy images of the respective cremaster muscles are shown below the IOI graph.

Adapted from Refs 74 and 410 with permission. (See page 4 in colour section at the back of the book)
Figure 14. Figure 14.

Translocation of eNOS. Top Panel: (A) Isolation of lipid rafts domains was done in control, Ach, or PAF‐treated cells. Fractions were probed against caveolin‐1 and eNOS. (A) ACh treatment. (B) PAF treatment. The blots represent three independent experiments. Bottom Panel: The influence of PAF and ACh on the distribution of eNOS‐GFP was assessed by fluorescence of GFP. The images are representative of three independent experiments. (A) ECV304‐eNOS‐GFP. (B) Human dermal microvascular cells stained with anti‐eNOS (red) and anti‐caveolin‐1 (green) antibodies. From Ref. 434 with permission. (See page 5 in colour section at the back of the book)

Figure 15. Figure 15.

Diagram of eNOS translocation process and eNOS activation. (A) Shows a simplified diagram of the eNOS translocation hypothesis. eNOS is anchored in caveolae through myristoylation and palmitoylation, and is associated with caveolin‐1. Upon stimulation with agonists, eNOS becomes phosphorylated, dissociates from caveolin‐1, and translocates from the caveolae to either the Golgi apparatus or the cytosol. It is postulated that preferential translocation to different compartments is related to vascular function and may allow eNOS to interact with specific function‐related target molecules. Even though the drawing depicts eNOS as being “free” in the cytosol, our recent‐unpublished data indicate that eNOS may be internalized to its intracellular targets via caveolae. (B). Several signals can regulate eNOS activity. While association with caveolin‐1 and phosphorylation of Thr‐497 decrease enzyme activity (NO production), interaction with Ca2+‐calmodulin and HSP90, phosphorylation on Ser‐1177 by Akt, and PKC activation can all increase activity. Panel A redrawn from 410 with permission.

Figure 16. Figure 16.

PAF‐induced eNOS phosphorylation and hyperpermeability in ECV304‐eNOSGFP cells. (A) ACh and PAF activate eNOS by Ser‐1177 phosphorylation and Thr495 dephosphorylation. (B) Inhibition of muscarinic ACh and PAF receptors blocks Ser‐1177 phosphorylation β‐actin served as a (loading) control in experiments shown in (A) and (B). (C) PAF increases permeability in ECVeNOS‐GFP cells. Monolayers of ECVeNOS‐GFP cells were treated with PAF and permeability to FITC‐Dx‐70 was measured. Data are expressed as permeability coefficients (mean ± SEM) for control and PAF‐stimulated monolayers. *P < 0.05, n = 6. From 434 with permission.

Figure 17. Figure 17.

Functions of Rap‐1. The diagram shows known and putative effectors and targets of Rap‐1. Arrows indicate stimulation, blunt ends denote inhibition.

Adapted from Ref. 457 with permission


Figure 1.

Microvascular hyperpermeability response to platelet activating factor (PAF: 1.0 × 10−7 M) in the hamster cheek pouch. The details of the microvascular network are observed by fluorescence intravital microscopy using a macromolecular fluorochrome (i.e. FITC‐dextran 150kDa). The branching pattern of the arterioles (a) and tributaries to venules (v) can be followed easily. The control panel (A) shows absence of FITC‐dextran 150 in the interstitium. Topical application of PAF (B) induced arteriolar constriction and permeation of the fluorescent macromolecular tracer into the interstitium across postcapillary venules (arrows). Modified from 66 with permission. (See page 3 in colour section at the back of the book)



Figure 2.

The Landis technique. A single microvessel is occluded with a glass micropipette and the distance between an erythrocyte trapped and in the microvessel lumen and the occluding probe is measured as a function of time. The microvessel diameter is also measured. Volume flux (J,JS) is estimated using the initial erythrocyte velocity. Hydraulic conductivity (Lp) is determined as the slope of J,JS as a function of mean occluded intraluminal pressure (P).



Figure 3.

The isolated venule technique. (A) Schematic of the experimental setup. A venule is dissected, excised, and mounted on two glass micropipettes and bathed with an albumin physiological saline solution at 37°C. Pressure and flow are controlled by altering the heights of the inflow and outflow reservoirs, which are mounted on manometers. (B) To assess permeability, the fluorescence intensity of a window containing the venule and the nearby extraluminal area is measured. The inflow is quickly switched from normal APSS to APSS containing a fluorochrome‐labeled tracer (e.g. FITC‐albumin), causing a step increase fluorescence intensity (ΔIfo) that is proportional to the number of solute tracer molecules that have entered the lumen. This is followed by a gradual increase in intensity (dIf/dt)o. The rate of (dIf/dt)o is proportional to solute transport across the microvascular wall. The intensity returns to the basal level when the tracer is washed out from the lumen.

Adapted from 34 and 79 with permission


Figure 4.

Cultured endothelial cell (EC) permeability models. An EC monolayer is grown on a porous membrane that is placed between two chambers. (A) A vertical chamber system is shown, in which the upper chamber represents the “luminal” side and the tower chamber represents the “abluminal” side of the endothelial monolayer. Note that the fluid levels of the upper and lower chambers are kept at the same height to minimize any hydrostatic differences between the two chambers. (B) A horizontal setup.



Figure 5.

Ultrastructure of canine cardiac postcapillary venule. The electron micrograph demonstrates the complex morphology of endothelial cells (ECs). Possible transport pathways for solutes are indicated by different arrows. Caveolae and vesicles are abundant throughout the EC. WP. Weibel—Palade body. Durán, WN and Berendsen P (unpublished). (See page 3 in colour section at the back of the book)



Figure 6.

Routes of fluid and solute transport across the endothelium. Two pathways, one paracellular and one transcellular, have been proposed. In the paracellular pathway, fluid and solutes pass between endothelial cells (ECs), with the major barrier being the tightness of junctions between ECs. Several molecules thought to regulate junctional integrity are shown. In the transcellular pathway, active vesicular transport, or the formation of channels of VVOs, is proposed as a route for fluid and solute passage.



Figure 7.

Signal transduction pathways in microvascular permeability. Receptors are activated upon binding to their respective agonists and interact with downstream signaling pathways. The diagram reflects a typical set of cascades activated by permeability agonists; VEGF is used as an example.



Figure 8.

Transfection of ca‐ROCK protein increases coronary venular permeability. Isolated porcine coronary venules were transfected with 3 μg/ml ca‐ROCK protein for 120min in absence (solid line, squares, n = 6) or presence (dashed line, triangles, n = 4) of the ROCK inhibitor Y‐27632 (5 × 10−6M). Y‐27632 was added to the bath 20min prior to the addition of ca‐ROCK. Transfection was initiated at 0min and continued throughout the time course. *Indicates P < 0.05 vs. baseline permeability (0‐min time point). ‡Indicates P < 0.05 vs. ca‐ROCK transfection when Y‐27632 is present. From Ref. 307 with permission.



Figure 9.

MLC phosphorylation as a mechanism of endothelial barrier regulation. Inflammatory stimuli cause elevated phosphorylation of MLC on Thr‐18/Ser‐19 in endothelial cells, which in turn elevates actin—myosin mediated contraction. This leads to increased cellular centrifugal force that promotes opening of endothelial cell—cell junctions, causing increased endothelial permeability. MLC phosphorylation increases when MLCK activity is elevated or MLCP activity is reduced. The Rho/ROCK pathway can inactivate MLCP through phosphorylation of the MLCP targeting subunit MYPT‐1 on Thr‐696. (See page 4 in colour section at the back of the book)



Figure 10.

ROCK‐mediated tension and barrier dysfunction in coronary venular endothelial cell (EC) monolayers. Transfection of ca‐ROCK (3 μg/ml) into cultured coronary venular EC (CVEC) monolayers increases isometric tension (triangles) and causes a drop in TER (solid line). A time course overlay of ca‐ROCK‐stimulated changes in TER and tension shows that these two events occur simultaneously. From Ref. 307 with permission.



Figure 11.

Adherens junctions in endothelial cells (EC). (A) Adhesion between ECs is achieved by homotypic binding of VE‐cadherin molecules at endothelial cell—cell junctions. Several catenins (p120, α, β, γ) are associated with VE‐cadherin and regulate its homotypic binding, as well as linkage to the cytoskeleton. (B) Under stimulated conditions, such as during VEGF treatment, VE‐cadherin, β‐catenin, and p120 catenin are phosphorylated on tyrosine residues, and a reorganization of endothelial cell—cell junctions occurs, characterized by the appearance of several VE‐cadherin rich finger‐like projections (arrows). Scale bar represents 20μm. Images from Ref. 207 with permission. (See page 4 in colour section at the back of the book)



Figure 12.

PAF‐induced NO production. (A) NO concentration was measured in the suffusate of the hamster cheek pouch using a Nitric Oxide Analyzer 407. (B) Perivenular NO concentration was determined using a NO‐sensitive electrode in the mouse cremaster muscle. NO concentration rose quickly upon topical administration of PAF in both tissues. “From 407 with permission.



Figure 13.

Obligatory role of eNOS‐mediated NO production in PAF‐induced hyperpermeability in vivo. PAF‐induced hyperpermeability is markedly reduced in eNOS−/−mice compared to wild‐type control mice. Typical intravital fluorescence microscopy images of the respective cremaster muscles are shown below the IOI graph.

Adapted from Refs 74 and 410 with permission. (See page 4 in colour section at the back of the book)


Figure 14.

Translocation of eNOS. Top Panel: (A) Isolation of lipid rafts domains was done in control, Ach, or PAF‐treated cells. Fractions were probed against caveolin‐1 and eNOS. (A) ACh treatment. (B) PAF treatment. The blots represent three independent experiments. Bottom Panel: The influence of PAF and ACh on the distribution of eNOS‐GFP was assessed by fluorescence of GFP. The images are representative of three independent experiments. (A) ECV304‐eNOS‐GFP. (B) Human dermal microvascular cells stained with anti‐eNOS (red) and anti‐caveolin‐1 (green) antibodies. From Ref. 434 with permission. (See page 5 in colour section at the back of the book)



Figure 15.

Diagram of eNOS translocation process and eNOS activation. (A) Shows a simplified diagram of the eNOS translocation hypothesis. eNOS is anchored in caveolae through myristoylation and palmitoylation, and is associated with caveolin‐1. Upon stimulation with agonists, eNOS becomes phosphorylated, dissociates from caveolin‐1, and translocates from the caveolae to either the Golgi apparatus or the cytosol. It is postulated that preferential translocation to different compartments is related to vascular function and may allow eNOS to interact with specific function‐related target molecules. Even though the drawing depicts eNOS as being “free” in the cytosol, our recent‐unpublished data indicate that eNOS may be internalized to its intracellular targets via caveolae. (B). Several signals can regulate eNOS activity. While association with caveolin‐1 and phosphorylation of Thr‐497 decrease enzyme activity (NO production), interaction with Ca2+‐calmodulin and HSP90, phosphorylation on Ser‐1177 by Akt, and PKC activation can all increase activity. Panel A redrawn from 410 with permission.



Figure 16.

PAF‐induced eNOS phosphorylation and hyperpermeability in ECV304‐eNOSGFP cells. (A) ACh and PAF activate eNOS by Ser‐1177 phosphorylation and Thr495 dephosphorylation. (B) Inhibition of muscarinic ACh and PAF receptors blocks Ser‐1177 phosphorylation β‐actin served as a (loading) control in experiments shown in (A) and (B). (C) PAF increases permeability in ECVeNOS‐GFP cells. Monolayers of ECVeNOS‐GFP cells were treated with PAF and permeability to FITC‐Dx‐70 was measured. Data are expressed as permeability coefficients (mean ± SEM) for control and PAF‐stimulated monolayers. *P < 0.05, n = 6. From 434 with permission.



Figure 17.

Functions of Rap‐1. The diagram shows known and putative effectors and targets of Rap‐1. Arrows indicate stimulation, blunt ends denote inhibition.

Adapted from Ref. 457 with permission
References
 1. Daniels BS. Regional versus systemic modulation of the microcirculation in diabetes mellitus. J Lab Clin Med 121: 375–376, 1993.
 2. Jaap AJ and Tooke JE. Pathophysiology of microvascular disease in non‐insulin‐dependent diabetes. Clin Sci (Lond) 89: 3–12, 1995.
 3. Mazzoni MC and Schmid‐Schonbein GW. Mechanisms and consequences of cell activation in the microcirculation. Cardiovasc Res 32: 709–719, 1996.
 4. McDonagh PF and Hokama JY. Microvascular perfusion and transport in the diabetic heart. Microcirculation 7: 163–181, 2000.
 5. Michel CC and Curry FE. Microvascular permeability. Physiol Rev 79: 703–761, 1999.
 6. Yamaji T, Fukuhara T and Kinoshita M. Increased capillary permeability to albumin in diabetic rat myocardium. Circ Res 72: 947–957, 1993.
 7. Yuan SY, Breslin JW, Perrin R, Gaudreault N, Guo M, Kargozaran H and Wu MH. Microvascular permeability in diabetes and insulin resistance. Microcirculation 14: 363–373, 2007.
 8. Yuan SY, Ustinova EE, Wu MH, Tinsley JH, Xu W, Korompai FL and Taulman AC. Protein kinase C activation contributes to microvascular barrier dysfunction in the heart at early stages of diabetes. Circ Res 87: 412–417, 2000.
 9. Groeneveld AB. Vascular pharmacology of acute lung injury and acute respiratory distress syndrome. Vascul Pharmacol 39: 247–256, 2002.
 10. Groeneveld AB, van Lambalgen TA and Thijs LG. Microvascular permeability in endotoxin and bacterial shock. Acute Care 12: 195–218, 1986.
 11. Lush CW and Kvietys PR. Microvascular dysfunction in sepsis. Microcirculation 7: 83–101, 2000.
 12. Arturson G. Forty years in burns research‐the postburn inflammatory response. Burns 26: 599–604, 2000.
 13. Cioffi WG. What's new in burns and metabolism. J Am Coll Surg 192: 241–254, 2001.
 14. Gibran NS and Heimbach DM. Current status of burn wound pathophysiology. Clin Plast Surg 27: 11–22, 2000.
 15. Lund T, Onarheim H and Reed RK. Pathogenesis of edema formation in burn injuries. World J Surg 16: 2–9, 1992.
 16. Renkin EM. Capillary transport of macromolecules: pores and other endothelial pathways. J Appl Physiol 58: 315–325, 1985.
 17. Bates DO and Harper SJ. Regulation of vascular permeability by vascular endothelial growth factors. Vascul Pharmacol 39: 225–237, 2003.
 18. Curry FE. Mechanics and thermodynamics of transcapillary exchange. In: Handbook of Physiology: The Cardiovascular System. Microcirculation, eds Renkin EM and Michel CC. Bethesda, MD: American Physiological Society, 1984, pp. 411–466.
 19. Crone C and Levitt DG. Capillary permeability to small solutes. Handbook of Physiology: The Cardiovascular System. Microcirculation. Bethesda, MD: American Physiological Society, 1984, pp. 411–466.
 20. Michel CC. Fluid movements through capillary walls. Handbook of Physiology: The Cardiovascular System. Microcirculation. Bethesda, MD: American Physiological Society, 1984, pp. 375–409.
 21. Taylor AE and Granger DN. Exchange of macrfomolecules across the microcirculation. In: Handbook of Physiology: The Cardiovascular System. Microcirculation, eds Renkin EM and Michel CC. Bethesda, MD: American Physiological Society, 1984, pp. 467–520.
 22. Starling EH. On the absorption of fluids from the connective tissue spaces. J Physiol 19: 312–326, 1896.
 23. Landis EM. Microinjection studies of capillary permeability. II. The relation between capillary pressure and the rate at which fluid passes through the walls of single capillaries. Am J Physiol 82: 217–238, 1927.
 24. Kedem O and Katchalsky A. Thermodynamic analysis of the permeability of biological membranes to non‐electrolytes. Biochim Biophys Acta 27: 229–246, 1958.
 25. Patlak CS, Goldstein DA and Hoffman JF. The flow of solute and solvent across a two‐membrane system. J Theor Biol 5: 426–442, 1963.
 26. Adamson RH, Lenz JF, Zhang X, Adamson GN, Weinbaum S and Curry FE. Oncotic pressures opposing filtration across nonfenestrated rat microvessels. J Physiol 557: 889–907, 2004.
 27. Huxley VH and Williams DA. Basal and adenosine‐mediated protein flux from isolated coronary arterioles. Am J Physiol 271: H1099–H1108, 1996.
 28. Bingaman S, Huxley VH and Rumbaut RE. Fluorescent dyes modify properties of proteins used in microvascular research. Microcirculation 10: 221–231, 2003.
 29. Huang Q and Yuan Y. Interaction of PKC and NOS in signal transduction of microvascular hyperpermeability. Am J Physiol 273: H2442–H2451, 1997.
 30. Wu HM, Huang Q, Yuan Y and Granger HJ. VEGF induces NO‐dependent hyperpermeability in coronary venules. Am J Physiol 271: H2735–H2739, 1996.
 31. Wu HM, Yuan Y, Zawieja DC, Tinsley J and Granger HJ. Role of phospholipase C, protein kinase C, and calcium in VEGF‐induced venular hyperpermeability. Am J Physiol 276: H535–H542, 1999.
 32. Yuan SY, Wu MH, Ustinova EE, Guo M, Tinsley JH, De Lanerolle P and Xu W. Myosin light chain phosphorylation in neutrophil‐stimulated coronary microvascular leakage. Circ Res 90: 1214–1221, 2002.
 33. Yuan Y, Chilian WM, Granger HJ and Zawieja DC. Permeability to albumin in isolated coronary venules. Am J Physiol 265: H543–H552, 1993.
 34. Yuan Y, Granger HJ, Zawieja DC and Chilian WM. Flow modulates coronary venular permeability by a nitric oxide‐related mechanism. Am J Physiol 263: H641–H646, 1992.
 35. Yuan Y, Granger HJ, Zawieja DC, DeFily DV and Chilian WM. Histamine increases venular permeability via a phospholipase C‐NO synthase‐guanylate cyclase cascade. Am J Physiol 264: H1734–H1739, 1993.
 36. Yuan Y, Huang Q and Wu HM. Myosin light chain phosphorylation: modulation of basal and agonist‐stimulated venular permeability. Am J Physiol 272: H1437–H1443, 1997.
 37. Yuan Y, Meng FY, Huang Q, Hawker J and Wu HM. Tyrosine phosphorylation of paxillin/pp125FAK and microvascular endothelial barrier function. Am J Physiol 275: H84–H93, 1998.
 38. Yuan Y, Mier RA, Chilian WM, Zawieja DC and Granger HJ. Interaction of neutrophils and endothelium in isolated coronary venules and arterioles. Am J Physiol 268: H490–H498, 1995.
 39. Crone C and Christensen O. Electrical resistance of a capillary endothelium. J Gen Physiol 77: 349–371, 1981.
 40. Olesen SP, de Saint‐Aubain ML and Bundgaard M. Permeabilities of single arterioles and venules in the frog skin: a functional and morphological study. Microvasc Res 28: 1–22, 1984.
 41. Majno G, Palade GE and Schoefl GI. Studies on inflammation. II. The site of action of histamine and serotonin along the vascular tree: a topographic study. J Biophys Biochem Cytol 11: 607–626, 1961.
 42. Cotran RS and Majno G. The delayed and prolonged vascular leakage in inflammation. I. topography of the leaking vessels after thermal injury. Am J Pathol 45: 261–281, 1964.
 43. Boric MP, Roblero JS and Durán WN. Quantitation of bradykinin‐induced microvascular leakage of FITC‐dextran in rat cremaster muscle. Microvasc Res 33: 397–412, 1987.
 44. Dillon PK and Durán WN. Effect of platelet‐activating factor on microvascular permselectivity: dose‐response relations and pathways of action in the hamster cheek pouch microcirculation. Circ Res 62: 732–740, 1988.
 45. Fox J, Galey F and Wayland H. Action of histamine on the mesenteric microvasculature. Microvasc Res 19: 108–126. 1980.
 46. Gawlowski DM and Durán WN. Dose‐related effects of adenosine and bradykinin on microvascular permselectivity to macromolecules in the hamster cheek pouch. Circ Res 58: 348–355, 1986.
 47. Gawlowski DM, Ritter AB and Durán WN. Reproducibility of microvascular permeability responses to successive topical applications of bradykinin in the hamster cheek pouch. Microvasc Res 24: 354–363, 1982.
 48. Horan KL, Adamski SW, Ayele W, Langone JJ and Grega GJ. Evidence that prolonged histamine suffusions produce transient increases in vascular permeability subsequent to the formation of venular macromolecular leakage sites. Proof of the Majno‐Palade hypothesis. Am J Pathol 123: 570–576, 1986.
 49. Lindbom L. Regulation of vascular permeability by neutrophils in acute inflammation. Chem Immunol Allergy 83: 146–166, 2003.
 50. Svensjo E. The hamster cheek pouch as a model in microcirculation research. Eur Respir J Suppl 12: 595s–600s, 1990, discussion, 600s‐601s.
 51. Svensjo E and Joyner WL. The effects of intermittent and continuous stimulation of microvessels in the cheek pouch of hamsters with histamine and bradykinin on the development of venular leaky sites. Microcirc Endothelium Lymphatics 1: 381–396, 1984.
 52. Wu NZ and Baldwin AL. Transient venular permeability increase and endothelial gap formation induced by histamine. Am J Physiol 262: H1238–H1247, 1992.
 53. Allen TH and Dralpvats PD. Spectrophotometric measurement of traces of the dye T‐1824 by extraction with cellophane from both blood serum and urine of normal dogs. Am J Physiol 154: 27–38, 1948.
 54. Rawson RA. The binding of T‐1824 and structurally related diazo dyes by plasma proteins. Am J Physiol 138: 708–717, 1943.
 55. Miles AA and Miles EM. Vascular reactions to histamine, histamine‐liberator and leukotaxine in the skin of guinea‐pigs. J Physiol 118: 228–257, 1952.
 56. Saria A and Lundberg JM. Evans blue fluorescence: quantitative and morphological evaluation of vascular permeability in animal tissues. J Neurosci Methods 8: 41–49, 1983.
 57. Udaka K, Takeuchi Y and Movat HZ. Simple method for quantitation of enhanced vascular permeability. Proc Soc Exp Biol Med 133: 1384–1387, 1970.
 58. Evans TW, Chung KF, Rogers DF and Barnes PJ. Effect of platelet‐activating factor on airway vascular permeability: possible mechanisms. J Appl Physiol 63: 479–484, 1987.
 59. Sheppard D, Scypinski L, Horn J, Gordon T and Thompson JE. Granulocyte‐mediated airway edema in guinea pigs. J Appl Physiol 60: 1213–1220, 1986.
 60. Sirois MG and Edelman ER. VEGF effect on vascular permeability is mediated by synthesis of platelet‐activating factor. Am J Physiol 272: H2746–H2756, 1997.
 61. Dvorak HF, Nagy JA, Feng D, Brown LF and Dvorak AM. Vascular permeability factor/vascular endothelial growth factor and the significance of microvascular hyperpermeability in angiogenesis. Curr Top Microbiol Immunol 237: 97–132, 1999.
 62. Bates DO and Curry FE. Vascular endothelial growth factor increases hydraulic conductivity of isolated perfused microvessels. Am J Physiol 271: H2520–H2528, 1996.
 63. Bates DO, Lodwick D and Williams B. Vascular endothelial growth factor and microvascular permeability. Microcirculation 6: 83–96, 1999.
 64. Taylor A, Townsley MI and Korthuis RJ. Macromolecule transport across the pulmonary microvessel walls. Exp Lung Res 8: 97–123, 1985.
 65. Levick JR and Michel CC. The permeability of individually perfused frog mesenteric capillaries to T1824 and T1824‐albumin as evidence for a large pore system. Q J Exp Physiol Cogn Med Sci 58: 67–85, 1973.
 66. Durán WN and Dillon PK. Acute microcirculatory effects of platelet‐activating factor. J Lipid Mediat 2 (Suppl): S215–S227, 1990.
 67. Mayhan WG and Joyner WL. The effect of altering the external calcium concentration and a calcium channel blocker, verapamil, on microvascular leaky sites and dextran clearance in the hamster cheek pouch. Microvasc Res 28: 159–179, 1984.
 68. Duling BR. The preparation and use of the hamster cheek pouch for studies of the microcirculation. Microvasc Res 5: 423–429, 1973.
 69. Kim D, Armenante PM and Durán WN. Transient analysis of macromolecular transport across microvascular wall and into interstitium. Am J Physiol 265: H993–H999, 1993.
 70. Ramirez MM, Quardt SM, Kim D, Oshiro H, Minnicozzi M and Durán WN. Platelet activating factor modulates microvascular permeability through nitric oxide synthesis. Microvasc Res 50: 223–234, 1995.
 71. Svensjo E, Arfors KE, Arturson G and Rutili G. The hamster cheek pouch preparation as a model for studies of macromolecular permeability of the microvasculature. Ups J Med Sci 83: 71–79, 1978.
 72. Bekker AY, Ritter AB and Durán WN. Analysis of microvascular permeability to macromolecules by video‐image digital processing. Microvasc Res 38: 200–216, 1989.
 73. Armenante PM, Kim D and Durán WN. Experimental determination of the linear correlation between in vivo TV fluorescence intensity and vascular and tissue FITC‐DX concentrations. Microvasc Res 42: 198–208, 1991.
 74. Hatakeyama T, Pappas PJ, Hobson RW, II, Boric MP, Sessa WC and Durán WN. Endothelial nitric oxide synthase regulates microvascular hyperpermeability in vivo. J Physiol 574: 275–281, 2006.
 75. Bekker AY, Ritter AB and Durán WN. Reduction of pressure in postcapillary venules induced by EPI‐fluorescent illumination of FITC‐dextrans. Microcirc Endothelium Lymphatics 3: 411–423, 1986.
 76. Bekker AY, Dillon PK, Paul J, Ritter AB and Durán WN. Dose‐response relationships between platelet activating factor and permeability‐surface area product of FITC‐dextran 150 in the hamster cheek pouch. Microcirc Endothelium Lymphatics 4: 433–446, 1988.
 77. Svensjo E, Adamski SW, Su K and Grega GJ. Quantitative physiological and morphological aspects of microvascular permeability changes induced by histamine and inhibited by terbutaline. Acta Physiol Scand 116: 265–273, 1982.
 78. Gerlowski LE and Jain RK. Microvascular permeability of normal and neoplastic tissues. Microvasc Res 31: 288–305, 1986.
 79. Huxley VH, Curry FE and Adamson RH. Quantitative fluorescence microscopy on single capillaries: alpha‐lactalbumin transport. Am J Physiol 252: H188–H197, 1987.
 80. Ley K and Arfors KE. Changes in macromolecular permeability by intravascular generation of oxygen‐derived free radicals. Microvasc Res 24: 25–33, 1982.
 81. Ley K and Arfors KE. Segmental differences of microvascular permeability for FITC‐dextrans measured in the hamster cheek pouch. Microvasc Res 31: 84–99, 1986.
 82. Ritter AB, Braun W, Stein A and Durán W. Visualization of the coronary microcirculation using digital image processing. Comput Biol Med 15: 361–374, 1985.
 83. Nugent LJ and Jain RK. Plasma pharmacokinetics and interstitial diffusion of macromolecules in a capillary bed. Am J Physiol 246: H129–H137, 1984.
 84. Fox JR and Wayland H. Interstitial diffusion of macromolecules in the rat mesentery. Microvasc Res 18: 255–276, 1979.
 85. Kim D, Armenante PM and Durán WN. Mathematical modeling of mass transfer in microvascular wall and interstitial space. Microvasc Res 40: 358–378, 1990.
 86. Noel AA, Fallek SR, Hobson RW, II and Durán WN. Inhibition of nitric oxide synthase attenuates primed microvascular permeability in the in vivo microcirculation. J Vasc Surg 22: 661–669, 1995, discussion 669‐670, 1995.
 87. Suval WD, Durán WN, Boric MP, Hobson RW, III, Berendsen PB and Ritter AB. Microvascular transport and endothelial cell alterations preceding skeletal muscle damage in ischemia and reperfusion injury. Am J Surg 154: 211–218, 1987.
 88. Suval WD, Hobson RW, II, Boric MP, Ritter AB and Durán WN. Assessment of ischemia reperfusion injury in skeletal muscle by macromolecular clearance. J Surg Res 42: 550–559, 1987.
 89. Yasuhara H, Hobson RW, II, Dillon PK and Durán WN. A new model for studying ischemia‐reperfusion injury in hamster cheek pouch. Am J Physiol 261: H1626–H1629, 1991.
 90. Suzuki Y, Deitch EA, Mishima S, Durán WN and Xu DZ. Endotoxin‐induced mesenteric microvascular changes involve iNOS‐derived nitric oxide: results from a study using iNOS knock out mice. Shock 13: 397–403, 2000.
 91. Yuan SY. Signal transduction pathways in enhanced microvascular permeability. Microcirculation 7: 395–403, 2000.
 92. Tinsley JH, Ustinova EE, Xu W and Yuan SY. Src‐dependent, neutrophil‐mediated vascular hyperpermeability and beta ‐catenin modification. Am J Physiol Cell Physiol 283: C1745–C1751, 2002.
 93. Tinsley JH, Zawieja DC, Wu MH, Ustinova EE, Xu W and Yuan SY. Protein transfection of intact microvessels specifically modulates vasoreactivity and permeability. J Vasc Res 38: 444–452, 2001.
 94. Jaffe EA, Nachman RL, Becker CG and Minick CR. Culture of human endothelial cells derived from umbilical veins. Identification by morphologic and immunologic criteria. J Clin Invest 52: 2745–2756, 1973.
 95. Gumbleton M and Audus KL. Progress and limitations in the use of in vitro cell cultures to serve as a permeability screen for the blood‐brain barrier. J Pharm Sci 90: 1681–1698, 2001.
 96. Albelda SM, Sampson PM, Haselton FR, McNiff JM, Mueller SN, Williams SK, Fishman AP and Levine EM. Permeability characteristics of cultured endothelial cell monolayers. J Appl Physiol 64: 308–322, 1988.
 97. Schelling ME, Meininger CJ, Hawker JR, Jr. and Granger HJ. Venular endothelial cells from bovine heart. Am J Physiol 254: H1211–H1217, 1988.
 98. Breslin JW and Yuan SY. Involvement of RhoA and Rho kinase in neutrophil‐stimulated endothelial hyperpermeability. Am J Physiol Heart Circ Physiol 286: H1057–H1062, 2004.
 99. Tinsley JH, Wu MH, Ma W, Taulman AC and Yuan SY. Activated neutrophils induce hyperpermeability and phosphorylation of adherens junction proteins in coronary venular endothelial cells. J Biol Chem 274: 24930–24934, 1999.
 100. Tilling T, Korte D, Hoheisel D and Galla HJ. Basement membrane proteins influence brain capillary endothelial barrier function in vitro. J Neurochem 71: 1151–1157, 1998.
 101. Furie MB, Cramer EB, Naprstek BL and Silverstein SC. Cultured endothelial cell monolayers that restrict the transendothelial passage of macromolecules and electrical current. J Cell Biol 98: 1033–1041, 1984.
 102. Navab M, Hough GP, Berliner JA, Frank JA, Fogelman AM, Haberland ME and Edwards PA. Rabbit beta‐migrating very low density lipoprotein increases endothelial macromolecular transport without altering electrical resistance. J Clin Invest 78: 389–397, 1986.
 103. Shasby DM and Shasby SS. Effects of calcium on transendothelial albumin transfer and electrical resistance. J Appl Physiol 60: 71–79, 1986.
 104. Territo M, Berliner JA and Fogelman AM. Effect of monocyte migration on low density lipoprotein transport across aortic endothelial cell monolayers. J Clin Invest 74: 2279–2284, 1984.
 105. Abbruscato TJ and Davis TP. Combination of hypoxia/aglycemia compromises in vitro blood‐brain barrier integrity. J Pharmacol Exp Ther 289: 668–675, 1999.
 106. Raub TJ, Kuentzel SL and Sawada GA. Permeability of bovine brain microvessel endothelial cells in vitro: barrier tightening by a factor released from astroglioma cells. Exp Cell Res 199: 330–340, 1992.
 107. Olesen SP and Crone C. Electrical resistance of muscle capillary endothelium. Biophys J 42: 31–41, 1983.
 108. Crone C and Olesen SP. Electrical resistance of brain microvascular endothelium. Brain Res 241: 49–55, 1982.
 109. Giaever I and Keese CR. Micromotion of mammalian cells measured electrically. Proc Natl Acad Sci USA 88: 7896–7900, 1991.
 110. Giaever I and Keese CR. A morphological biosensor for mammalian cells. Nature 366: 591–592, 1993.
 111. Moy AB, Blackwell K and Kamath A. Differential effects of histamine and thrombin on endothelial barrier function through actin‐myosin tension. Am J Physiol Heart Circ Physiol 282: H21–H29, 2002.
 112. Moy AB, Blackwell K, Wang N, Haxhinasto K, Kasiske MK, Bodmer J, Reyes G and English A. Phorbol ester‐mediated pulmonary artery endothelial barrier dysfunction through regulation of actin cytoskeletal mechanics. Am J Physiol Lung Cell Mol Physiol 287: L153–L167, 2004.
 113. Moy AB, Van Engelenhoven J, Bodmer J, Kamath J, Keese C, Giaever I, Shasby S and Shasby DM. Histamine and thrombin modulate endothelial focal adhesion through centripetal and centrifugal forces. J Clin Invest 97: 1020–1027, 1996.
 114. Moy AB, Winter M. Kamath A, Blackwell K, Reyes G, Giaever I, Keese C and Shasby DM. Histamine alters endothelial barrier function at cell‐cell and cell‐matrix sites. Am J Physiol Lung Cell Mol Physiol 278: L888–L898, 2000.
 115. Tiruppathi C, Malik AB, Del Vecchio PJ, Keese CR and Giaever I. Electrical method for detection of endothelial cell shape change in real time: assessment of endothelial barrier function. Proc Natl Acad Sci USA 89: 7919–7923, 1992.
 116. Lum H and Malik AB. Regulation of vascular endothelial barrier function. Am J Physiol 267: L223–L241, 1994.
 117. McDonagh PF and Williams SK. The preparation and use of fluorescent‐protein conjugates for microvascular research. Microvasc Res 27: 14–27, 1984.
 118. Martin JP and Logsdon N. Oxygen radicals mediate cell inactivation by acridine dyes, fluorescein, and lucifer yellow CH. Photochem Photobiol 46: 45–53, 1987.
 119. Gawlowski DM, Harding NR and Granger HJ. Leukocyte phagocytosis and alterations in microvascular integrity elicited by FITC‐dextran 150 and epi‐illumination in the microcirculation of the hamster cheek pouch. Microvasc Res 37: 1–15, 1989.
 120. Reed MW and Miller FN. Importance of light dose in fluorescent microscopy. Microvasc Res 36: 104–107, 1988.
 121. Vink H and Duling BR. Identification of distinct luminal domains for macromolecules, erythrocytes, and leukocytes within mammalian capillaries. Circ Res 79: 581–589, 1996.
 122. Zhang JL, Yokoyama S and Ohhashi T. Inhibitory effects of fluorescein isothiocyanate photoactivation on lymphatic pump activity. Microvasc Res 54: 99–107, 1997.
 123. Rumbaut RE, Harris NR, Sial AJ, Huxley VH and Granger DN. Leakage responses to L‐NAME differ with the fluorescent dye used to label albumin. Am J Physiol 276: H333–H339, 1999.
 124. Rumbaut RE and Sial AJ. Differential phototoxicity of fluorescent dye‐labeled albumin conjugates. Microcirculation 6: 205–213, 1999.
 125. Pappenheimer JR, Renkin EM and Borrero LM. Filtration, diffusion and molecular sieving through peripheral capillary membranes; a contribution to the pore theory of capillary permeability. Am J Physiol 167: 13–46, 1951.
 126. Renkin EM. Cellular aspects of transvascular exchange: a 40‐year perspective. Microcirculation 1: 157–167, 1994.
 127. Rippe B and Haraldsson B. Transport of macromolecules across microvascular walls: the two‐pore theory. Physiol Rev 74: 163–219. 1994.
 128. Renkin EM, Gustafson Sgro M and Sibley L. Coupling of albumin flux to volume flow in skin and muscles of anesthetized rats. Am J Physiol 255: H458–H466, 1998.
 129. Curry FE and Michel CC. A fiber matrix model of capillary permeability. Microvasc Res 20: 96–99, 1980.
 130. Palade GE. Transport in quanta across the endothelium of blood capillaries. Anat Rec 116: 254, 1960, (Abstract).
 131. Bruns RR and Palade GE. Studies on blood capillaries. II. Transport of ferritin molecules across the wall of muscle capillaries. J Cell Biol 37: 277–299, 1968.
 132. Clough G and Michel CC. The role of vesicles in the transport of ferritin through frog endothelium. J Physiol 315: 127–142, 1981.
 133. Jennings MA and Florey L. An investigation of some properties of endothelium related to capillary permeability. Proc R Soc Lond B Biol Sci 167: 39–63, 1967.
 134. Johansson BR. Size and distribution of endothelial plasmalemmal vesicles in consecutive segments of the microvasculature in cat skeletal muscle. Microvasc Res 17: 107–117, 1979.
 135. Simionescu N, Siminoescu M and Palade GE. Permeability of muscle capillaries to small heme‐peptides. Evidence for the existence of patent transendothelial channels. J Cell Biol 64: 586–607, 1975.
 136. Predescu D, Horvat R, Predescu S and Palade GE. Transcytosis in the continuous endothelium of the myocardial microvasculature is inhibited by N‐ethylmaleimide. Proc Natl Acad Sci USA 91: 3014–3018, 1994.
 137. Schnitzer JE, Oh P, Pinney E and Allard J. Filipin‐sensitive caveolae‐mediated transport in endothelium: reduced transcytosis. scavenger endocytosis, and capillary permeability of select macromolecules. J Cell Biol 127: 1217–1232, 1994.
 138. Simionescu M and Simionescu N. Ultrastructure of the microvascular wall: functional correlations. In: Handbook of Physiology: The Cardiovascular System. Microcirculation, 2nd Ed, eds Renkin EM and Michel CC. Bethesda, MD: American Physiological Society, 1984, pp. 41–101.
 139. Bundgaard M, Frokjaer‐Jensen J and Crone C. Endothelial plasmalemmal vesicles as elements in a system of branching invaginations from the cell surface. Proc Natl Acad Sci USA 76: 6439–6442, 1979.
 140. Frokjaer‐Jensen J. Three‐dimensional organization of plasmalemmal vesicles in endothelial cells. An analysis by serial sectioning of frog mesenteric capillaries. J Ultrastruct Res 73: 9–20, 1980.
 141. Crone C. Recent Advances in Physiology. London: Chruchill Livingstone, 1984.
 142. Reed RK. Transcapillary albumin extravasation in rat skin and skeletal muscle: effect of increased venous pressure. Acta Physiol Scand 134: 375–382, 1988.
 143. Carlsson O, Rosengren BI and Rippe B. Transcytosis inhibitor N‐ethylmaleimide increases microvascular permeability in rat muscle. Am J Physiol Heart Circ Physiol 281: H1728–H1733, 2001.
 144. Rippe B and Taylor A. NEM and filipin increase albumin transport in lung microvessels. Am J Physiol Heart Circ Physiol 280: H34–H41, 2001.
 145. Haraldsson B and Johansson BR. Changes in transcapillary exchange induced by perfusion fixation with glutaraldehyde, followed by measurements of capillary filtration coefficient, diffusion capacity and albumin clearance. Acta Physiol Scand 124: 99–106, 1985.
 146. Clough G. Relationship between microvascular permeability and ultrastructure. Prog Biophys Mol Biol 55: 47–69, 1991.
 147. Dvorak AM and Feng D. The vesiculo‐vacuolar organelle (VVO). A new endothelial cell permeability organelle. J Histochem Cytochem 49: 419–432, 2001.
 148. Feng D, Nagy JA, Brekken RA, Pettersson A, Manseau EJ, Pyne K, Mulligan R, Thorpe PE, Dvorak HF and Dvorak AM. Ultrastructural localization of the vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) receptor‐2 (FLK‐1, KDR) in normal mouse kidney and in the hyperpermeable vessels induced by VPF/VEGF‐expressing tumors and adenoviral vectors. J Histochem Cytochem 48: 545–556, 2000.
 149. Vasile E, Qu H, Dvorak HF and Dvorak AM. Caveolae and vesiculo‐vacuolar organelles in bovine capillary endothelial cells cultured with VPF/VEGF on floating Matrigel‐collagen gels. J Histochem Cytochem 47: 159–167, 1999.
 150. Minshall RD, Tiruppathi C, Vogel SM, Niles WD, Gilchrist A, Hamm HE and Malik AB. Endothelial cell‐surface gp60 activates vesicle formation and trafficking via G(i)‐coupled Src kinase signaling pathway. J Cell Biol 150: 1057–1070, 2000.
 151. Vogel SM, Minshall RD, Pilipovic M, Tiruppathi C and Malik AB. Albumin uptake and transcytosis in endothelial cells in vivo induced by albumin‐binding protein. Am J Physiol Lung Cell Mol Physiol 281: L1512–L1522, 2001.
 152. Holmang A, Bjorntorp P and Rippe B. Tissue uptake of insulin and inulin in red and white skeletal muscle in vivo. Am J Physiol 263: H1170–H1176, 1992.
 153. Vasile E, Simionescu M and Simionescu N. Visualization of the binding, endocytosis. and transcytosis of low‐density lipoprotein in the arterial endothelium in situ. J Cell Biol 96: 1677–1689, 1983.
 154. Rutledge JC. Temperature and hydrostatic pressure‐dependent pathways of low‐density lipoprotein transport across microvascular barrier. Am J Physiol 262: H234–H245, 1992.
 155. Chambers R and Zweifach BW. Intercellular cement and capillary permeability. Physiol Rev 27: 436–463, 1947.
 156. Majno G and Palade G. Studies on inflammation I. The effect of histamine and serotonin on vascular permeability: An electron microscopic study. J Biophys Cytol 11: 597–605, 1961.
 157. Dudek SM and Garcia JG. Cytoskeletal regulation of pulmonary vascular permeability. J Appl Physiol 91: 1487–1500, 2001.
 158. Garcia JG, Davis HW and Patterson CE. Regulation of endothelial cell gap formation and barrier dysfunction: role of myosin light chain phosphorylation. J Cell Physiol 163: 510–522, 1995.
 159. Moy AB, Sheldon R, Lindsley K, Shasby S and Shasby DM. Centripetal tension and endothelial retraction. Chest 105: 107S–108S, 1994.
 160. Yuan SY. Protein kinase signaling in the modulation of microvascular permeability. Vascul Pharmacol 39: 213–223, 2003.
 161. Hixenbaugh EA, Goeckeler ZM, Papaiya NN, Wysolmerski RB, Silverstein SC and Huang AJ. Stimulated neutrophils induce myosin light chain phosphorylation and isometric tension in endothelial cells. Am J Physiol 213: H981–H988, 1997.
 162. Kolodney MS and Wysolmerski RB. Isometric contraction by fibroblasts and endothelial cells in tissue culture: A quantitative study. J Cell Biol 117: 73–82, 1992.
 163. Harhaj NS and Antonetti DA. Regulation of tight junctions and loss of barrier function in pathophysiology. Int J Biochem Cell Biol 36: 1206–1237, 2004.
 164. Bazzoni G and Dejana E. Endothelial cell‐to‐cell junctions: molecular organization and role in vascular homeostasis. Physiol Rev 84: 869–901, 2004.
 165. Beyer EC, Gemel J, Seul KH, Larson DM, Banach K and Brink PR. Modulation of intercellular communication by differential regulation and heteromeric mixing of co‐expressed connexins. Braz J Med Biol Res 33: 391–397, 2000.
 166. Kevil CG, Okayama N, Trocha SD, Kalogeris TJ, Coe LL, Specian RD, Davis CP and Alexander JS. Expression of zonula occludens and adherens junctional proteins in human venous and arterial endothelial cells: role of occludin in endothelial solute barriers. Microcirculation 5: 197–210, 1998.
 167. Hirase T, Staddon JM, Saitou M, Ando‐Akatsuka Y, Itoh M, Furuse M, Fujimoto K, Tsukita S and Rubin LL. Occludin as a possible determinant of tight junction permeability in endothelial cells. J Cell Sci 110 (Pt 14): 1603–1613, 1997.
 168. DeMaio L, Chang YS, Gardner TW, Tarbell JM and Antonetti DA. Shear stress regulates occludin content and phosphorylation. Am J Physiol Heart Circ Physiol 281: H105–H113, 2001.
 169. Antonetti DA, Barber AJ, Khin S, Lieth E, Tarbell JM and Gardner TW. Vascular permeability in experimental diabetes is associated with reduced endothelial occludin content: vascular endothelial growth factor decreases occludin in retinal endothelial cells. Penn State Retina Research Group. Diabetes 47: 1953–1959, 1998.
 170. Muresan Z, Paul DL and Goodenough DA. Occludin 1B, a variant of the tight junction protein occludin. Mol Biol Cell 11: 627–634, 2000.
 171. Antonetti DA, Barber AJ, Hollinger LA, Wolpert EB and Gardner TW. Vascular endothelial growth factor induces rapid phosphorylation of tight junction proteins occludin and zonula occluden 1. A potential mechanism for vascular permeability in diabetic retinopathy and tumors. J Biol Chem 274: 23463–23467, 1999.
 172. Nitta T, Hata M, Gotoh S, Seo Y, Sasaki H, Hashimoto N, Furuse M and Tsukita S. Size‐selective loosening of the blood‐brain barrier in claudin‐5‐deficient mice. J Cell Biol 161: 653–660, 2003.
 173. Corada M, Liao F, Lindgren M, Lampugnani MG, Breviario F, Frank R, Muller WA, Hicklin DJ, Bohlen P and Dejana E. Monoclonal antibodies directed to different regions of vascular endothelial cadherin extracellular domain affect adhesion and clustering of the protein and modulate endothelial permeability. Blood 97: 1679–1684, 2001.
 174. Corada M, Mariotti M, Thurston G, Smith K, Kunkel R, Brockhaus M, Lampugnani MG, Martin‐Padura I, Stoppacciaro A, Ruco L, McDonald DM, Ward PA and Dejana E. Vascular endothelial‐cadherin is an important determinant of microvascular integrity in vivo. Proc Natl Acad Sci USA 96: 9815–9820, 1999.
 175. Guo M, Wu MH, Granger HJ and Yuan SY. Transference of recombinant VE‐cadherin cytoplasmic domain alters endothelial junctional integrity and porcine microvascular permeability. J Physiol 554: 78–88, 2004.
 176. Simon AM and McWhorter AR. Role of connexin37 and connexin40 in vascular development. Cell Commun Adhes 10: 379–385, 2003.
 177. Simon AM and McWhorter AR. Vascular abnormalities in mice lacking the endothelial gap junction proteins connexin37 and connexin40. Dev Biol 251: 206–220, 2002.
 178. Suarez S and Ballmer‐Hofer K. VEGF transiently disrupts gap junctional communication in endothelial cells. J Cell Sci 114: 1229–1235, 2001.
 179. Thuringer D. The vascular endothelial growth factor‐induced disruption of gap junctions is relayed by an autocrine communication via ATP release in coronary capillary endothelium. Ann N Y Acad Sci 1030: 14–27, 2004.
 180. Agre P, Homer, W. Smith award lecture. Aquaporin water channels in kidney. J Am Soc Nephrol 11: 764–777, 2000.
 181. King LS, Nielsen S, Agre P and Brown RH. Decreased pulmonary vascular permeability in aquaporin‐1‐null humans. Proc Natl Acad Sci USA 99: 1059–1063, 2002.
 182. Verkman AS. Aquaporin water channels and endothelial cell function. J Anat 200: 617–627, 2002.
 183. de Weerd WF and Leeb‐Lundberg LM. Bradykinin sequesters B2 bradykinin receptors and the receptor‐coupled Galpha subunits Galphaq and Galphai in caveolae in DDT1 MF‐2 smooth muscle cells. J Biol Chem 272: 17858–17866, 1997.
 184. Snyder F. Platelet‐activating factor and related acetylated lipids as potent biologically active cellular mediators. Am J Physiol 259: C697–C708, 1990.
 185. Millauer B, Wizigmann‐Voos S, Schnurch H, Martinez R, Moller NP, Risau W and Ullrich A. High affinity VEGF binding and developmental expression suggest Flk‐1 as a major regulator of vasculogenesis and angiogenesis. Cell 72: 835–846, 1993.
 186. Quinn TP, Peters KG, De Vries C, Ferrara N and Williams LT. Fetal liver kinase 1 is a receptor for vascular endothelial growth factor and is selectively expressed in vascular endothelium. Proc Natl Acad Sci USA 90: 7533–7537, 1993.
 187. Izumi T and Shimizu T. Platelet‐activating factor receptor: gene expression and signal transduction. Biochim Biophys Acta 1259: 317–333, 1995.
 188. Kuruvilla A, Pielop C and Shearer WT. Platelet‐activating factor induces the tyrosine phosphorylation and activation of phospholipase C‐gamma 1, Fyn and Lyn kinases, and phosphatidylinositol 3‐kinase in a human B cell line. J Immunol 153: 5433–5442, 1994.
 189. Kroll J and Waltenberger J. The vascular endothelial growth factor receptor KDR activates multiple signal transduction pathways in porcine aortic endothelial cells. J Biol Chem 272: 32521–32527, 1997.
 190. Guo D, Jia Q, Song HY, Warren RS and Donner DB. Vascular endothelial cell growth factor promotes tyrosine phosphorylation of mediators of signal transduction that contain SH2 domains. Association with endothelial cell proliferation. J Biol Chem 270: 6729–6733, 1995.
 191. Mukhopadhyay D, Nagy JA, Manseau EJ and Dvorak HF. Vascular permeability factor/vascular endothelial growth factor‐mediated signaling in mouse mesentery vascular endothelium. Cancer Res 58: 1278–1284, 1998.
 192. Breslin JW, Gaudreault N, Watson KD, Reynoso R, Yuan SY and Wu MH. Vascular endothelial growth factor‐C stimulates the lymphatic pump by a VEGF receptor‐3‐dependent mechanism. Am J Physiol Heart Circ Physiol 293: H709–H718, 2007.
 193. Breslin JW, Yuan SY and Wu MH. VEGF‐C alters barrier function of cultured lymphatic endothelial cells through a vEGFR‐3 dependent mechanism. Lymphat Res Biol 5: 105–113, 2007.
 194. Hillman NJ, Whittles CE, Pocock TM, Williams B and Bates DO. Differential effects of vascular endothelial growth factor‐C and placental growth factor‐1 on the hydraulic conductivity of frog mesenteric capillaries. J Vasc Res 38: 176–186, 2001.
 195. Tomeo AC and Durán WN. Resistance and exchange microvessels are modulated by different PAF receptors. Am J Physiol 261: H1648–H1652, 1991.
 196. McLeod RL, Mingo GG, Herczku C, DeGennaro‐Culver F, Kreutner W, Egan RW and Hey JA. Combined histamine H1 and H3 receptor blockade produces nasal decongestion in an experimental model of nasal congestion. Am J Rhinol 13: 391–399, 1999.
 197. McLeod RL, Mingo GG, Kreutner W and Hey JA. Effect of combined histamine H1 and H3 receptor blockade on cutaneous microvascular permeability elicited by compound 48/80. Life Sci 76: 1787–1794, 2005.
 198. Lampugnani MG, Zanetti A, Breviario F, Balconi G, Orsenigo F, Corada M, Spagnuolo R, Betson M, Braga V and Dejana E. VE‐cadherin regulates endothelial actin activating Rac and increasing membrane association of Tiam. Mol Biol Cell 13: 1175–1189, 2002.
 199. Kim D and Durán WN. Platelet‐activating factor stimulates protein tyrosine kinase in hamster cheek pouch microcirculation. Am J Physiol 268: H399–H403, 1995.
 200. Newton AC. Regulation of protein kinase C. Curr Opin Cell Biol 9: 161–167, 1997.
 201. Tinsley JH, Teasdale NR and Yuan SY. Involvement of PKCdelta and PKD in pulmonary microvascular endothelial cell hyperpermeability. Am J Physiol Cell Physiol 286: C105–C111, 2004.
 202. Johnson A, Hocking DC and Ferro TJ. Mechanisms of pulmonary edema induced by a diacylglycerol second messenger. Am J Physiol 258: H85–H91, 1990.
 203. Kobayashi I, Kim D, Hobson RW, II and Durán WN. Platelet‐activating factor modulates microvascular transport by stimulation of protein kinase C. Am J Physiol 266: H1214–H1220, 1994.
 204. Lynch JJ, Ferro TJ, Blumenstock FA, Brockenauer AM and Malik AB. Increased endothelial albumin permeability mediated by protein kinase C activation. J Clin Invest 85: 1991–1998, 1990.
 205. Pocock TM, Foster RR and Bates DO. Evidence of a role for TRPC channels in VEGF‐mediated increased vascular permeability in vivo. Am J Physiol Heart Circ Physiol 286: H1015–H1026, 2004.
 206. Ramirez MM, Kim DD and Durán WN. Protein kinase C modulates microvascular permeability through nitric oxide synthase. Am J Physiol 271: H1702–H1705, 1996.
 207. Aramoto H, Breslin JW, Pappas PJ, Hobson RW, II and Durán WN. Vascular endothelial growth factor stimulates differential signaling pathways in in vivo microcirculation. Am J Physiol Heart Circ Physiol 287: H1590–H1598, 2004.
 208. Murray MA, Heistad DD and Mayhan WG. Role of protein kinase C in bradykinin‐induced increases in microvascular permeability. Circ Res 68: 1340–1348, 1991.
 209. Johnson A, Phillips P, Hocking D, Tsan MF and Ferro T. Protein kinase inhibitor prevents pulmonary edema in response to H2O2. Am J Physiol 256: H1012–H1022, 1989.
 210. Tanita T, Song C, Kubo H, Ono S and Fujimura S. Endothelial signal transduction system enhances neutrophil‐induced pulmonary vascular permeability. Eur Respir J 15: 452–458, 2000.
 211. Breslin JW, Pappas PJ, Cerveira JJ, Hobson RW, II and Durán WN. VEGF Increases endothelial permeability by separate signaling pathways involving ERK‐1/2 and nitric oxide. Am J Physiol Heart Circ Physiol 284: H92–H100, 2003.
 212. Siflinger‐Birnboim A, Goligorsky MS, Del Vecchio PJ and Malik AB. Activation of protein kinase C pathway contributes to hydrogen peroxide‐induced increase in endothelial permeability. Lab Invest 67: 24–30, 1992.
 213. Tinsley JH, Breslin JW, Teasdale NR and Yuan SY. PKC‐dependent. burn‐induced adherens junction reorganization and barrier dysfunction in pulmonary microvascular endothelial cells. Am J Physiol. 2005, ePub ahead of print, PMID: 15821015, 2005.
 214. Xia P, Aiello LP, Ishii H, Jiang ZY, Park DJ, Robinson GS, Takagi H, Newsome WP, Jirousek MR and King GL. Characterization of vascular endothelial growth factor's effect on the activation of protein kinase C, its isoforms, and endothelial cell growth. J Clin Invest 98: 2018–2026, 1996.
 215. Kim DD, Ramirez MM and Durán WN. Platelet‐activating factor modulates microvascular dynamics through phospholipase C in the hamster cheek pouch. Microvasc Res 59: 7–13, 2000.
 216. Bates DO and Curry FE. Vascular endothelial growth factor increases microvascular permeability via a Ca(2 + (‐dependent pathway. Am J Physiol 273: H687–H694, 1997.
 217. Sandoval R, Malik AB, Minshall RD, Kouklis P, Ellis CA and Tiruppathi C. Ca(2 +) signalling and PKCalpha activate increased endothelial permeability by disassembly of VE‐cadherin junctions. J Physiol 533: 433–445, 2001.
 218. Sandoval R, Malik AB, Naqvi T, Mehta D and Tiruppathi C. Requirement for Ca2+ signaling in the mechanism of thrombin‐induced increase in endothelial permeability. Am J Physiol Lung Cell Mol Physiol 280: L239–L247, 2001.
 219. He P, Pagakis SN and Curry FE. Measurement of cytoplasmic calcium in single microvessels with increased permeability. Am J Physiol 258: H1366–H1374, 1990.
 220. Glass CA, Pocock TM, Curry FE and Bates DO. Cytosolic Ca2+ concentration and the rate of increase of the cytosolic Ca2+ concentration in the regulation of vascular permeability in Rana in vivo. J Physiol, 2005.
 221. Pocock TM, Williams B, Curry FE and Bates DO. VEGF and ATP act by different mechanisms to increase microvascular permeability and endothelial [Ca(2 +](i). Am J Physiol Heart Circ Physiol 279: H1625–H1634, 2000.
 222. Ferro T, Neumann P, Gertzberg N, Clements R and Johnson A. Protein kinase C‐alpha mediates endothelial barrier dysfunction induced by TNF‐alpha. Am J Physiol Lung Cell Mol Physiol 278: L1107–L1117, 2000.
 223. Mehta D, Rahman A and Malik AB. Protein kinase C‐alpha signals rho‐guanine nucleotide dissociation inhibitor phosphorylation and rho activation and regulates the endothelial cell barrier function. J Biol Chem 276: 22614–22620, 2001.
 224. Nagpala PG, Malik AB, Vuong PT and Lum H. Protein kinase C beta 1 overexpression augments phorbol ester‐induced increase in endothelial permeability. J Cell Physiol 166: 249–255, 1996.
 225. Jirousek MR, Gillig JR, Gonzalez CM, Heath WF, McDonald JH, III, Neel DA, Rito CJ, Singh U, Stramm LE, Melikian‐Badalian A, Baevsky M, Ballas LM, Hall SE, Winneroski LL and Faul MM. (S)‐13‐[dimethylamino)methyl]‐10,11,14,15‐tetrahydro‐4,9:16, 21‐dimetheno‐1H, 13H‐dibenzo[e,k]pyrrolo[3,4‐h][1,4,13]oxadi azacyclohexadecene‐l,3(2H)‐d ione (LY333531) and related analogues: isozyme selective inhibitors of protein kinase C beta. J Med Chem 39: 2664–2671, 1979.
 226. Aiello LP, Bursell SE, Clermont A, Duh E, Ishii H, Takagi C, Mori F, Ciulla TA, Ways K, Jirousek M, Smith LE and King GL. Vascular endothelial growth factor‐induced retinal permeability is mediated by protein kinase C in vivo and suppressed by an orally effective beta‐isoform‐selective inhibitor. Diabetes 46: 1473–1480, 1997.
 227. Ishii H, Jirousek MR, Koya D, Takagi C, Xia P, Clermont A, Bursell SE, Kern TS, Ballas LM, Heath WF, Stramm LE, Feener EP and King GL. Amelioration of vascular dysfunctions in diabetic rats by an oral PKC beta inhibitor. Science 272: 728–731, 1996.
 228. Koya D, Haneda M, Nakagawa H, Isshiki K, Sato H, Maeda S, Sugimoto T, Yasuda H, Kashiwagi A, Ways DK, King GL and Kikkawa R. Amelioration of accelerated diabetic mesangial expansion by treatment with a PKC beta inhibitor in diabetic db/db mice, a rodent model for type 2 diabetes. FASEB J 14: 439–447, 2000.
 229. Guo M, Wu MH, Korompai F and Yuan SY. Upregulation of PKC genes and isozymes in cardiovascular tissues during early stages of experimental diabetes. Physiol Genomics 12: 139–146, 2003.
 230. Comer GM and Ciulla TA. Pharmacotherapy for diabetic retinopathy. Curr Opin Ophthalmol 15: 508–518, 2004.
 231. Tuttle KR and Anderson PW. A novel potential therapy for diabetic nephropathy and vascular complications: protein kinase C beta inhibition. Am J Kidney Dis 42: 456–465, 2003.
 232. Kim H, Sasaki T, Maeda K, Koya D, Kashiwagi A and Yasuda H. Protein kinase Cbeta selective inhibitor LY333531 attenuates diabetic hyperalgesia through ameliorating cGMP level of dorsal root ganglion neurons. Diabetes 52: 2102–2109, 2003.
 233. Stasek JE, Jr., Patterson CE and Garcia JG. Protein kinase C phosphorylates caldesmon77 and vimentin and enhances albumin permeability across cultured bovine pulmonary artery endothelial cell monolayers. J Cell Physiol 153: 62–75, 1992.
 234. Abedi H and Zachary I. Vascular endothelial growth factor stimulates tyrosine phosphorylation and recruitment to new focal adhesions of focal adhesion kinase and paxillin in endothelial cells. J Biol Chem 272: 15442–15451, 1997.
 235. Alexander JS, Jackson SA, Chaney E, Kevil CG and Haselton FR. The role of cadherin endocytosis in endothelial barrier regulation: involvement of protein kinase C and actin‐cadherin interactions. Inflammation 22: 419–433, 1998.
 236. Moore TM, Chetham PM, Kelly JJ and Stevens T. Signal transduction and regulation of lung endothelial cell permeability. Interaction between calcium and cAMP. Am J Physiol 275: L203–L222, 1998.
 237. He P, Zeng M and Curry FE. Dominant role of cAMP in regulation of microvessel permeability. Am J Physiol Heart Circ Physiol 278: H1124–H1133, 2000.
 238. Adamson RH, Liu B, Fry GN, Rubin LL and Curry FE. Microvascular permeability and number of tight junctions are modulated by cAMP. Am J Physiol 274: H1885–H1894, 1998.
 239. Cobb MH. MAP kinase pathways. Prog Biophys Mol Biol 71: 479–500, 1999.
 240. Hood J and Granger HJ. Protein kinase G mediates vascular endothelial growth factor‐induced Raf‐1 activation and proliferation in human endothelial cells. J Biol Chem 273: 23504–23508, 1998.
 241. Parenti A. Morbidelli L, Cui XL, Douglas JG, Hood JD, Granger HJ, Ledda F and Ziche M. Nitric oxide is an upstream signal of vascular endothelial growth factor‐induced extracellular signal‐regulated kinase 1/2 activation in postcapillary endothelium. J Biol Chem 273: 4220–4226, 1998.
 242. Kooistra MR, Corada M, Dejana E and Bos JL. Epacl regulates integrity of endothelial cell junctions through VE‐cadherin. FEBS Lett 579: 4966–4972, 2005.
 243. DeFouw LM and DeFouw DO. Differential phosphodiesterase activity contributes to restrictive endothelial barrier function during angiogenesis. Microvasc Res 62: 263–270, 2001.
 244. Sarker MH and Fraser PA. The role of guanylyl cyclases in the permeability response to inflammatory mediators in pial venular capillaries in the rat. J Physiol 540: 209–218, 2002.
 245. Mayhan WG. Nitric oxide accounts for histamine‐induced increases in macromolecular extravasation. Am J Physiol 266: H2369–H2373, 1994.
 246. Meyer DJ, Jr. and Huxley VH. Capillary hydraulic conductivity is elevated by cGMP‐dependent vasodilators. Circ Res 70: 382–391, 1992.
 247. He P, Zeng M and Curry FE. cGMP modulates basal and activated microvessel permeability independently of [Ca2 +]i. Am J Physiol 274: H1865–H1874, 1998.
 248. Varma S, Breslin JW, Lal BK, Pappas PJ, Hobson RW, II and Durán WN. p42/44MAPK regulates baseline permeability and cGMP‐induced hyperpermeability in endothelial cells. Microvasc Res 63: 172–178, 2002.
 249. Thakker GD, Hajjar DP, Muller WA and Rosengart TK. The role of phosphatidylinositol 3‐kinase in vascular endothelial growth factor signaling. J Biol Chem 274: 10002–10007, 1999.
 250. Conus NM, Hannan KM, Cristiano BE, Hemmings BA and Pearson RB. Direct identification of tyrosine 474 as a regulatory phosphorylation site for the Akt protein kinase. J Biol Chem 277: 38021–38028, 2002.
 251. Downward J. Mechanisms and consequences of activation of protein kinase B/Akt. Curr Opin Cell Biol 10: 262–267, 1998.
 252. Hemmings BA. Akt signaling: linking membrane events to life and death decisions. Science 275: 628–630, 1997.
 253. Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell 103: 211–225, 2000.
 254. Lal BK, Varma S, Pappas PJ, Hobson RW, II and Durán WN. VEGF increases permeability of the endothelial cell monolayer by activation of PKB/akt, endothelial nitric‐oxide synthase, and MAP kinase pathways. Microvasc Res 62: 252–262, 2001.
 255. Bauer PM, Yu J, Chen Y, Hickey R, Bernatchez PN, Looft‐Wilson R, Huang Y, Giordano F, Stan RV and Sessa WC. Endothelial‐specific expression of caveolin‐1 impairs microvascular permeability and angiogenesis. Proc Natl Acad Sci USA 102: 204–209, 2005.
 256. Chang L and Karin M. Mammalian MAP kinase signalling cascades. Nature 410: 37–40, 2001.
 257. Chuang E, Barnard D, Hettich L, Zhang XF, Avruch J and Marshall MS. Critical binding and regulatory interactions between Ras and Raf occur through a small, stable N‐terminal domain of Raf and specific Ras effector residues. Mol Cell Biol 14: 5318–5325, 1994.
 258. English J, Pearson G, Wilsbacher J, Swantek, Karandikar M, Xu S and Cobb MH. New insights into the control of MAP kinase pathways. Exp Cell Res 253: 255–270, 1999.
 259. Garrington TP and Johnson GL. Organization and regulation of mitogen‐activated protein kinase signaling pathways. Curr Opin Cell Biol 11: 211–218, 1999.
 260. Mischak H, Seitz T, Janosch P, Eulitz M, Steen H, Schellerer M, Philipp A and Kolch W. Negative regulation of Raf‐1 by phosphorylation of serine 621. Mol Cell Biol 16: 5409–5418, 1996.
 261. Wu J, Dent P, Jelinek T, Wolfman A, Weber MJ and Sturgill TW. Inhibition of the EGF‐activated MAP kinase signaling pathway by adenosine 3′.5′‐monophosphate. Science 262: 1065–1069, 1993.
 262. Hagemann C and Rapp UR. Isotype‐specific functions of Raf kinases. Exp Cell Res 253: 34–46, 1999.
 263. Becker PM, Verin AD, Booth MA, Liu F, Birukova A and Garcia JG. Differential regulation of diverse physiological responses to VEGF in pulmonary endothelial cells. Am J Physiol Lung Cell Mol Physiol 281: L1500–1511, 2001.
 264. Kevil CG, Oshima T, Alexander B, Coe LL and Alexander JS. H(2)O(2)‐mediated permeability: role of MAPK and occludin. Am J Physiol Cell Physiol 279: C21–30, 2000.
 265. Kevil CG, Payne DK, Mire E and Alexander JS. Vascular permeability factor/vascular endothelial cell growth factor‐mediated permeability occurs through disorganization of endothelial junctional proteins. J Biol Chem 273: 15099–15103, 1998.
 266. Yu P, Hatakeyama T, Aramoto H, Miyata T, Shigematsu H, Nagawa H, Hobson RW and Durán WN. Mitogen‐activated protein kinases regulate platelet‐activating factor‐induced hyperpermeability. Microcirculation 12: 637–643, 2005.
 267. Verin AD, Liu F, Bogatcheva N, Borbiev T, Hershenson MB, Wang P and Garcia JG. Role of ras‐dependent ERK activation in phorbol ester‐induced endothelial cell barrier dysfunction. Am J Physiol Lung Cell Mol Physiol 279: L360–370, 2000.
 268. Wu MH, Yuan SY and Granger HJ. The protein kinase MEK1/2 mediate vascular endothelial growth factor‐ and histamine‐induced hyperpermeability in porcine coronary venules. J Physiol 563: 95–104, 2005.
 269. Garcia JG, Wang P, Schaphorst KL, Becker PM, Borbiev T, Liu F, Birukova A, Jacobs K, Bogatcheva N and Verin AD. Critical involvement of p38 MAP kinase in pertussis toxin‐induced cytoskeletal reorganization and lung permeability. FASEB J 16: 1064–1076, 2002.
 270. Issbrucker K, Marti HH, Hippenstiel S, Springmann G, Voswinckel R, Gaumann A, Breier G, Drexler HC, Suttorp N and Clauss M. p38 MAP kinase ‐ a molecular switch between VEGF‐induced angiogenesis and vascular hyperpermeability. FASEB J 17: 262–264, 2003.
 271. Niwa K, Inanami O, Ohta T, Ito S, Karino T and Kuwabara M. p38 MAPK and Ca2+ contribute to hydrogen peroxide‐induced increase of permeability in vascular endothelial cells but ERK does not. Free Radic Res 35: 519–527, 2001.
 272. Nwariaku FE, Chang J, Zhu X, Liu Z, Duffy SL, Halaihel NH, Terada L and Turnage RH. The role of p38 map kinase in tumor necrosis factor‐induced redistribution of vascular endothelial cadherin and increased endothelial permeability. Shock 18: 82–85. 2002.
 273. Kiemer AK, Weber NC, Furst R, Bildner N, Kulhanek‐Heinze S and Vollmar AM. Inhibition of p38 MAPK activation via induction of MKP‐1: atrial natriuretic peptide reduces TNF‐alpha‐induced actin polymerization and endothelial permeability. Circ Res 90: 874–881, 2002.
 274. Shi S, Garcia JG, Roy S, Parinandi NL and Natarajan V. Involvement of c‐Src in diperoxovanadate‐induced endothelial cell barrier dysfunction. Am J Physiol Lung Cell Mol Physiol 279: L441–451, 2000.
 275. Thomas SM and Brugge JS. Cellular functions regulated by Src family kinases. Annu Rev Cell Dev Biol 13: 513–609, 1997.
 276. Kevil CG, Okayama N and Alexander JS. H(2)O(2)‐mediated permeability II: importance of tyrosine phosphatase and kinase activity. Am J Physiol Cell Physiol 281: C1940–1947, 2001.
 277. Nwariaku FE, Liu Z, Zhu X, Turnage RH, Sarosi GA and Terada LS. Tyrosine phosphorylation of vascular endothelial cadherin and the regulation of microvascular permeability. Surgery 132: 180–185, 2002.
 278. Eliceiri BP, Paul R, Schwartzberg PL, Hood JD, Leng J and Cheresh DA. Selective requirement for Src kinases during VEGF‐induced angiogenesis and vascular permeability. Mol Cell 4: 915–924, 1999.
 279. Paul R, Zhang ZG, Eliceiri BP, Jiang Q, Boccia AD, Zhang RL, Chopp M and Cheresh DA. Src deficiency or blockade of Src activity in mice provides cerebral protection following stroke. Nat Med 7: 222–227, 2001.
 280. Tiruppathi C, Song W, Bergenfeldt M, Sass P and Malik AB. Gp60 activation mediates albumin transcytosis in endothelial cells by tyrosine kinase‐dependent pathway. J Biol Chem 272: 25968–25975, 1997.
 281. Shajahan AN, Timblin BK, Sandoval R, Tiruppathi C, Malik AB and Minshall RD. Role of Src‐induced dynamin‐2 phosphorylation in caveolae‐mediated endocytosis in endothelial cells. J Biol Chem 279: 20392–20400, 2004.
 282. Birukov KG, Csortos C, Marzilli L, Dudek S. Ma SF, Bresnick AR. Verin AD, Cotter RJ and Garcia JG. Differential regulation of alternatively spliced endothelial cell myosin light chain kinase isoforms by p60(Src). J Biol Chem 276: 8567–8573, 2001.
 283. Dudek SM, Birukov KG, Zhan X and Garcia JG. Novel interaction of cortactin with endothelial cell myosin light chain kinase. Biochem Biophys Res Commun 298: 511–519, 2002.
 284. Garcia JG, Verin AD, Schaphorst K, Siddiqui R, Patterson CE, Csortos C and Natarajan V. Regulation of endothelial cell myosin light chain kinase by Rho, cortactin, and p60(src). Am J Physiol 276: L989–998, 1999.
 285. Mucha DR, Myers CL and Schaeffer RC, Jr. Endothelial contraction and monolayer hyperpermeability are regulated by Src kinase. Am J Physiol Heart Circ Physiol 284: H994–H1002, 2003.
 286. Gumbiner BM. Signal transduction of beta‐catenin. Curr Opin Cell Biol 7: 634–640, 1995.
 287. Eliceiri BP, Puente XS, Hood JD, Stupack DG, Schlaepfer DD, Huang XZ, Sheppard D and Cheresh DA. Src‐mediated coupling of focal adhesion kinase to integrin alpha(v)beta5 in vascular endothelial growth factor signaling. J Cell Biol 157: 149–160, 2002.
 288. Garcia JG, Schaphorst KL, Verin AD, Vepa S, Patterson CE and Natarajan V. Diperoxovanadate alters endothelial cell focal contacts and barrier function: role of tyrosine phosphorylation. J Appl Physiol 89: 2333–2343, 2000.
 289. Wang P, Verin AD, Birukova A, Gilbert‐McClain LI, Jacobs K and Garcia JG. Mechanisms of sodium fluoride‐induced endothelial cell barrier dysfunction: role of MLC phosphorylation. Am J Physiol Lung Cell Mol Physiol 281: L1472–1483, 2001.
 290. Cruz A, DeFouw LM and DeFouw DO. Restrictive endothelial barrier function during normal angiogenesis in vivo: partial dependence on tyrosine dephosphorylation of beta‐catenin. Microvasc Res 59: 195–203, 2000.
 291. Birukova AA, Smurova K, Birukov KG, Kaibuchi K, Garcia JG and Verin AD. Role of Rho GTPases in thrombin‐induced lung vascular endothelial cells barrier dysfunction. Microvasc Res 67: 64–77, 2004.
 292. Verin AD, Patterson CE, Day MA and Garcia JG. Regulation of endothelial cell gap formation and barrier function by myosin‐associated phosphatase activities. Am J Physiol 269: L99–108, 1995.
 293. Tar K, Birukova AA, Csortos C, Bako E, Garcia JG and Verin AD. Phosphatase 2A is involved in endothelial cell microtubule remodeling and barrier regulation. J Cell Biochem 92: 534–546, 2004.
 294. Lum H, Podolski JL, Gurnack ME, Schulz IT, Huang F and Holian O. Protein phosphatase 2B inhibitor potentiates endothelial PKC activity and barrier dysfunction. Am J Physiol Lung Cell Mol Physiol 281: L546–555, 2001.
 295. Abbi S and Guan JL. Focal adhesion kinase: protein interactions and cellular functions. Histol Histopathol 17: 1163–1171, 2002.
 296. Wojciak‐Stothard B and Ridley AJ. Rho GTPases and the regulation of endothelial permeability. Vascul Pharmacol 39: 187–199, 2002.
 297. Wojciak‐Stothard B, Potempa S, Eichholtz T and Ridley AJ. Rho and Rac but not Cdc42 regulate endothelial cell permeability. J Cell Sci 114: 1343–1355, 2001.
 298. Kouklis P, Konstantoulaki M, Vogel S, Broman M and Malik AB. Cdc42 regulates the restoration of endothelial barrier function. Circ Res 94: 159–166, 2004.
 299. Amano M, Chihara K, Kimura K, Fukata Y, Nakamura N, Matsuura Y and Kaibuchi K. Formation of actin stress fibers and focal adhesions enhanced by Rho‐kinase. Science 275: 1308–1311, 1997.
 300. Flinn HM and Ridley AJ. Rho stimulates tyrosine phosphorylation of focal adhesion kinase, p130 and paxillin. J Cell Sci 109 (Pt 5): 1133–1141, 1996.
 301. Hall A. Rho GTPases and the actin cytoskeleton. Science 279: 509–514, 1998.
 302. Needham LK and Rozengurt E. Galpha12 and Galpha13 stimulate Rho‐dependent tyrosine phosphorylation of focal adhesion kinase, paxillin, and p130 Crk‐associated substrate. J Biol Chem 273: 14626–14632, 1998.
 303. Sinnett‐Smith J, Lunn JA, Leopoldt D and Rozengurt E. Y‐27632, an inhibitor of Rho‐associated kinases, prevents tyrosine phosphorylation of focal adhesion kinase and paxillin induced by bombesin: dissociation from tyrosine phosphorylation of p130(CAS). Exp Cell Res 266: 292–302, 2001.
 304. Carbajal JM and Schaeffer RC, Jr. RhoA inactivation enhances endothelial barrier function. Am J Physiol 277: C955–964, 1999.
 305. Clements RT, Minnear FL, Singer HA, Keller RS and Vincent PA. RhoA and Rho‐kinase dependent and independent signals mediate TGF‐β‐induced pulmonary endothelial cytoskeletal reorganization and permeability. Am J Physiol Lung Cell Mol Physiol 288: 294–306, 2005.
 306. Nwariaku FE, Rothenbach P, Liu Z, Zhu X, Turnage RH and Terada LS. Rho inhibition decreases TNF‐induced endothelial MAPK activation and monolayer permeability. J Appl Physiol 95: 1889–1895, 2002.
 307. Breslin JW. Sun H, Xu W, Rodarte C, Moy AB, Wu MH and Yuan SY. Involvement of ROCK‐mediated endothelial tension development in neutrophil‐stimulated microvascular leakage. Am J Physiol Heart Circ Physiol 290: H741–750, 2006.
 308. Carbajal JM, Gratrix ML, Yu CH and Schaeffer RC, Jr., ROCK mediates thrombin's endothelial barrier dysfunction. Am J Physiol Cell Physiol 279: C195–204, 2000.
 309. van Nieuw Amerongen GP, van Delft S, Vermeer MA, Collard JG and van Hinsbergh VW. Activation of RhoA by thrombin in endothelial hyperpermeability: Role of Rho kinase and protein tyrosine kinases. Circ Res 87: 335–340, 2000.
 310. Adamson RH, Curry FE, Adamson G, Liu B, Jiang Y, Aktories K, Barth H, Daigeler A, Golenhofen N, Ness W and Drenckhahn D. Rho and rho kinase modulation of barrier properties: cultured endothelial cells and intact microvessels of rats and mice. J Physiol 539: 295–308, 2002.
 311. Tokuyama K, Nishimura H, Iizuka K, Kato M, Arakawa H, Saga R, Mochizuki H and Morikawa A. Effects of Y‐27632, a Rho/Rho kinase inhibitor, on leukotriene D(4)‐ and histamine‐induced airflow obstruction and airway microvascular leakage in guinea pigs in vivo. Pharmacology 64: 189–195, 2002.
 312. Sun H, Breslin JW, Zhu J, Yuan SY and Wu MH. Rho and ROCK signaling in VEGF‐induced microvascular endothelial hyperpermeability. Microcirculation 13: 237–247, 2006.
 313. Tinsley JH, Teasdale NR and Yuan SY. Myosin light chain phosphorylation and pulmonary endothelial cell hyperpermeability in burns. Am J Physiol Lung Cell Mol Physiol 286: L841–847, 2004.
 314. Zheng HZ, Zhao KS, Zhou BY and Huang QB. Role of Rho kinase and actin filament in the increased vascular permeability of skin venules in rats after scalding. Burns 29: 820–827, 2003.
 315. Hirase T, Kawashima S, Wong EY, Ueyama T, Rikitake Y, Tsukita S, Yokoyama M and Staddon JM. Regulation of tight junction permeability and occludin phosphorylation by Rhoa‐p160ROCK‐dependent and ‐independent mechanisms. J Biol Chem 276: 10423–10431, 2001.
 316. Petrache I, Verin AD, Crow MT, Birukova A, Liu F and Garcia JG. Differential effect of MLC kinase in TNF‐alpha‐induced endothelial cell apoptosis and barrier dysfunction. Am J Physiol Lung Cell Mol Physiol 280: L1168–1178, 2001.
 317. Wojciak‐Stothard B, Entwistle A, Garg R and Ridley AJ. Regulation of TNF‐alpha‐induced reorganization of the actin cytoskeleton and cell‐cell junctions by Rho, Rac, and Cdc42 in human endothelial cells. J Cell Physiol 176: 150–165, 1998.
 318. Garcia JG, Liu F, Verin AD, Birukova A, Dechert MA, Gerthoffer WT, Bamberg JR and English D. Sphingosine 1‐phosphate promotes endothelial cell barrier integrity by Edg‐dependent cytoskeletal rearrangement. J Clin Invest 108: 689–701, 2001.
 319. Hippenstiel S, Tannert‐Otto S, Vollrath N, Krull M, Just I, Aktories K, von Eichel‐Streiber C and Suttorp N. Glucosylation of small GTP‐binding Rho proteins disrupts endothelial barrier function. Am J Physiol 272: L38–43, 1997.
 320. Waschke J, Baumgartner W, Adamson RH, Zeng M, Aktories K, Barth H, Wilde C, Curry FE and Drenckhahn D. Requirement of Rac activity for maintenance of capillary endothelial barrier properties. Am J Physiol Heart Circ Physiol 286: H394–401, 2004.
 321. Thurston G and Baldwin AL. Endothelial actin cytoskeleton in rat mesentery microvasculature. Am J Physiol 266: H1896–1909, 1994.
 322. Baldwin AL and Thurston G. Changes in endothelial actin cytoskeleton in venules with time after histamine treatment. Am J Physiol 269: H1528–1537, 1995.
 323. Thurston G, Baldwin AL and Wilson LM. Changes in endothelial actin cytoskeleton at leakage sites in the rat mesenteric microvasculature. Am J Physiol 268: H316–329, 1995.
 324. Korthuis RJ, Carden DL, Kvietys PR, Shepro D and Fuseler J. Phalloidin attenuates postischemic neutrophil infiltration and increased microvascular permeability. J Appl Physiol 71: 1261–1269, 1991.
 325. Baldwin AL and Thurston G. Mechanics of endothelial cell architecture and vascular permeability. Crit Rev Biomed Eng 29: 247–278, 2001.
 326. McDonald DM, Thurston G and Baluk P. Endothelial gaps as sites for plasma leakage in inflammation. Microcirculation 6: 7–22, 1999.
 327. Schnittler HJ, Wilke A, Gress T, Suttorp N and Drenckhahn D. Role of actin and myosin in the control of paracellular permeability in pig. rat and human vascular endothelium. J Physiol 431: 379–401, 1990.
 328. Moy AB, Shasby SS, Scott BD and Shasby DM. The effect of histamine and cyclic adenosine monophosphate on myosin light chain phosphorylation in human umbilical vein endothelial cells. J Clin Invest 92: 1198–1206, 1993.
 329. Sheldon R, Moy A, Lindsley K, Shasby S and Shasby DM. Role of myosin light‐chain phosphorylation in endothelial cell retraction. Am J Physiol 265: L606–612, 1993.
 330. Shi S, Verin AD, Schaphorst KL, Gilbert‐McClain LI, Patterson CE, Irwin RP, Natarajan V and Garcia JG. Role of tyrosine phosphorylation in thrombin‐induced endothelial cell contraction and barrier function. Endothelium 6: 153–171, 1998.
 331. Reynoso R, Perrin RM, Breslin JW, Daines DA, Watson KD, Watterson DM, Wu MH and Yuan S. A Role for Long Chain Myosin Light Chain Kinase (Mlck‐210) in Microvascular Hyperpermeability During Severe Burns. Shock 28: 589–595, 2007.
 332. Tinsley JH, De Lanerolle P, Wilson E, Ma W and Yuan SY. Myosin light chain kinase transference induces myosin light chain activation and endothelial hyperpermeability. Am J Physiol Cell Physiol 279: C1285–1289, 2000.
 333. Amano M, Fukata Y and Kaibuchi K. Regulation and functions of Rho‐associated kinase. Exp Cell Res 261: 44–51, 2000.
 334. Wong RK, Baldwin AL and Heimark RL. Cadherin‐5 redistribution at sites of TNF‐alpha and IFN‐gamma‐induced permeability in mesenteric venules. Am J Physiol 276: H736–748, 1999.
 335. Shasby DM, Ries DR, Shasby SS and Winter MC. Histamine stimulates phosphorylation of adherens junction proteins and alters their link to vimentin. Am J Physiol Lung Cell Mol Physiol 282: L1330–1338, 2002.
 336. Birukova AA, Smurova K, Birukov KG, Usatyuk P, Liu F, Kaibuchi K, Ricks‐Cord A, Natarajan V, Alieva I, Garcia JG and Verin AD. Microtubule disassembly induces cytoskeletal remodeling and lung vascular barrier dysfunction: role of Rho‐dependent mechanisms. J Cell Physiol 201: 55–70, 2004.
 337. Cattan CE and Oberg KC. Vinorelbine tartrate‐induced pulmonary edema confirmed on rechallenge. Pharmacotherapy 19: 992–994, 1999.
 338. Birukova AA, Birukov KG, Smurova K, Adyshev D, Kaibuchi K, Alieva I, Garcia JG and Verin AD. Novel role of microtubules in thrombin‐induced endothelial barrier dysfunction. FASEB J 18: 1879–1890, 2004.
 339. Birukova AA, Adyshev D, Gorshkov B, Bokoch GM, Birukov KG and Verin AD. GEF‐H1 is involved in agonist‐induced human pulmonary endothelial barrier dysfunction. Am J Physiol Lung Cell Mol Physiol 290: L540–548, 2006.
 340. Dejana E, Lampugnani MG, Martinez‐Estrada O and Bazzoni G. The molecular organization of endothelial junctions and their functional role in vascular morphogenesis and permeability. Int J Dev Biol 44: 743–748, 2000.
 341. Vittet D, Buchou T, Schweitzer A, Dejana E and Huber P. Targeted null‐mutation in the vascular endothelial‐cadherin gene impairs the organization of vascular‐like structures in embryoid bodies. Proc Natl Acad Sci USA 94: 6273–6278, 1997.
 342. Caveda L, Martin‐Padura I, Navarro P, Breviario F, Corada M, Gulino D, Lampugnani MG and Dejana E. Inhibition of cultured cell growth by vascular endothelial cadherin (cadherin‐5/VE‐cadherin). J Clin Invest 98: 886–893, 1996.
 343. Shay‐Salit A, Shushy M, Wolfovitz E, Yahav H, Breviario F, Dejana E and Resnick N. VEGF receptor 2 and the adherens junction as a mechanical transducer in vascular endothelial cells. Proc Natl Acad Sci USA 99: 9462–9467, 2002.
 344. Lampugnani MG, Corada M, Caveda L, Breviario F, Ayalon O, Geiger B and Dejana E. The molecular organization of endothelial cell to cell junctions: differential association of plakoglobin, beta‐catenin, and alpha‐catenin with vascular endothelial cadherin (VE‐cadherin). J Cell Biol 129: 203–217, 1995.
 345. Lampugnani MG, Corada M, Andriopoulou P, Esser S, Risau W and Dejana E. Cell confluence regulates tyrosine phosphorylation of adherens junction components in endothelial cells. J Cell Sci 110 (Pt 17): 2065–2077, 1997.
 346. Andriopoulou P, Navarro P, Zanetti A, Lampugnani MG and Dejana E. Histamine induces tyrosine phosphorylation of endothelial cell‐to‐cell adherens junctions. Arterioscler Thromb Vasc Biol 19: 2286–2297, 1999.
 347. Cohen AW, Carbajal JM and Schaeffer RC, Jr. VEGF stimulates tyrosine phosphorylation of beta‐catenin and small‐pore endothelial barrier dysfunction. Am J Physiol 277: H2038–2049, 1999.
 348. Esser S, Lampugnani MG, Corada M, Dejana E and Risau W. Vascular endothelial growth factor induces VE‐cadherin tyrosine phosphorylation in endothelial cells. J Cell Sci 111 (Pt 13): 1853–1865, 1998.
 349. Alexander JS, Alexander BC, Eppihimer LA, Goodyear N, Haque R, Davis CP, Kalogeris TJ, Carden DL, Zhu YN and Kevil CG. Inflammatory mediators induce sequestration of VE‐cadherin in cultured human endothelial cells. Inflammation 24: 99–113, 2000.
 350. Tinsley JH, Hawker J and Yuan Y. Efficient protein transfection of cultured coronary venular endothelial cells. Am J Physiol 275: H1873–1878, 1998.
 351. Brady‐Kalnay SM, Mourton T, Nixon JP, Pietz GE, Kinch M, Chen H, Brackenbury R, Rimm DL, Del Vecchio RL and Tonks NK. Dynamic interaction of PTPmu with multiple cadherins in vivo. J Cell Biol 141: 287–296, 1998.
 352. Downing JR and Reynolds AB. PDGF, CSF‐1, and EGF induce tyrosine phosphorylation of p120. a pp60src transformation‐associated substrate. Oncogene 6: 607–613, 1991.
 353. Hoschuetzky H, Aberle H and Kemler R. Beta‐catenin mediates the interaction of the cadherin‐catenin complex with epidermal growth factor receptor. J Cell Biol 127: 1375–1380, 1994.
 354. Tsukita S, Oishi K, Akiyama T, Yamanashi Y and Yamamoto T. Specific proto‐oncogenic tyrosine kinases of src family are enriched in cell‐to‐cell adherens junctions where the level of tyrosine phosphorylation is elevated. J Cell Biol 113: 867–879, 1991.
 355. Ukropec JA, Hollinger MK, Salva SM and Woolkalis MJ. SHP2 association with VE‐cadherin complexes in human endothelial cells is regulated by thrombin. J Biol Chem 275: 5983–5986, 2000.
 356. Carmeliet P, Lampugnani MG, Moons L, Breviario F, Compernolle V, Bono F, Balconi G, Spagnuolo R, Oostuyse B, Dewerchin M, Zanetti A, Angellilo A, Mattot V, Nuyens D, Lutgens E, Clotman F, de Ruiter MC, Gittenberger‐de Groot A, Poelmann R, Lupu F, Herbert JM, Collen D and Dejana E. Targeted deficiency or cytosolic truncation of the VE‐cadherin gene in mice impairs VEGF‐mediated endothelial survival and angiogenesis. Cell 98: 147–157, 1999.
 357. Zanetti A, Lampugnani MG, Balconi G, Breviario F, Corada M, Lanfrancone L and Dejana E. Vascular endothelial growth factor induces SHC association with vascular endothelial cadherin: a potential feedback mechanism to control vascular endothelial growth factor receptor‐2 signaling. Arterioscler Thromb Vasc Biol 22: 617–622, 2002.
 358. Anastasiadis PZ and Reynolds AB. Regulation of Rho GTPases by p120‐catenin. Curr Opin Cell Biol 13: 604–610, 2001.
 359. Sander EE, ten Klooster JP, van Delft S, van der Kammen RA and Collard JG. Rac downregulates Rho activity: reciprocal balance between both GTPases determines cellular morphology and migratory behavior. J Cell Biol 147: 1009–1022, 1999.
 360. Zondag GC, Evers EE, ten Klooster JP, Janssen L, van der Kammen RA and Collard JG. Oncogenic Ras downregulates Rac activity, which leads to increased Rho activity and epithelial‐mesenchymal transition. J Cell Biol 149: 775–782, 2000.
 361. Braga VM, Betson M, Li X and Lamarche‐Vane N. Activation of the small GTPase Rac is sufficient to disrupt cadherin‐dependent cell‐cell adhesion in normal human keratinocytes. Mol Biol Cell 11: 3703–3721, 2000.
 362. Braga VM, Machesky LM, Hall A and Hotchin NA. The small GTPases Rho and Rac are required for the establishment of cadherin‐dependent cell‐cell contacts. J Cell Biol 137: 1421–1431, 1997.
 363. Kaibuchi K, Kuroda S, Fukata M and Nakagawa M. Regulation of cadherin‐mediated cell‐cell adhesion by the Rho family GTPases. Curr Opin Cell Biol 11: 591–596, 1999.
 364. Takaishi K, Sasaki T, Kotani H, Nishioka H and Takai Y. Regulation of cell‐cell adhesion by rac and rho small G proteins in MDCK cells. J Cell Biol 139: 1047–1059, 1997.
 365. Mariner DJ, Anastasiadis P, Keilhack H, Bohmer FD, Wang J and Reynolds AB. Identification of Src phosphorylation sites in the catenin p120ctn. J Biol Chem 276: 28006–28013, 2001.
 366. Murphy JT and Duffy S. ZO‐1 redistribution and F‐actin stress fiber formation in pulmonary endothelial cells after thermal injury. J Trauma 54: 81–89, 2003, discussion 89‐90.
 367. Leeb‐Lundberg LM, Song XH and Mathis SA. Focal adhesion‐associated proteins p125FAK and paxillin are substrates for bradykinin‐stimulated tyrosine phosphorylation in Swiss 3T3 cells. J Biol Chem 269: 24328–24334, 1994.
 368. Guo M, Wu MH, Granger HJ and Yuan SY. Focal adhesion kinase in neutrophil‐induced microvascular hyperpermeability. Microcirculation 12: 223–232, 2005.
 369. Wu MH, Guo M, Yuan SY and Granger HJ. Focal adhesion kinase mediates porcine venular hyperpermeability elicited by vascular endothelial growth factor. J Physiol 552: 691–699, 2003.
 370. Guan JL and Shalloway D. Regulation of focal adhesion‐associated protein tyrosine kinase by both cellular adhesion and oncogenic transformation. Nature 358: 690–692, 1992.
 371. Hanks SK, Calalb MB, Harper MC and Patel SK. Focal adhesion protein‐tyrosine kinase phosphorylated in response to cell attachment to fibronectin. Proc Natl Acad Sci USA 89: 8487–8491, 1992.
 372. Huang MM, Lipfert L, Cunningham M, Brugge JS, Ginsberg MH and Shattil SJ. Adhesive ligand binding to integrin alpha IIb beta 3 stimulates tyrosine phosphorylation of novel protein substrates before phosphorylation of pp125FAK. J Cell Biol 122: 473–483, 1993.
 373. Frisch SM, Vuori K, Ruoslahti E and Chan‐Hui PY. Control of adhesion‐dependent cell survival by focal adhesion kinase. J Cell Biol 134: 793–799, 1996.
 374. van Nieuw Amerongen GP, Natarajan K, Yin G, Hoefen RJ, Osawa M, Haendeler J, Ridley AJ, Fujiwara K, van Hinsbergh VW and Berk BC. GIT1 mediates thrombin signaling in endothelial cells: role in turnover of RhoA‐type focal adhesions. Circ Res 94: 1041–1049, 2004.
 375. Govers R and Rabelink TJ. Cellular regulation of endothelial nitric oxide synthase. Am J Physiol Renal Physiol 280: F193–206, 2001.
 376. Liu J, Hughes TE and Sessa WC. The first 35 amino acids and fatty acylation sites determine the molecular targeting of endothelial nitric oxide synthase into the Golgi region of cells: a green fluorescent protein study. J Cell Biol 137: 1525–1535, 1997.
 377. Sessa WC, Garcia‐Cardena G, Liu J, Keh A, Pollock JS, Bradley J, Thiru S, Braverman IM and Desai KM. The Golgi association of endothelial nitric oxide synthase is necessary for the efficient synthesis of nitric oxide. J Biol Chem 270: 17641–17644, 1995.
 378. Shaul PW, Smart EJ, Robinson U, German Z, Yuhanna IS, Ying Y, Anderson RG and Michel T. Acylation targets emdothelial nitric‐oxide synthase to plasmalemmal caveolae. J Biol Chem 271: 6518–6522, 1996.
 379. Zhang Q, Church JE, Jagnandan D, Catravas JD, Sessa WC and Fulton D. Functional relevance of Golgi‐ and plasma membrane‐localized endothelial NO synthase in reconstituted endothelial cells. Arterioscler Thromb Vasc Biol 26: 1015–1021, 2006.
 380. Liu J, Garcia‐Cardena G and Sessa WC. Palmitoylation of endothelial nitric oxide synthase is necessary for optimal stimulated release of nitric oxide: Implications for caveolae localization. Biochemistry 35: 13277–13281, 1996.
 381. Sakoda T, Hirata K, Kuroda R, Miki N, Suematsu M, Kawashima S and Yokoyama M. Myristoylation of endothelial cell nitric oxide synthase is important for extracellular release of nitric oxide. Mol Cell Biochem 152: 143–148, 1995.
 382. Garcia‐Cardena G, Oh P, Liu J, Schnitzer JE and Sessa WC. Targeting of nitric oxide synthase to endothelial cell caveolae via palmitoylation: implications for nitric oxide signaling. Proc Natl Acad Sci USA 93: 6448–6453, 1996.
 383. Pobiner BF, Northup JK, Bauer PH, Fraser ED and Garrison JC. Inhibitory GTP‐binding regulatory protein Gi3 can couple angiotensin II receptors to inhibition of adenylyl cyclase in hepatocytes. Mol Pharmacol 40: 156–167, 1991.
 384. Fleming I and Busse R. Signal transduction of eNOS activation. Cardiovasc Res 43: 532–541, 1999.
 385. Feron O, Saldana F, Michel JB and Michel T. The endothelial nitric‐oxide synthase‐caveolin regulatory cycle. J Biol Chem 273: 3125–3128, 1998.
 386. Garcia‐Cardena G, Martasek P, Masters BS, Skidd PM, Couet J, Li S, Lisanti MP and Sessa WC. Dissecting the interaction between nitric oxide synthase (NOS) and caveolin. Functional significance of the nos caveolin binding domain in vivo. J Biol Chem 272: 25437–25440, 1997.
 387. Michel JB, Feron O, Sacks D and Michel T. Reciprocal regulation of endothelial nitric‐oxide synthase by Ca2+‐calmodulin and caveolin. J Biol Chem 272: 15583–15586, 1997.
 388. Michel JB, Feron O, Sase K, Prabhakar P and Michel T. Caveolin versus calmodulin. Counterbalancing allosteric modulators of endothelial nitric oxide synthase. J Biol Chem 272: 25907–25912, 1997.
 389. Blatter LA, Taha Z, Mesaros S, Shacklock PS, Wier WG and Malinski T. Simultaneous measurements of Ca2+ and nitric oxide in bradykinin‐stimulated vascular endothelial cells. Circ Res 76: 922–924, 1995.
 390. Kanai AJ, Strauss HC, Truskey GA, Crews AL, Grunfeld S and Malinski T. Shear stress induces ATP‐independent transient nitric oxide release from vascular endothelial cells, measured directly with a porphyrinic microsensor. Circ Res 77: 284–293, 1995.
 391. Feng Y, Venema VJ, Venema RC, Tsai N and Caldwell RB. VEGF induces nuclear translocation of Flk‐1/KDR, endothelial nitric oxide synthase, and caveolin‐1 in vascular endothelial cells. Biochem Biophys Res Commun 256: 192–197, 1999.
 392. Goetz RM, Thatte HS, Prabhakar P, Cho MR, Michel T and Golan DE. Estradiol induces the calcium‐dependent translocation of endothelial nitric oxide synthase. Proc Natl Acad Sci USA 96: 2788–2793, 1999.
 393. Prabhakar P, Thatte HS, Goetz RM, Cho MR, Golan DE and Michel T. Receptor‐regulated translocation of endothelial nitric‐oxide synthase. J Biol Chem 273: 27383–27388, 1998.
 394. Venema VJ, Marrero MB and Venema RC. Bradykinin‐stimulated protein tyrosine phosphorylation promotes endothelial nitric oxide synthase translocation to the cytoskeleton. Biochem Biophys Res Commun 226: 703–710, 1996.
 395. Wang Q, Patton WF, Hechtman HB and Shepro D. A novel antiinflammatory peptide inhibits endothelial cell cytoskeletal rearrangement, nitric oxide synthase translocation, and paracellular permeability increases. J Cell Physiol 172: 171–182, 1997.
 396. Yeh DC, Duncan JA, Yamashita S and Michel T. Depalmitoylation of endothelial nitric‐oxide synthase by acyl‐protein thioesterase 1 is potentiated by Ca(2+(‐calmodulin. J Biol Chem 274: 33148–33154, 1999.
 397. Dedio J, Konig P, Wohlfart P, Schroeder C, Kummer W and Muller‐Esterl W. NOSIP, a novel modulator of endothelial nitric oxide synthase activity. FASEB J 15: 79–89, 2001.
 398. Zimmermann K, Opitz N, Dedio J, Renne C, Muller‐Esterl W and Oess S. NOSTRIN: a protein modulating nitric oxide release and subcellular distribution of endothelial nitric oxide synthase. Proc Natl Acad Sci USA 99: 17167–17172, 2002.
 399. Furchgott RF. The role of endothelium in the responses of vascular smooth muscle to drugs. Annu Rev Pharmacol Toxicol 24: 175–197, 1984.
 400. Furchgott RF and Zawadzki JV. The obligatory role of endothelial cells in the relaxation of arterial smooth muscle by acetylcholine. Nature 288: 373–376, 1980.
 401. Ignarro LJ. Biosynthesis and metabolism of endothelium‐derived nitric oxide. Annu Rev Pharmacol Toxicol 30: 535–560, 1990.
 402. Crone C. The Malpighi lecture. From “Porositates carnis” to cellular microcirculation. Int J Microcirc Clin Exp 6: 101–122, 1987.
 403. Ialenti A, Ianaro A, Moncada S and Di Rosa M. Modulation of acute inflammation by endogenous nitric oxide. Eur J Pharmacol 211: 177–182, 1992.
 404. Kurose I, Kubes P, Wolf R, Anderson DC, Paulson J, Miyasaka M and Granger DN. Inhibition of nitric oxide production. Mechanisms of vascular albumin leakage. Circ Res 73: 164–171, 1993.
 405. Kurose I, Wolf R, Grisham MB, Aw TY, Specian RD and Granger DN. Microvascular responses to inhibition of nitric oxide production. Role of active oxidants. Circ Res 76: 30–39, 1995.
 406. Kurose I, Wolf R, Grisham MB and Granger DN. Modulation of ischemia/reperfusion‐induced microvascular dysfunction by nitric oxide. Circ Res 74: 376–382, 1994.
 407. Durán WN, Seyama A, Yoshimura K, Gonzalez DR, Jara PI, Figueroa XF and Boric MP. Stimulation of NO production and of eNOS phosphorylation in the microcirculation in vivo. Microvasc Res 60: 104–111, 2000.
 408. Zhu L, Castranova V and He P. fMLP‐stimulated neutrophils increase endothelial [Ca2+]i and microvessel permeability in the absence of adhesion: role of reactive oxygen species. Am J Physiol Heart Circ Physiol 288: H1331–1338, 2005.
 409. Fukumura D, Gohongi T, Kadambi A, Izumi Y, Ang J, Yun CO, Buerk DG, Huang PL and Jain RK. Predominant role of endothelial nitric oxide synthase in vascular endothelial growth factor‐induced angiogenesis and vascular permeability. Proc Natl Acad Sci USA 98: 2604–2609, 2001.
 410. Durán WN, Hatakeyama T and Sanchez FA. Endothelial nitric oxide synthase‐derived NO signals regulate microvascular permeability. Physiol News 65: 21–23, 2006.
 411. Arnal JF, Dinh‐Xuan AT, Pueyo M, Darblade B and Rami J. Endothelium‐derived nitric oxide and vascular physiology and pathology. Cell Mol Life Sci 55: 1078–1087, 1999.
 412. Garcia‐Cardena G, Fan R, Shah V, Sorrentino R, Cirino G, Papapetropoulos A and Sessa WC. Dynamic activation of endothelial nitric oxide synthase by Hsp90. Nature 392: 821–824, 1998.
 413. He H, Venema VJ, Gu X, Venema RC, Marrero MB and Caldwell RB. Vascular endothelial growth factor signals endothelial cell production of nitric oxide and prostacyclin through flk‐1/KDR activation of c‐Src. J Biol Chem 274: 25130–25135, 1999.
 414. Hood JD, Meininger CJ, Ziche M and Granger HJ. VEGF upregulates ecNOS message, protein, and NO production in human endothelial cells. Am J Physiol 274: H1054–1058, 1998.
 415. Ju H, Venema VJ, Marrero MB and Venema RC. Inhibitory interactions of the bradykinin B2 receptor with endothelial nitric‐oxide synthase. J Biol Chem 273: 24025–24029, 1998.
 416. Dimmeler S, Fleming I, Fisslthaler B, Hermann C, Busse R and Zeiher AM. Activation of nitric oxide synthase in endothelial cells by Akt‐dependent phosphorylation. Nature 399: 601–605, 1999.
 417. Fleming I, Fisslthaler B, Dimmeler S, Kemp BE and Busse R. Phosphorylation of Thr(495) regulates Ca(2 +)/calmodulin‐dependentendothelial nitric oxide synthase activity. Circ Res 88: E68–75, 2001.
 418. Fulton D, Gratton JP, McCabe TJ, Fontana J, Fujio Y, Walsh K, Franke TF, Papapetropoulos A and Sessa WC. Regulation of endothelium‐derived nitric oxide production by the protein kinase Akt. Nature 399: 597–601, 1999.
 419. Garcia‐Cardena G, Fan R, Stern DF, Liu J and Sessa WC. Endothelial nitric oxide synthase is regulated by tyrosine phosphorylation and interacts with caveolin‐1. J Biol Chem 271: 27237–27240, 1996.
 420. Gratton JP, Fontana J, O'Connor DS, Garcia‐Cardena G, McCabe TJ and Sessa WC. Reconstitution of an endothelial nitric‐oxide synthase (eNOS), hsp90, and caveolin‐1 complex in vitro. Evidence that hsp90 facilitates calmodulin stimulated displacement of eNOS from caveolin‐1. J Biol Chem 275: 22268–22272, 2000.
 421. Luo Z, Fujio Y, Kureishi Y, Rudic RD, Daumerie G, Fulton D, Sessa WC and Walsh K. Acute modulation of endothelial Akt/PKB activity alters nitric oxide‐dependent vasomotor activity in vivo. J Clin Invest 106: 493–499, 2000.
 422. Michell BJ, Griffiths JE, Mitchelhill KI, Rodriguez‐Crespo I, Tiganis T, Bozinovski S, de Montellano PR, Kemp BE and Pearson RB. The Akt kinase signals directly to endothelial nitric oxide synthase. Curr Biol 9: 845–848, 1999.
 423. Michell BJ, Chen Z, Tiganis T, Stapleton D, Katsis F, Power DA, Sim AT and Kemp BE. Coordinated control of endothelial nitric‐oxide synthase phosphorylation by protein kinase C and the cAMP‐dependent protein kinase. J Biol Chem 276: 17625–17628, 2001.
 424. Marsen TA, Egink G, Suckau G and Baldamus CA. Tyrosine‐kinase‐dependent regulation of the nitric oxide synthase gene by endothelin‐1 in human endothelial cells. Pflugers Arch 438: 538–544, 1999.
 425. Ohara Y, Sayegh HS, Yamin JJ and Harrison DG. Regulation of endothelial constitutive nitric oxide synthase by protein kinase C. Hypertension 25: 415–420, 1995.
 426. Shen BQ, Lee DY and Zioncheck TF. Vascular endothelial growth factor governs endothelial nitric‐oxide synthase expression via a KDR/Flk‐1 receptor and a protein kinase C signaling pathway. J Biol Chem 274: 33057–33063, 1999.
 427. Wedgwood S, Bekker JM and Black SM. Shear stress regulation of endothelial NOS in fetal pulmonary arterial endothelial cells involves PKC. Am J Physiol Lung Cell Mol Physiol 281: L490–498, 2001.
 428. Ignarro LJ, Cirino G, Casini A and Napoli C. Nitric oxide as a signaling molecule in the vascular system: an overview. J Cardiovasc Pharmacol 34: 879–886, 1999.
 429. Yonemaru M, Ishii K, Murad F and Raffin TA. Atriopeptin‐induced increases in endothelial cell permeability are associated with elevated cGMP levels. Am J Physiol 263: L363–369, 1992.
 430. Mayhan WG. VEGF increases permeability of the blood‐brain barrier via a nitric oxide synthase/cGMP‐dependent pathway. Am J Physiol 276: C1148–1153, 1999.
 431. Corson MA, James NL, Latta SE, Nerem RM, Berk BC and Harrison DG. Phosphorylation of endothelial nitric oxide synthase in response to fluid shear stress. Circ Res 79: 984–991, 1996.
 432. Michel T and Feron O. Nitric oxide synthases: which, where, how, and why? J Clin Invest 100: 2146–2152, 1997.
 433. Michel T, Li GK and Busconi L. Phosphorylation and subcellular translocation of endothelial nitric oxide synthase. Proc Natl Acad Sci USA 90: 6252–6256, 1993.
 434. Sanchez FA, Savalia NB, Durán RG, Lal BK, Boric MP and Durán WN. Functional significance of differential eNOS translocation. Am J Physiol Heart Circ Physiol 291: H1058–1064, 2006.
 435. Erwin PA, Lin AJ, Golan DE and Michel T. Receptor‐regulated dynamic S‐nitrosylation of endothelial nitric‐oxide synthase in vascular endothelial cells. J Biol Chem 280: 19888–19894, 2005.
 436. Figueroa XF, Gonzalez DR, Martinez AD, Durán WN and Boric MP. ACh‐induced endothelial NO synthase translocation, NO release and vasodilatation in the hamster microcirculation in vivo. J Physiol 544: 883–896, 2002.
 437. Pober JS, Kluger MS and Schechner JS. Human endothelial cell presentation of antigen and the homing of memory/effector T cells to skin. Ann N Y Acad Sci 941: 12–25, 2001.
 438. Beynon HL, Davies KA, Haskard DO and Walport MJ. Erythrocyte complement receptor type 1 and interactions between immune complexes, neutrophils, and endothelium. J Immunol 153: 3160–3167, 1994.
 439. Burns AR, Walker DC, Brown ES, Thurmon LT, Bowden RA, Keese CR, Simon SI, Entman ML and Smith CW. Neutrophil transendothelial migration is independent of tight junctions and occurs preferentially at tricellular corners. J Immunol 159: 2893–2903, 1997.
 440. Kubes P, Grisham MB, Barrowman JA, Gaginella T and Granger DN. Leukocyte‐induced vascular protein leakage in cat mesentery. Am J Physiol 261: H1872–1879, 1991.
 441. Minnicozzi M, Durán WN, Gleich GJ and Egan RW. Eosinophil granule proteins increase microvascular macromolecular transport in the hamster cheek pouch. J Immunol 153: 2664–2670, 1994.
 442. Minnicozzi M, Ramirez MM, Egan RW, Gleich GJ, Kobayashi I, Kim D and Durán WN. Polyarginine and eosinophil‐derived major basic protein increase microvascular permeability independently of histamine or nitric oxide release. Microvasc Res 50: 56–70, 1995.
 443. Garcia JG, Verin AD, Herenyiova M and English D. Adherent neutrophils activate endothelial myosin light chain kinase: role in transendothelial migration. J Appl Physiol 84: 1817–1821, 1998.
 444. Tomeo AC, Egan RW and Durán WN. Priming interactions between platelet activating factor and histamine in the in vivo microcirculation. FASEB J 5: 2850–2855, 1991.
 445. Curry FE, Zeng M and Adamson RH. Thrombin increases permeability only in venules exposed to inflammatory conditions. Am J Physiol Heart Circ Physiol 285: H2446–H2453, 2003.
 446. Fan J and Malik AB. Toll‐like receptor‐4 (TLR4) signaling augments chemokine‐induced neutrophil migration by modulating cell surface expression of chemokine receptors. Nat Med 9: 315–321, 2003.
 447. Mollen KP, Anand RJ, Tsung A, Prince JM, Levy RM and Billiar TR. Emerging paradigm: toll‐like receptor 4‐sentinel for the detection of tissue damage. Shock 26: 430–437, 2006.
 448. Breslin JW, Wu MH, Guo M, Reynoso R and Yuan SY. Toll‐like receptor‐4 contributes to microvascular inflammation and barrier dysfunction in thermal injury. Shock 29: 349–355, 2008.
 449. Dudzinski DM, Igarashi J, Greif D and Michel T. The regulation and pharmacology of endothelial nitric oxide synthase. Annu Rev Pharmacol Toxicol 46: 235–276, 2006.
 450. de Rooij J, Zwartkruis FJ, Verheijen MH, Cool RH, Nijman SM, Wittinghofer A and Bos JL. Epac is a Rap1 guanine‐nucleotide‐exchange factor directly activated by cyclic AMP. Nature 396: 474–477, 1998.
 451. Christensen AE, Selheim F, de Rooij J, Dremier S, Schwede F, Dao KK, Martinez A, Maenhaut C, Bos JL, Genieser HG and Doskeland SO. cAMP analog mapping of Epac 1 and cAMP kinase. Discriminating analogs demonstrate that Epac and cAMP kinase act synergistically to promote PC‐12 cell neurite extension. J Biol Chem 278: 35394–35402, 2003.
 452. DiPilato LM, Cheng X and Zhang J. Fluorescent indicators of cAMP and Epac activation reveal differential dynamics of cAMP signaling within discrete subcellular compartments. Proc Natl Acad Sci USA 101: 16513–16518, 2004.
 453. Enserink JM, Christensen AE, de Rooij J, van Triest M, Schwede F, Genieser HG, Doskeland SO, Blank JL and Bos JL. A novel Epac‐specific cAMP analogue demonstrates independent regulation of Rap1 and ERK. Nat Cell Biol 4: 901–906, 2002.
 454. Kramer GC, Harms BA, Bodai BI, Demling RH and Renkin EM. Mechanisms for redistribution of plasma protein following acute protein depletion. Am J Physiol 243: H803–809, 1982.
 455. Cullere X, Shaw SK, Andersson L, Hirahashi J, Luscinskas FW and Mayadas TN. Regulation of vascular endothelial barrier function by Epac, a cAMP‐activated exchange factor for Rap GTPase. Blood 105: 1950–1955, 2005.
 456. Fukuhara S, Sakurai A, Sano H, Yamagishi A, Somekawa S, Takakura N, Saito Y, Kangawa K and Mochizuki N. Cyclic AMP potentiates vascular endothelial cadherin‐mediated cell‐cell contact to enhance endothelial barrier function through an Epac‐Rap1 signaling pathway. Mol Cell Biol 25: 136–146, 2005.
 457. Zwartkruis FJ and Bos JL. Ras and Rap1: two highly related small GTPases with distinct function. Exp Cell Res 253: 157–165, 1999.
 458. Minnear FL, Zhu L and He P. Sphingosine 1‐phosphate prevents platelet‐activating factor‐induced increase in hydraulic conductivity in rat mesenteric venules: pertussis toxin sensitive. Am J Physiol Heart Circ Physiol 289: H840–844, 2005.
 459. Kim DD, Pica AM, Durán RG and Durán WN. Acupuncture reduces experimental renovascular hypertension through mechanisms involving nitric oxide synthases. Microcirculation 13: 577–585, 2006.
 460. Kim DD, Sanchez FA, Durán RG, Kanetaka T and Durán WN. Endothelial nitric oxide synthase is a molecular vascular target for the Chinese herb Danshen in hypertension. Am J Physiol Heart Circ Physiol 292: H2131–2137, 2007.
 461. Huang Q, Wu M. Meininger C, Kelly K, and Yuan Y. Neutrophil‐dependent augmentation of PAF‐induced vasoconstriction and albumin flux in coronary arterioles. Am J Physiol 275: H1138–1147, 1998.
 462. Huxley VH and Williams DA. Role of a glycocalyx on coronary arteriole permeability to proteins: evidence from enzyme treatments. Am J Physiol Heart Circ Physiol 278: H1177–1185, 2000.
 463. Wu MH, Ustinova E, and Granger HJ. Integrin binding to fibronectin and vitronectin maintains the barrier function of isolated porcine coronary venules. J Physiol 532: 785–791, 2001.
 464. Rumbaut RE and Huxley VH. Similar permeability responses to nitric oxide synthase inhibitors of venules from three animal species. Microvasc Res 64: 21–31, 2002.
 465. Garlick DG and Renkin EM. Transport of large molecules from plasma to interstitial fluid and lymph in dogs. Am J Physiol 219: 1595–1605, 1970.
 466. Baxter LT, Jain RK, Svensjö E. Vascular permeability and interstitial diffusion of macromolecules in the hamster cheek pouch: effects of vasoactive drugs. Microvasc Res 34: 336–348, 1987.

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Walter N. Durán, Fabiola A. Sánchez, Jerome W. Breslin. Microcirculatory Exchange Function. Compr Physiol 2011, Supplement 9: Handbook of Physiology, The Cardiovascular System, Microcirculation: 81-124. First published in print 2008. doi: 10.1002/cphy.cp020404