Comprehensive Physiology Wiley Online Library

Local Regulation of Microvascular Perfusion

Full Article on Wiley Online Library



Abstract

The sections in this article are:

1 Historical Perspectives
2 Autoregulation
2.1 Blood Flow Regulation
2.2 Capillary Pressure Regulation
3 Mechanisms of Blood Flow Regulation
3.1 Myogenic Mechanism of Blood Flow Regulation
3.2 Signaling Mechanisms Underlying the Myogenic Response
3.3 Metabolic Mechanisms of Blood Flow Regulation
3.4 Interactions Between Local Blood Flow Control Mechanisms
3.5 Interactions Between Microvascular Control Mechanisms
3.6 Converging Pathways for the Interaction of Local Regulatory Mechanisms
4 Glossary of Abbreviations
Figure 1. Figure 1.

Whole‐organ pressure‐flow relationships. Data recalculated from Refs 34,35,36,37,38,1378. Flows are normalized to flow at P = 100 mmHg, except pulmonary data 1378 are normalized to Ppulmonary artery = 21 mmHg. SKM. skeletal muscle.

Figure 2. Figure 2.

(A) Time course of 2A hemodynamic changes in response to Pp reduction; redrawn from Ref. 52 and used by permission; Pp calculated as (digital artery pressure minus box pressure). Flow decreased initially, but returned to control (arrow) within 2–3 min as the arteriole dilated. (B) Regulation of bat wing 1A flow during mild reductions in Pp; redrawn from Ref. 52 and used by permission; mean data calculated from individual points and fit with third‐order polynomial, weighted by standard error and forced through Pp = 90 mmHg.

Figure 3. Figure 3.

(A) Time course of arteriolar dilation in response to 50‐mmHg reduction in box pressure. Plotted from data in Ref. 52. Pp calculated from digital artery pressure minus box pressure. Values represent averages for 8–15 vessels (error bars omitted for clarity). (B) Average steady‐state bat wing arteriolar diameters as a function of perfusion pressure. Error bars and two points for TAs are omitted for clarity. AA, arcuate arteriole; TA, terminal arteriole. Plotted from data in 52.

Figure 4. Figure 4.

(A) Venular diameters during lowered Pp in cat sartorius muscle. Redrawn from Ref. 70; used by permission. (B) Responses of bat wing venules to lowered pressure. Top: in vivo 2V (unpublished); bottom: in vitro IV from Ref. 73; used by permission. IV, first‐order venule; 2V, second‐order venule.

Figure 5. Figure 5.

(A) Steady‐state capillary diameter changes in bat wing capillaries (control D = 7.6 ± 0.4 μm) as a function of box pressure. Replotted from data in Refs 52,53. (B) Capillary diameter changes during aortic occlusion to Pa = 17 mmHg and subsequent phase of reactive hyperemia. Redrawn from Ref. 79.

Figure 6. Figure 6.

Maintenance of relatively stable tissue volume in cat skeletal muscle during reduced Pp 87. (A) Sample recording; (B) summary data. From 87; used by permission.

Figure 7. Figure 7.

(A) PCvenule measurement during reduction in Pp 40. (B) Stability of transvascular fluid flux and PCvenule pressure as a function of Pa. Solid line = normal tone: dotted line = after papaverine (from Ref. 104); used by permission.

Figure 8. Figure 8.

(A) Bat wing Pc as a function of box pressure (Pbox); redrawn from Ref. 52. Solid line is linear regression line of entire data set; line “a” represents perfect regulation; line “b” represents no regulation. Different symbols represent data for individual capillaries. (B) Single Pc recording analysis redrawn from Ref. 54; used by permission. Solid line represents limits of pressure excursion due to regional vasomotion; line “a” represents perfect regulation; line “b” represents no regulation.

Figure 9. Figure 9.

Relative degrees of (A) Pc compensation and (B) flow compensation during Pp changes (from Ref. 104); used by permission. Numbers on graph are calculated open‐loop gains.

Figure 10. Figure 10.

Types of myogenic behavior. (A) pressure‐induced constriction (133]; used by permission (B) Development of basal arteriolar tone at constant pressure 190: used by permission (C) Negative slope of steady‐state, pressure‐diameter curve 128: used by permission (D) Active pressure‐diameter curve with less positive slope than passive curve 1379: used by permission. Filled symbols denote the passive curve. (E) Increased amplitude and frequency of spontaneous contractions with vessel stretch 139; used by permission. (F) Secondary force development (after initial stretch and stress relaxation) in an isometric smooth muscle preparation The response is evident after a 25% stretch but not after a 10% stretch. Modified from 141: used by permisison.

Figure 11. Figure 11.

The wall tension hypothesis for myogenic regulation of vessel radius (from Ref. 30): used by permission.

Figure 12. Figure 12.

Predicted relationships for myogenically active arterioles functioning as a series‐coupled unit, as homogeneously dilating vessels, or as passive vessels. Top panels show predicted diameter responses for the 3 models, bottom panels show predicted microvascular pressure responses. Dotted lines in insets indicate reference lines for perfect pressure regulation. Top panels from [1380); used by permission. Bottom panels modified from 54 and 1380; used by permission.

Figure 13. Figure 13.

(A) Compilation of myogenic index data, redrawn from Ref. 128; used by permission. See 128 for symbol key. (B) Pressure‐diameter relationships of four different branching orders of rat mesenteric arterioles (from Ref. 130); used by permission.

Figure 14. Figure 14.

Schematic diagram illustrating general signaling events underlying the arteriolar myogenic response. An increase in intraluminal pressure provides the initial stimulus through stretch of a smooth muscle membrane or cytoskeletal element or a change in wall tension. Detection of the stimulus occurs either directly at the level of the membrane or as a result of extracellular matrix‐integrin interactions. Subsequent ion channel‐based mechanisms lead to membrane depolarization, increased conductance of voltage‐gated Ca2+ channels, and increased intracellular Ca2+ levels. Ca2+‐ mediated activation of the contractile proteins then initiates contraction. This basic mechanism may also be acted upon by various second messengers to further enhance the level of contraction. Negative feedback mechanisms may also be initiated to limit the extent of contraction thus preventing an unstable feed‐forward system (dotted lines).

Figure 15. Figure 15.

Relationships between membrane potential (Em) and mechanical stimulation. (A) The change in Em for isolated arteriolar smooth muscle cells subjected to defined length increases applied by the movement of glass micropipettes 250; the degree of cell stretch is comparable to that exerted by a modest pressure increase in isolated arterioles. From 250; used by permission. (B) The effect of intraluminal pressure on Em for cannulated cerebral (squares; 280) and cremaster muscle arterioles (triangles. 262; used by permission. Numbers in parentheses indicate pressure in mmHg. (C) Data from panel B plotted as a function of the calculated level of active myogenic tone. From 261; used by permission. Numbers in parenthesis indicate pressure in mmHg. The data suggest a sigmoidal relationship between Em and myogenic tone and suggest differences between vessels from different vascular beds.

Figure 16. Figure 16.

Postulated ion channel mechanisms linking a pressure or stretch stimulus to depolarization and ultimately contraction. Modified from 134; used by permission.

Figure 17. Figure 17.

Involvement of K+ channels in the regulation of myogenic contraction. The figure illustrates two concepts whereby firstly K+ channel inhibition, via generation of the eicosanoid 20‐HETE, leads to depolarization and contraction and secondly by virtue of restricted domains formed by close apposition of the plasma and SR membranes where activation of K+ channels provides a hyperpolarization and relaxation. The two pathways modulate voltage‐gated Ca2+ entry via their effects on Em.

Figure 18. Figure 18.

Relationships between intraluminal pressure, [Ca2+]i, and wall tension for cannulated, fura 2‐loaded, rat cremaster muscle first‐order arterioles. (A) Effect of intraluminal pressure on global smooth muscle [Ca2+]i levels calculated from the 340/380 nm fluorescent ratio (R 340/380). Data shown for active and passive states; n, number of vessels. From 172; used by permission. (B) Relationship between calculated wall tension and [Ca2+]i. A significant (r2 = 0.72, P < 0.001) linear correlation was obtained between tension and [Ca2+]i. From 172; used by permission.

Figure 19. Figure 19.

Connectivity between arteriolar smooth muscle and endothelial cells. (A) Electromicrograph of a rat cremaster first‐order arteriole highlighting junctional connections between endothelial cells and between smooth muscle and endothelial cells. This supports the syncytial nature of the arteriolar wall and the likelihood of gap junctional communication between the two cell types. Modified from 1381; used by permission. (B) Schematic of interactions between smooth muscle and endothelial cells at the biochemical level. Communication is viewed to occur both through gap junctions and the localized release of paracrine factors. McSherry and Dora, personal communication.

Figure 20. Figure 20.

Temporal nature of signaling events following an increase in arteriolar pressure. Multiple pathways are hypothesized to be initiated by the mechanical stimulus, with progression to intermediate and longer term responses being a function of the effectiveness of the initial contractile response. From 1382: used by permission from IOS Press.

Figure 21. Figure 21.

(A) Reactive hyperemia in the dog coronary circulation. CF = coronary flow; RH = reactive hyperemia. From 593; used by permission. (B) Reactive hyperemia in single cat sartorius muscle capillaries. From 592; used by permission.

Figure 22. Figure 22.

Time course of diameter. PO2 and pressure changes during and after local arteriolar occlusion. 1‐min occlusions. Whalen‐style PO2 microelectrodes 602 were used to measure PO2 levels in the tissue (from Ref. 600): used by permission

Figure 23. Figure 23.

(A) Reactive dilation in isolated rat gracilis arterioles (B) Endothelium‐derived NO mediates ∼35% of the reactive dilation 610; used by permission.

Figure 24. Figure 24.

Little effect of superfusate O2 on reactive hyperemia of hamster cheek pouch arterioles (from Ref. [597)); used by permission.

Figure 25. Figure 25.

(A) Capillary velocity and tissue PO2 changes during and after arterial occlusion from Ref. 618; used by permission. (B) Time course of changes in tissue oxidative metabolism (NADH fluorescence; diamonds) and PO2 (circles) in resting rat spinotrapezius after occlusion of feed artery/vein. Modified from Ref. 616; used by permission.

Figure 26. Figure 26.

Proposed contributions of different factors to the various phases of reactive hyperemia 610.

Figure 27. Figure 27.

(A) Functional capillary units in hamster cremaster muscle (from Ref. 967). (B) Changes in indexes of capillary perfusion in hamster cremaster muscle during electrical stimulation of single fibers (from Ref. 680); used by permission.

Figure 28. Figure 28.

(A) Distribution of tissue PO2 measurements made blindly in cat myocardium: modified from Ref. 633; used by permission. Note increase in number of sites with PO2 < 5 mmHg at lower Pp. (B) Change in tissue PO2 during electrical stimulation of the rat spinotrapezius muscle. Open symbols denote points that are significantly different from control value (mean PO2 = 28 mmHg). Suffusate PO2 was ∼14 mmHg From 640; used by permission.

Figure 29. Figure 29.

A) Graded arteriolar dilation as a function of muscle fiber stimulation frequency from Ref. 712; used by permission. (B) Arteriolar dilation precedes fall in tissue PO2 in hamster cheek pouch microcirculation. Modified from Ref. 764; used by permission.

Figure 30. Figure 30.

(A) Longitudinal distribution of perivascular PO2 at various levels of suffusate PO2. Symbols indicate different solution PO2. PCO2 = 32mmHg in all cases. Modified from Ref. [1035); used by permission. (B) Longitudinal distribution of perivascular PO2 in rat cremaster muscle when superfusate PO2 <10 mmHg. Modified from Ref. 631; used by permission.

Figure 31. Figure 31.

(A) Effects of local (micropipette) and global (suffusion) oxygenated solutions on diameter of an aparenchymal cheek pouch arteriole. From 785; used by permission. (B) Summary data for free‐flowing and occluded (no‐flow) aparenchymal arterioles at high and low PO2. Replotted from Ref. 785; used by permission.

Figure 32. Figure 32.

(A) Responses of isolated cerebral arterioles to various concentrations of K+ (B) Responses of same vessels to elevated [K+] in the presence of 50μM Ba2+ to block KIR channels. From Ref. 918: used by permission.

Figure 33. Figure 33.

Effect of extravascular acidosis on coronary arteriolar diameter. (A) Hydrogen ions dilated the coronary arteriole in a concentration‐dependent manner. After replacing with normal pH solution (i.e. washout with pH = 7.4), the diameter returned to the baseline level. (B) Exposure of the vessel to KATP channel inhibitor glibenclamide (5 μM) for 20 min did not alter the baseline diameter of the vessel, but the dilation in response to an increase in hydrogen ion concentration (pH = 7.2) was abolished. Modified from Ref. 1056; used by permission.

Figure 34. Figure 34.

Effect of hyperosmolarity on vascular tone. (A) A coronary arteriole dilated to an increase in extravascular osmolarity by replacing the vessel bath solution containing a high concentration of glucose. Hyperosmolarity produced a sustained arteriolar dilation and vascular tone recovered after washout. (B) Coronary arteriolar dilation to the hyperosmotic glucose solution was inhibited after denudation. (C): Intraluminal KCl (80mM) or KATP channel inhibitor glibenclamide (Glib. 1 μM) significantly attenuated the vasodilation to hyperosmotic glucose. Lumenal diameters were normalized to maximum dilation in response to sodium nitroprusside (0.1 mM). Modified from Ref. 1085; used by permission.

Figure 35. Figure 35.

Segmental coronary microvascular dilation to increased flow (shear stress) at constant mean luminal pressure. Luminal diameters were normalized to resting diameter in the absence of flow. The hierarchy of shear stress‐induced response was large arterioles > intermediate arterioles > small arterioles = small arteries. Modified from Ref. 1186; used by permission.

Figure 36. Figure 36.

Segmental coronary microvascular dilation to adenosine at constant mean luminal pressure without flow. Luminal diameters were normalized to maximum dilation in response to sodium nitroprusside (0.1 mM). The hierarchy of adenosine‐induced response was small arterioles > intermediate arterioles > large arterioles = small arteries. Modified from Ref. 1186; used by permission.

Figure 37. Figure 37.

Scheme for series‐coupled, segmental responsiveness of an arterial network to flow, pressure, metabolic, and adrenergic stimuli. Note that each vasoactive mechanism has a dominant site of action in a particular microvascular segment. All responses are normalized to their maximum. Modified from Ref. 1383; used by permission.

Figure 38. Figure 38.

Integrative regulation of coronary flow by metabolic, myogenic, and shear stress‐induced mechanisms in response to metabolic activation. The sequential, series recruitment of metabolic, myogenic, and flow‐induced dilation in the arteriolar network would optimize functional hyperemia.

Figure 39. Figure 39.

(A) Diagram showing preparation for study of conducted responses using an arteriole from the mouse cremaster muscle in vivo. ACh or KCl is applied locally from a micropipette while observing the arteriolar diameter change either locally or at various remote sites upstream in the flow path. Suffusate flow is in the same direction as blood flow. (B) Arteriolar dilation is observed at the local site of ACh application (time = 0) and the dilation is rapidly conducted to sites 660 and 1320 μm upstream, with partial attenuation of the response. (C) In response to KCl application, arteriolar constriction is observed at the local site and the constriction is conducted upstream, with substantial attenuation at the 660 μm observation site, and complete attenuation of the response 1320 μm away. From Ref. 1261; used by permission.

Figure 40. Figure 40.

Conducted dilation to adenosine in an isolated coronary arteriole (50‐110 μm, ID). Diagram at top shows the experimental set‐up. (A) Adenosine (10 μM) application from a micropipette induces a substantial dilation at the application site that is rapidly conducted upstream to the remote site (>800 μm away). (B) The conducted response but not the local response is blocked by Ba2+ (30μM) in the suffsate. Modified from Ref. 1246; used by permission.

Figure 41. Figure 41.

(A) Segmental resistance changes in cat skeletal muscle in response to transmural pressure elevation using a plethysmograph. Resistances were calculated continuously from total flow and segmental pressures measured by indwelling microcatheters. From 159; used by permission. Vertical lines and A, B labels were added. (B) Constriction of bat wing 2A in response to transmural pressure elevation. Arteriolar pressure increase is sustained, but flow initially increases (dotted line indicates reference flow level), then decreases secondarily modified from Ref. 53; used by permission.

Figure 42. Figure 42.

(A) Arteriolar dilations under free flow and no‐flow conditions; Modified from Ref. 169; used by permission. (B) Arteriolar dilations upstream and downstream from an occlusion. Superfusion solution equilibrated with 0% O2. Modified from Ref. 597; used by permission.

Figure 43. Figure 43.

Interaction between pressure‐induced myogenic responses and flow‐induced vasodilation in isolated subepicardial arterioles. (A) Myogenic constriction (initiated by an increase in intraluminal pressure. (IP) was attenuated in the presence of flow (initiated by a pressure gradient. ΔP, longitudinally across the vessel). (B) Myogenic dilation was enhanced by flow. (C) Flow‐induced dilation was attenuated by elevating IP (D) Flow‐induced dilation was potentiated by lowering IP from Ref. 1174; used by permission.

Figure 44. Figure 44.

(A) Pressure‐diameter relations (myogenic response) of coronary arterioles in the presence and absence of lumenal flow. Myogenic responsiveness was attenuated in the presence of flow. With sodium nitroprusside (10−4M), vessels showed passive responses to intraluminal pressure changes. Lumen diameters were normalized to diameters at intraluminal pressure of 60cmH2O in presence of sodium nitroprusside. Modified from Ref. 1174. (B) How‐induced dilation at different levels of myogenic tone (i.e. 20, 60, and 100cmH2O intraluminal pressure). Intraluminal flow was initiated by increasing pressure gradient longitudinally across the vessel. Lumenal diameters were normalized to passive diameter at respective intraluminal pressure in the presence of sodium nitroprusside (10−4M). Modified from Ref. 1174; used by permission.

Figure 45. Figure 45.

Arteriolar myogenic reactivity during α1‐ and α2‐adrenoceptor constriction. Box‐pressure technique was used to study myogenic responsiveness in first‐order rat cremaster arterioles. Myogenic constriction was insignificant under control conditions (no α tone). In the presence of either type of adrenoceptor tone, myogenic constriction was augmented. With α2 tone, myogenic vasodilation was 3‐ to 4‐fold more pronounced than with α tone. Modified from Ref. 1306; used by permission.



Figure 1.

Whole‐organ pressure‐flow relationships. Data recalculated from Refs 34,35,36,37,38,1378. Flows are normalized to flow at P = 100 mmHg, except pulmonary data 1378 are normalized to Ppulmonary artery = 21 mmHg. SKM. skeletal muscle.



Figure 2.

(A) Time course of 2A hemodynamic changes in response to Pp reduction; redrawn from Ref. 52 and used by permission; Pp calculated as (digital artery pressure minus box pressure). Flow decreased initially, but returned to control (arrow) within 2–3 min as the arteriole dilated. (B) Regulation of bat wing 1A flow during mild reductions in Pp; redrawn from Ref. 52 and used by permission; mean data calculated from individual points and fit with third‐order polynomial, weighted by standard error and forced through Pp = 90 mmHg.



Figure 3.

(A) Time course of arteriolar dilation in response to 50‐mmHg reduction in box pressure. Plotted from data in Ref. 52. Pp calculated from digital artery pressure minus box pressure. Values represent averages for 8–15 vessels (error bars omitted for clarity). (B) Average steady‐state bat wing arteriolar diameters as a function of perfusion pressure. Error bars and two points for TAs are omitted for clarity. AA, arcuate arteriole; TA, terminal arteriole. Plotted from data in 52.



Figure 4.

(A) Venular diameters during lowered Pp in cat sartorius muscle. Redrawn from Ref. 70; used by permission. (B) Responses of bat wing venules to lowered pressure. Top: in vivo 2V (unpublished); bottom: in vitro IV from Ref. 73; used by permission. IV, first‐order venule; 2V, second‐order venule.



Figure 5.

(A) Steady‐state capillary diameter changes in bat wing capillaries (control D = 7.6 ± 0.4 μm) as a function of box pressure. Replotted from data in Refs 52,53. (B) Capillary diameter changes during aortic occlusion to Pa = 17 mmHg and subsequent phase of reactive hyperemia. Redrawn from Ref. 79.



Figure 6.

Maintenance of relatively stable tissue volume in cat skeletal muscle during reduced Pp 87. (A) Sample recording; (B) summary data. From 87; used by permission.



Figure 7.

(A) PCvenule measurement during reduction in Pp 40. (B) Stability of transvascular fluid flux and PCvenule pressure as a function of Pa. Solid line = normal tone: dotted line = after papaverine (from Ref. 104); used by permission.



Figure 8.

(A) Bat wing Pc as a function of box pressure (Pbox); redrawn from Ref. 52. Solid line is linear regression line of entire data set; line “a” represents perfect regulation; line “b” represents no regulation. Different symbols represent data for individual capillaries. (B) Single Pc recording analysis redrawn from Ref. 54; used by permission. Solid line represents limits of pressure excursion due to regional vasomotion; line “a” represents perfect regulation; line “b” represents no regulation.



Figure 9.

Relative degrees of (A) Pc compensation and (B) flow compensation during Pp changes (from Ref. 104); used by permission. Numbers on graph are calculated open‐loop gains.



Figure 10.

Types of myogenic behavior. (A) pressure‐induced constriction (133]; used by permission (B) Development of basal arteriolar tone at constant pressure 190: used by permission (C) Negative slope of steady‐state, pressure‐diameter curve 128: used by permission (D) Active pressure‐diameter curve with less positive slope than passive curve 1379: used by permission. Filled symbols denote the passive curve. (E) Increased amplitude and frequency of spontaneous contractions with vessel stretch 139; used by permission. (F) Secondary force development (after initial stretch and stress relaxation) in an isometric smooth muscle preparation The response is evident after a 25% stretch but not after a 10% stretch. Modified from 141: used by permisison.



Figure 11.

The wall tension hypothesis for myogenic regulation of vessel radius (from Ref. 30): used by permission.



Figure 12.

Predicted relationships for myogenically active arterioles functioning as a series‐coupled unit, as homogeneously dilating vessels, or as passive vessels. Top panels show predicted diameter responses for the 3 models, bottom panels show predicted microvascular pressure responses. Dotted lines in insets indicate reference lines for perfect pressure regulation. Top panels from [1380); used by permission. Bottom panels modified from 54 and 1380; used by permission.



Figure 13.

(A) Compilation of myogenic index data, redrawn from Ref. 128; used by permission. See 128 for symbol key. (B) Pressure‐diameter relationships of four different branching orders of rat mesenteric arterioles (from Ref. 130); used by permission.



Figure 14.

Schematic diagram illustrating general signaling events underlying the arteriolar myogenic response. An increase in intraluminal pressure provides the initial stimulus through stretch of a smooth muscle membrane or cytoskeletal element or a change in wall tension. Detection of the stimulus occurs either directly at the level of the membrane or as a result of extracellular matrix‐integrin interactions. Subsequent ion channel‐based mechanisms lead to membrane depolarization, increased conductance of voltage‐gated Ca2+ channels, and increased intracellular Ca2+ levels. Ca2+‐ mediated activation of the contractile proteins then initiates contraction. This basic mechanism may also be acted upon by various second messengers to further enhance the level of contraction. Negative feedback mechanisms may also be initiated to limit the extent of contraction thus preventing an unstable feed‐forward system (dotted lines).



Figure 15.

Relationships between membrane potential (Em) and mechanical stimulation. (A) The change in Em for isolated arteriolar smooth muscle cells subjected to defined length increases applied by the movement of glass micropipettes 250; the degree of cell stretch is comparable to that exerted by a modest pressure increase in isolated arterioles. From 250; used by permission. (B) The effect of intraluminal pressure on Em for cannulated cerebral (squares; 280) and cremaster muscle arterioles (triangles. 262; used by permission. Numbers in parentheses indicate pressure in mmHg. (C) Data from panel B plotted as a function of the calculated level of active myogenic tone. From 261; used by permission. Numbers in parenthesis indicate pressure in mmHg. The data suggest a sigmoidal relationship between Em and myogenic tone and suggest differences between vessels from different vascular beds.



Figure 16.

Postulated ion channel mechanisms linking a pressure or stretch stimulus to depolarization and ultimately contraction. Modified from 134; used by permission.



Figure 17.

Involvement of K+ channels in the regulation of myogenic contraction. The figure illustrates two concepts whereby firstly K+ channel inhibition, via generation of the eicosanoid 20‐HETE, leads to depolarization and contraction and secondly by virtue of restricted domains formed by close apposition of the plasma and SR membranes where activation of K+ channels provides a hyperpolarization and relaxation. The two pathways modulate voltage‐gated Ca2+ entry via their effects on Em.



Figure 18.

Relationships between intraluminal pressure, [Ca2+]i, and wall tension for cannulated, fura 2‐loaded, rat cremaster muscle first‐order arterioles. (A) Effect of intraluminal pressure on global smooth muscle [Ca2+]i levels calculated from the 340/380 nm fluorescent ratio (R 340/380). Data shown for active and passive states; n, number of vessels. From 172; used by permission. (B) Relationship between calculated wall tension and [Ca2+]i. A significant (r2 = 0.72, P < 0.001) linear correlation was obtained between tension and [Ca2+]i. From 172; used by permission.



Figure 19.

Connectivity between arteriolar smooth muscle and endothelial cells. (A) Electromicrograph of a rat cremaster first‐order arteriole highlighting junctional connections between endothelial cells and between smooth muscle and endothelial cells. This supports the syncytial nature of the arteriolar wall and the likelihood of gap junctional communication between the two cell types. Modified from 1381; used by permission. (B) Schematic of interactions between smooth muscle and endothelial cells at the biochemical level. Communication is viewed to occur both through gap junctions and the localized release of paracrine factors. McSherry and Dora, personal communication.



Figure 20.

Temporal nature of signaling events following an increase in arteriolar pressure. Multiple pathways are hypothesized to be initiated by the mechanical stimulus, with progression to intermediate and longer term responses being a function of the effectiveness of the initial contractile response. From 1382: used by permission from IOS Press.



Figure 21.

(A) Reactive hyperemia in the dog coronary circulation. CF = coronary flow; RH = reactive hyperemia. From 593; used by permission. (B) Reactive hyperemia in single cat sartorius muscle capillaries. From 592; used by permission.



Figure 22.

Time course of diameter. PO2 and pressure changes during and after local arteriolar occlusion. 1‐min occlusions. Whalen‐style PO2 microelectrodes 602 were used to measure PO2 levels in the tissue (from Ref. 600): used by permission



Figure 23.

(A) Reactive dilation in isolated rat gracilis arterioles (B) Endothelium‐derived NO mediates ∼35% of the reactive dilation 610; used by permission.



Figure 24.

Little effect of superfusate O2 on reactive hyperemia of hamster cheek pouch arterioles (from Ref. [597)); used by permission.



Figure 25.

(A) Capillary velocity and tissue PO2 changes during and after arterial occlusion from Ref. 618; used by permission. (B) Time course of changes in tissue oxidative metabolism (NADH fluorescence; diamonds) and PO2 (circles) in resting rat spinotrapezius after occlusion of feed artery/vein. Modified from Ref. 616; used by permission.



Figure 26.

Proposed contributions of different factors to the various phases of reactive hyperemia 610.



Figure 27.

(A) Functional capillary units in hamster cremaster muscle (from Ref. 967). (B) Changes in indexes of capillary perfusion in hamster cremaster muscle during electrical stimulation of single fibers (from Ref. 680); used by permission.



Figure 28.

(A) Distribution of tissue PO2 measurements made blindly in cat myocardium: modified from Ref. 633; used by permission. Note increase in number of sites with PO2 < 5 mmHg at lower Pp. (B) Change in tissue PO2 during electrical stimulation of the rat spinotrapezius muscle. Open symbols denote points that are significantly different from control value (mean PO2 = 28 mmHg). Suffusate PO2 was ∼14 mmHg From 640; used by permission.



Figure 29.

A) Graded arteriolar dilation as a function of muscle fiber stimulation frequency from Ref. 712; used by permission. (B) Arteriolar dilation precedes fall in tissue PO2 in hamster cheek pouch microcirculation. Modified from Ref. 764; used by permission.



Figure 30.

(A) Longitudinal distribution of perivascular PO2 at various levels of suffusate PO2. Symbols indicate different solution PO2. PCO2 = 32mmHg in all cases. Modified from Ref. [1035); used by permission. (B) Longitudinal distribution of perivascular PO2 in rat cremaster muscle when superfusate PO2 <10 mmHg. Modified from Ref. 631; used by permission.



Figure 31.

(A) Effects of local (micropipette) and global (suffusion) oxygenated solutions on diameter of an aparenchymal cheek pouch arteriole. From 785; used by permission. (B) Summary data for free‐flowing and occluded (no‐flow) aparenchymal arterioles at high and low PO2. Replotted from Ref. 785; used by permission.



Figure 32.

(A) Responses of isolated cerebral arterioles to various concentrations of K+ (B) Responses of same vessels to elevated [K+] in the presence of 50μM Ba2+ to block KIR channels. From Ref. 918: used by permission.



Figure 33.

Effect of extravascular acidosis on coronary arteriolar diameter. (A) Hydrogen ions dilated the coronary arteriole in a concentration‐dependent manner. After replacing with normal pH solution (i.e. washout with pH = 7.4), the diameter returned to the baseline level. (B) Exposure of the vessel to KATP channel inhibitor glibenclamide (5 μM) for 20 min did not alter the baseline diameter of the vessel, but the dilation in response to an increase in hydrogen ion concentration (pH = 7.2) was abolished. Modified from Ref. 1056; used by permission.



Figure 34.

Effect of hyperosmolarity on vascular tone. (A) A coronary arteriole dilated to an increase in extravascular osmolarity by replacing the vessel bath solution containing a high concentration of glucose. Hyperosmolarity produced a sustained arteriolar dilation and vascular tone recovered after washout. (B) Coronary arteriolar dilation to the hyperosmotic glucose solution was inhibited after denudation. (C): Intraluminal KCl (80mM) or KATP channel inhibitor glibenclamide (Glib. 1 μM) significantly attenuated the vasodilation to hyperosmotic glucose. Lumenal diameters were normalized to maximum dilation in response to sodium nitroprusside (0.1 mM). Modified from Ref. 1085; used by permission.



Figure 35.

Segmental coronary microvascular dilation to increased flow (shear stress) at constant mean luminal pressure. Luminal diameters were normalized to resting diameter in the absence of flow. The hierarchy of shear stress‐induced response was large arterioles > intermediate arterioles > small arterioles = small arteries. Modified from Ref. 1186; used by permission.



Figure 36.

Segmental coronary microvascular dilation to adenosine at constant mean luminal pressure without flow. Luminal diameters were normalized to maximum dilation in response to sodium nitroprusside (0.1 mM). The hierarchy of adenosine‐induced response was small arterioles > intermediate arterioles > large arterioles = small arteries. Modified from Ref. 1186; used by permission.



Figure 37.

Scheme for series‐coupled, segmental responsiveness of an arterial network to flow, pressure, metabolic, and adrenergic stimuli. Note that each vasoactive mechanism has a dominant site of action in a particular microvascular segment. All responses are normalized to their maximum. Modified from Ref. 1383; used by permission.



Figure 38.

Integrative regulation of coronary flow by metabolic, myogenic, and shear stress‐induced mechanisms in response to metabolic activation. The sequential, series recruitment of metabolic, myogenic, and flow‐induced dilation in the arteriolar network would optimize functional hyperemia.



Figure 39.

(A) Diagram showing preparation for study of conducted responses using an arteriole from the mouse cremaster muscle in vivo. ACh or KCl is applied locally from a micropipette while observing the arteriolar diameter change either locally or at various remote sites upstream in the flow path. Suffusate flow is in the same direction as blood flow. (B) Arteriolar dilation is observed at the local site of ACh application (time = 0) and the dilation is rapidly conducted to sites 660 and 1320 μm upstream, with partial attenuation of the response. (C) In response to KCl application, arteriolar constriction is observed at the local site and the constriction is conducted upstream, with substantial attenuation at the 660 μm observation site, and complete attenuation of the response 1320 μm away. From Ref. 1261; used by permission.



Figure 40.

Conducted dilation to adenosine in an isolated coronary arteriole (50‐110 μm, ID). Diagram at top shows the experimental set‐up. (A) Adenosine (10 μM) application from a micropipette induces a substantial dilation at the application site that is rapidly conducted upstream to the remote site (>800 μm away). (B) The conducted response but not the local response is blocked by Ba2+ (30μM) in the suffsate. Modified from Ref. 1246; used by permission.



Figure 41.

(A) Segmental resistance changes in cat skeletal muscle in response to transmural pressure elevation using a plethysmograph. Resistances were calculated continuously from total flow and segmental pressures measured by indwelling microcatheters. From 159; used by permission. Vertical lines and A, B labels were added. (B) Constriction of bat wing 2A in response to transmural pressure elevation. Arteriolar pressure increase is sustained, but flow initially increases (dotted line indicates reference flow level), then decreases secondarily modified from Ref. 53; used by permission.



Figure 42.

(A) Arteriolar dilations under free flow and no‐flow conditions; Modified from Ref. 169; used by permission. (B) Arteriolar dilations upstream and downstream from an occlusion. Superfusion solution equilibrated with 0% O2. Modified from Ref. 597; used by permission.



Figure 43.

Interaction between pressure‐induced myogenic responses and flow‐induced vasodilation in isolated subepicardial arterioles. (A) Myogenic constriction (initiated by an increase in intraluminal pressure. (IP) was attenuated in the presence of flow (initiated by a pressure gradient. ΔP, longitudinally across the vessel). (B) Myogenic dilation was enhanced by flow. (C) Flow‐induced dilation was attenuated by elevating IP (D) Flow‐induced dilation was potentiated by lowering IP from Ref. 1174; used by permission.



Figure 44.

(A) Pressure‐diameter relations (myogenic response) of coronary arterioles in the presence and absence of lumenal flow. Myogenic responsiveness was attenuated in the presence of flow. With sodium nitroprusside (10−4M), vessels showed passive responses to intraluminal pressure changes. Lumen diameters were normalized to diameters at intraluminal pressure of 60cmH2O in presence of sodium nitroprusside. Modified from Ref. 1174. (B) How‐induced dilation at different levels of myogenic tone (i.e. 20, 60, and 100cmH2O intraluminal pressure). Intraluminal flow was initiated by increasing pressure gradient longitudinally across the vessel. Lumenal diameters were normalized to passive diameter at respective intraluminal pressure in the presence of sodium nitroprusside (10−4M). Modified from Ref. 1174; used by permission.



Figure 45.

Arteriolar myogenic reactivity during α1‐ and α2‐adrenoceptor constriction. Box‐pressure technique was used to study myogenic responsiveness in first‐order rat cremaster arterioles. Myogenic constriction was insignificant under control conditions (no α tone). In the presence of either type of adrenoceptor tone, myogenic constriction was augmented. With α2 tone, myogenic vasodilation was 3‐ to 4‐fold more pronounced than with α tone. Modified from Ref. 1306; used by permission.

References
 1. Guyton AC, Ross JM, Carrier O and Walker JR. Evidence for tissue oxygen demand as the major factor causing autoregulation. Circ Res 15 (Suppl.): I60–I69, 1964.
 2. Johnson PC. Measurement of microvascular dimensions in vivo. J Appl Physiol 23: 593–596, 1967.
 3. Johnson PC and Wayland H. Regulation of blood flow in single capillaries. Am J Physiol 212: 1405–1415, 1967.
 4. Duling BR and Berne RM. Propagated vasodilation in the microcirculation of the hamster cheek pouch. Circ Res 26: 163–170, 1970.
 5. Duling BR and Berne RM. Microiontophoretic application of vasoactive agents to the microcirculation of the hamster cheek pouch. Microvasc Res 1: 158–173, 1968.
 6. Duling BR and Berne RM. Longitudinal gradients in periarteriolar oxygen tension: a possible mechanism for the participation of oxygen in local regulation of blood flow. Circ Res 27: 669–678, 1970.
 7. Duling BR and Berne RM. Microcirculation of the hamster cheek pouch. Circ Res 26: 163–170, 1970.
 8. Wiederhielm CA, Woodbury JW, Kirk S and Rushmer RF. Pulsatile pressures in the microcirculation of frog's mesentery. Am J Physiol 207: 173–176, 1964.
 9. Wiederhielm CA and Pelroy GE. A TV‐micro‐densitometer for the study of diffusion processes in tissues. Proceedings 636‐637, 1963.
 10. Wiederhielm CA. Physiologic characteristics of small vessels. In: Proceedings 5th National Symposium on Microcirculation. Springfield, Il: Charles C. Thomas. 1969, pp. 75–88.
 11. Fox JR and Wiederhielm CA. Characteristics of the servo‐controlled micropipet pressure system. Microvasc Res 5: 324–335, 1973.
 12. Wiederhielm CA, Lee DR and Stromberg DD. A membrane osmometer for microliter samples. J Appl Physiol 35: 432–435, 1973.
 13. Wiederhielm CA, Shaw ML, Kehl TH and Fox JR. A digital system for studying interstitial transport of dye molecules. Microvasc Res 5: 243–250, 1973.
 14. Wayland H and Johnson PC. Erythrocyte velocity measurement in microvessels by a correlation method. In: 4th European Conference on Microcirculation, ed. Harder H. Basel: S. Karger, 1966, pp. 160–163.
 15. Intaglietta M, Topkins WR and Richardson DR. Velocity Measurements in the microvasculature of the cat omentum by on‐line method. Microvasc Res 2: 462–473, 1970.
 16. Intaglietta M, Pawula RF and Tompkins WR. Pressure measurements in the mammalian microvasculature. Microvasc Res 2: 212–220, 1970.
 17. Intaglietta M and Tompkins WR. Micropressure measurement with 1 mu and smaller cannulae. Microvasc Res 3: 211–214, 1971.
 18. Yin FC, Tompkins WR, Peterson KL and Intaglietta M. A video‐dimension analyzer. IEEE Transactions on Biomedical Engineering 19: 376–381, 1972.
 19. Intaglietta M. Pressure measurements in the microcirculation with active and passive transducers. Microvasc Res 5: 317–323, 1973.
 20. Intaglietta M and Tompkins WR. Microvascular measurements by video image shearing and splitting. Microvasc Res 5: 309–312, 1973.
 21. Intaglietta M, Silverman NR and Tompkins WR. Capillary flow velocity measurements in vivo and in situ by television methods. Microvasc Res 10: 165–179, 1975.
 22. Johnson PC. Autoregulation of blood flow. Circ Res 15: 11–291, 1964.
 23. Johnson PC. Autoregulation of blood flow. Circ Res 59: 483–495, 1986.
 24. Watts SW, Kanagy NL and Lombard JH. Receptor‐mediated events in the microcirculation. Handbook of Physiology, 2008.
 25. Navar G. Handbook of Physiology, 2008.
 26. Fleming I. Biology of nitric oxide synthases, Chapter 3. In: Handbook of Physiology: Microcirculation, 2nd Edition, eds Tuma RF, Durán WN and Ley K. Elsevier, 2008 (this volume).
 27. Sparks HV. Effect of local metabolic factors on vascular smooth muscle. In: Handbook of Physiology, Sec 2: The Cardiovascular System, Vol. 2: Vascular Smooth Muscle, eds Bohr DF, Somlyo AP and Sparks HV. Bethesda, MD: American Physiological Society, 1980, pp. 475–513.
 28. Renkin EM. Control of microcirculation and blood‐tissue exchange. In: Handbook of Physiology, Sec 2: The Cardiovascular System, Vol. IV, Microcirculation, Part 2, eds Renkin EM, Michel CC and Geiger SR. Bethesda, MD: American Physiological Society, 1984, pp. 627–687.
 29. Zweifach BW and Lipowsky HH. Pressure‐flow relations in blood and lymph microcirculation. In: Handbook of Physiology, Sec 2: The Cardiovascular System, Vol. IV, eds Renkin EM and Michel CC. Bethesda, Maryland: American Physiological Society, 1984. pp. 251–307.
 30. Johnson PC. The myogenic response. In: Handbook of Physiology The Cardiovascular System Vascular Smooth Muscle, eds Bohr DF, Somlyo AP and Sparks HV, Jr. Bethesda, MD: American Physiological Society. 1980, pp. 409–442.
 31. Folkow B. Intravascular pressure as a factor regulating the tone of the small vessels. Acta Physiol Scand 17: 289–310, 1949.
 32. Folkow B. A study of the factors influencing the tone of denervated blood vessels performed at various pressures. Acta Physiol Scand 27: 99–117, 1952.
 33. Folkow B. Transmural pressure and vascular tone ‐ some aspects of an old controversy. Archives Internationales de Pharmacodynamie et de Therapie 3‐4: 455–469, 1962.
 34. Kirchheim HR, Ehmke H, Hackenthal E, Lowe W and Persoon P. Autoregulation of renal blood flow. Pflügers Arch Eur J Physiol 410: 441–449, 1987.
 35. Laird JD, Breuls PN, van der Meer P and Spaan JA. Can a single vasodilator be responsible for both coronary autoregulation and metabolic vasodilation? Basic Res Cardiol 76: 354–358, 1981.
 36. Held K, Gottstein U and Niedermayer W. Cerebral blood flow in nonpulsatile perfusion. In: Cerebral Blood Flow, eds Brock M, Fieshi C, Ingvar DH, Lassen NA and Schürmann K. Berlin: Springer, 1969, pp. 94–95.
 37. Stainsby WN and Renkin EM. Autoregulation of blood flow in resting skeletal muscle. Am J Physiol 201: 117–122, 1961.
 38. Norris CP, Barnes GE, Smith EE and Granger HJ. Autoregulation of superior mesenteric flow in fasted and fed dogs. Am J Physiol (Heart Circ Physiol) 237: H174–H177, 1979.
 39. Haddy FJ, Richards AG, Alden JL and Visscher MB. Small vein and artery pressure in normal and edematous extremities of dogs under local and general anesthesia. Am J Physiol 176: 355–360, 1954.
 40. Mellander S, Björnberg J, Maspers M and Myrhage R. Method for continuous recording of hydrostatic exchange vessel pressure in cat skeletal muscle. Acta Physiol Scand 129: 325–335, 1987.
 41. Johnson PC and Hanson KM. Effect of arterial pressure on arterial and venous resistance of intestine. J Appl Physiol 17: 503–508, 1962.
 42. Davis MJ and Gore RW. Pressure distribution in the microvascular network of the hamster cheek pouch. In: Microvascular Networks‐Theoretical and Experimental Studies, eds Popel AS and Johnson PC. Basel: Karger, 1986, pp. 142–154.
 43. Lang DJ and Johnson PC. Size dependence of arteriolar responses to arterial pressure redution. In: Microvascular Networks‐Experimental and Theoretical Studies, eds Popel AS and Johnson PC. Basel: Karger, 1985, pp. 112–122.
 44. Meininger GA, Mack CA, Fehr KL and Bohlen HG. Myogenic vaso‐regulation overrides local metabolic control in resting rat skeletal muscle. Circ Res 60: 861–870, 1987.
 45. Wiederhielm CA, Bouskela E, Heald R and Black L. A method for varying arterial and venous pressures in intact, unanesthetized mammals. Microvasc Res 18: 124–128, 1979.
 46. Morff RJ and Granger HJ. Autoregulation of blood flow within individual arterioles in the rat cremaster muscle. Circ Res 51: 43–55, 1982.
 47. Sullivan SM and Johnson PC. Effect of oxygen on arteriolar dimensions and blood flow in cat sartorius muscle. Am J Physiol (Heart Circ Physiol) 241: H547–H556, 1981.
 48. Pries AR, Secomb TW and Gaehtgens P. Design principles of vascular beds. Circ Res 77: 1017–1023, 1995.
 49. Pries AR, Secomb TW, Gaehtgens P and Gross JF. Blood flow in microvascular networks: Experiments and simulation. Circ Res 67: 826–834, 1990.
 50. Sweeney TE and Sarelius IH. Spatial heterogeneity in striated muscle arteriolar tone, cell flow, and capillarity. Am J Physiol (Heart Circ Physiol) 259: H124–H136, 1990.
 51. Intaglietta M. Arteriolar vasomotion: implications for tissue ischemia. Blood Vessels 28 (Suppl. 1): 1–7, 1991.
 52. Davis MJ. Conlrol of bat wing capillary pressure and blood flow during reduced perfusion pressure. Am J Physiol (Heart Circ Physiol) 255: H1114–H1129, 1988.
 53. Davis MJ. Microvascular control of capillary pressure during increases in local arterial and venous pressure. Am J Physiol (Heart Circ Physiol) 254: H772–H784, 1988.
 54. Slaaf DW, Reneman RS and Wiederhielm CA. Pressure regulation in muscle of unanesthetized bats. Microvasc Res 33: 315–326, 1987.
 55. Bouskela E and Wiederhielm CA. Microvascular myogenic reaction in the wing of the intact unanesthetized bat. Am J Physiol (Heart Circ Physiol) 237: H59–H65, 1979.
 56. Colantuoni A, Bertuglia S and Intaglietta M. Effects of anesthesia on the spontaneous activity of the microvasculature. Int J Microcirc Clin Exp 3: 13–38, 1984.
 57. Funk W, Endrich B, Messmer K and Intaglietta M. Spontaneous arteriolar vasomotion as a determinant of peripheral vascular resistance. Int J Microcirc Clin Exp 2: 11–25, 1983.
 58. Colantuoni A, Bertuglia S, Coppini G and Donato L. Superposition of arteriolar vasomotion waves and regulation of blood flow in skeletal muscle microcirculation. Adv Exp Med Biol 277: 549–558, 1990.
 59. Jackson WF, Mülsch A and Busse R. Rhythmic smooth muscle activity in hamster aortas is mediated by continuous release of NO from the endothelium. Am J Physiol (Heart Circ Physiol) 260: H248–H253, 1991.
 60. Slaaf DW, Oude Vrielink HHE, Tangelder GJ and Reneman RS. Effective diameter as a determinant of local vascular resistance in presence of vasomotion. Am J Physiol (Heart Circ Physiol) 255: H1240–H1243, 1988.
 61. Slaaf DW, Tangelder GJ and Reneman RS. Arteriolar vasomotion and arterial pressure reduction in rabbit tenuissimus muscle. Microvasc Res 33: 71–80, 1987.
 62. Tsai AG and Intaglietta M. Evidence of flowmotion induced changes in local tissue oxygenation. Int J Microcirc Clin Exp 12: 75–88, 1993.
 63. Schmidt JA, Borgström P and Intaglietta M. The vascular origin of slow wave flowmotion in skeletal muscle during local hypotension. Int J Microcirc Clin Exp 12: 287–297, 1993.
 64. Bertuglia S, Colantuoni A, Arnold M and Witte H. Dynamic coherence analysis of vasomotion and flow motion in skeletal muscle microcirculation. Microvasc Res 52: 235–244, 1996.
 65. Nilsson H and Aalkjaer C. Vasomotion: mechanisms and physiological importance. Molecular Interventions 3: 79–89, 51, 2003.
 66. Ursino M, Colantuoni A and Bertuglia S. Vasomotion and blood flow regulation in hamster skeletal muscle microcirculation: A theoretical and experimental study. Microvasc Res 56: 233–252, 1998.
 67. Meyer JU, Borgstrom P, Lindbon L and Intaglietta M. Vasomotion patterns in skeletal muscle arterioles during changes in arterial pressure. Microvasc Res 35: 193–203, 1988.
 68. Oude Vrielink HHE, Slaaf DW, Tangelder GJ and Reneman RS. Changes in vasomotion pattern and local arteriolar resistance during stepwise pressure reduction. Pflügers Arch Eur J Physiol 414: 571–578, 1989.
 69. Vrielink HHEO, Slaaf DW, Tangelder GJ, Weijmer‐Van Velzen S and Reneman RS. Analysis of vasomotion waveform changes during pressure reduction and adenosine application. Am J Physiol 258: H29–H37, 1990.
 70. House SD and Johnson PC. Diameter and blood flow of skeletal muscle venules during local flow regulation. Am J Physiol (Heart Circ Physiol) 250: H828–H837, 1986.
 71. Bishop JJ, Nance PR, Popel AS, Intaglietta M and Johnson PC. Diameter changes in skeletal muscle venules during arterial pressure reduction. Am J Physiol (Heart Circ Physiol) 279: H47–H57, 2000.
 72. Kuo L, Arko F, Chilian WM and Davis MJ. Coronary venular responses to flow and pressure. Circ Res 72: 607–615, 1993.
 73. Davis MJ, Shi X and Sikes PJ. Modulation of bat wing venule contraction by transmural pressure changes. Am J Physiol (Heart Circ Physiol) 262: H625–H634, 1992.
 74. Noddeland H, Nicolaysen G and Aukland K. Rhythmic variations of venous pressure in the human foot. Acta Physiol Scand 113: 125–127, 1981.
 75. Benoit JN, Zawieja DC, Goodman AH and Granger HJ. Characterization of intact mesenteric lymphatic pump and its responsiveness to acute edemagenic stress. Am J Physiol (Heart Circ Physiol) 257: H2059–H2069, 1989.
 76. Zawieja DC, Davis KL, Schuster R, Hinds WM and Granger HJ. Distribution, propagation, and coordination of contractile activity in lymphatics. Am J Physiol (Heart Circ Physiol) 264: H1283–H1291, 1993.
 77. Bosman J, Tangelder GJ, Oude Egbrink MG, Reneman RS and Slaaf DW. Capillary diameter changes during low perfusion pressure and reactive hyperemia in rabbit skeletal muscle. Am J Physiol (Heart Circ Physiol) 269: H1048–H1055, 1995.
 78. Skalak TC and Schmid‐Schonbein GW. Viscoelastic properties of microvessels in rat spinotrapezius muscle. Biomech Eng 108: 193–200, 1986.
 79. Baldwin AL and Gore RW. Simultaneous measurement of capillary distensibility and hydraulic conductance. Microvasc Res 38: 1–22, 1989.
 80. Groebe K. Diameter control in the arteriolar tree by changes in post‐capillary resistance ‐ A theoretical study. Adv Exp Med Biol 428: 687–694, 1997.
 81. Bishop JJ, Popel AS, Intaglietta M and Johnson PC. Rheological effects of red blood cell aggregation in the venous network: a review of recent studies. Biorheology 38: 263–274, 2001.
 82. Bishop JJ, Nance PR, Popel AS, Intaglietta M and Johnson PC. Effect of erythrocyte aggregation on velocity profiles in venules. Am J Physiol (Heart Circ Physiol) 280: H222–H236, 2001.
 83. Murrant CL and Sarelius IH. Coupling of muscle metabolism and muscle blood flow in capillary units during contraction. Acta Physiol Scand 168: 531–541, 2000.
 84. Lindbom L and Arfors KE. Mechanisms and site of control for variation in the number of perfused capillaries in skeletal muscle. Int J Microcirc Clin Exp 4: 19–30, 1985.
 85. Slaaf DW, Reneman RS and Wiederhielm CA. Cessation and onset of muscle capillary flow at simultaneously reduced perfusion and transmural pressure. Int J Microcirc Clin Exp 6: 215–224, 1987.
 86. Michel CC. Fluid movements through capillary walls. In: Handbook of Physiology, eds Renkin EM and Michel CC. Bethesda, MD: American Physiological Society, 1984, pp. 375–409.
 87. Järhult J and Mellander S. Autoregulation of capillary hydrostatic pressure in skeletal muscle during regional arterial hypo‐ and hypertension. Acta Physiol Scand 91: 32–41, 1974.
 88. Granger HJ, Goodman AH and Granger DN. Role of resistance and exchange vessels in local microvascular control of skeletal muscle oxygenation in the dog. Circ Res 38: 379–385, 1976.
 89. Cobbold A, Folkow B, Kjellmer I and Mellander S. Nervous and local chemical control of pre‐capillary sphincters in skeletal muscle as measured by changes in filtration coefficient. Acta Physiol Scand 57: 180–192, 1963.
 90. Mellander S and Johansson B. Control of resistance, exchange, and capacitance functions in the peripheral circulation. Pharmacological Rev 20: 117–196, 1968.
 91. Maspers M and Björnberg J. Beta2‐adrenergic attenuation of capillary pressure autoregulation during haemorrhagic hypotension, a mechanism promoting transcapillary fluid absorption in skeletal muscle. Acta Physiol Scand 142: 11–20, 1991.
 92. Pappenheimer JR and Soto‐Rivera A. Effective osmotic pressure of the plasma proteins and other quantities associated with the capillary circulation in the hindlimbs of cats and dogs. Am J Physiol 152: 471–491, 1948.
 93. Hanson KM and Johnson PC. Vascular resistance and arterial pressure in autoperfused dog hind limb. Am J Physiol 203: 615–620, 1962.
 94. Thulesius O and Johnson PC. Pre‐ and postcapillary resistance in skeletal muscle. Am J Physiol 210: 869–872, 1966.
 95. Krogh A, Landis EM and Turner AH. The movement of fluid through the human capillary wall in relation to venous pressure and to the colloid osmotic pressure of the blood. J Clin Invest 11: 63–95, 1932.
 96. Landis EM and Pappenheimer JR. Exchange of substances through capillary walls. In: Handbook of Physiology The Cardiovascular System, eds Hamilton WF. Bethesda, MD: American Physiological Society, 1963, pp. 961–1034.
 97. Richardson PDI, Granger DN and Taylor AE. Capillary filtration coefficient: the technique and its application to the small intestine. Cardiovasc Res 13: 547–561, 1979.
 98. Aukland K and Nicolaysen G. Interstitial fluid volume: local regulatory mechanisms. Physiological Rev 61: 556–643, 1981.
 99. Levick JR. Capillary filtration‐absorption balance reconsidered in light of dynamic extravascular factors. Exp Physiol 76: 825–857, 1991.
 100. Swanson JA and Kern DF. Effect of common vasodilators on lung microvascular permeability. J Appl Physiol 75: 2326–2331, 1993.
 101. Maron MB and Pilati CF. Effect of papaverine on pulmonary vascular permeability to proteins. J Appl Physiol 65: 1367–1371, 1988.
 102. Korthuis RJ, Granger DN, Townsley MI and Taylor AE. Autoregulation of capillary pressure and filtration rate in isolated rat hindquarters. Am J Physiol (Heart Circ Physiol) 248: H835–H842, 1985.
 103. Björnberg J, Grände P‐O, Maspers M and Mellander S. Site of autoregulatory reactions in the vascular bed of cat skeletal muscle as determined with a new technique for segmental vascular resistance recordings. Acta Physiol Scand 133: 199–210, 1988.
 104. Mellander S, Maspers M, Björnberg J and Andersson LO. Autoregulation of capillary pressure and filtration in cat skeletal muscle in states of normal and reduced vascular tone. Acta Physiol Scand 129: 337–351, 1987.
 105. Grände P‐O. Lundvall J and Mellander S. Evidence for a rate‐sensitive regulatory mechanism in myogenic microvascular control. Acta Physiol Scand 99: 432–447, 1977.
 106. Gore RW and Bohlen HG. Pressure regulation in the microcirculation. FASEB J 34: 2031–2037, 1975.
 107. Bohlen HG and Gore RW. Comparison of microvascular pressures and diameters in the innervated and denervated rat intestine. Microvasc Res 14: 251–264, 1977.
 108. Gore RW. Pressures in cat mesenteric arterioles and capillaries during changes in systemic arterial blood pressure. Circ Res 34: 581–591, 1974.
 109. Zweifach BW. Quantitative studies of microcirculatory structure and function. I. Analysis of pressure distribution in the terminal vascular bed in cat mesentery. Circ Res 34: 843–857, 1974.
 110. Zweifach BW. Local regulation of capillary pressure. Circ Res Suppl. 1: I‐29–I‐134, 1971.
 111. Mellander S. On the control of capillary fluid transfer by precapillary and postcapillary vascular adjustments. Microvasc Res 15: 319–330, 1978.
 112. Davis MJ and Sikes PJ. A rate‐sensitive component to the myogenic response is absent from bat wing arterioles. Am J Physiol (Heart Circ Physiol) 256: H32–H40, 1989.
 113. Proctor KG and Busija DW. Relationships among arteriolar, regional, and whole organ blood flow in cremaster muscle. Am J Physiol (Heart Circ Physiol) 249: H34–H41, 1985.
 114. Nicoll PA and Webb RL. Vascular patterns and active vasomotion as determiners of flow through minute vessels. Angiology 6: 291–308, 1955.
 115. Clark WE and Blomfield LB. The efficiency of intramuscular anastomoses with observations on the regeneration of devascularized muscle. J Anatomy 79: 15–32, 1945.
 116. Spalteholz W. Die vertheilung der blutgefasse im muskel. Ab Math Phys Cl Konigl Sach Ges Wiss Leipzig 14: 509–528, 1888.
 117. Meininger GA, Fehr KL and Yates MB. Anatomic and hemodyanamic characteristics of the blood vessels feeding the cremaster skeletal muscle in the rat. Microvasc Res 33: 81–97, 1987.
 118. Davis MJ, Ferrer PN and Gore RW. Vascular anatomy and hydrostatic pressure profile in the hamster cheek pouch. Am J Physiol (Heart Circ Physiol) 250: H291–H303, 1986.
 119. Schmid‐Schönbein GW, Firestone G and Zweifach BW. Network anatomy of arteries feeding the spinotrapezius muscle in normotensive and hypertensive rats. Blood Vessels 23: 34–49, 1986.
 120. Mayrovitz HN. Hemodynamic significance of microvascular arteriolar anastomosing. In: Microvascular Networks: Experimental and Theoretical Studies, eds Popel AS and Johnson PC. Basel: Karger, 1986, pp. 197–209.
 121. Greene AS, Tonellato PJ and Davis MJ. Autoregulation and network functionality in the bat wing microvascular network. FASEB J 3: A1388, 1989.
 122. Greene AS, Tonellato PJ, Lui J, Lombard JH and Cowley AW, Jr. Microvascular rarefaction and tissue vascular resistance in hypertension. Am J Physiol (Heart Circ Physiol) 256: H126–H131, 1989.
 123. Nagle FJ, Scott JB, Swindall BT and Haddy FJ. Venous resistances in skeletal muscle and skin during local blood flow regulation. Am J Physiol 214: 885–891, 1968.
 124. House SD and Johnson PC. Microvascular pressure in venules of skeletal muscle during arterial pressure reduction. Am J Physiol (Heart Circ Physiol) 250: H838–H845, 1986.
 125. Fronek K and Zweifach BW. Pre‐ and postcapillary resistances in cat mesentery. Microvasc Res 7: 351–361, 1974.
 126. Dörnyei G, Monos E, Kaley G and Koller A. Myogenic responses of isolated rat skeletal muscle venules: Modulation by norepinephrine and endothelium. Am J Physiol (Heart Circ Physiol) 271: H267–H272, 1996.
 127. Speden RN. The maintenance of arterial constriction at different transmural pressures. J Physiol 229: 361–381, 1973.
 128. Davis MJ. Myogenic response gradient in an arteriolar network. Am J Physiol (Heart Circ Physiol) 264: H2168–H2179, 1993.
 129. Sun D, Kaley G and Koller A. Characteristics and origin of myogenic response in isolated gracilis muscle arterioles. Am J Physiol (Heart Circ Physiol) 266: H1177–H1183, 1994.
 130. Sun D, Messina EJ, Kaley G and Koller A. Characteristics and origin of myogenic response in isolated mesenteric arterioles. Am J Physiol (Heart Circ Physiol) 263: H1486–H1491, 1992.
 131. Mellander S. Control of capillary fluid transfer by precapillary and postcapillary vascular adjustments. Bibl Anat 16: 287–293, 1977.
 132. Bayliss WM. On the local reactions of the arterial wall to changes of internal pressure. J Physiol 28: 220–231, 1902.
 133. Davis MJ and Sikes PJ. Myogenic response of isolated arterioles: test for a rate‐sensitive mechanism. Am J Physiol (Heart Circ Physiol) 259: H1890–H1900, 1990.
 134. Davis MJ and Hill MA. Signaling mechanisms underlying the vascular myogenic response. Physiological Rev 79: 387–423, 1999.
 135. Osol G, Brekke JF, McElroy‐Yaggy K and Gokina NI. Myogenic tone, reactivity and forced dilatation: a three‐phase model of in vitro arterial myogenic behavior. Am J Physiol (Heart Circ Physiol) 283: H2260–H2267, 2002.
 136. Schubert R, Kalentchuk VU and Krien U. Rho kinase inhibition partly weakens myogenic reactivity in rat small arteries by changing calcium sensitivity. Am J Physiol (Heart Circ Physiol) 283: H2288–H2295, 2002.
 137. Lagaud G, Gaudreault N, Moore ED, Van Breemen C and Laher I. Pressure‐dependent myogenic constriction of cerebral arteries occurs independently of voltage‐dependent activation. Am J Physiol (Heart Circ Physiol) 283: H2187–H2195, 2002.
 138. Lombard JH, Eskinder H, Kauser K, Osborn JL and Harder DR. Enhanced norepinephrine sensitivity in renal arteries at elevated transmural pressure. Am J Physiol (Heart Circ Physiol) 259: H29–H33, 1990.
 139. Johansson B and Mellander S. Static and dynamic components in the vascular myogenic response to passive changes in length as revealed by electrical and mechanical recordings from the rat portal vein. Circ Res 36: 76–83, 1975.
 140. Burnstock G and Prosser CL. Responses of smooth muscles to quick stretch; relation of stretch to conduction. Am J Physiol 198: 921–925, 1960.
 141. Sparks HV, Jr. Effect of quick stretch on isolated vascular smooth muscle. Circ Res 15 (Suppl. 1): I‐254–I‐260, 1964.
 142. Simmons RM and Jewell BR. Mechanics and models of muscular contraction. Recent Adv Physiol 31: 87–147, 1974.
 143. Rhodin JAG. Architecture of the vessel wall. In: Handbook of Physiology, The Cardiovascular System, Vol. II ‐ Vascular Smooth Muscle, eds Bohr DF, Somlyo AP and Sparks HV, Jr. Bethesda, MD: American Physiological Society, 1980, pp. 1–31.
 144. Kargacin GJ, Cooke H, Abramson SB and Fay FS. Periodic organization of the contractile apparatus in smooth muscle revealed by the motion of dense bodies in single cells. J Cell Biol 108: 1465–1475, 1989.
 145. Johansson B. Myogenic tone and reactivity: Definitions based on muscle physiology. J Hypertens 7 (Suppl. 4): S5–S8, 1989.
 146. Johnson PC. Autoregulatory responses of cat mesenteric arterioles measured in vivo. Circ Res 22: 199–212, 1968.
 147. Jackson PA and Duling BR. Myogenic response and wall mechanics of arterioles. Am J Physiol (Heart Circ Physiol) 257: H1147–H1155, 1989.
 148. Gunst SJ. Effect of length history on contractile behavior of canine tracheal smooth muscle. Am J Physiol (Cell Physiol) 250: C146–C154, 1986.
 149. Davis MJ and Davidson J. Force‐velocity relationship of myogenically active arterioles. Am J Physiol (Heart Circ Physiol) 282: H165–H174, 2002.
 150. Johansson B. Active state in vascular smooth muscle. Acta Physiol Scand 84: 17A–18A, 1972.
 151. Sigurdsson SB, Johansson B and Mellander S. Rate‐dependent myogenic response of vascular smooth muscle during imposed changes in length and force. Acta Physiol Scand 99: 183–189, 1977.
 152. Smiesko V. Unidirectional rate sensitivity component in local control of vascular tone. Pflügers Arch Eur J Physiol 327: 324–336, 1971.
 153. Dobrin PB. Influence of initial length on length‐tension relationship of vascular smooth muscle. Am J Physiol 225: 664–667, 1973.
 154. Levick JR and Michel CC. The effects of position and skin temperature on the capillary pressures in the fingers and toes. J Physiol 274: 97–109, 1978.
 155. Mellander S, Oberg B and Odelram H. Vascular adjustments to increased transmural pressure in cat and man with special reference to shifts in capillary fluid transfer. Acta Physiol Scand 61: 34–48, 1964.
 156. Nakayama K. Calcium‐dependent contractile activation of cerebral artery produced by quick stretch. Am J Physiol (Heart Circ Physiol) 242: H760–H768, 1982.
 157. Lane MM, Davis AM, Gashev AA, Zawieja DC and Davis MJ. Rate‐sensitive contractile responses of rat mesenteric lymphatic vessels to circumferential stretch. FASEB J 21: A85 (Abstract 587), 2007.
 158. Grände P‐O and Mellander S. Characteristics of static and dynamic regulatory mechanisms in myogenic microvascular control. Acta Physiol Scand 102: 231–245, 1978.
 159. Grände P‐O, Borgstrom P and Mellander S. On the nature of basal vascular tone in cat skeletal muscle and its dependence on transmural pressure stimuli. Acta Physiol Scand 107: 365–376, 1979.
 160. Borgstrom P, Grände P‐O and Lindbom L. Responses of single arterioles in vivo in cat skeletal muscle to change in arterial pressure applied at different rates. Acta Physiol Scand 113: 207–212, 1981.
 161. Greensmith JE and Duling BR. Morphology of the constricted arteriolar wall: physiological implications. Am J Physiol (Heart Circ Physiol) 247: H687–H698, 1984.
 162. Rovick AA and Robertson PA. Interaction of mean and pulse pressures in the circulation of the isolated dog tongue. Circ Res 15: 208–215, 1964.
 163. Shepherd AP and Riedel GL. Effect of pulsatile pressure and metabolic rate on intestinal autoregulation. Am J Physiol (Heart Circ Physiol) 242: H769–H775, 1982.
 164. Speden RN and Warren DM. Myogenic adaptation of rabbit ear arteries to pulsatile internal pressures. J Physiol 391: 313–323, 1987.
 165. Davis MJ and Davidson J. Responses of hamster cheek pouch arterioles to pulsatile pressure in vitro. FASEB J 7: A882 (abstract #M836), 1993.
 166. Goto M, VanBavel E, Giezeman MJMM and Spaan JAE. Vasodilatory effect of pulsatile pressure on coronary resistance vessels. Circ Res 79: 1039–1045, 1996.
 167. Sorop O, Spaan JA and VanBavel E. Pulsation‐induced dilation of subendocardial and subepicardial arterioles: effect on vasodilator sensitivity. Am J Physiol (Heart Circ Physiol) 282: H311–H319, 2002.
 168. Sorop O, Spaan JA, Sweeney TE and VanBavel E. Effect of steady versus oscillating flow on porcine coronary arterioles: involvement of NO and superoxide anion. Circ Res 92: 1344–1351, 2003.
 169. Johnson PC and Intaglietta M. Contributions of pressure and flow sensitivity to autoregulation in mesenteric arterioles. Am J Physiol 231: 1686–1698, 1976.
 170. Burrows ME and Johnson PC. Arteriolar responses to elevation of venous and arterial pressures in cat mesentery. Am J Physiol (Heart Circ Physiol) 245: H796–H807, 1983.
 171. Burrows ME and Johnson PC. Diameter, wall tension and flow in mesenteric arterioles during autoregulation. Am J Physiol (Heart Circ Physiol) 241: H829–H837, 1981.
 172. Zou H, Ratz PH and Hill MA. Role of myosin phosphorylation and [Ca 2+]i in myogenic reactivity and arteriolar tone. Am J Physiol (Heart Circ Physiol) 269: H1590–H1596, 1995.
 173. Oien AH and Aukland K. A mathematical analysis of the myogenic hypothesis with special reference to autoregulation of renal blood flow. Circ Res 52: 241–252, 1983.
 174. Carmines PK, Inscho EW and Gensure RC. Arterial pressure effects on preglomerular microvasculature of juxtamedullary nephrons. Am J Physiol (Ren Fluid Electrolyte Physiol) 258: F94–F102, 1990.
 175. Loutzenhiser R, Bidani A and Chilton L. Renal myogenic response: kinetic attributes and physiological role. Circ Res 90: 1316–1324, 2002.
 176. Björnberg J, Albert U and Mellander S. Resistance responses in proximal arterial vessels, arterioles and veins during reactive hyperaemia in skeletal muscle and their underlying regulatory mechanisms. Acta Physiol Scand 139: 535–550, 1990.
 177. Johnson PC. The myogenic response in the microcirculation and its interaction with other control systems. J Hypertens 7 (Suppl. 4): S33–S39, 1989.
 178. Bevan JA and Ljung B. Longitudinal propagation of myogenic activity in rabbit arteries and in the rat portal vein. Acta Physiol Scand 90: 703–715, 1974.
 179. Rivers RJ. Remote effects of pressure changes in arterioles. Am J Physiol (Heart Circ Physiol) 268: H1379–H1382, 1995.
 180. Casellas D, Carmines PK and Navar LG. Microvascular reactivity of in vitro blood perfused juxtamedullary nephrons from rats. Kidney Int 28: 752–759, 1985.
 181. Kirton CA and Loutzenhiser R. Alterations in basal protein kinase C activity modulate renal afferent arteriolar myogenic reactivity. Am J Physiol (Heart Circ Physiol) 275: H467–H475, 1998.
 182. Koch AR. Some mathematical forms of autoregulatory models. Circ Res 15 (Suppl. 1): I‐269–I‐278, 1964.
 183. Hall JE. Regulation of renal hemodynamics. Int Rev Physiol 26: 243–321, 1982.
 184. Folkow B. Description of the myogenic hypothesis. Circ Res Suppl. 1: I‐279–I‐287, 1964.
 185. Cipolla MJ and Osol G. Vascular smooth muscle actin cytoskeleton in cerebral artery forced dilatation. Stroke 29: 1223–1228, 1998.
 186. Cipolla MJ, Gokina NI and Osol G. Pressure‐induced actin polymerization in vascular smooth muscle as a mechanism underlying myogenic behavior. FASEB J 16: 72–76, 2002.
 187. Fronek K and Zweifach BW. Microvascular blood flow in cat tenuissimus muscle. Microvasc Res 14: 181–189, 1977.
 188. Lindbom L, Tuma RF and Arfors KE. Blood flow in the rabbit tenuissimus muscle influence of preparative procedures for intravital microscopic observation. Acta Physiol Scand 114: 121–127, 1982.
 189. Duling BR. The preparation and use of the hamster cheek pouch for studies of the microcirculation. Microvasc Res 5: 423–429, 1973.
 190. Duling BR, Gore RW, Dacey RG, Jr. and Damon DN. Methods for isolation, cannulation and in vitro study of single microvessels. Am J Physiol (Heart Circ Physiol) 241: H108–H116, 1981.
 191. Davis MJ, Kuo L, Chilian WM and Muller JM. Isolated. Perfused Microvessels. In: Clinically Applied Microcirculation Research. eds Barker JH Anderson GL and Menger MD, Boca Raton, FL: CRC Press, 1995, pp. 435–456.
 192. Duling BR and Rivers RJ. Isolation, cannulation and perfusion of microvessels. In: Microcirculatory Technology, eds Baker CH and Nastuk WL. New York: Academic Press, 1986, pp. 265–280.
 193. D'Agrosa LS. Patterns of venous vasomotion in the bat wing. Am J Physiol 218: 530–535, 1970.
 194. Crowe MJ, von der Weid PY, Brock JA and Van Helden DF. Coordination of contractile activity in guinea‐pig mesenteric lymphatics. J Physiol 500: 235–244, 1997.
 195. McHale N, Hollywood M, Sergeant G and Thornbury K. Origin of spontaneous rhythmicity in smooth muscle. J Physiol 570: 23–28, 2006.
 196. Muthuchamy M, Gashev A, Boswell N, Dawson N and Zawieja D. Molecular and functional analyses of the contractile apparatus in lymphatic muscle. FASEB J 17: 920–922, 2003.
 197. Kuo L, Chilian WM and Davis MJ. Coronary arteriolar myogenic response is independent of endothelium. Circ Res 66: 860–866, 1990.
 198. Davis MJ and Gore RW. Capillary pressures in rat intestinal muscle and mucosal villi during venous pressure elevation. Am J Physiol (Heart Circ Physiol) 249: H174–H187, 1985.
 199. Lash JM, Bohlen HG and Waite L. Mechanical characteristics and active tension generation in rat intestinal arterioles. Am J Physiol (Heart Circ Physiol) 260: H1561–H1574, 1991.
 200. Mellander S and Albert U. Effects of increased and decreased tissue pressure on haemodynamic and capillary events in cat skeletal muscle. J Physiol 481 (1): 163–175, 1994.
 201. Mahy IR, Tooke JE and Shore AC. Capillary pressure during and after incremental venous pressure elevation in man. J Physiol 485: 213–219, 1995.
 202. Shore AC, Sandeman DD and Tooke JE. Effect of an increase in systemic blood pressure on nailfold capillary pressure in humans. Am J Physiol (Heart Circ Physiol) 265: H820–H823, 1993.
 203. Maspers M, Björnberg and Mellander S. Relation between capillary pressure and vascular tone over the range from maximum dilatation to maximum constriction in cat skeletal muscle. Acta Physiol Scand 140: 73–83, 1990.
 204. Meininger GA and Davis MJ. Cellular mechanisms involved in the vascular myogenic response. Am J Physiol (Heart Circ Physiol) 263: H647–H659, 1992.
 205. Harder DR, Narayanan J, Gebremedhin D and Roman RJ. Transduction of physical force by the vascular wall: Role of phospholipase C and cytochrome P450 metabolites of arachidonic acid. Trends Cardiovasc Med 5: 7–14, 1995.
 206. Osol G. Mechanotransduction by vascular smooth muscle. J Vasc Res 32: 275–292, 1995.
 207. Schubert R and Mulvany MJ. The myogenic response: established facts and attractive hypotheses. Clin Sci 96: 313–326, 1999.
 208. Uchida E and Bohr DF. Myogenic tone in isolated perfused resistance vessels from rats. Am J Physiol 216: 1343–1350, 1969.
 209. Uchida E and Bohr DF. Myogenic tone in isolated perfused vessels. Occurrence among vascular beds and along vascular trees. Circ Res 25: 549–555, 1969.
 210. Bulbring E. Correlation between membrane potential, spike discharge and tension in smooth muscle. J Physiol 128: 200–221, 1955.
 211. Knot HJ and Nelson MT. Regulation of membrane potential and diameter by voltage‐dependent K + channels in rabbit myogenic cerebral arteries. Am J Physiol (Heart Circ Physiol) 269: H348–H355, 1995.
 212. Brayden JE and Nelson MT. Regulation of arterial tone by activation of calcium‐dependent potassium channels. Science 256: 532–535, 1992.
 213. Nelson MT and Quayle JM. Physiological roles and properties of potassium channels in arterial smooth muscle. Am J Physiol (Cell Physiol) 268: C799–C822, 1995.
 214. Zou H, Ratz PH and Hill MA. Temporal aspects of Ca 2+ and myosin phosphorylation during myogenic and norepinephrine‐induced arteriolar constriction. J Vasc Res 37: 556–567, 2000.
 215. Harder DR. Pressure‐dependent membrane depolarization in cat middle cerebral artery. Circ Res 55: 197–202, 1984.
 216. Eskinder H, Harder DR and Lombard JH. Role of the vascular endothelium in regulating the response of small arteries of the dog kidney to transmural pressure elevation and reduced PO2. Circ Res 66: 1427–1435, 1990.
 217. Bérczi V, Greene AS, Dörnyei G, Csengódy J, Hódi G, Kádár A and Monos E. Venous myogenic tone: Studies in human and canine vessels. Am J Physiol (Heart Circ Physiol) 263: H315–H320, 1992.
 218. Scotland RS, Chauhan S, Davis C, De Felipe C, Hunt S, Kabir J, Kotsonis P, Oh U and Ahluwalia A. Vanilloid receptor TRPV1, sensory C‐fibers and vascular autoregulation: a novel mechanism involved in myogenic constriction. Circ Res 95: 1027–1034, 2004.
 219. McCarron JG, Osol G and Halpern W. Myogenic responses are independent of the endothelium in rat pressurized posterior cerebral arteries. Blood Vessels 26: 315–319, 1989.
 220. Falcone JC, Davis MJ and Meininger GA. Endothelial independence of myogenic response in isolated skeletal muscle arterioles. Am J Physiol (Heart Circ Physiol) 260: H130–H135, 1991.
 221. Harder DR, Sanchez‐Ferrer C, Kauser K, Stekiel WJ and Rubanyi GM. Pressure releases a transferable endothelial contractile factor in cat cerebral arteries. Circ Res 65: 193–198, 1989.
 222. Scotland RS, Chauhan S, Vallance PJ and Ahluwalia A. An endothelium‐derived hyperpolarizing factor‐like factor moderates myogenic constriction of mesenteric resistance arteries in the absence of endothelial nitric oxide synthase‐derived nitric oxide. Hypertension 38: 833–839, 2001.
 223. Katusic ZS, Shepherd JT and Vanhoutte PM. Endothelium‐dependent contraction to stretch in canine basilar arteries. Am J Physiol (Heart Circ Physiol) 252: H671–H673, 1987.
 224. Kauser K, Stekiel WJ, Rubanyi G and Harder DR. Mechanism of action of EDRF on pressurized arteries: effect on K + conductance. Circ Res 65: 199–204, 1989.
 225. Harder DR. Pressure‐induced myogenic activation of cat cerebral arteries is dependent on intact endothelium. Circ Res 60: 102–107, 1987.
 226. de Jong BW, Bakker Schut TC, Wolffenbuttel KP, Nijman JM, Kok DJ and Puppels GJ. Identification of bladder wall layers by Raman spectroscopy. J Urol 168: 1771–1778, 2002.
 227. Humphries MJ, Travis MA, Clark K and Mould AP. Mechanisms of integration of cells and extracellular matrices by integrins. Biochem Soc Trans 32: 822–825, 2004.
 228. Hynes RO. The emergence of integrins: a personal and historical perspective. Matrix Biol 23: 333–340, 2004.
 229. Katsumi A, Orr AW, Tzima E and Schwartz MA. Integrins in mechanotransduction. J Biol Chem 279: 12001–12004, 2004.
 230. Schwartz MA. Integrin signaling revisited. Trends Cell Biol 11: 466–470, 2001.
 231. Sheetz MP, Felsenfeld DP and Galbraith CG. Cell migration: Regulation of force on extracellular‐matrix‐integrin complexes. Trends Cell Biol 8: 51–54, 1998.
 232. Schlaepfer DD and Hunter T. Integrin signalling and tyrosine phosphorylation: just the FAKs? Trends Cell Biol 8: 151–157, 1998.
 233. Wu X, Mogford JE, Platts SH, Davis GE, Meininger GA and Davis MJ. Modulation of calcium current in arteriolar smooth muscle by αv β3 and α5 β1 integrin ligands. J Cell Biol 143: 241–252, 1998.
 234. Wu X, Davis GE, Meininger GA, Wilson E and Davis MJ. Regulation of the L‐type calcium channel by α5 β1 integrin requires signaling between focal adhesion proteins. J Biol Chem 276: 30285–30292, 2001.
 235. Hofmann G, Bemabei PA, Crociani O, Cherubini A, Guasti L, Pillozzi S, Lastraioli E, Polvani S, Bartolozzi B, Solazzo V, Gragnani L, Defilippi P, Rosati B, Wanke E, Olivotto M and Arcangeli A. HERG K + channels activation during β1 integrin‐mediated adhesion to fibronectin induces an up‐regulation of αv β3 integrin in the preosteoclastic leukemia cell line FLG 29.1. J Biol Chem 276: 4923–4931, 2001.
 236. Browe DM and Baumgarten CM. Stretch of β1 integrin activates an outwardly rectifying chloride current via FAK and Src in rabbit ventricular myocytes. J Gen Physiol 122: 689–702, 2003.
 237. Davis MJ, Wu X, Nurkiewicz TR, Kawasaki J, Davis GE, Hill MA and Meininger GA. Integrins and mechanotransduction of the vascular myogenic response. Am J Physiol (Heart Circ Physiol) 280: H1427–H1433., 2001.
 238. Martinez‐Lemus LA, Wu X, Wilson E, Hill MA, Davis GE, Davis MJ and Meininger GA. Integrins as unique receptors for vascular control. J Vasc Res 40: 211–233, 2003.
 239. Martinez‐Lemus LA, Sun Z, Trache A, Trzciakowski JP and Meininger GA. Integrins and regulation of the microcirculation: from arterioles to molecular studies using atomic force microscopy. Microcirculation 12: 99–112, 2005.
 240. Mogford JE, Davis GE, Platts SH and Meininger GA. Vascular smooth muscle αv β3 integrin mediates arteriolar vasodilation in response to RGD peptides. Circ Res 79: 821–826, 1996.
 241. D'Angelo G, Mogford JE, Davis GE, Davis MJ and Meininger GA. Integrin‐mediated reduction in vascular smooth muscle Ca 2+i induced by RGD‐containing peptide. Am J Physiol (Heart Circ Physiol) 272: H2065–H2070, 1997.
 242. Hein TW, Platts SH, Waitkus‐Edwards KR, Kuo L, Mousa SA and Meininger GA. Integrin‐binding peptides containing RGD produce coronary arteriolar dilation via cyclooxygenase activation. Am J Physiol (Heart Circ Physiol) 281: H2378–H2384, 2001.
 243. Waitkus‐Edwards KR, Martinez‐Lemus LA, Wu X, Trzeciakowski JP, Davis MJ, Davis GE and Meininger GA. α4 β1 integrin activation of L‐type calcium channels in vascular smooth muscle causes arteriole vasoconstriction. Circ Res 90: 473–480, 2002.
 244. Martinez‐Lemus LA, Crow T, Davis MJ and Meininger GA. αv β3 ‐and α5 β1 ‐integrin blockade inhibits myogenic constriction of skeletal muscle resistance arterioles. Am J Physiol (Heart Circ Physiol) 289: H322–H329, 2005.
 245. Diez‐Marques ML, Ruiz‐Torres MP, Griera M, Lopez‐Ongil S, Saura M, Rodriguez‐Puyol D and Rodriguez‐Puyol M. Arg‐Gly‐Asp (RGD)‐containing peptides increase soluble guanylate cyclase in contractile cells. Cardiovasc Res 69: 359–369, 2006.
 246. Gui P, Wu X, Ling S, Stotz SC, Winkfein RJ, Wilson E, Davis GE, Braun AP, Zamponi GW and Davis MJ. Integrin receptor activation triggers converging regulation of Cav1.2 calcium channels by c‐Src and protein kinase A pathways. J Biol Chem 281: 14015–14025, 2006.
 247. Baneyx G, Baugh L and Vogel V. Fibronectin extension and unfolding within cell matrix fibrils controlled by cytoskeletal tension. Proc Natl Acad Sci USA 99: 5139–5143, 2002.
 248. Davis GE, Bayless KJ, Davis MJ and Meininger GA. Regulation of tissue injury responses by the exposure of matricryptic sites within extracellular matrix molecules. Am J Pathol 156: 1489–1498, 2000.
 249. Pichard V, Honore S, Kovacic H, Li C, Prevot C, Briand C and Rognoni JB. Adhesion, actin cytoskeleton organisation and the spreading of colon adenocarcinoma cells induced by EGF are mediated by α2 β1 integrin low clustering through focal adhesion kinase. Histochem Cell Biol 116: 337–348, 2001.
 250. Davis MJ, Donovitz JA and Hood JD. Stretch‐activated single‐channel and whole‐cell currents in vascular smooth muscle cells. Am J Physiol (Cell Physiol) 262: C1083–C1088, 1992.
 251. Wellner MC and Isenberg G. Properties of stretch‐activated channels in myocytes from the guinea‐pig urinary bladder. J Physiol 466: 213–227, 1993.
 252. Wellner M‐C and Isenberg G. Stretch effects on whole‐cell currents of guinea‐pig urinary bladder myocytes. J Physiol 480: 439–448, 1994.
 253. Setoguchi M, Ohya Y, Abe I and Fujishima M. Stretch‐activated whole‐cell currents in smooth muscle cells from mesenteric resistance artery of guinea‐pig. J Physiol 501: 343–353, 1997.
 254. Ohya Y, Adachi N, Nakamura Y, Setoguchi M, Abe I and Fujishima M. Stretch‐activated channels in arterial smooth muscle of genetic hypertensive rats. Hypertension 31: 254–258, 1998.
 255. Laher I and Bevan JA. Stretch of vascular smooth muscle activates tone and 45 Ca 2+ influx. J Hypertens 7 (Suppl. 4): S17–S20, 1989.
 256. McCarron JG, Crichton CA, Langton PD, MacKenzie A and Smith GL. Myogenic contraction by modulation of voltage‐dependent calcium currents in isolated rat cerebral arteries. J Physiol 498: 371–379, 1997.
 257. Yanagisawa T and Okada Y. KCl depolarization increases Ca 2+ sensitivity of contractile elements in coronary arterial smooth muscle. Am J Physiol (Heart Circ Physiol) 267: H614–H621, 1994.
 258. Mita M, Yanagihara H, Hishinuma S, Saito M and Walsh MP. Membrane depolarization‐induced contraction of rat caudal arterial smooth muscle involves Rho‐associated kinase. Biochem J 364: 431–440, 2002.
 259. Urban NH, Berg KM and Ratz PH. K + depolarization induces RhoA kinase translocation to caveolae and Ca 2+ sensitization of arterial muscle. Am J Physiol (Cell Physiol) 285: C1377–1385, 2003.
 260. Hill MA, Davis MJ, Song J and Zou H. Calcium dependence of indolactam‐mediated contractions in resistance vessels. J Pharmacol Exp Therapeut 276: 867–874, 1996.
 261. Jackson WF and Blair KL. Characterization and function of Ca 2+ ‐activated K + channels in arteriolar muscle cells. Am J Physiol (Heart Circ Physiol) 274: H27–H34, 1998.
 262. Kotecha N and Hill MA. Myogenic contraction in rat skeletal muscle arterioles: smooth muscle membrane potential and Ca 2+ signaling. Am J Physiol (Heart Circ Physiol) 289: H1326–H1334, 2005.
 263. Shaul PW and Anderson RG. Role of plasmalemmal caveolae in signal transduction. Am J Physiol (Lung Respir Physiol) 275: L843–L851, 1998.
 264. Everson WV and Smart EJ. Influence of caveolin, cholesterol and lipoproteins on nitric oxide synthase: implications for vascular disease. Trends Cardiovasc Med 11: 246–250, 2001.
 265. Taggart MJ. Smooth muscle excitation‐contraction coupling: a role for caveolae and caveolins? News Physiol Sci 16: 61–65, 2001.
 266. Ushio‐Fukai M, Hilenski L, Santanam N, Becker PL, Ma Y, Griendling KK and Alexander RW. Cholesterol depletion inhibits epidermal growth factor receptor transactivation by angiotensin II in vascular smooth muscle cells: role of cholesterol‐rich microdomains and focal adhesions in angiotensin II signaling. J Biol Chem 276: 48269–48275, 2001.
 267. Bergdahl A and Sward K. Caveolae‐associated signalling in smooth muscle. Can J Physiol Pharmacol 82: 289–299, 2004.
 268. Sedding DG, Hermsen J, Seay U, Eickelberg O, Kummer W, Schwencke C, Strasser RH, Tillmanns H and Braun‐Dullaeus RC. Caveolin‐1 facilitates mechanosensitive protein kinase B (Akt) signaling in vitro and in vivo. Circ Res 96: 635–642, 2005.
 269. Yu J, Bergaya S, Murata T, Alp IF, Bauer MP, Lin MI, Drab M, Kurzchalia TV, Stan RV and Sessa WC. Direct evidence for the role of caveolin‐1 and caveolae in mechanotransduction and remodeling of blood vessels. J Clin Invest 116: 1284–1291, 2006.
 270. Drab M, Verkade P, Elger M, Kasper M, Lohn M, Lauterbach B, Menne J, Lindschau C, Mende F, Luft FC, Schedl A, Haller H and Kurzchalia TV. Loss of caveolae, vascular dysfunction and pulmonary defects in caveolin‐1 gene‐disrupted mice. Science 293: 2449–2452, 2001.
 271. Razani B, Engelman JA, Wang XB, Schubert W, Zhang XL, Marks CB, Macaluso F, Russell RG, Li M, Pestell RG, Di Vizio D, Hou H. Jr., Kneitz B, Lagaud G, Christ GJ, Edelmann W and Lisanti MP. Caveolin‐1 null mice are viable but show evidence of hyperproliferative and vascular abnormalities. J Biol Chem 276: 38121–38138, 2001.
 272. Adebiyi A, Zhao G, Cheranov SY, Ahmed A and Jaggar JH. Caveolin‐1 abolishment attenuates the myogenic response in murine cerebral arteries. Am J Physiol (Heart Circ Physiol) 292: H1584–H1592, 2007.
 273. Potocnik SJ, Jenkins N, Murphy TV and Hill MA. Membrane cholesterol depletion with beta‐cyclodextran impairs pressure‐induced contraction and calcium signalling in isolated skeletal muscle arterioles. J Vasc Res 44: 292–302, 2007.
 274. Park H, Go YM, St John PL, Maland MC, Lisanti MP, Abrahamson DR and Jo H. Plasma membrane cholesterol is a key molecule in shear stress‐dependent activation of extracellular signal‐regulated kinase. J Biol Chem 273: 32304–32311, 1998.
 275. Hamill OP and Martinac B. Molecular basis of mechanotransduction in living cells. Physiol Rev 81: 685–740, 2001.
 276. Sukharev S and Corey DP. Mechanosensitive channels: multiplicity of families and gating paradigms. Sci STKE 2004: re4, 2004.
 277. Langton PD. Calcium channel currents recorded from isolated myocytes of rat basilar artery are stretch sensitive. J Physiol 471: 1–11, 1993.
 278. Sachs F. Mechanical transduction by ion channels: How forces reach the channel. Soc Gen Physiol Ser 52: 209–218, 1997.
 279. Martinac B and Hamill OP. Gramicidin A channels switch between stretch activation and stretch inactivation depending on bilayer thickness. Proc Natl Acad Sci USA 99: 4308–4312, 2002.
 280. Knot HJ and Nelson MT. Regulation of arterial diameter and wall [Ca 2+] in cerebral arteries of rat by membrane potential and intravascular pressure. J Physiol 508: 199–209, 1998.
 281. Kirber MT, Ordway RW, Clapp LH, Walsh JV, Jr. and Singer JJ. Both membrane stretch and fatty acids directly activate large conductance Ca 2+ ‐activated K + channels in vascular smooth muscle cells. FEBS Lett 297: 24–28, 1992.
 282. Wu X and Davis MJ. Characterization of stretch‐activated cation current in coronary smooth muscle cells. Am J Physiol (Heart Circ Physiol) 280: H1751–H1761., 2001.
 283. Barritt G and Rychkov G. TRPs as mechanosensitive channels. Nat Cell Biol 7: 105–107, 2005.
 284. Maroto R, Raso A, Wood TG, Kurosky A, Martinac B and Hamill OP. TRPC1 forms the stretch‐activated cation channel in vertebrate cells. Nat Cell Biol 7: 179–185, 2005.
 285. Corey DP, Garcia‐Anoveros J, Holt JR, Kwan KY, Lin SY, Vollrath MA, Amalfitano A, Cheung EL, Derfler BH, Duggan A, Geleoc GS, Gray PA, Hoffman MP, Rehm HL, Tamasauskas D and Zhang DS. TRPA1 is a candidate for the mechanosensitive transduction channel of vertebrate hair cells. Nature 432: 723–730, 2004.
 286. Zhang Y, Gao F, Popov VL, Wen JW and Hamill OP. Mechanically gated channel activity in cytoskeleton‐deficient plasma membrane blebs and vesicles from Xenopus oocytes. J Physiol 523: 117–130, 2000.
 287. Welsh DG, Morielli AD, Nelson MT and Brayden JE. Transient receptor potential channels regulate myogenic tone of resistance arteries. Circ Res 90: 248–250, 2002.
 288. Earley S, Waldron BJ and Brayden JE. Critical role for transient receptor potential channel TRPM4 in myogenic constriction of cerebral arteries. Circ Res 95: 922–929, 2004.
 289. Flavahan NA, Bailey SR, Flavahan WA, Mitra S and Flavahan S. Imaging remodeling of the actin cytoskeleton in vascular smooth muscle cells after mechanosensitive arteriolar constriction. Am J Physiol (Heart Circ Physiol) 288: H660–669, 2005.
 290. Lin SY and Corey DP. TRP channels in mechanosensation. Curr Opin Neurobiol 15: 350–357, 2005.
 291. Minke B and Cook B. TRP channel proteins and signal transduction. Physiol Rev 82: 429–472, 2002.
 292. Clapham DE. TRP channels as cellular sensors. Nature 426: 517–524, 2003.
 293. Beech DJ, Muraki K and Flemming R. Non‐selective cationic channels of smooth muscle and the mammalian homologues of Drosophila TRP. J Physiol 559: 685–706, 2004.
 294. Owsianik G, Talavera K, Voets T and Nilius B. Permeation and Selectivity of TRP Channels. Annu Rev Physiol 68: 685–717, 2006.
 295. Nilius B and Droogmans G. Ion channels and their functional role in vascular endothelium. Physiol Rev 81: 1415–1459, 2001.
 296. Narayanan J, Imig M, Roman RJ and Harder DR. Pressurization of isolated renal arteries increases inositol trisphosphate and diacylglycerol. Am J Physiol (Heart Circ Physiol) 266: H1840–H1845, 1994.
 297. Albert AP and Large WA. Synergism between inositol phosphates and diacylglycerol on native TRPC6‐like channels in rabbit portal vein myocytes. J Physiol 552: 789–795, 2003.
 298. Basora N, Boulay G, Bilodeau L, Rousseau E and Payet MD. 20‐hydroxyeicosatetraenoic acid (20‐HETE) activates mouse TRPC6 channels expressed in HEK293 cells. J Biol Chem 278: 31709–31716, 2003.
 299. Albert AP and Large WA. Signal transduction pathways and gating mechanisms of native TRP‐like cation channels in vascular myocytes. J Physiol 570: 45–51, 2006.
 300. Nilius B, Prenen J, Droogmans G, Voets T, Vennekens R, Freichel M, Wissenbach U and Flockerzi V. Voltage dependence of the Ca 2+ ‐activated cation channel TRPM4. J Biol Chem 278: 30813–30820, 2003.
 301. Earley S, Straub SV and Brayden J. Protein kinase C regulates vascular myogenic tone through activation of TRPM4. Am J Physiol (Heart Circ Physiol) 292: H2613–H2622, 2007.
 302. Hill MA and Davis MJ. Coupling a change in intraluminal pressure to vascular smooth muscle depolarization: still stretching for an explanation. Am J Physiol (Heart Circ Physiol) 292: H2570–H2572, 2007.
 303. Beech DJ. TRPC1: store‐operated channel and more. Pflugers Arch Eur J Physiol 451: 53–60, 2005.
 304. Spassova MA, Hewavitharana T, Xu W, Soboloff J and Gill DL. A common mechanism underlies stretch activation and receptor activation of TRPC6 channels. Proc Natl Acad Sci USA 103: 16586–16591, 2006.
 305. Reading SA, Earley S, Waldron BJ, Welsh DG and Brayden JE. TRPC3 mediates pyrimidine receptor‐induced depolarization of cerebral arteries. Am J Physiol (Heart Circ Physiol) 288: H2055–H2061, 2005.
 306. Earley S, Heppner TJ, Nelson MT and Brayden JE. TRPV4 forms a novel Ca 2+ signaling complex with ryanodine receptors and BKCa channels. Circ Res 97: 1270–1279, 2005.
 307. Drummond HA, Gebremedhin D and Harder DR. Degenerin/epithelial Na + channel proteins: components of a vascular mechanosensor. Hypertension 44: 643–648, 2004.
 308. Mano I and Driscoll M. DEG/ENaC channels: a touchy superfamily that watches its salt. Bioessays 21: 568–578, 1999.
 309. Carattino MD, Sheng S and Kleyman TR. Epithelial Na+ channels are activated by laminar shear stress. J Biol Chem 279: 4120–4126, 2004.
 310. Satlin LM, Sheng S, Woda CB and Kleyman TR. Epithelial Na + channels are regulated by flow. Am J Physiol (Renal, Fluid, Elect Physiol) 280: F1010–1018, 2001.
 311. Shakibaei M and Mobasheri A. β1‐integrins co‐localize with Na, K‐ATPase, epithelial sodium channels (ENaC) and voltage activated calcium channels (VACC) in mechanoreceptor complexes of mouse limb‐bud chondrocytes. Histol Histopathol 18: 343–351, 2003.
 312. Awayda MS, Shao W, Guo F, Zeidel M and Hill WG. ENaC‐membrane interactions: regulation of channel activity by membrane order. J Gen Physiol 123: 709–727, 2004.
 313. McKnight NL and Frangos JA. Strain rate mechanotransduction in aligned human vascular smooth muscle cells. Annu Biomed Eng 31: 239–249, 2003.
 314. Gudi S, Nolan JP and Frangos JA. Modulation of GTPase activity of G proteins by fluid shear stress and phospholipid composition. Proc Natl Acad Sci USA 95: 2515–2519, 1998.
 315. Benos DJ. Sensing tension: recognizing ENaC as a stretch sensor. Hypertension 44: 616–617, 2004.
 316. Canessa CM, Schild L, Buell G, Thorens B, Gautschi I, Horisberger JD and Rossier BC. Amiloride‐sensitive epithelial Na + channel is made of three homologous subunits. Nature 367: 463–467, 1994.
 317. Jernigan NL and Drummond HA. Vascular ENaC proteins are required for renal myogenic constriction. Am J Physiol (Renal Physiol) 289: F891–F901, 2005.
 318. Jernigan NL and Drummond HA. Myogenic vasoconstriction in mouse renal interlobar arteries: role of endogenous beta and gamma ENaC. Am J Physiol (Renal Physiol) 291: F1184–F1191, 2006.
 319. Nelson MT, Conway MA, Knot HJ and Brayden JE. Chloride channel blockers inhibit myogenic tone in rat cerebral arteries. J Physiol 502: 259–264, 1997.
 320. Raina H, Ella SR and Hill MA. Decreased activity of the smooth muscle Na + /Ca + ‐exchanger impairs arteriolar myogenic reactivity. J Physiol 586: 1669–1681, 2008.
 321. Pacaud P, Loirand G, Baron A, Mironneau C and Mironneau J. Ca 2+ channel activation and membrane depolarization mediated by Cl − channels in response to noradrenaline in vascular myocytes. Br J Pharmacol 104: 1000–1006, 1991.
 322. Yamazaki J, Duan D, Janiak J, Kuenzli K, Horowitz B and Hume JR. Functional and molecular expression of volume regulated chloride channels in canine vascular smooth muscle cells. J Physiol 507: 729–736, 1998.
 323. Doughty JM and Langton PD. Measurement of chloride flux associated with the myogenic response in rat cerebral arteries. J Physiol 534: 753–761, 2001.
 324. Doughty JM and Langton PD. A transient dilatation of pressurised rat cerebral arteries during rapid pressure increases is mediated by nitric oxide. Pflügers Arch Eur J Physiol 436: 220–226, 1998.
 325. Doughty JM, Miller AL and Langton PD. Non‐specificity of chloride channel blockers in rat cerebral arteries: block of the L‐type calcium channel. J Physiol 507: 433–439, 1998.
 326. Remillard CV, Lupien MA, Crepeau V and Leblanc N. Role of Ca 2+ ‐ and swelling‐activated Cl − channels in α1 ‐adrenoceptor‐mediated tone in pressurized rabbit mesenteric arterioles. Cardiovasc Res 46: 557–568, 2000.
 327. Nelson MT. Bayliss, myogenic tone and volume‐regulated chloride channels in arterial smooth muscle. J Physiol 507: 629, 1998.
 328. ZhuGe R, Sims SM, Tuft RA, Fogarty KE and Walsh JV, Jr. Ca 2+ sparks activate K + and Cl − channels, resulting in spontaneous transient currents in guinea‐pig tracheal myocytes. J Physiol 513: 711–718, 1998.
 329. Duan D, Winter C, Cowley S, Hume JR and Horowitz B. Molecular identification of a volume‐regulated chloride channel. Nature 390: 417–421, 1997.
 330. Masuda T, Tomiyama Y, Kitahata H, Kuroda Y and Oshita S. Effect of propofol on hypotonic swelling‐induced membrane depolarization in human coronary artery smooth muscle cells. Anesthesiology 100: 648–656, 2004.
 331. Welsh DG, Nelson MT, Eckman DM and Brayden JE. Swelling‐activated cation channels mediate depolarization of rat cerebrovascular smooth muscle by hyposmolarity and intravascular pressure. J Physiol 527 139–148, 2000.
 332. Bryan RM, You J, Phillips SC, Andresen JJ, Lloyd EE, Rogers PA, Dryer SE and Marrelli SP. Evidence for two‐pore domain potassium channels in rat cerebral arteries. Am J Physiol (Heart Circ Physiol) 291: H770–H780, 2006.
 333. Jackson WF. Potassium channels and regulation of the microcirculation. Microcirculation 5: 85–90, 1998.
 334. Jackson WF. Potassium channels in the peripheral microcirculation. Microcirculation 12: 113–127, 2005.
 335. Sobey CG. Potassium channel function in vascular disease. Arterioscl Thromb Vascul Biol 21: 28–38, 2001.
 336. Albarwani S, Nemetz LT, Madden JA, Tobin AA, England SK, Pratt PF and Rusch NJ. Voltage‐gated K + channels in rat small cerebral arteries: molecular identity of the functional channels. J Physiol 551: 751–763, 2003.
 337. Cheong A, Dedman AM and Beech DJ. Expression and function of native potassium channel [KV α1] subunits in terminal arterioles of rabbit. J Physiol 534: 691–700, 2001.
 338. Cheong A, Dedman AM, Xu SZ and Beech DJ. KVal channels in murine arterioles: differential cellular expression and regulation of diameter. Am J Physiol (Heart Circ Physiol) 281: H1057–H1065, 2001.
 339. Plane F, Johnson R, Kerr P, Wiehler W, Thorneloe K, Ishii K, Chen T and Cole W. Heteromultimeric Kvl channels contribute to myogenic control of arterial diameter. Circ Res 96: 216–224, 2005.
 340. Cheong A, Quinn K, Dedman AM and Beech DJ. Activation thresholds of KV, BK and ClCa channels in smooth muscle cells in pial precapillary arterioles. J Vasc Res 39: 122–130, 2002.
 341. Ahmed A, Waters CM, Leffler CW and Jaggar JH. Ionic mechanisms mediating the myogenic response in newborn porcine cerebral arteries. Am J Physiol (Heart Circ Physiol) 287: H2061–2069, 2004.
 342. Cox RH, Folander K and Swanson R. Differential expression of voltage‐gated K + channel genes in arteries from spontaneously hypertensive and Wistar‐Kyoto rats. Hypertension 37: 1315–1322, 2001.
 343. Li H, Chai Q, Gutterman DD and Liu Y. Elevated glucose impairs cAMP‐mediated dilation by reducing Kv channel activity in rat small coronary smooth muscle cells. Am J Physiol (Heart Circ Physiol) 285: H1213–H1219, 2003.
 344. VanBavel E and Mulvany MJ. Role of wall tension in the vasoconstrictor response of cannulated rat mesenteric small arteries. J Physiol 477: 103–115, 1994.
 345. Chen TT, Luykenaar KD, Walsh EJ, Walsh MP and Cole WC. Key role of Kv 1 channels in vasoregulation. Circ Res 99: 53–60, 2006.
 346. Aiello EA, Clement‐Chomienne O, Sontag DP, Walsh MP and Cole WC. Protein kinase C inhibits delayed rectifier K + current in rabbit vascular smooth muscle cells. Am J Physiol (Heart Circ Physiol) 271: H109–H119, 1996.
 347. Cole WC, Clement‐Chomienne O and Aiello EA. Regulation of 4‐aminopyridine‐sensitive, delayed rectifier K + channels in vascular smooth muscle by phosphorylation. Biochem Cell Biol 74: 439–447, 1996.
 348. Sobey CG and Faraci FM. Inhibitory effect of 4‐aminopyridine on responses of the basilar artery to nitric oxide. Br J Pharmacol 126: 1437–1443, 1999.
 349. Hirst GD, Silverberg GD and van Helden DF. The action potential and underlying ionic currents in proximal rat middle cerebral arterioles. J Physiol 371: 289–304, 1986.
 350. Edwards FR, Hirst GDS and Silverberg GD. Inward rectification in rat cerebral arterioles: involvement of potassium ions in autoregulation. J Physiol 404: 455–466, 1988.
 351. Quayle JM, McCarron JG, Brayden JE and Nelson MT. Inward rectifier K + currents in smooth muscle cells from rat resistance‐sized cerebral arteries. Am J Physiol (Cell Physiol) 265: C1363–C1370, 1993.
 352. Robertson BE, Bonev AD and Nelson MT. Inward rectifier K + currents in smooth muscle cells from rat coronary arteries: block by Mg 2+, Ca 2+, and Ba 2+. Am J Physiol (Heart Circ Physiol) 271: H696–705, 1996.
 353. Knot HJ, Zimmermann PA and Nelson MT. Extracellular K + ‐induced hyperpolarizations and dilatations of rat coronary and cerebral arteries involve inward rectifier K + channels. J Physiol 492: 419–430, 1996.
 354. Johnson TD, Marrelli SP, Steenberg ML, Childres WF and Bryan RM, Jr. Inward rectifier potassium channels in the rat middle cerebral artery. Am J Physiol (Regul, Integr Comp Physiol) 274: R541–R547, 1998.
 355. Chrissobolis S, Ziogas J, Chu Y, Faraci FM and Sobey CG. Role of inwardly rectifying K + channels in K + ‐induced cerebral vasodilatation in vivo. Am J Physiol (Heart Circ Physiol) 279: H2704–2712, 2000.
 356. Brayden JE. Membrane hyperpolarization is a mechanism of endothelium‐dependent cerebral vasodilation. Am J Physiol (Heart Circ Physiol) 259: H668–H673, 1990.
 357. Zaritsky JJ, Eckman DM, Wellman GC, Nelson MT and Schwarz TL. Targeted disruption of Kir2.1 and Kir2.2 genes reveals the essential role of the inwardly rectifying K + current in K + ‐mediated vasodilation. Circ Res 87: 160–166, 2000.
 358. Yamada M, Isomoto S, Matsumoto S, Kondo C, Shindo T, Horio Y and Kurachi Y. Sulphonylurea receptor 2B and Kir6.1 form a sulphonylurea‐sensitive but ATP‐insensitive K + channel. J Physiol 499: 715–720, 1997.
 359. Standen NB, Quayle JM, Davies NW, Brayden JE, Huang Y and Nelson MT. Hyperpolarizing vasodilators activate ATP‐sensitive K + channels in arterial smooth muscle. Science 245: 177–180, 1989.
 360. Kleppisch T and Nelson MT. ATP‐sensitive K + currents in cerebral arterial smooth muscle: Pharmacological and hormonal modulation. Am J Physiol (Heart Circ Physiol) 269: H1634–H1640, 1995.
 361. Quayle JM, Nelson MT and Standen NB. ATP‐sensitive and inwardly rectifying potassium channels in smooth muscle. Physiological Rev 77: 1165, 1997.
 362. Jackson WF, König A, Dambacher T and Busse R. Prostacyclin‐induced vasodilation in rabbit heart is mediated by ATP‐sensitive potassium channels. Am J Physiol (Heart Circ Physiol) 264: H238–H243, 1993.
 363. Daut J, Klieber HG, Cyrys S and Noack T. KATP channels and basal coronary vascular tone. Cardiovasc Res 28: 811–817, 1994.
 364. Farouque HM, Worthley SG, Meredith IT, Skyrme‐Jones RA and Zhang MJ. Effect of ATP‐sensitive potassium channel inhibition on resting coronary vascular responses in humans. Circ Res 90: 231–236, 2002.
 365. Vanelli G and Hussain SN. Effects of potassium channel blockers on basal vascular tone and reactive hyperemia of canine diaphragm. Am J Physiol (Heart Circ Physiol) 266: H43–H51, 1994.
 366. Jackson WF, Huebner JM and Rusch NJ. Enzymatic isolation and characterization of single vascular smooth muscle cells from cremasteric arterioles. Microcirculation 4: 35–50, 1997.
 367. Brayden JE. Functional roles of KATP channels in vascular smooth muscle. Clin Exp Pharmacol Physiol 29: 312–316, 2002.
 368. Bonev AD and Nelson MT. Vasoconstrictors inhibit ATP‐sensitive K + channels in arterial smooth muscle through protein kinase C. J Gen Physiol 108: 315–323, 1996.
 369. Cox DH, Cui J and Aldrich RW. Allosteric gating of a large conductance Ca‐activated K + channel. J Gen Physiol 110: 257–281, 1997.
 370. Brenner R, Perez GJ, Bonev AD, Eckman DM, Kosek JC, Wiler SW, Patterson AJ, Nelson MT and Aldrich RW. Vasoregulation by the β1 subunit of the calcium‐activated potassium channel. Nature 407: 870–876, 2000.
 371. Pluger S, Faulhaber J, Furstenau M, Lohn M, Waldschutz R, Gollasch M, Haller H, Luft FC, Ehmke H and Pongs O. Mice with disrupted BK channel betal subunit gene feature abnormal Ca 2+ spark/STOC coupling and elevated blood pressure. Circ Res 87: E53–60, 2000.
 372. Busse R, Edwards G, Feletou M, Fleming I, Vanhoutte PM and Weston AH. EDHF: bringing the concepts together. Trends Pharmacol Sci 23: 374–380, 2002.
 373. Dong H, Waldron GJ, Cole WC and Triggle CR. Roles of calcium‐activated and voltage‐gated delayed rectifier potassium channels in endothelium‐dependent vasorelaxation of the rabbit middle cerebral artery. Br J Pharmacol 123: 821–832, 1998.
 374. Harder DR, Lange AR, Gebremedhin D, Birks EK and Roman RJ. Cytochrome P450 metabolites of arachidonic acid as intracellular signaling molecules in vascular tissue. J Vasc Res 34: 237–243, 1997.
 375. Dopico AM, Kirber MT, Singer JJ and Walsh JV, Jr. Membrane stretch directly activates large conductance Ca 2+ ‐activated K + channels in mesenteric artery smooth muscle cells. Am J Hypertens 7: 82–89, 1994.
 376. Sadoshima J, Akaike N, Kanaide H and Nakamura M. Cyclic AMP modulates Ca‐activated K + channel in cultured smooth muscle cells of rat aortas. Am J Physiol (Heart Circul) 255: H754–H759, 1988.
 377. Robertson BE, Schubert R, Hescheler J and Nelson MT. cGMP‐dependent protein kinase activates Ca‐activated K + channels in cerebral artery smooth muscle cells. Am J Physiol (Cell Physiol) 265: C299–C303, 1993.
 378. Minami K, Fukuzawa K and Nakaya Y. Protein kinase C inhibits the Ca 2+ ‐activated K + channel of cultured porcine coronary artery smooth muscle cells. Biochem Biophys Res Commun 190: 263–269, 1993.
 379. Ma YH, Gebremedhin D, Schwartzman ML, Falck JR, Clark JE, Masters BS, Harder DR and Roman RJ. 20‐Hydroxyeicosatetraenoic acid is an endogenous vasoconstrictor of canine renal arcuate arteries. Circ Res 72: 126–136, 1993.
 380. Zou AP, Fleming JT, Falck JR, Jacobs ER, Gebremedhin D, Harder DR and Roman RJ. 20‐HETE is an endogenous inhibitor of the large‐conductance Ca 2+ ‐activated K + channel in renal arterioles. Am J Physiol (Regul Integr Comparat Physiol) 270: R228–R237, 1996.
 381. Randriamboavonjy V, Busse R and Fleming I. 20‐HETE‐induced contraction of small coronary arteries depends on the activation of Rho‐kinase. Hypertension 41: 801–806, 2003.
 382. Lange A, Gebremedhin D, Narayanan J and Harder D. 20‐Hydroxyeicosatetraenoic acid‐induced vasoconstriction and inhibition of potassium current in cerebral vascular smooth muscle is dependent on activation of protein kinase C. J Biol Chem 272: 27345–27352, 1997.
 383. Sun CW, Falck JR, Harder DR and Roman RJ. Role of tyrosine kinase and PKC in the vasoconstrictor response to 20‐HETE in renal arterioles. Hypertension 33 (Suppl.): 414–418, 1999.
 384. Roman RJ. P‐450 metabolites of arachidonic acid in the control of cardiovascular function. Physiological Rev 82: 131–185, 2002.
 385. Gebremedhin D, Lange AR, Lowry TF, Taheri MR, Birks EK, Hudetz AG, Narayanan J, Falck JR, Okamoto H, Roman RJ, Nithipatikom K, Campbell WB and Harder DR. Production of 20‐HETE and its role in autoregulation of cerebral blood flow. Circ Res 87: 60–65, 2000.
 386. Looft‐Wilson RC, Falck JR, Krishna UM and Gisolfi CV. 20‐HETE pathway antagonists inhibit rat small mesenteric artery tone. Microvasc Res 64: 349–352, 2002.
 387. Frisbee JC, Roman RJ, Falck JR, Krishna UM and Lombard JH. 20‐HETE contributes to myogenic activation of skeletal muscle resistance arteries in Brown Norway and Sprague‐Dawley rats. Microcirculation 8: 45–55, 2001.
 388. Zhang F, Wang MH, Wang JS, Zand B, Gopal VR, Falck JR, Laniado‐Schwartzman M and Nasjletti A. Transfection of CYP4A1 cDNA decreases diameter and increases responsiveness of gracilis muscle arterioles to constrictor stimuli. Am J Physiol (Heart Circ Physiol) 287: H1089–H1095, 2004.
 389. Huang A, Sun D, Yan C, Falck JR and Kaley G. Contribution of 20‐HETE to Augmented Myogenic Constriction in Coronary Arteries of Endothelial NO Synthase Knockout Mice. Hypertension 46: 1–7, 2005.
 390. Frisbee JC, Falck JR and Lombard JH. Contribution of cytochrome P‐450 omega‐hydroxylase to altered arteriolar reactivity with high‐salt diet and hypertension. Am J Physiol (Heart Circ Physiol) 278: H1517–H1526, 2000.
 391. Schubert R and Nelson MT. Protein kinases: tuners of the BKCa channel in smooth muscle. Trends Pharmacol Sci 22: 505–512, 2001.
 392. Chen L, Tian L, MacDonald SH, McClafferty H, Hammond MS, Huibant JM, Ruth P, Knaus HG and Shipston MJ. Functionally diverse complement of large conductance calcium‐ and voltage‐activated potassium channel (BK) alpha‐subunits generated from a single site of splicing. J Biol Chem 280: 33599–33609, 2005.
 393. Nelson MT, Patlak JB, Worley JF and Standen NB. Calcium channels, potassium channels and voltage dependence of arterial smooth muscle tone. Am J Physiol (Cell Physiol) 259: C3–C18, 1990.
 394. Bean BP, Sturek M, Puga A and Hermsmeyer K. Calcium channels in muscle cells isolated from rat mesenteric arteries: modulation by dihydropyridine drugs. Circ Res 59: 229–235, 1986.
 395. Morita H, Cousins H, Onoue H, Ito Y and Inoue R. Predominant distribution of nifedipine‐insensitive, high voltage‐activated Ca 2+ channels in the terminal mesenteric artery of guinea pig. Circ Res 85: 596–605, 1999.
 396. Loutzenhiser R, Hayashi K and Epstein M. Divergent effects of KCl‐induced depolarization on afferent and efferent arterioles. Am J Physiol (Renal Physiol) 257: F561–F564, 1989.
 397. Gustafsson F, Andreasen D, Salomonsson M, Jensen BL and Holstein‐Rathlou N. Conducted vasoconstriction in rat mesenteric arterioles: role for dihydropyridine‐insensitive Ca 2+ channels. Am J Physiol (Heart Circ Physiol) 280: H582–H590, 2001.
 398. Aaronson PI, Bolton TB, Lang RJ and MacKenzie I. Calcium currents in single isolated smooth muscle cells from the rabbit ear artery in normal‐calcium and high‐barium solutions. J Physiol 405: 57–75, 1988.
 399. Benham CD and Tsien RW. Noradrenaline modulation of calcium channels in single smooth muscle cells from rabbit ear artery. J Physiol 404: 767–784, 1988.
 400. Hofmann F, Lacinova L and Klugbauer N. Voltage‐dependent calcium channels: from structure to function. Rev Physiol Biochem Pharmacol 139: 33–87, 1999.
 401. Moosmang S, Lenhardt P, Haider N, Hofmann F and Wegener JW. Mouse models to study L‐type calcium channel function. Pharmacol Therapeut 106: 347–355, 2005.
 402. Hill MA and Meininger GA. Calcium entry and myogenic phenomena in skeletal muscle arterioles. Am J Physiol 267: H1085–H1092, 1994.
 403. Wesselman JPM, VanBavel E, Pfaffendorf M and Spaan JAE. Voltage‐operated calcium channels are essential for the myogenic responsiveness of cannulated rat mesenteric small arteries. J Vasc Res 33: 32–41, 1996.
 404. Moosmang S, Schulla V, Welling A, Feil R, Feil S, Wegener JW, Hofmann F and Klugbauer N. Dominant role of smooth muscle L‐type calcium channel Cav1.2 for blood pressure regulation. EMBO J 22: 6027–6034, 2003.
 405. Sinnegger‐Brauns MJ, Hetzenauer A, Huber IG, Renstrom E, Wietzorrek G, Berjukov S, Cavalli M, Walter D, Koschak A, Waldschutz R, Hering S, Bova S, Rorsman P, Pongs O, Singewald N and Striessnig JJ. Isoform‐specific regulation of mood behavior and pancreatic beta cell and cardiovascular function by L‐type Ca 2+ channels. J Clin Invest 113: 1430–1439, 2004.
 406. Matsuda N, Hagiwara N, Shoda M, Kasanuki H and Hosoda S. Enhancement of the L‐type Ca 2+ current by mechanical stimulation in single rabbit cardiac myocytes. Circ Res 78: 650–659, 1996.
 407. Xu WX, Kim SJ, Kim SJ, So I, Kang TM, Rhee JC and Kim KW. Effect of stretch on calcium channel currents recorded from the antral circular myocytes of guinea‐pig stomach. Pflügers Arch Eur J Physiol 432: 159–164, 1996.
 408. Lyford GL, Strege PR, Shepard A, Ou Y, Ermilov L, Miller SM, Gibbons SJ, Rae JL, Szurszewski JH and Farrugia G. α1C (Cav 1.2) L‐type calcium channel mediates mechanosensitive calcium regulation. Am J Physiol (Cell Physiol) 283: C1001–C1008, 2002.
 409. Ben‐Tabou S, Keller E and Nussinovitch I. Mechanosensitivity of voltage‐gated calcium currents in rat anterior pituitary cells. J Physiol 476: 29–39, 1994.
 410. Keef KD, Hume JR and Zhong J. Regulation of cardiac and smooth muscle Ca 2+ channels (Cav 1.2a,b) by protein kinases. Am J Physiol (Cell Physiol) 281: C1743–C1756, 2001.
 411. Kamp TJ and Hell JW. Regulation of cardiac L‐type calcium channels by protein kinase A and protein kinase C. Circ Res 87: 1095–1102, 2000.
 412. Gebremedhin D, Lange AR, Narayanan J, Aebly MR, Jacobs ER and Harder DR. Cat cerebral arterial smooth muscle cells express cytochrome P450 4A2 enzyme and produce the vasoconstrictor 20‐HETE which enhances L‐type Ca 2+ current. J Physiol 507: 771–781, 1998.
 413. Mishra SK and Hermsmeyer K. Inhibition of signal Ca 2+ in dog coronary arterial vascular muscle cells by Ro 40‐5967. J Cardiovasc Pharmacol 24: 1–7, 1994.
 414. Mishra SK and Hermsmeyer K. Resting state block and use independence of rat vascular muscle Ca ++ channels by Ro 40‐5967. J Pharmacol Exp Therapeut 269: 178–183, 1994.
 415. VanBavel E, Sorop O, Andreasen D, Pfaffendorf M and Jensen BL. Role of T‐type calcium channels in myogenic tone of skeletal muscle resistance arteries. Am J Physiol (Heart Circ Physiol) 283: H2239–H2243, 2002.
 416. Hermsmeyer K and Erne P. Cellular calcium regulation in hypertension. Am J Hypertens 2: 655–658, 1989.
 417. Hansen PB, Jensen BL, Andreasen D and Skott O. Differential expression of T‐ and L‐type voltage‐dependent calcium channels in renal resistance vessels. Circ Res 89: 630–638, 2001.
 418. Chen XL, Bayliss DA, Fern RJ and Barrett PQ. A role for T‐type Ca 2+ channels in the synergistic control of aldosterone production by ANG II and K +. Am J Physiol (Heart Circ Physiol) 276: F674–F683, 1999.
 419. Potocnik SJ, Murphy TV, Kotecha N and Hill MA. Effects of mibefradil and nifedipine on arteriolar myogenic responsiveness and intracellular Ca 2+. Br J Pharmacol 131: 1065–1072, 2000.
 420. Lam E, Skarsgard P and Laher I. Inhibition of myogenic tone by mibefradil in rat cerebral arteries. Eur J Pharmacol 358: 165–168, 1998.
 421. Moosmang S, Haider N, Bruderl B, Welling A and Hofmann F. Antihypertensive effects of the putative T‐type calcium channel antagonist mibefradil are mediated by the L‐type calcium channel Cav 1.2. Circ Res 98: 105–110, 2006.
 422. Chen CC, Lamping KG, Nuno DW, Barresi R, Prouty SJ, Lavoie JL, Cribbs LL, England SK, Sigmund CD, Weiss RM, Williamson RA, Hill JA and Campbell KP. Abnormal coronary function in mice deficient in α1H T‐type Ca 2+ channels. Science 302: 1416–1418, 2003.
 423. Ishiguro M, Wellman TL, Honda A, Russell SR, Tranmer BI and Wellman GC. Emergence of a R‐type Ca 2+ channel (Cav 2.3) contributes to cerebral artery constriction after subarachnoid hemorrhage. Circ Res 96: 419–426, 2005.
 424. Powell RJ, Carruth JA, Basson MD, Bloodgood R and Sumpio BE. Matrix‐specific effect of endothelial control of smooth muscle cell migration. J Vasc Surg 24: 51–57, 1996.
 425. Pesic A, Madden JA, Pesic M and Rusch NJ High blood pressure upregulates arterial L‐type Ca 2+ channels: is membrane depolarization the signal? Circ Res 94: 97–104, 2004.
 426. Adler KB, Krill J, Alberghini TV and Evans JN. Effect of cytochalasin D on smooth muscle contraction. Cell Motil 3: 545–551, 1983.
 427. Ingber DE. Mechanical signaling and the cellular response to extracellular matrix in angiogenesis and cardiovascular physiology. Circ Res 91: 877–887, 2002.
 428. Banes AJ, Tsuzaki M, Yamamoto J, Fischer T, Brigman B, Brown T and Miller L. Mechanoreception at the cellular level: the detection, interpretation, and diversity of responses to mechanical signals. Biochem Cell Biol 73: 349–365, 1995.
 429. Small JV and Gimona M. The cytoskeleton of the vertebrate smooth muscle cell. Acta Physiol Scand 164: 341–348, 1998.
 430. Mehta D and Gunst SJ. Actin polymerization stimulated by contractile activation regulates force development in canine tracheal smooth muscle. J Physiol 519: 829–840, 1999.
 431. Battistella‐Patterson AS, Wang S and Wright GL. Effect of disruption of the cytoskeleton on smooth muscle contraction. Can J Physiol Pharmacol 75: 1287–1299, 1997.
 432. Holda JR and Blatter LA. Capacitative calcium entry is inhibited in vascular endothelial cells by disruption of cytoskeletal microfilaments. FEBS Lett 403: 191–196, 1997.
 433. Bishara NB, Murphy TV and Hill MA. Capacitative Ca 2+ entry in vascular endothelial cells is mediated via pathways sensitive to 2 aminoethoxydiphenyl borate and xestospongin C. Br J Pharmacol 135: 119–128, 2002.
 434. Gokina NI and Osol G. Actin cytoskeletal modulation of pressure‐induced depolarization and Ca 2+ influx in cerebral arteries. Am J Physiol (Heart Circ Physiol) 282: H1410–H1420, 2002.
 435. Platts SH, Falcone JC, Holton WT, Hill MA and Meininger GA. Alteration of microtubule polymerization modulates arteriolar vasomotor tone. Am J Physiol (Heart Circ Physiol) 277: H100–H106, 1999.
 436. Paul RJ, Bowman PS and Kolodney MS. Effects of microtubule disruption on force, velocity, stiffness and [Ca 2+]i in porcine coronary arteries. Am J Physiol (Heart Circ Physiol) 279: H2493–H2501, 2000.
 437. Chitaley K and Webb RC. Microtubule depolymerization facilitates contraction of rat aorta via activation of Rho‐kinase. Vasc Pharmacol 38: 157–161, 2002.
 438. Henrion D, Terzi F, Matrougui K, Duriez M, Boulanger CM, Colucci‐Guyon E, Babinet C, Briand P, Friedlander G, Poitevin P and Levy BI. Impaired flow‐induced dilation in mesenteric resistance arteries from mice lacking vimentin. J Clin invest 100: 2909–2914, 1997.
 439. Loufrani L, Matrougui K, Li Z, Levy BI, Lacolley P, Paulin D and Henrion D. Selective microvascular dysfunction in mice lacking the gene encoding for desmin. FASEB J 16: 117–119, 2002.
 440. Laher I, Van Breemen C and Bevan JA. Stretch‐dependent calcium uptake associated with myogenic tone in rabbit facial vein. Circ Res 63: 669–672, 1988.
 441. Tanaka Y, Imai T, Igarashi T, Takayanagi K, Otsuka K, Yamaki F, Tanaka H and Shigenobu K. Comparison of the Ca 2+ entry channels responsible for mechanical responses of guinea‐pig aorta to noradrenaline and thapsigargin using SK&F 96365 and LOE 908. Naunyn‐Schmiedebergs Arch Pharmacol 362: 160–168, 2000.
 442. Potocnik SJ and Hill MA. Pharmacological evidence for capacitative Ca 2+ entry in cannulated and pressurized skeletal muscle arterioles. Br J Pharmacol 134: 247–256, 2001.
 443. Meininger GA, Zawieja DC, Falcone JC, Hill MA and Davey JP. Calcium measurement in isolated arterioles during myogenic and agonist stimulation. Am J Physiol (Heart Circ Physiol) 261: H950–H959, 1991.
 444. Hill MA, Zou H, Davis MJ, Potocnik SJ and Price S. Transient increases in diameter and [Ca 2+]i are not obligatory for myogenic constriction. Am J Physiol (Heart Circ Physiol) 278: H345–H352, 2000.
 445. Kirber MT, Guerrero‐Hernández A, Bowman DS, Fogarty KE, Tuft RA, Singer JJ and Fay FS. Multiple pathways responsible for the stretch‐induced increase in Ca 2+ concentration in toad stomach smooth muscle cells. J Physiol 524: 3–17, 2000.
 446. Osol G, Laher I and Kelley M. Myogenic tone is coupled to phospholipase C and G protein activation in small cerebral arteries. Am J Physiol (Heart Circ Physiol) 265: H415–H420, 1993.
 447. Zang WJ, Balke CW and Wier WG. Graded α1 ‐adrenoceptor activation of arteries involves recruitment of smooth muscle cells to produce ‘all or none’ Ca 2+ signals. Cell Calcium 29: 327–334, 2001.
 448. Shaw L, Ahmed S, Austin C and Taggart MJ. Inhibitors of actin filament polymerisation attenuate force but not global intracellular calcium in isolated pressurised resistance arteries. J Vasc Res 40: 1–10, 2003.
 449. Cheng H, Lederer WJ and Cannell MB. Calcium sparks: Elementary events underlying excitation‐contraction coupling in heart muscle. Science 262: 740–744, 1993.
 450. ZhuGe R, Fogarty KE, Tuft RA and Walsh JV, Jr. Spontaneous transient outward currents arise from microdomains where BK channels are exposed to a mean Ca 2+ concentration on the order of 10 μM during a Ca 2+ spark. J Gen Physiol 120: 15–27, 2002.
 451. Heppner TJ, Bonev AD, Santana LF and Nelson MT. Alkaline pH shifts Ca 2+ sparks to Ca 2+ waves in smooth muscle cells of pressurized cerebral arteries. Am J Physiol (Heart Circ Physiol) 283: H2169–2176, 2002.
 452. Bolton TB. Calcium events in smooth muscles and their interstitial cells; physiological roles of sparks. J Physiol 570: 5–11, 2006.
 453. Nelson MT, Cheng H, Rubart M, Santana LF, Bonev AD, Knot HJ and Lederer WJ. Relaxation of arterial smooth muscle by calcium sparks. Science 270: 633–637, 1995.
 454. Arnaudeau S, Macrez‐Lepretre N and Mironneau J. Activation of calcium sparks by angiotensin II in vascular myocytes. Biochem Biophys Res 222: 809–815, 1996.
 455. Mironneau J, Arnaudeau S, Macrez‐Lepretre N and Boittin FX. Ca 2+ sparks and Ca 2+ waves activate different Ca 2+ ‐dependent ion channels in single myocytes from rat portal vein. Cell Calc 20: 153–160, 1996.
 456. Miriel VA, Mauban JR, Blaustein MP and Wier WG. Local and cellular Ca 2+ transients in smooth muscle of pressurized rat resistance arteries during myogenic and agonist stimulation. J Physiol 518: 815–824, 1999.
 457. Bolton TB and Gordienko DV. Confocal imaging of calcium release events in single smooth muscle cells. Acta Physiol Scand 164: 567–575, 1998.
 458. Kotlikoff MI and Wang YX. Calcium release and calcium‐activated chloride channels in airway smooth muscle cells. Am J Respir Crit Care Med 158 (Suppl. S2): S109–S114, 1998.
 459. Jaggar JH, Stevenson AS and Nelson MT. Voltage dependence of Ca 2+ sparks in intact cerebral arteries. Am J Physiol (Cell Physiol) 274: C1755–C1761, 1998.
 460. Jaggar JH, Wellman GC, Heppner TJ, Porter VA, Perez GJ, Gollasch M, Kleppisch T, Rubart M, Stevenson AS, Lederer WJ, Knot HJ, Bonev AD and Nelson MT. Ca 2+ channels, ryanodine receptors and Ca 2+ ‐activated K + channels: a functional unit for regulating arterial tone. Acta Physiol Scand 164: 577–587, 1998.
 461. Benham CD and Bolton TB. Spontaneous transient outward currents in single visceral and vascular smooth muscle cells of the rabbit. J Physiol 381: 385–406, 1986.
 462. Ganitkevich V and Isenberg G. Isolated guinea pig coronary smooth muscle cells. Aceylcholine induces hyperpolarization due to sarcoplasmic reticulum calcium release activating potassium channels. Circ Res 67: 525–528, 1990.
 463. Porter VA, Bonev AD, Knot HJ, Heppner TJ, Stevenson AS, Kleppisch T, Lederer WJ and Nelson MT. Frequency modulation of Ca 2+ sparks is involved in regulation of arterial diameter by cyclic nucleotides. Am J Physiol (Cell Physiol) 274: C1346–C1355, 1998.
 464. ZhuGe R, Fogarty KE, Baker SP, McCarron JG, Tuft RA, Lifshitz LM and Walsh JV, Jr. Ca 2+ spark sites in smooth muscle cells are numerous and differ in number of ryanodine receptors, large‐conductance K + channels and coupling ratio between them. Am J Physiol (Cell Physiol) 287: C1577–C1588, 2004.
 465. Jaggar JH. Intravascular pressure regulates local and global Ca 2+ signaling in cerebral artery smooth muscle cells. Am J Physiol (Cell Physiol) 281: C439–448, 2001.
 466. Wellman GC, Nathan DJ, Saundry CM, Perez G, Bonev AD, Penar PL, Tranmer BI and Nelson MT. Ca 2+ sparks and their function in human cerebral arteries. Stroke 33: 802–808, 2002.
 467. Van Helden DF. Spontaneous and noradrenaline‐induced transient depolarizations in the smooth muscle of guinea‐pig mesenteric vein. J Physiol 437: 511–541, 1991.
 468. Hogg RC, Wang Q and Large WA. Time course of spontaneous calcium‐activated chloride currents in smooth muscle cells from the rabbit portal vein. J Physiol 464: 15–31, 1993.
 469. Helliwell RM and Large WA. Effect of temperature on spontaneous Ca 2+ ‐activated Cl − currents in rabbit portal vein cells. Pflügers Arch Eur J Physiol 431: 28–31, 1995.
 470. Peng H, Matchkov V, Ivarsen A, Aalkjaer C and Nilsson H. Hypothesis for the initiation of vasomotion. Circ Res 88: 810–815, 2001.
 471. Dolmetsch RE, Xu K and Lewis RS. Calcium oscillations increase the efficiency and specificity of gene expression. Nature 392: 933–936, 1998.
 472. Shaw L, O'Neill S, Jones CJ, Austin C and Taggart MJ. Comparison of U46619‐, endothelin‐1‐ or phenylephrine‐induced changes in cellular Ca 2+ profiles and Ca 2+ sensitisation of constriction of pressurised rat resistance arteries. Br J Pharmacol 141: 678–688, 2004.
 473. Zacharia J, Zhang J and Wier WG. Ca 2+ signaling in mouse mesenteric small arteries: myogenic tone and vasoconstriction. Am J Physiol (Heart Circ Physiol) 292: H1523–H1532, 2007.
 474. Hai CM and Phair RD. Kinetic identification of an intracellular calcium compartment sensitive to phosphate and dinitrophenol in intact isolated rabbit aorta. Circ Res 59: 85–92, 1986.
 475. Phair RD and Hai CM. Resolution of intracellular calcium metabolism in intact segments of rabbit aorta. Circ Res 59: 74–84, 1986.
 476. Hai CM and Phair RD. Forskolin and caffeine induce Ca 2+ release from intracellular stores in rabbit aorta. Am J Physiol (Cell Physiol) 257: C413–C418, 1989.
 477. Stehno‐Bittel L, Perez‐Terzic C, Luckhoff A and Clapham DE. Nuclear ion channels and regulation of the nuclear pore. Soc Gen Physiol Ser 51: 195–207, 1996.
 478. Yamamoto H and van Breemen C. Ca 2+ compartments in saponin‐skinned cultured vascular smooth muscle cells. J Gen Physiol 87: 369–389, 1986.
 479. Somlyo AP and Somlyo AV. Electron probe analysis of calcium content and movements in sarcoplasmic reticulum, endoplasmic reticulum, mitochondria and cytoplasm. J Cardiovasc Pharmacol 8 (Suppl. 8): S42–S47, 1986.
 480. Somlyo AP and Himpens B. Cell calcium and its regulation in smooth muscle. FASEB J 3: 2266–2276, 1989.
 481. Pozzan T, Magalhaes P and Rizzuto R. The comeback of mitochondria to calcium signalling. Cell Calc 28: 279–283, 2000.
 482. Hajnoczky G, Csordas G, Madesh M and Pacher P. The machinery of local Ca 2+ signalling between sarco‐endoplasmic reticulum and mitochondria. J Physiol 529: 69–81, 2000.
 483. Drummond RM and Fay FS. Mitochondria contribute to Ca 2+ removal in smooth muscle cells. Pflügers Arch Eur J Physiol 431: 473–482, 1996.
 484. Drummond RM and Tuft RA. Release of Ca 2+ from the sarcoplasmic reticulum increases mitochondrial Ca 2+ in rat pulmonary artery smooth muscle cells. J Physiol 516 (1): 139–147, 1999.
 485. Szado T, Kuo KH, Bernard‐Helary K, Poburko D, Lee CH, Seow C, Ruegg UT and van Breemen C. Agonist‐induced mitochondrial Ca 2+ transients in smooth muscle. FASEB J 17: 28–37, 2003.
 486. Monteith GR and Blaustein MP. Heterogeneity of mitochondrial matrix free Ca 2+ : resolution of Ca 2+ dynamics in individual mitochondria in situ. Am J Physiol (Cell Physiol) 276: C1193–C1204, 1999.
 487. Kamishima T and Quayle JM. Mitochondrial Ca 2+ uptake is important over low [Ca 2+]i range in arterial smooth muscle. Am J Physiol (Heart Circ Physiol) 283: H2431–H2439, 2002.
 488. Cheranov SY and Jaggar JH. Mitochondrial modulation of Ca 2+ sparks and transient KCa currents in smooth muscle cells of rat cerebral arteries. J Physiol 556: 755–771, 2004.
 489. Greenwood IA, Helliwell RM and Large WA. Modulation of Ca 2+ ‐activated Cl − currents in rabbit portal vein smooth muscle by an inhibitor of mitochondrial Ca 2+ uptake. J Physiol 505: 53–64, 1997.
 490. Poburko D, Lee CH and van Breemen C. Vascular smooth muscle mitochondria at the cross roads of Ca 2+ regulation. Cell Calc 35: 509–521, 2004.
 491. Somlyo AP and Somlyo AV. Ca 2+ sensitivity of smooth muscle and nonmuscle myosin II: modulated by G proteins, kinases, and myosin phosphatase. Physiological Rev 83: 1325–1358, 2003.
 492. Woodsome TP, Eto M, Everett A, Brautigan DL and Kitazawa T. Expression of CPI‐17 and myosin phosphatase correlates with Ca 2+ sensitivity of protein kinase C‐induced contraction in rabbit smooth muscle. J Physiol 535: 553–564, 2001.
 493. Hill MA, Falcone JA and Meininger GA. Evidence for protein kinase C involvement in arteriolar myogenic activity. Am J Physiol (Heart Circ Physiol) 259: H1586–H1594, 1990.
 494. Osol G, Laher I and Cipolla M. Protein kinase C modulates basal myogenic tone in resistance arteries from the cerebral circulation. Circ Res 68: 359–367, 1991.
 495. VanBavel E, Wesselman JP and Spaan JA. Myogenic activation and calcium sensitivity of cannulated rat mesenteric small arteries. Circ Res 82: 210–220, 1998.
 496. Gokina NI, Park KM, McElroy‐Yaggy K and Osol G. Effects of Rho kinase inhibition on cerebral artery myogenic tone and reactivity. J Appl Physiol 98: 1940–1948, 2005.
 497. Uehata M, Ishizaki T, Satoh H, Ono T, Kawahara T, Morishita T, Tamakawa H, Yamagami K, Inui J, Maekawa M and Narumiya S. Calcium sensitization of smooth muscle mediated by a Rho‐associated protein kinase in hypertension. Nature 389: 990–994, 1997.
 498. Smith RH, Palmer RM and Reeds PJ. Protein synthesis in isolated rabbit forelimb muscles. The possible role of metabolites of arachidonic acid in the response to intermittent stretching. Biochem J 214: 153–161, 1983.
 499. Revtyak GE, Johnson AR and Campbell WB. Cultured bovine coronary arterial endothelial cells synthesize HETEs and prostacyclin. Am J Physiol (Cell Physiol) 254: C8–19, 1988.
 500. He Y and Grinnell F. Stress relaxation of fibroblasts activates a cyclic AMP signaling pathway. J Cell Biol 126: 457–464, 1994.
 501. Hardie RC. Regulation of TRP channels via lipid second messengers. Annu Rev Physiol 65: 735–759, 2003.
 502. Harder DR, Gebremedhin D, Narayanan J, Jefcoat C, Falck JR, Campbell WB and Roman R. Formation and action of a P‐450 4A metabolite of arachidonic acid in cat cerebral microvessels. Am J Physiol (Heart Circ Physiol) 266: H2098–H2107, 1994.
 503. Miyata N and Roman RJ. Role of 20‐hydroxyeicosatetraenoic acid (20‐HETE) in vascular system. J Smooth Muscle Res 41: 175–193, 2005.
 504. Kaide J, Wang MH, Wang JS, Zhang F, Gopal VR, Falck JR, Nasjletti A and Laniado‐Schwartzman M. Transfection of CYP4A1 cDNA increases vascular reactivity in renal interlobar arteries. Am J Physiol (Renal Physiol) 284: F51–56, 2003.
 505. Obara K, Koide M and Nakayama K. 20‐Hydroxyeicosatetraenoic acid potentiates stretch‐induced contraction of canine basilar artery via PKC alpha‐mediated inhibition of KCa channel. Br J Pharmacol 137: 1362–1370, 2002.
 506. Bolz SS, Fisslthaler B, Pieperhoff S, De Wit C, Fleming I, Busse R and Pohl U. Antisense oligonucleotides against cytochrome P450 2C8 attenuate EDHF‐mediated Ca 2+ changes and dilation in isolated resistance arteries. FASEB J 14: 255–260, 2000.
 507. Fleming I. Cytochrome p450 enzymes in vascular homeostasis. Circ Res 89: 753–762, 2001.
 508. Kuo L, Davis MJ and Chilian WM. Endothelium‐dependent, flow‐induced dilation of isolated coronary arterioles. Am J Physiol (Heart Circ Physiol) 259: H1063–H1070, 1990.
 509. Barany K, Ledvora RF, Mougios V and Barany M. Stretch‐induced myosin light chain phosphorylation and stretch‐release‐induced tension development in arterial smooth muscle. J Biol Chem 260: 7126–7130, 1985.
 510. Ledvora RF, Barany K, Vander Muelen DL, Barron JT and Barany M. Stretch‐induced phosphorylation of the 20,000‐dalton light chain of myosin in arterial smooth muscle. J Biol Chem 258: 14080–14083, 1983.
 511. Ledvora RF, Barany M and Barany K. Myosin light chain phosphorylation and tension development in stretch‐activated arterial smooth muscle. Clinical Chem 30: 2063–2068, 1984.
 512. Barany K, Rokolya A and Barany M. Stretch activates myosin light chain kinase in arterial smooth muscle. Biochem Biophys Res Commun 173: 164–171, 1990.
 513. Rembold CM and Murphy RA. Muscle length, shortening, myoplasmic [Ca 2+], and activation of arterial smooth muscle. Circ Res 66: 1354–1361, 1990.
 514. Hai CM. Length‐dependent myosin phosphorylation and contraction of arterial smooth muscle. Pflügers Arch Eur J Physiol 418: 564–571, 1991.
 515. Szeto B and Hai CM. Length‐dependent modulation of myosin phosphorylation and contractile force in coronary arterial smooth muscle. Arch Biochem Biophys 329: 241–248, 1996.
 516. Gunst SJ and Tang DD. The contractile apparatus and mechanical properties of airway smooth muscle. Eur Respir J 15: 600–616, 2000.
 517. Laporte R, Haeberle JR and Laher I. Phorbol ester‐induced potentiation of myogenic tone is not associated with increases in Ca 2+ influx, myoplasmic free Ca 2+ concentration, or 20‐kDa myosin light chain phosphorylation. J Molecul Cell Cardiol 26: 297–302, 1994.
 518. Dessy C, Matsuda N, Hulvershorn J, Sougnez CL, Sellke FW and Morgan KG. Evidence for involvement of the PKC‐α isoform in myogenic contractions of the coronary microcirculation. Am J Physiol (Heart Circ Physiol) 279: H916–H923, 2000.
 519. Deng JT, Van Lierop JE, Sutherland C and Walsh MP. Ca 2+ ‐independent smooth muscle contraction, a novel function for integrin‐linked kinase. J Biol Chem 276: 16365–16373, 2001.
 520. Weber LP, Van Lierop JE and Walsh MP. Ca 2+ independent phosphorylation of myosin in rat caudal artery and chicken gizzard myofilaments. J Physiol 516: 805–824, 1999.
 521. Somlyo AV, Wang H, Choudhury N, Khromov AS, Majesky M, Owens GK and Somlyo AP. Myosin light chain kinase knockout. J Muscle Res Cell Motil 25: 241–242, 2004.
 522. Meininger GA and Faber JE. Adrenergic facilitation of myogenic response in skeletal muscle arterioles. Am J Physiol (Heart Circ Physiol) 260: H1424–H1432, 1991.
 523. Kitazawa TT, Kobayashi S, Horiuti K, Somlyo AV and Somlyo AP. Receptor‐coupled, permeabilized smooth muscle. Role of the phosphatidylinositol cascade, G‐proteins and modulation of the contractile response to Ca 2+. J Biol Chem 264: 5339–5342, 1989.
 524. Nishimura J and Van Breemen C. Direct regulation of smooth muscle contractile elements by second messengers. Biochem Biophys Res Commun 163: 929–935, 1989.
 525. Salamanca DA and Khalil RA. Protein kinase C isoforms as specific targets for modulation of vascular smooth muscle function in hypertension. Biochem Pharmacol 70: 1537–1547, 2005.
 526. Massett MP, Ungvari Z, Csiszar A, Kaley G and Koller A. Different roles of PKC and MAP kinases in arteriolar constrictions to pressure and agonists. Am J Physiol (Heart Circ Physiol) 283: H2282–2287, 2002.
 527. Korzick DH, Laughlin MH and Bowles DK. Alterations in PKC signaling underlie enhanced myogenic tone in exercise‐trained porcine coronary resistance arteries. J Appl Physiol 96: 1425–1432, 2004.
 528. Slish DF, Welsh DG and Brayden JE. Diacylglycerol and protein kinase C activate cation channels involved in myogenic tone. Am J Physiol (Heart Circ Physiol) 283: H2196–H2201, 2002.
 529. Inagaki M, Yokokura H, Itoh T, Kanmura Y, Kuriyama H and Hidaka H. Purified rabbit brain protein kinase C relaxes skinned vascular smooth muscle and phosphorylates myosin light chain. Arch Biochem Biophys 254: 136–141, 1987.
 530. Morgan KG and Gangopadhyay SS. Invited review: cross‐bridge regulation by thin filament‐associated proteins. J Appl Physiol 91: 953–962, 2001.
 531. Bonev AD, Jaggar JH, Rubart M and Nelson MT. Activators of protein kinase C decrease Ca 2+ spark frequency in smooth muscle cells from cerebral arteries. Am J Physiol 273: C2090–C2095, 1997.
 532. VanBavel E, van der Meulen ET and Spaan JA. Role of Rho‐associated protein kinase in tone and calcium sensitivity of cannulated rat mesenteric small arteries. Exp Physiol 86: 585–592, 2001.
 533. Nakamura A, Hayashi K, Ozawa Y, Fujiwara K, Okubo K, Kanda T, Wakino S and Saruta T. Vessel‐ and vasoconstrictor‐dependent role of Rho/Rho‐kinase in renal microvascular tone. J Vasc Res 40: 244–251, 2003.
 534. Yeon DS, Kim JS, Ahn DS, Kwon SC, Kang BS, Morgan KG and Lee YH. Role of protein kinase C‐ or RhoA‐induced Ca 2+ sensitization in stretch‐induced myogenic tone. Cardiovasc Res 53: 431–438, 2002.
 535. Dubroca C, You D, Levy BI, Loufrani L and Henrion D. Involvement of RhoA/Rho kinase pathway in myogenic tone in the rabbit facial vein. Hypertension 45: 974–979, 2005.
 536. Bolz SS, Vogel L, Sollinger D, Derwand R, Boer C, Pitson SM, Spiegel S and Pohl U. Sphingosine kinase modulates microvascular tone and myogenic responses through activation of RhoA/Rho kinase. Circulation 108: 342–347, 2003.
 537. Shabir S, Borisova L, Wray S and Burdyga T. Rho‐kinase inhibition and electromechanical coupling in rat and guinea‐pig ureter smooth muscle: Ca 2+ ‐dependent and ‐independent mechanisms. J Physiol 560: 839–855, 2004.
 538. Wang P and Bitar KN. Rho A regulates sustained smooth muscle contraction through cytoskeletal reorganization of HSP27. Am J Physiol (Gastrointestin Liver Physiol) 275: G1454–G1462, 1998.
 539. Yamboliev IA. Hedges JC, Mutnick JL, Adam LP and Gerthoffer WT. Evidence for modulation of smooth muscle force by the p38 MAP kinase/HSP27 pathway. Am J Physiol (Heart Circ Physiol) 278: H1899–H1907, 2000.
 540. Jarajapu YP and Knot HJ. Relative contribution of Rho kinase and protein kinase C to myogenic tone in rat cerebral arteries in hypertension. Am J Physiol (Heart Circ Physiol) 289: H1917–H1922, 2005.
 541. Saba JD and Hla T. Point‐counterpoint of sphingosine 1‐phosphate metabolism. Circ Res 94: 724–734, 2004.
 542. Wang F, Van Brocklyn JR, Hobson JP, Movafagh S, Zukowska‐Grojec Z, Milstien S and Spiegel S. Sphingosine 1‐phosphate stimulates cell migration through a G(i)‐coupled cell surface receptor. Potential involvement in angiogenesis. J Biol Chem 274: 35343–35350, 1999.
 543. Liu Y, Wada R, Yamashita T, Mi Y, Deng CX, Hobson JP, Rosenfeldt HM, Nava VE, Chae SS, Lee MJ, Liu CH, Hla T, Spiegel S and Proia RL. Edg‐1, the G protein‐coupled receptor for sphingosine‐1‐phosphate, is essential for vascular maturation. J Clin Invest 106: 951–961, 2000.
 544. Zhou H and Murthy KS. Identification of the G protein‐activating sequence of the single‐transmembrane natriuretic peptide receptor C (NPR‐C). Am J Physiol (Cell Physiol) 284: C1255–1261, 2003.
 545. Ohmori T, Yatomi Y, Osada M, Kazama F, Takafuta T, Ikeda H and Ozaki Y. Sphingosine 1‐phosphate induces contraction of coronary artery smooth muscle cells via S1P2. Cardiovasc Res 58: 170–177, 2003.
 546. Alemany R, Kleuser B, Ruwisch L, Danneberg K, Lass H, Hashemi R, Spiegel S, Jakobs KH and Meyer zu Heringdorf D. Depolarisation induces rapid and transient formation of intracellular sphingosine‐1‐phosphate. FEBS Lett 509: 239–244, 2001.
 547. Bolz SS and Pohl U. Highly effective non‐viral gene transfer into vascular smooth muscle cells of cultured resistance arteries demonstrated by genetic inhibition of sphingosine‐1‐phosphate‐induced vasoconstriction. J Vasc Res 40: 399–405, 2003.
 548. Semenchuk LA and DiSalvo J. Receptor‐activated increases in intracellular calcium and protein tyrosine phosphorylation in vascular smooth muscle cells. FEBS Lett 370: 127–130, 1995.
 549. Vuori K. Integrin signaling: Tyrosine phosphorylation events in focal adhesions. J Membr Biol 165: 191–199, 1998.
 550. Parsons JT. Focal adhesion kinase: the first ten years. J Cell Sci 116: 1409–1416, 2003.
 551. Franklin MT, Wang CL and Adam LP. Stretch‐dependent activation and desensitization of mitogen‐activated protein kinase in carotid arteries. Am J Physiol (Cell Physiol) 273: C1819–C1827, 1997.
 552. Tang D, Mehta D and Gunst SJ. Mechanosensitive tyrosine phosphorylation of paxillin and focal adhesion kinase in tracheal smooth muscle. Am J Physiol (Cell Physiol) 276: C250–C258, 1999.
 553. Tang DD and Gunst SJ. Selected contribution: roles of focal adhesion kinase and paxillin in the mechanosensitive regulation of myosin phosphorylation in smooth muscle. J Appl Physiol 91: 1452–1459, 2001.
 554. Spurrell BE, Murphy TV and Hill MA. Tyrosine phosphorylation modulates arteriolar tone but is not fundamental to myogenic response. Am J Physiol (Heart Circ Physiol) 278: H373–382, 2000.
 555. Spurrell BE, Murphy TV and Hill MA. Intraluminal pressure stimulates MAPK phosphorylation in arterioles: temporal dissociation from myogenic contractile response. Am J Physiol (Heart Circ Physiol) 285: H1764–1773, 2003.
 556. Murphy TV, Spurrell BE and Hill MA. Tyrosine phosphorylation following alterations in arteriolar intraluminal pressure and wall tension. Am J Physiol (Heart Circ Physiol) 281: H1047–1056, 2001.
 557. Nowicki PT, Flavahan S, Hassanain H, Mitra S, Holland S, Goldschmidt‐Clermont PJ and Flavahan NA. Redox signaling of the arteriolar myogenic response. Circ Res 89: 114–116, 2001.
 558. Paravicini TM, Chrissobolis S, Drummond GR and Sobey CG. Increased NADPH‐oxidase activity and Nox4 expression during chronic hypertension is associated with enhanced cerebral vasodilatation to NADPH in vivo. Stroke 35: 584–589, 2004.
 559. Paravicini TM, Drummond GR and Sobey CG. Reactive oxygen species in the cerebral circulation: physiological roles and therapeutic implications for hypertension and stroke. Drugs 64: 2143–2157, 2004.
 560. Volk T, Hensel M, and Kox WJ. Transient Ca 2+ changes in endothelial cells induced by low doses of reactive oxygen species: role of hydrogen peroxide. Molecul Cell Biochem 171: 11–21, 1997.
 561. Matoba T, Shimokawa H, Nakashima M, Hirakawa Y, Mukai Y, Hirano K, Kanaide H and Takeshita A. Hydrogen peroxide is an endothelium‐derived hyperpolarizing factor in mice. J Clin Invest 106: 1521–1530, 2000.
 562. Ellis A, Pannirselvam M Anderson TJ and Triggle CR. Catalase has negligible inhibitory effects on endothelium‐dependent relaxations in mouse isolated aorta and small mesenteric artery. Br J Pharmacol 140: 1193–1200, 2003.
 563. Thengchaisri N and Kuo L. Hydrogen peroxide induces endothelium‐dependent and ‐independent coronary arteriolar dilation: role of cyclooxygenase and potassium channels. Am J Heart Circ Physiol 285: H2255–H2263, 2003.
 564. Nagase S, Takemura K, Ueda A, Hirayama A, Aoyagi K, Kondoh M and Koyama A. A novel nonenzymatic pathway for the generation of nitric oxide by the reaction of hydrogen peroxide and D‐ or L‐arginine. Biochem Biophys Res Commun 233: 150–153, 1997.
 565. Newby AC. Dual role of matrix metalloproteinases (matrixins) in intimal thickening and atherosclerotic plaque rupture. Physiological Rev 85: 1–31, 2005.
 566. Chesler NC, Ku DN and Galis ZS. Transmural pressure induces matrix‐degrading activity in porcine arteries ex vivo. Am J Physiol (Heart Circ Physiol) 277: H2002–2009, 1999.
 567. Lucchesi PA, Sabri A, Belmadani S and Matrougui K. Involvement of metalloproteinases 2/9 in epidermal growth factor receptor trans‐activation in pressure‐induced myogenic tone in mouse mesenteric resistance arteries. Circulation 110: 3587–3593, 2004.
 568. Chew DK, Conte MS and Khalil RA. Matrix metalloproteinase‐specific inhibition of Ca 2+ entry mechanisms of vascular contraction. J Vasc Surg 40: 1001–1010, 2004.
 569. Galis ZS and Khatri SS. Matrix mettaloproteinases in vasular remodeling. Circ Res 90: 251–262, 2002.
 570. Wellman GC, Santana LF, Bonev AD and Nelson MT. Role of phospholamban in the modulation of arterial Ca 2+ sparks and Ca 2+ ‐activated K + channels by cAMP. Am J Physiol (Cell Physiol) 281: C1029–1037, 2001.
 571. Ungvari Z and Koller A. Selected contribution: NO released to flow reduces myogenic tone of skeletal muscle arterioles by decreasing smooth muscle Ca 2+ sensitivity. J Appl Physiol 91: 522–527; Discussion 504‐525, 2001.
 572. Martinez‐Lemus LA, Hill MA, Bolz SS, Pohl U and Meininger GA. Acute mechanoadaptation of vascular smooth muscle cells in response to continuous arteriolar vasoconstriction: implications for functional remodeling. FASEB J 18: 708–710, 2004.
 573. Sandow SL and Hill CE. Incidence of myoendothelial gap junctions in the proximal and distal mesenteric arteries of the rat is suggestive of a role in endothelium‐derived hyperpolarizing factor‐mediated responses. Circ Res 86: 341–346, 2000.
 574. Dora KA, Sandow SL, Gallagher NT, Takano H, Rummery NM, Hill CE and Garland CJ. Myoendothelial gap junctions may provide the pathway for EDHF in mouse mesenteric artery. J Vasc Res 40: 480–490, 2003.
 575. Sandow SL, Looft‐Wilson R, Doran B, Grayson TH, Segal SS and Hill CE. Expression of homocellular and heterocellular gap junctions in hamster arterioles and feed arteries. Cardiovasc Res 60: 643–653, 2003.
 576. Pannirselvam M, Verma S, Anderson TJ and Triggle CR. Cellular basis of endothelial dysfunction in small mesenteric arteries from spontaneously diabetic (db/db −/−) mice: role of decreased tetrahydrobiopterin bioavailability. Br J Pharmacol 136: 255–263, 2002.
 577. Sandow SL. Factors, fiction and endothelium‐derived hyperpolarizing factor. Clin Exp Pharmacol Physiol 31: 563–570, 2004.
 578. Griffith TM, Chaytor AT and Edwards DH. The obligatory link: role of gap junctional communication in endothelium‐dependent smooth muscle hyperpolarization. Pharmacol Res 49: 551–564, 2004.
 579. Segal SS and Duling BR. Flow control among microvessels coordinated by intercellular conduction. Science 234: 868–870, 1986.
 580. Segal SS and Duling BR. Conduction of vasomotor respones in arterioles: a role for cell‐to‐cell coupling? Am J Physiol (Heart Circ Physiol) 256: H838–H845, 1989.
 581. Emerson GG and Segal SS. Electrical coupling between endothelial cells and smooth muscle cells in hamster feed arteries: role in vasomotor control Circ Res 87: 474–479, 2000.
 582. Segal SS and Bény J‐L. Intracellular recording and dye transfer in arterioles during blood flow control. Am J Physiol (Heart Circ Physiol) 263: H1–H7, 1992.
 583. Little TL, Xia J and Duling BR. Dye tracers define differential endothelial and smooth muscle coupling patterns within the arteriolar wall. Circ Res 76: 498–504, 1995.
 584. Kruger O, Bény J‐L. Chabaud F, Traub O, Theis M, Brix K, Kirchhoff S and Willecke K. Altered dye diffusion and upregulation of connexin37 in mouse aortic endothelium deficient in connexin40. J Vasc Res 39: 160–172, 2002.
 585. Zou Y, Akazawa H, Qin Y, Sano M, Takano H, Minamino T, Makita N, Iwanaga K, Zhu W, Kudoh S, Toko H, Tamura K, Kihara M, Nagai T, Fukamizu A, Umemura S, Iiri T, Fujita T and Komuro I. Mechanical stress activates angiotensin II type I receptor without the involvement of angiotensin II. Nature Cell Biol 6: 499–506, 2004.
 586. Murphy TV, Kotecha N and Hill MA. Endothelium‐independent constriction of isolated, pressurized arterioles by N(omega)‐nitro‐L‐arginine methyl ester (L‐NAME). Br J Pharmacol 151: 602–609, 2007.
 587. Taylor MS, Bonev AD, Gross TP, Eckman DM, Brayden JE, Bond CT, Adelman JP and Nelson MT. Altered expression of small‐conductance Ca 2+ ‐activated K + (SK3) channels modulates arterial tone and blood pressure. Circ Res 93: 124–131, 2003.
 588. Dora KA, Hinton JM, Walker SD and Garland CJ. An indirect influence of phenylephrine on the release of endothelium‐derived vasodilators in rat small mesenteric artery. Br J Pharmacol 129: 381–387, 2000.
 589. Solan JL and Lampe PD. Connexin phosphorylation as a regulatory event linked to gap junction channel assembly. Biochem Biophys Acta 1711: 154–163, 2005.
 590. Prewitt RL, Rice DC and Dobrian AD. Adaptation of resistance arteries to increases in pressure. Microcirculation 9: 295–304, 2002.
 591. Borgstrom P and Grände P‐O. Myogenic microvascular responses to change of transmural pressure. A mathematical approach. Acta Physiol Scand 106: 411–423, 1979.
 592. Johnson PC, Burton KS, Henrich H and Henrich U. Effect of occlusion duration on reactive hyperemia in sartorius muscle capillaries. Am J Physiol 230: 715–719, 1976.
 593. Coffman JD and Gregg DE. Reactive hyperemia characteristics of the myocardium. Am J Physiol 199: 1143–1149, 1960.
 594. Olsson RA and Gregg DE. Myocardial reactive hyperemia in the unanesthetized dog. Am J Physiol 208: 224–230, 1965.
 595. Lombard JH and Duling BR. Multiple mechanisms of reactive hyperemia in arterioles of the hamster cheek pouch. Am J Physiol (Heart Circ Physiol) 241: H748–H755, 1981.
 596. Lombard JH and Duling BR. Relative contributions of passive and myogenic factors to diameter changes during single arteriole occlusion in the hamster cheek pouch. Circ Res 41: 365–373, 1977.
 597. Lombard JH and Duling BR. Relative importance of tissue oxygenation and vascular smooth muscle hypoxia in determining arteriolar responses to occlusion in the hamster cheek pouch. Circ Res 40: 546–551, 1977.
 598. Lombard JH, Kaminski RP and Stekiel WJ. Arteriolar responses to changes in oxygen availability following single withdrawal hemorrhage. Microvasc Res 21: 332–342, 1981.
 599. Hintze TH and Vatner SF. Reactive dilation of large coronary arteries in conscious dogs. Circ Res 54: 50–57, 1984.
 600. Eikens E and Wilcken DEL. Reactive hyperemia in the dog heart. Effects of temporarily restricting arterial inflow and of coronary occlusions lasting one and two cardiac cycles. Circ Res 35: 702–712, 1974.
 601. Eikens E and Wilcken DE. Myocardial reactive hyperemia and coronary vascular reactivity in the dog. Circ Res 33: 267–274, 1973.
 602. Konradi GP and Levtov VA. The relation between skeletal muscle reactive hyperemia and the duration of circulatory interruption. Fiziol Zh SSSR Im I M Sechenova 56: 366–374, 1970.
 603. Patterson GC. The role of intravascular pressure in the causation of reactive hyperaemia in the human forearm. Clin Sci 15: 17–25, 1956.
 604. Wood JE, Litter J and Wilkins RW. The mechanism of limb segment reactive hyperemia in man. Circ Res 3: 581–587, 1955.
 605. Kontos HA, Mauck HP, Jr. and Patterson JL, Jr. Mechanism of reactive hyperemia in limbs of anesthetized dogs. Am J Physiol 209: 1106–1114, 1965.
 606. Meininger GA. Responses of sequentially branching macro‐and microvessels during reactive hyperemia in skeletal muscle. Microvasc Res 34: 29–45, 1987.
 607. Kanatsuka H, Sekiguchi N, Sato K, Akai K, Wang Y, Komaru T, Ashikawa K and Takishima T. Microvascular sites and mechanisms responsible for reactive hyperemia in the coronary circulation of the beating canine heart. Circ Res 71: 912–922, 1992.
 608. Hilton SM. Experiments on the post‐contraction hyperaemia of skeletal muscle. J Physiol 120: 230–245, 1953.
 609. Koller A and Kaley G. Role of endothelium in reactive dilation of skeletal muscle arterioles. Am J Physiol (Heart Circ Physiol) 259: H1313–H1316, 1990.
 610. Koller A and Bagi Z. On the role of mechanosensitive mechanisms eliciting reactive hyperemia. Am J Physiol (Heart Circ Physiol) 283: H2250–H2259, 2002.
 611. Sun D, Huang A, Recchia FA, Cui Y, Messina EJ, Koller A and Kaley G. Nitric oxide‐mediated arteriolar dilation after endothelial deformation. Am J Physiol (Heart Circ Physiol) 280: H714–H721, 2001.
 612. Dora KA, Doyle MP and Duling BR. Elevation of intracellular calcium in smooth muscle causes endothelial cell generation of NO in arterioles. Proc Natl Acad Sci USA 94: 6529–6534, 1997.
 613. Zatta AJ and Headrick JP. Mediators of coronary reactive hyperaemia in isolated mouse heart. Br J Anaesthesiol 144: 576–587, 2005.
 614. Pohl U, Holtz J, Busse R and Bassenge E. Crucial role of endothelium in the vasodilator response to increased flow in vivo. Hypertension 8: 37–44, 1986.
 615. Koller A and Bagi Z. Nitric oxide and H2 O2 contribute to reactive dilation of isolated coronary arterioles. Am J Physiol (Heart Circ Physiol) 287: H2461–H2467, 2004.
 616. Richmond KN, Shonat RD, Lynch RM and Johnson PC. Critical PO2 of skeletal muscle in vivo. Am J Physiol (Heart Circ Physiol) 277: H1831–H1840, 1999.
 617. Wang W, Winlove CP and Michel CC. Oxygen partial pressure in outer layers of skin of human finger nail folds. J Physiol 549: 855–863, 2003.
 618. Klabunde RE and Johnson PC. Capillary velocity and tissue PO2 changes during reactive hyperemia in skeletal muscle. Am J Physiol 233: H379–H383, 1977.
 619. Barcroft H. An enquiry into the nature of the mediator of the vasodilatation in skeletal muscle in exercise and during circulatory arrest. J Physiol 222: 99P–118P, 1972.
 620. Shepherd JT. Circulation to skeletal muscle. In: Handbook of Physiology: Sec 2: The Cardiovascular System. Vol. 3: Peripheral Circulation and Organ Blood Flow, Part 1, ed. Shepherd JT. Bethesda, MD: American Physiological Society, 1983, pp. 319–370.
 621. Tominaga S, Watanabe K and Nakamura T. Role of adenosine or AMP as a probable mediator of blood flow regulation in canine hindlimb muscles. Tokushima J Exp Med 115: 185–195, 1975.
 622. Scott JB, Rudko M, Radawski D and Haddy FJ. Role of osmolarity, K +, H +, Mg ++, and O2 in local blood flow regulation. Am J Physiol 218: 338–345, 1970.
 623. Shepherd JT. Prolegomenon. In: Mechanisms of Vasodilatation. eds Vanhoutte PM and Leusen I. Basel: Karger, 1978, pp. 1–11.
 624. Tuma RF, Lindbom L and Arfors KE. Dependence of reactive hyperemia in skeletal muscle on oxygen tension. Am J Physiol (Heart Circ Physiol) 233: H289–H294, 1977.
 625. Chance B, Mayevsky A, Goodwin C and Mela L. Factors in oxygen delivery to tissue. Microvasc Res 8: 276–282, 1974.
 626. Wilson DF, Erecinska M, Drown C and Silver IA. The oxygen dependence of cellular energy metabolism. Arch Biochem Biophys 195: 485–493, 1979.
 627. Pittman RN and Duling BR. Oxygen sensitivity of vascular smooth muscle. I. In vitro studies. Microvasc Res 6: 202–211, 1973.
 628. Whalen WJ and Nair P. Intracellular PO2 and its regulation in resting skeletal muscle of the guinea pig. Circ Res 21: 251–261, 1967.
 629. Whalen WJ. Intracellular PO2 in heart and skeletal muscle. Physiologist 14: 69–82, 1971.
 630. Whalen WJ, Nair P, Buerk D and Thuning CA. Tissue PO2 in normal and denervated cat skeletal muscle. Am J Physiol 227: 1221–1225, 1974.
 631. Lash JM and Bohlen HG. Perivascular and tissue PO2 in contracting rat spinotrapezius muscle. Am J Physiol (Heart Circ Physiol) 252: H1192–H1202, 1987.
 632. Duling BR and Pittman RN. Oxygen tension: dependent or independent variable in local control of blood flow. FASEB J 34: 2012–2019, 1975.
 633. Schubert RW, Whalen WJ and Nair P. Myocardial PO2 distribution: relationship to coronary autoregulation. Am J Physiol (Heart Circ Physiol) 234: H361–H370, 1978.
 634. Klitzman B, Damon DN, Gorczynski RJ and Duling BR. Augmented tissue oxygen supply during striated muscle contraction in the hamster. Relative contributions of capillary recruitment. Circ Res 51: 711–721, 1982.
 635. Vanderkooi JM, Maniara G, Green TJ and Wilson DF. An optical method for measurement of dioxygen concentration based upon quenching of phosphorescence. J Biol Chem 262: 5476–5482, 1987.
 636. Shonat RD, Richmond KN and Johnson PC. Phosphorescence quenching and the microcirculation: an automated multipoint oxygen tension measuring instrument. Rev Sci Instr 66: 5075–5084, 1995.
 637. Richmond KN, Burnite S and Lynch RM. Oxygen sensitivity of mitochondrial metabolic state in isolated skeletal and cardiac myocytes. Am J Physiol (Cell Physiol) 273: C1613–C1622, 1997.
 638. Tamura M, Oshino N, Chance B and Silver IA. Optical measurements of intracellular oxygen concentration of rat heart in vitro. Arch Biochem Biophys 191: 8–22, 1978.
 639. Rubio R and Berne RM. Release of adenosine by the normal myocardium in dogs and its relationship to the regulation of coronary resistance. Circ Res 25: 407–415, 1969.
 640. Kingsbury MP, Robinson H, Flores NA and Sheridan DJ. Investigation of mechanisms that mediate reactive hyperaemia in guinea‐pig hearts: role of KATP channels, adenosine, nitric oxide and prostaglandins. Br J Pharmacol 132: 1209–1216, 2001.
 641. Kirkeboen KA, Aksnes G, Lande K and Ilebekk A. Role of adenosine for reactive hyperemia in normal and stunned porcine myocardium. Am J Physiol (Heart Circ Physiol) 263: H1119–H1127, 1992.
 642. Olsson RA, Snow JA and Gentry MK. Adenosine metabolism in canine myocardial reactive hyperemia. Circ Res 42: 358–362, 1978.
 643. Costa F, Sulur P, Angel M, Cavalcante J, Haile V, Christman B and Biaggioni I. Intravascular source of adenosine during forearm ischemia in humans: implications for reactive hyperemia. Hypertension 33: 1453–1457, 1999.
 644. Yamabe H, Okumura K, Ishizaka H, Tsuchiya T and Yasue H. Role of endothelium‐derived nitric oxide in myocardial reactive hyperemia. Am J Physiol (Heart Circ Physiol) 263: H8–H14, 1992.
 645. Clayton FC, Hess TA, Smith MA and Grover GJ. Coronary reactive hyperemia and adenosine‐induced vasodilation are mediated partially by a glyburide‐sensitive mechanism. Pharmacology 44: 92–100, 1992.
 646. Messina EJ, Weiner R and Kaley G. Arteriolar reactive hyperemia: modification by inhibitors of prostaglandin synthesis. Am J Physiol (Heart Circ Physiol) 232: H571–H575, 1977.
 647. Herbaczynska‐Cedro K and Vane JR. Prostaglandins as mediators of reactive hyperaemia in kidney. Nature 247: 492, 1974.
 648. Alexander RW, Kent KM, Pisano JJ, Keiser HR and Cooper T. Regulation of postocclusive hyperemia by endogenously synthesized prostaglandins in the dog heart. J Clin Invest 55: 1174–1181, 1975.
 649. VanTeeffelen JW, Constantinescu AA, Vink H and Spaan JA. Hypercholesterolemia impairs reactive hyperemic vasodilation of 2A but not 3A arterioles in mouse cremaster muscle. Am J Physiol (Heart Circ Physiol) 289: H447–H454, 2005.
 650. Kostic MM and Schrader J. Role of nitric oxide in reactive hyperemia of the guinea pig. Circ Res 70: 208–212, 1992.
 651. Wolin MS, Rodenburg JM, Messina EJ and Kaley G. Similarities in the pharmacological modulation of reactive hyperemia and vasodilation to hydrogen peroxide in rat skeletal muscle arterioles: effects of probes for endothelium‐derived mediators. J Pharmacol Exp Therapeut 253: 508–512, 1990.
 652. Ray CJ, Abbas MR, Coney AM and Marshall JM. Interactions of adenosine, prostaglandins and nitric oxide in hypoxia‐induced vasodilatation: in vivo and in vitro studies. J Physiol 544: 195–209, 2002.
 653. Upmacis RK, Deeb RS and Hajjar DP. Regulation of prostaglandin H2 synthase activity by nitrogen oxides. Biochemistry 38: 12505–12513, 1999.
 654. Salvemini D, Misko TP, Masferrer JL, Seibert K, Currie MG and Needleman P. Nitric oxide activates cyclooxygenase enzymes. Proc Natl Acad Sci USA 90: 7240–7244, 1993.
 655. Vassalle C, Domenici C, Lubrano V and L'Abbate A. Interaction between nitric oxide and cyclooxygenase pathways in endothelial cells. J Vasc Res 40: 491–499, 2003.
 656. Goodwin DC, Landino LM and Marnett LJ. Reactions of prostaglandin endoperoxide synthase with nitric oxide and peroxynitrite. Drug Metabol Rev 31: 273–294, 1999.
 657. Fredricks KT, Liu Y, Rusch NJ and Lombard JH. Role of endothelium and arterial K + channels in mediating hypoxic dilation of middle cerebral arteries. Am J Physiol (Heart Circ Physiol) 267: H580–H586, 1994.
 658. Busse R, Fotstermann R, Matsuda H and Pohl U. The role of prostaglandins in the endothelium‐mediated vasodilatory response to hypoxia. Pflügers Arch Eur J Physiol 401: 77–83, 1984.
 659. Pohl U and Busse R. Hypoxia stimulates release of endothelium‐derived relaxant factor. Am J Physiol (Heart Circ Physiol) 256: H1595–H1600, 1989.
 660. Koller A and Kaley G. Prostaglandins mediate arteriolar dilation to increased blood flow velocity in skeletal muscle microcirculation. Circ Res 67: 529–534, 1990.
 661. Messina EJ, Sun D, Koller A, Wolin MS and Kaley G. Increases in oxygen tension evoke arteriolar constriction by inhibiting endothelial prostaglandin synthesis. Microvasc Res 48: 151–160, 1994.
 662. Saito Y, Eraslan A and Hester RL. Role of endothelium‐derived relaxing factors in arteriolar dilation during muscle contraction elecited by electrical field stimulation. Microcirculation 1 (3): 195–201, 1994.
 663. Laughlin MH, Korthuis RJ, Duncker DJ and Bache RJ. Control of blood flow to cardiac and skeletal muscle during exercise. In: Handbook of Physiology. Bethesda, MD: American Physiological Society, 1996, pp. 705–769.
 664. Folkow B, Haglund U, Jodal M and Lundgren O. Blood flow in the calf muscle of man during heavy rhythmic exercise. Acta Physiol Scand 81: 157–163, 1971.
 665. Sheriff DD and Van Bibber R. Flow‐generating capability of the isolated skeletal muscle pump. Am J Physiol (Heart Circ Physiol) 274: H1502–H1508, 1998.
 666. Laughlin MH. Skeletal muscle blood flow capacity: role of muscle pump in exercise hyperemia. Am J Physiol 253: H993–1004, 1987.
 667. Delp MD. Control of skeletal muscle perfusion at the onset of dynamic exercise. Med Sci Sports Exer 31: 1011–1018, 1999.
 668. Shoemaker JK, Tschakovsky ME and Hughson RL. Vasodilation contributes to the rapid hyperemia with rhythmic contractions in humans. Can J Physiol Pharmacol 76: 418–427, 1998.
 669. Mohrman DE and Sparks HV. Myogenic hyperemia following brief tetanus of canine skeletal muscle. Am J Physiol 227: 531–535, 1974.
 670. Lundvall J, Mellander S and Sparks H. Myogenic response of resistance vessels and precapillary sphincters in skeletal muscle during exercise. Acta Physiol Scand 70: 257–268, 1967.
 671. Anrep GV, Blalock A and Samaan A. The effect of muscular contraction on the blood flow in skeletal muscle. Proc Roy Soc Lond Series B Biol Sci 114: 233–245, 1933.
 672. Mellander S. Interaction of local and nervous factors in vascular control. Angiologia 8: 187–201, 1970.
 673. Tune JD, Gorman MW and Feigl EO. Matching coronary blood flow to myocardial oxygen consumption. J Appl Physiol 97: 404–415, 2004.
 674. Delp MD and O'Leary DS. Integrative control of the skeletal muscle microcirculation in the maintenance of arterial pressure during exercise. J Appl Physiol 97: 1112–1118, 2004.
 675. Saltin B, Radegran G, Koskolou MD and Roach RC. Skeletal muscle blood flow in humans and its regulation during exercise. Acta Physiol Scand 162: 421–436, 1998.
 676. Hudlická O and el Khelly F. Metabolic factors involved in regulation of muscle blood flow. J Cardiovasc Pharmacol 7 (Suppl. 3): S59–S72, 1985.
 677. Bookman EL and McKenzie JE. Tissue adenosine content in active soleus and gracilis muscles of cats. Am J Physiol (Heart Circ Physiol) 244: H552–H559, 1983.
 678. Schwartz LM and McKenzie JE. Adenosine and active hyperemia in soleus and gracilis muscle of cats. Am J Physiol (Heart Circ Physiol) 259: H1295–H1304, 1990.
 679. Hester RL, Eraslan A and Saito Y. Differences in EDNO contribution to arteriolar diameters at rest and during functional dilation in striated muscle. Am J Physiol (Heart Circ Physiol) 265: H146–H151, 1993.
 680. Berg BR, Cohen KD and Sarelius IH. Direct coupling between blood flow and metabolism at the capillary level in striated muscle. Am J Physiol (Heart Circ Physiol) 272: H2693–H2700, 1997.
 681. Hnik P, Vyskocil F, Kriz N and Holas M. Work‐induced increase of extracellular potassium concentration in muscle measured by ion‐specific electrodes. Brain Res 40: 559–562, 1972.
 682. Hnik P, Holas M, Krekule I, Kuriz N, Mejsnar J, Smiesko V, Ujec E and Vyskocil F. Work‐induced potassium changes in skeletal muscle and effluent venous blood assessed by liquid ion‐exchanger microelectrodes. Pflügers Arch Eur J Physiol 362: 85–94, 1976.
 683. Hill MA, Trippe KM, Li QX and Meininger GA. Arteriolar arcades and pressure distribution in cremaster muscle microcirculation. Microvasc Res 44: 117–124, 1992.
 684. DeLano FA, Schmid‐Schönbein GW, Skalak TC and Zweifach BW. Penetration of the systemic blood pressure into the microvasculature of rat skeletal muscle. Microvasc Res 41: 92–110, 1991.
 685. Rosendal L, Blangsted AK, Kristiansen J, Sogaard K, Langberg H, Sjogaard G and Kjaer M. Interstitial muscle lactate, pyruvate and potassium dynamics in the trapezius muscle during repetitive low‐force arm movements, measured with microdialysis. Acta Physiol Stand 182: 379–388, 2004.
 686. Zweifach BW and Metz DB. Regional differences in response of terminal vascular bed to vasoactive agents. Am J Physiol 182: 155–165, 1955.
 687. Baez S. Bayliss response in the microcirculation. FASEB J 27: 1410–1415, 1968.
 688. Gore RW. Wall stress: a determinant of regional differences in response of frog microvessels to norepinephrine. Am J Physiol 222: 82–91, 1972.
 689. Price JM, Davis DL and Knauss EB. Length‐dependent sensitivity in vascular smooth muscle. Am J Physiol (Heart Circ Physiol) 241: H557–H563, 1981.
 690. Halpern W, Mongeon SA and Root DT. Stress, tension, and myogenic aspects of small isolated extraparenchymal rat arteries. In: Smooth Muscle Contraction, ed. Stephens NL. New York: Marcel Decker, 1984, pp. 427–456.
 691. Marshall JM and Tandon HC. Direct observations of muscle arterioles and venules following contraction of skeletal muscle fibres in the rat. J Physiol 350: 447–459, 1984.
 692. Berg BR and Sarelius IH. Functional capillary organization in striated muscle. Am J Physiol (Heart Circ Physiol) 268: H1215–H1222, 1995.
 693. Honig CR, Odoroff CL and Frierson JL. Capillary recruitment in exercise: rate, extent, uniformity, and relation to blood flow. Am J Physiol (Heart Circ Physiol) 238: H31–H42, 1980.
 694. Wiedeman MP, Tuma RF and Mayrovitz HN. Defining the precapillary sphincter. Microvasc Res 12: 71–75, 1977.
 695. Honig CR, Odoroff CL and Frierson JL. Active and passive capillary control in red muscle at rest and in exercise. Am J Physiol (Heart Circ Physiol) 243: H196–H206, 1982.
 696. Krogh A. The Anatomy and Physiology of Capillaries. New Haven, CT: Yale University Press, 1922, p. 422.
 697. Weigelt H and Schwarzmann V. A new method for the simultaneous presentation of low and high magnifications of microscopic specimens: application to in vivo studies of mesenterial capillaries. Microscopica Acta 85: 161–173, 1981.
 698. McCuskey RS. Sphincters in the microvascular system. Microvasc Res 2: 428–433, 1971.
 699. Wiedeman MP. Blood flow through terminal arterial vessels after denervation of the bat wing. Circ Res 22: 83–89, 1968.
 700. Eriksson E and Myrhage R. Microvascular dimensions and blood flow in skeletal muscle. Acta Physiol Scand 86: 211–222, 1972.
 701. Tyml K, Weigelt H and Lubbers DW. Occurrence of the “capillary contractility” phenomenon and its significance in the distribution of microvascular flow in frog skeletal muscle. Microvasc Res 27: 135–151, 1984.
 702. Baez S, Feldman M and Gootman PM. Central neural influence on precapillary microvessels and sphincter. Am J Physiol (Heart Circ Physiol) 233: H141–H147, 1977.
 703. Lutz BR and Fulton GP. Smooth muscle and blood flow in small blood vesdsles. In: Factors Regulating Blood Flow, eds Fulton GP and Zweifach BW Washington, DC: American Physiological Society, 1958, pp. 13–24.
 704. Rhodin JAG. The ultrastructure of mammaliam arterioles and precapillary sphincters. J Ultrastruct Res 18: 181–223, 1967.
 705. Bosman J. Flow cessation and capillary diameter changes in skeletal muscle. Maastricht, Netherlands: University of Limborg, 1996.
 706. Secomb TW and Hsu R. Resistance to blood flow in nonuniform capillaries. Microcirculation 4: 421–427, 1997.
 707. Fitzal F, DeLano FA, Young C and Schmid‐Schonbein GW. Early capillary no‐reflow during low‐flow reperfusion after hind limb ischemia in the rat. Annu Plastic Surg 49: 170–180, 2002.
 708. Rhodin JAG. Ultrastructure of the microvascular bed. In: The Microcirculation in Clinical Medicine, ed. Wells R. New York: Academic Press, 1972.
 709. Delashaw JB and Duling BR. A study of the functional elements regulating capillary perfusion in striated muscle. Microvasc Res 36: 162–171, 1988.
 710. Lund N, Damon DH, Damon DN and Duling BR. Capillary grouping in hamster tibials anterior muscles: flow patterns, and physiological significance. Int J Microcirc Clin Exp 5: 359–372, 1987.
 711. Skalak TC and Schmid‐Schönbein GW. The microvasculature in skeletal muscle. IV. A model of the capillary network. Microvasc Res 32: 333–347, 1986.
 712. Gorczynski RJ, Klitzman B and Duling BR. Interrelations between contracting striated muscle and precapillary microvessels. Am J Physiol (Heart Circ Physiol) 235: H494–H504, 1978.
 713. Wiedeman MP. Lengths and diameters of peripheral arterial vessels in the living animal. Circ Res 10: 686–690, 1962.
 714. Ellis CG, Ellsworth ML, Pittman RN and Burgess WL. Application of image analysis for evaluation of red blood cell dynamics in capillaries. Microvasc Res 44: 214–225, 1992.
 715. Japee SA, Ellis CG and Pittman RN. Flow visualization tools for image analysis of capillary networks. Microcirculation 11: 39–54, 2004.
 716. Harder DR, Zhang C and Gebremedhin D. Astrocytes function in matching blood flow to metabolic activity. News Physiol Sci 17: 27–31, 2002.
 717. Harder DR, Alkayed NJ, Lange AR, Gebremedhin D and Roman RJ. Functional hyperemia in the brain ‐ Hypothesis for astrocyte‐derived vasodilator metabolites. Stroke 29: 229–234, 1998.
 718. Filosa JA, Bonev AD, Straub SV, Meredith AL, Wilkerson MK, Aldrich RW and Nelson MT. Local potassium signaling couples neuronal activity to vasodilation in the brain. Nat Neurosci 9: 1397–1403, 2006.
 719. Bohlen HG. Microvascualr consequences of obesity and diabetes, Chapter 19. In: Handbook of Physiology: Microcirculation, 2nd Edition, eds Tuma RF, Durän WN and Ley K. Elsevier, 2008 (this volume).
 720. Folkow B, Sonnenschein RR and Wright DL. Loci of neurogenic and metabolic effects on pricapillary vessels of skeletal muscle. Acta Physiol Scand 81: 459–471, 1971.
 721. Goodman AH, Einstein R and Granger HJ. Effect of changing metabolic rate or local blood flow control in the canine hindlimb. Circ Res 43: 769–776, 1978.
 722. Proctor KG and Bohlen GG. Exercise hyperemia in the absence of a tissue PO2 decrease. Blood Vessels 18: 58–66, 1981.
 723. Segal SS and Duling BR. Communication between feed arteries and microvessels in hamster striated muscle: segmental vascular responses are functionally coordinated. Circ Res 59: 283–290, 1986.
 724. Kurjiaka DT and Segal SS. Interaction between conducted vasodilation and sympathetic nerve activation in arterioles of hamster striated muscle. Circ Res 76: 885–891, 1995.
 725. Beach JM, McGahren ED and Duling BR. Capillaries and arterioles are electrically coupled in hamster cheek pouch. Am J Physiol (Heart Circ Physiol) 275: H1489–H1496, 1998.
 726. Dietrich HH. Effect of locally applied epinephrine and norepinephrine on blood flow and diameter in capillaries of rat mesentery. Microvasc Res 38: 125–135, 1989.
 727. Mitchell D, Yu J and Tyml K. Comparable effects of arteriolar and capillary stimuli on blood flow in rat skeletal muscle. Microvasc Res 53: 22–32, 1997.
 728. Dietrich HH and Tyml K. Microvascular flow response to localized application of norepinephrine on capillaries in rat and frog skeletal muscle. Microvasc Res 43: 73–86, 1992.
 729. Ellsworth ML, Forrester T, Ellis CG and Dietrich HH. The erythrocyte as a regulator of vascular tone. Am J Physiol (Heart Circ Physiol) 269: H2155–2161, 1995.
 730. Tyml K and Groom AC. Regulation of blood flow in individual capillaries of resting skeletal muscle in frogs. Microvasc Res 20: 346–357, 1980.
 731. Collins DM, McCullough WT and Ellsworth ML. Conducted vascular responses: Communication across the capillary bed. Microvasc Res 56: 43–53, 1998.
 732. Hester RL. Uptake of metabolites by postcapillary venules: Mechanism for the control of arteriolar diameter. Microvasc Res 46: 254–261, 1993.
 733. Falcone JC and Bohlen HG. EDRF from rat intestine and skeletal muscle venules causes dilation of arterioles. Am J Physiol (Heart Circ Physiol) 258: H1515–H1523, 1990.
 734. Frame MDS and Sarelius IH. Regulation of capillary perfusion by small arterioles is spatially organized. Circ Res 73: 155–163, 1993.
 735. Song H and Tyml K. Evidence for sensing and integration of biological signals by the capillary network. Am J Physiol (Heart Circ Physiol) 265: H1235–H1242, 1993.
 736. Murrant CL and Sarelius IH. Local and remote arteriolar dilations initiated by skeletal muscle contraction. Am J Physiol (Heart Circ Physiol) 279: H2285–H2294, 2000.
 737. Rivers RJ. Cumulative conducted vasodilation within a single arteriole and the maximum conducted response. Am J Physiol (Heart Circ Physiol) 273: H310–H316, 1997.
 738. Laughlin MH and Armstrong RB. Rat muscle blood flows as a function of time during prolonged slow treadmill exercise. Am J Physiol (Heart Circ Physiol) 244: H814–H824, 1983.
 739. Folkow B and Halicka HD. A comparison betwen “red” and “white” muscle with respect to blood supply, capillary surface area and oxygen uptake during rest and exercise. Microvasc Res 1: 1–14, 1968.
 740. Hudlicka O. Resting and post‐contraction blood flow in slow and fast muscles of the chick during development. Microvasc Res 1: 390–402, 1969.
 741. Fuglevand AJ and Segal SS. Simulation of motor unit recruitment and microvascular unit perfusion: spatial considerations. J Appl Physiol 83: 1223–1234, 1997.
 742. Stainsby WN and Otis AB. Blood flow, blood oxygen tension, oxygen uptake, and oxygen transport in skeletal muscle. Am J Physiol 206: 858–866, 1964.
 743. Sullivan SM and Johnson PC. Effect of oxygen on blood flow autoregulation in cat sartorius muscle. Am J Physiol (Heart Circ Physiol) 241: H807–H815, 1981.
 744. Jones RD and Berne RM. Evidence for a metabolic mechanism in autoregulation of blood flow in skeletal muscle. Circ Res 17: 540–554, 1965.
 745. Kontos HA, Wei EP, Raper AJ, Rosenblum WI, Navari RM and Patterson JL, Jr. Role of tissue hypoxia in local regulation of cerebral microcirculation. Am J Physiol (Heart Circ Physiol) 234: H582–H591, 1978.
 746. Walker JR and Guyton AC. Influence of blood oxygen saturation on pressure‐flow curve of dog hindleg. Am J Physiol 212: 506–509, 1967.
 747. Bond RF, Blackard RF and Taxis JA. Evidence against oxygen being the primary factor governing autoregulation. Am J Physiol 216: 788–793, 1969.
 748. Kanatsuka H, Lamping KG, Eastham CL, Dellsperger KC and Marcus ML. Comparison of the effects of increased myocardial oxygen consumption and adenosine on the coronary microvascular resistance. Circ Res 65: 1296–1305, 1989.
 749. Mohrman DE and Regal RR. Relation of blood flow to VO2 . PO2, and PCO2 in dog gastrocnemius muscle. Am J Physiol (Heart Circ Physiol) 255: H1004–H1010, 1988.
 750. Stainsby WN. Autoregulation of blood flow in skeletal muscle during increased metabolic activity. Am J Physiol 202: 273–276, 1962.
 751. Shepherd AP. Intestinal capillary blood flow during metabolic hyperemia. Am J Physiol (Endocrinol Metabol) 6: E548–E554, 1979.
 752. Lo A, Fuglevand AJ and Secomb TW. Oxygen delivery to skeletal muscle fibers: effects of microvascular unit structure and control mechanisms. Am J Physiol (Heart Circ Physiol) 285: H955–H963, 2003.
 753. Tevald MA, Golub AS, Carvalho H, Lowman JD and Pittman RN. Microscopic assessment of skeletal muscle interstitial PO2 kinetics at the onset of contraction. FASEB J 19: A136, 2005.
 754. Welch HG and Stainsby WN. Oxygen debt in contracting dog skeletal muscle in situ. Respir Physiol 3: 229–242, 1967.
 755. Kramer K, Obal F and Quensel W. Untersuchungen uber den Muskelstoffwechsel des Warmebluters. III. Mitteilung. Die Sauerstoffaufnahme des Muskels wahrend rhythmischer Tatigkeit. Pflügers Archives Fur Die Gesamte Physiologie Des Menschen Und Der Tiere 241: 717–729, 1939.
 756. Mohrman DE, Cant JR and Sparks HV. Time course of vascular resistance and venous oxygen changes following brief tetanus of dog skeletal muscle. Circ Res 33: 323–336, 1973.
 757. Belloni FL and Sparks HV. Dynamics of myocardial oxygen consumption and coronary vascular resistance. Am J Physiol (Heart Circ Physiol) 233: H34–H43, 1977.
 758. Koskolou MD, Calbet JA, Radegran G and Roach RC. Hypoxia and the cardiovascular response to dynamic knee‐extensor exercise. Am J Physiol (Heart Circ Physiol) 272: H2655–H2663, 1997.
 759. Roach RC, Koskolou MD, Calbet JA and Saltin B. Arterial O2 content and tension in regulation of cardiac output and leg blood flow during exercise in humans. Am J Physiol (Heart Circ Physiol) 276: H438–H445, 1999.
 760. Rowell LB, Saltin B, Kiens B and Christensen NJ. Is peak quadriceps blood flow in humans even higher during exercise with hypoxemia? Am J Physiol (Heart Circ Physiol) 251: H1038–H1044, 1986.
 761. Stowe DF, Owen TL, Anderson DK, Haddy FJ and Scott JB. Interaction of O2 and CO2 in sustained exercise hyperemia of canine skeletal muscle. Am J Physiol 229: 28–33, 1975.
 762. Richardson RS, Grassi B, Gavin TP, Haseler LJ, Tagore K, Roca J and Wagner PD. Evidence of O2 supply‐dependent VO2 max in the exercise‐trained human quadriceps. J Appl Physiol 86: 1048–1053, 1999.
 763. Knight DR, Schaffartzik W, Poole DC, Hogan MC, Bebout DE and Wagner PD. Effects of hyperoxia on maximal leg O2 supply and utilization in men. J Appl Physiol 75: 2586–2594, 1993.
 764. Gorczynski RJ and Duling BR. Role of oxygen in arteriolar functional vasodilation in hamster striated muscle. Am J Physiol (Heart Circ Physiol) 235: H505–H515, 1978.
 765. Torres Filho IP and Intaglietta M. Microvessel PO2 measurements by phosphorescence decay method. Am J Physiol (Heart Circ Physiol) 265: H1434–H1438, 1993.
 766. Smith LM, Barbee RW, Ward KR and Pittman RN. Prolonged tissue PO2 reduction after contraction in spinotrapezius muscle of spontaneously hypertensive rats. Am J Physiol (Heart Circ Physiol) 287: H401–H407, 2004.
 767. Detar R and Bohr DF. Oxygen and vascular smooth muscle contraction. Am J Physiol 214: 241–244, 1968.
 768. Carrier O, Walker JR and Guyton AC. Role of oxygen in autoregulation of blood flow in isolated vessels. Am J Physiol 206: 951–954, 1964.
 769. Harder DR, Narayanan J, Birks EK, Liard JF, Imig JD, Lombard JH, Lange AR and Roman RJ. Identification of a putative microvascular oxygen sensor. Circ Res 79: 54–61, 1996.
 770. Katayama Y, Coburn RF, Fillers WS and Baron CB. Oxygen sensors in vascular smooth muscle. J Appl Physiol 77 (5): 2086–2092, 1994.
 771. Lombard JH, Smeda J, Madden JA and Harder DR. Effect of reduced oxygen availability upon myogenic depolarization and contraction of cat middle cerebral artery. Circ Res 58: 565–569, 1986.
 772. Fredricks KT, Liu Y and Lombard JH. Response of extraparenchymal resistance arteries of rat skeletal muscle to reduced PO2. Am J Physiol (Heart Circ Physiol) 267: H706–H715, 1994.
 773. Duling BR. Microvascular responses to alterations in oxygen tension. Circ Res 31: 481–489, 1972.
 774. Duling BR. Oxygen sensitivity of vascular smooth muscle. II. In vivo studies. Am J Physiol 227: 42–49, 1974.
 775. Hilton R and Eichholtz F. The influence of chemical factors in the coronary circulation. J Physiol 59: 413–425, 1924.
 776. Lammerant J, De Schryver C, Becsei I, Camphyn M and Mertens‐Strijthagen J. Coronary circulation response to hyperoxia after vagotomy and combined alpha and beta adrenergic receptors blockade in the anesthetized intact dog. Pflügers Arch Eur J Physiol 308: 185–196, 1969.
 777. Berne RM, Blackmon JR and Gardner TH. Hypoxemia and coronary blood flow. J Clin Invest 36: 1101–1106, 1957.
 778. Kuo L and Pittman RN. Effect of hemodilution on oxygen transport in arteriolar networks of hamster striated muscle. Am J Physiol (Heart Circ Physiol) 254: H331–H339, 1988.
 779. Torres Filho IP, Kerger H and Intaglietta M. pO2 measurements in arteriolar networks. Microvasc Res 51: 202–212, 1996.
 780. Kuo L and Pittman RN. Influence of hemoconcentration on arteriolar oxygen transport in hamster striated muscle. Am J Physiol (Heart Circ Physiol) 259: H1694–H1702, 1990.
 781. Popel AS and Gross JF. Analysis of oxygen diffusion from arteriolar networks. Am J Physiol (Heart Circ Physiol) 237: H681–H689, 1979.
 782. Popel AS, Pittman RN and Ellsworth ML. Rate of oxygen loss from arterioles is an order of magnitude higher than expected. Am J Physiol (Heart Circ Physiol) 256: H921–H924, 1989.
 783. Stein JC, Ellis CG and Ellsworth ML. Relationship between capillary and systemic venous PO2 during nonhypoxic and hypoxic ventilation. Am J Physiol (Heart Circ Physiol) 265: H537–H542, 1993.
 784. Jackson WF and Duling BR. The oxygen sensitivity of hamster cheek pouch arterioles. In vitro and in situ studies. Circ Res 53: 515–525, 1983.
 785. Jackson WF. Arteriolar oxygen reactivity: where is the sensor? Am J Physiol (Heart Circ Physiol) 253: H1120–H1126, 1987.
 786. Lynch FM, Austin C, Heagerty AM and Izzard AS. Adenosine and hypoxic dilation of rat coronary small arteries: roles of the ATP‐sensitive potassium channel, endothelium and nitric oxide. Am J Physiol (Heart Circ Physiol) 290: H1145–H1150, 2006.
 787. Liu Q and Flavahan NA. Hypoxic dilatation of porcine small coronary arteries: Role of endothelium and KATP ‐channels. Br J Pharmacol 120: 728–734, 1997.
 788. Messina EJ, Sun D, Koller A, Wolin MS and Kaley G. Role of endothelium‐derived prostaglandins in hypoxia‐elicited arteriolar dilation in rat skeletal muscle. Circ Res 71: 790–796, 1992.
 789. Frisbee JC, Maier KG, Falck JR, Roman RJ and Lombard JH. Integration of hypoxic dilation signaling pathways for skeletal muscle resistance arteries. Am J Physiol (Regul Integral Comparat Physiol) 283: R309–R319, 2002.
 790. Myers PR, Banitt PF, Guerra R, Jr. and Harrison DG. Role of the endothelium in modulation of the acetylcholine vasoconstrictor response in porcine coronary microvessels. Cardiovasc Res 25: 129–137, 1991.
 791. Miura H, Wachtel RE, Loberiza FR, Jr., Saito T, Miura M, Nicolosi AC and Gutterman DD. Diabetes mellitus impairs vasodilation to hypoxia in human coronary arterioles: reduced activity of ATP‐sensitive potassium channels. Circ Res 92: 151–158, 2003.
 792. Myers PR, Muller JM and Tanner MA. Effects of oxygen tension on endothelium dependent responses in canine coronary microvessels. Cardiovasc Res 25: 885–894, 1991.
 793. Lombard JH, Kunert MP, Roman RJ, Falck JR, Harder DR and Jackson WF. Cytochrome P‐450 omega‐hydroxylase senses O2 in hamster muscle, but not cheek pouch epithelium, microcirculation. Am J Physiol (Heart Circ Physiol) 276: H503–H508, 1999.
 794. Jackson WF. Regional differences in mechanism of action of oxygen on hamster arterioles. Am J Physiol (Heart Circ Physiol) 265: H599–H603, 1993.
 795. Tigno XT, Ley K, Pries AR and Gaehtgens P. Venulo‐arteriolar communication and propagated response. A possible mechanism for local control of blood flow. Pflügers Arch Eur J Physiol 414: 450–456, 1989.
 796. Hester RL. Venular‐arteriolar diffusion of adenosine in hamster cremaster microcirculation. Am J Physiol (Heart Circ Physiol) 258: H1918–H1924, 1990.
 797. Ellsworth ML and Pittman RN. Arterioles supply oxygen to capillaries by diffusion as well as by convection. Am J Physiol (Heart Circ Physiol) 258: H1240–H1243, 1990.
 798. Busse R, Pohl U, Klemm U and Kellner C. Endothelial cells are involved in the vasodilatory response to hypoxia. Pflügers Arch Eur J Physiol 397: 78–80, 1983.
 799. Kunert MP, Roman RJ, Alonso‐Galicia M, Falck JR and Lombard JH. Cytochrome P‐450 omega‐hydroxylase: a potential O2 sensor in rat arterioles and skeletal muscle cells. Am J Physiol (Heart Circ Physiol) 280: H1840–H1845, 2001.
 800. Frisbee JC, Roman RJ, Krishna UM, Falck JR and Lombard JH. Relative contributions of cyclooxygenase‐ and cytochrome P450 omega‐hydroxylase‐dependent pathways to hypoxic dilation of skeletal muscle resistance arteries. J Vasc Res 38: 305–314, 2001.
 801. Nase GP, Tuttle J and Bohlen HG. Reduced perivascular PO2 increases nitric oxide release from endothelial cells. Am J Physiol (Heart Circ Physiol) 285: H507–H515, 2003.
 802. Welsh DG, Jackson WF and Segal SS. Oxygen‐induces electromechanical coupling in arteriolar smooth muscle cells: a role for L‐type Ca2+ channels. Am J Physiol (Heart Circ Physiol) 274: H2018–H2024, 1998.
 803. Lopez‐Barneo J, Pardal R and Ortega‐Saenz P. Cellular mechanism of oxygen sensing. Annual Review of Physiology 63: 259–287, 2001.
 804. McCulloch KM, Osipenko ON and Gurney AM. Oxygen‐sensing potassium currents in pulmonary artery. Gen Pharmacol 32: 403–411, 1999.
 805. Noma A. ATP‐regulated K+ channels in cardiac muscle. Nature 305: 147–148, 1983.
 806. Ashcroft SJ and Ashcroft FM. Properties and functions of ATP‐sensitive K+‐channels. Cell Signal 2: 197–214, 1990.
 807. Daut J, Maier‐Rudolph W, Von Beckerath N, Mehrke G, Günther K and Goedel‐Meinen L. Hypoxic dilation of coronary arteries is mediated by ATP‐sensitive potassium channels. Science 247: 1341–1344, 1990.
 808. Decking UK, Reffelmann T, Schrader J and Kammermeier H. Hypoxia‐induced activation of KATP channels limits energy depletion in the guinea pig heart. Am J Physiol (Heart Circ Physiol) 269: H734–H742, 1995.
 809. Ho K, Nichols CG, Lederer WJ, Lytton J, Vassilev PM, Kanazirska MV and Hebert SC. Cloning and expression of an inwardly rectifying ATP‐regulated potassium channel. Nature 362: 31–38, 1993.
 810. Nakhostine N and Lamontagne D. Adenosine contributes to hypoxia‐induced vasodilation through ATP‐sensitive K+ channel activation. Am J Physiol (Heart Circ Physiol) 265: H1289–H1293, 1993.
 811. Lombard JH, Liu YP, Fredricks KT, Bizub DM, Roman RJ and Rusch NJ. Electrical and mechanical responses of rat middle cerebral arteries to reduced PO2 and prostacyclin. Am J Physiol (Heart Circ Physiol) 276: H509–H516, 1999.
 812. Duncker DJ, Van Zon N, Altman JD, Pavek TJ and Bache RJ. Role of K+ATP channels in coronary vasodilation during exercise. Circulation 88: 1245–1253, 1993.
 813. Richmond KN, Tune JD, Gorman MW and Feigl EO. Role of K+ATP channels in local metabolic coronary vasodilation. Am J Physiol (Heart Circ Physiol) 277: H2115–H2123, 1999.
 814. Richmond KN, Tune JD, Gorman MW and Feigl EO. Role of KATP+ channels and adenosine in the control of coronary blood flow during exercise. J Appl Physiol 89: 529–536, 2000.
 815. Kontos HA and Wei EP. Arginine analogues inhibit responses mediated by ATP‐sensitive K+ channels. Am J Physiol (Heart Circ Physiol) 271: H1498–H1506, 1996.
 816. Wei EP and Kontos HA. Blockade of ATP‐sensitive potassium channels in cerebral arterioles inhibits vasoconstriction from hypocapnic alkalosis in cats. Stroke 30: 851–853; Discussion p. 854, 1999.
 817. Sakaguchi M, Matsuura H and Ehara T. Swelling‐induced Cl− current in guinea‐pig atrial myocytes: inhibition by glibenclamide. J Physiol 505: 41–52, 1997.
 818. Schaffer P, Pelzmann B, Bernhart E, Lang P, Mächler H, Rigler B and Koidl B. The sulphonylurea glibenclamide inhibits voltage dependent potassium currents in human atrial and ventricular myocytes. Br J Pharmacol 128: 1175–1180, 1999.
 819. Cocks TM, King SJ and Angus JA. Glibenclamide is a competitive antagonist of the thromboxane A2 receptor in dog coronary artery in vitro. Br J Pharmacol 100: 375–378, 1990.
 820. Duncker DJ, van Zon NS, Pavek TJ, Herrlinger SK and Bache RJ. Endogenous adenosine mediates coronary vasodilation during exercise after KATP+ channel blockade. J Clin Invest 95: 285–295, 1995.
 821. Duncker DJ, van Zon NS, Ishibashi Y and Bache RJ. Role of K+ ATP channels and adenosine in the regulation of coronary blood flow during exercise with normal and restricted coronary blood flow. J Clin Invest 97: 996–1009, 1996.
 822. Ishibashi Y, Duncker DJ, Zhang J and Bache RJ. ATP‐sensitive K+ channels, adenosine and nitric oxide‐mediated mechanisms account for coronary vasodilation during exercise. Circ Res 82: 346–359, 1998.
 823. Hardie DG. AMP‐activated protein kinase: a key system mediating metabolic responses to exercise. Med Sci Sports Exer 36: 28–34, 2004.
 824. Rubin LJ, Magliola L, Feng X, Jones AW and Hale CC. Metabolic activation of AMP kinase in vascular smooth muscle. J Appl Physiol 98: 296–306, 2005.
 825. Boyd IA and Forrester T. The release of adenosine triphosphate from frog skeletal muscle in vitro. J Physiol 199: 115–135, 1968.
 826. Forrester T. Adenosine or adenosine triphosphate? In: Vasodilatation, eds Vanhoutte PM and Leusen I. New York: Raven Press, 1981, pp. 205–209.
 827. Forrester T and Lind AR. Identification of adenosine triphosphate in human plasma and the concentration in the venous effluent of forearm muscles before, during and after sustained contractions. J Physiol 204: 347–364, 1969.
 828. Forrester T. An estimate of adenosine triphosphate release into the venous effluent from exercising human forearm muscle. J Physiol 224: 611–628, 1972.
 829. Forrester T, Harper AM, MacKenzie ET and Thomson EM. Effect of adenosine triphosphate and some derivatives on cerebral blood flow and metabolism. J Physiol 296: 343–355, 1979.
 830. Rossi N, Churchill P, Ellis V and Amore B. Mechanism of adenosine receptor‐induced renal vasoconstriction in rats. Am J Physiol (Heart Circ Physiol) 255: H885–H890, 1988.
 831. Duff F, Patterson GC and Shepherd JT. A quantitative study of the response to adenosine triphosphate of the blood vessels of the human hand and forearm. J Physiol 125: 581–589, 1954.
 832. Phillis JW. Adenosine and adenine nucleotides as regulators of cerebral blood flow: roles of acidosis, cell swelling, and KATP+ channels. Crit Rev Neurobiol 16: 237–270, 2004.
 833. Benham CD. ATP‐gated channels in vascular smooth muscle cells. Ann NY Acad Sci 603: 275–286, 1990.
 834. McCullough WT, Collins DM and Ellsworth ML. Arteriolar responses to extracellular ATP in striated muscle. Am J Physiol (Heart Circ Physiol) 272: H1886–H1891, 1997.
 835. Ralevic V and Burnstock G. Receptors for purines and pyrimidines. Pharmacological Rev 50: 413–492, 1998.
 836. De Mey JG and Vanhoutte PM. Role of the intima in cholinergic and purinergic relaxation of isolated canine femoral arteries. J Physiol 316: 347–355, 1981.
 837. Berne RM. The role of adenosine in the regulation of coronary blood flow. Circ Res 47: 807–824, 1980.
 838. Lynge J, Juel C and Hellsten Y. Extracellular formation and uptake of adenosine during skeletal muscle contraction in the rat: role of adenosine transporters. J Physiol 537: 597–605, 2001.
 839. De Mey J, Burnstock G and Vanhoutte PM. Modulation of the evoked release of noradrenaline in canine saphenous vein via presynaptic receptors for adenosine but not ATP. Eur J Pharmacol 55: 401–405, 1979.
 840. Verhaeghe RH. Action of adenosine and adenine nucleotides on dogs' isolated veins. Am J Physiol (Heart Circ Physiol) 233: H114–H121, 1977.
 841. Rubio R, Wiedmeier VT and Berne RM. Relationship between coronary flow and adenosine production and release. J Molec Cell Cardiol 6: 561–566, 1974.
 842. Bockman EL, Berne RM and Rubio R. Release of adenosine and lack of release of ATP from contracting skeletal muscle. Pflügers Arch Eur J Physiol 355: 229–241, 1975.
 843. Benham CD. Neurotransmitters: ATP joins the fast lane. Nature 359: 103–104, 1992.
 844. Miseta A, Bogner P, Berenyi E, Kellermayer M, Galambos C, Wheatley DN and Cameron IL. Relationship between cellular ATP, potassium, sodium and magnesium concentrations in mammalian and avian erythrocytes. Biochim Biophys Acta 1175: 133–139, 1993.
 845. Bergfeld GR and Forrester T. Release of ATP from human erythrocytes in response to a brief period of hypoxia and hypercapnia. Cardiovasc Res 26: 40–47, 1992.
 846. Sprague RS, Ellsworth ML, Stephenson AH and Lonigro AJ. ATP: the red blood cell link to NO and local control of the pulmonary circulation. Am J Physiol (Heart Circ Physiol) 271: H2717–H2722, 1996.
 847. Bodin P, Bailey D and Burnstock G. Increased flow‐induced ATP release from isolated vascular endothelial cells but not smooth muscle cells. Br J Pharmacol 103: 1203–1205, 1991.
 848. Schrage WG, Wilkins BW, Dean VL, Scott JP, Henry NK, Wylam ME and Joyner MJ. Exercise hyperemia and vasoconstrictor responses in humans with cystic fibrosis. J Appl Physiol 99: 1866–1871, 2005.
 849. Berne RM. Cardiac nucleotides in hypoxia: possible role in regulation of coronary blood flow. Am J Physiol 204: 317–322, 1963.
 850. Honig CR and Frierson JL. Role of adenosine in exercise vasodilation in dog gracilis muscle. Am J Physiol (Heart Circ Physiol) 238: H703–H715, 1980.
 851. Belloni FL, Phair RD and Sparks HV. The role of adenosine in prolonged vasodilation following flow‐restricted exercise of canine skeletal muscle. Circ Res 44: 759–766, 1979.
 852. Poucher SM, Nowell CG and Collis MG. The role of adenosine in exercise hyperaemia of the gracilis muscle in anaesthetized cats. J Physiol 427: 19–29, 1990.
 853. Rubio R, Berne RM and Dobson JG, Jr. Sites of adenosine production in cardiac and skeletal muscle. Am J Physiol 225: 938–953, 1973.
 854. Sparks HV, Jr. and Bardenheuer H. Regulation of adenosine formation by the heart. Circ Res 58: 193–201, 1986.
 855. DeWitt DF, Wangler RD, Thompson CI and Sparks HV, Jr. Phasic release of adenosine during steady state metabolic stimulation in the isolated guinea pig heart. Circ Res 53: 636–643, 1983.
 856. Hellsten Y, Maclean D, Radegran G, Saltin B and Bangsbo J. Adenosine concentrations in the interstitium of resting and contracting human skeletal muscle. Circulation 98: 6–8, 1998.
 857. Schrader J, Haddy FJ and Gerlach E. Release of adenosine, inosine and hypoxanthine from the isolated guinea pig heart during hypoxia, flow‐autoregulation and reactive hyperemia. Pflügers Arch Eur J Physiol 369: 1–6, 1977.
 858. Wiedmeier VT, Rubio R and Berne RM. Inosine incorporation into myocardial nucleotides. J Molecul Cell Cardiol 4: 445–452, 1972.
 859. Kuo L and Chancellor JD. Adenosine potentiates flow‐induced dilation of coronary arterioles by activating KATP channels in endothelium. Am J Physiol (Heart Circ Physiol) 269: H541–H549, 1995.
 860. Hein TW, Belardinelli L and Kuo L. Adenosine A2A receptors mediate coronary microvascular dilation to adenosine: role of nitric oxide and ATP‐sensitive potassium channels. J Pharmacol Exp Therapeut 291: 655–664, 1999.
 861. Rivers RJ and Frame MD. Network vascular communication initiated by increases in tissue adenosine. J Vasc Res 36: 193–200, 1999.
 862. Thengchaisri N and Rivers RJ. Adenosine initiates cascular network response via activation of sensory nerve: role of CGRP and cGMP. FASEB J 18: A643, 2004.
 863. Juhran W, Voss EM, Dietmann K and Schaumann W. Pharmacological effects on coronary reactive hyperemia in conscious dogs. Naunyn‐Schmiedebergs Arch Pharmacol 269: 32–47, 1971.
 864. Merrill GF, Downey HF and Jones CE. Adenosine deaminase attenuates canine coronary vasodilation during systemic hypoxia. Am J Physiol (Heart Circ Physiol) 250: H579–H583, 1986.
 865. Proctor KG. Reduction of contraction‐induced arteriolar vasodilation by adenosine deaminase or theophylline. Am J Physiol (Heart Circ Physiol) 247: H195–H205, 1984.
 866. Proctor KG and Duling BR. Adenosine and free‐flow functional hyperemia in striated muscle. Am J Physiol (Heart Circ Physiol) 242: H688–H697, 1982.
 867. Tune JD, Richmond KN, Gorman MW, Olsson RA and Feigl EO. Adenosine is not responsible for local metabolic control of coronary blood flow in dogs during exercise. Am J Physiol (Heart Circ Physiol) 278: H74–H84, 2000.
 868. Bittar N and Pauly TJ. Myocardial reactive hyperemia responses in the dog after aminophylline and lidoflazine. Am J Physiol 220: 812–815, 1971.
 869. Bache RJ, Dai X‐Z, Schwartz JS and Homans DC. Role of adenosine in coronary vasodilation during exercise. Circ Res 62: 846–853, 1988.
 870. Komaru T, Lamping KG and Dellsperger KC. Role of adenosine in vasodilation of epimyocardial coronary microvessels during reduction in perfusion pressure. J Cardiovasc Pharmacol 24: 434–442, 1994.
 871. Yada T, Richmond KN, Van Bibber R, Kroll K and Feigl EO. Role of adenosine in local metabolic coronary vasodilation. Am J Physiol (Heart Circ Physiol) 276: H1425–H1433, 1999.
 872. Duncker DJ, Stubenitsky R and Verdouw PD. Role of adenosine in the regulation of coronary blood flow in swine at rest and during treadmill exercise. Am J Physiol (Heart Circ Physiol) 275: H1663–H1672, 1998.
 873. Merrill GF, Haddy FJ and Dabney JM. Adenosine, theophylline, and perfusate pH in the isolated, perfused guinea pig heart. Circ Res 42: 225–229, 1978.
 874. Miura M, Tominaga S and Hashimoto K. Potentiation of reactive hyperemia in the coronary and femoral circulation by the selective use of 2.6‐bis(diethanolamino)‐4,8‐dipiperidino‐pyrimodo[5,4‐d]pyrimidine. Arzneimittelforschung 17: 976–979, 1967.
 875. Saito D, Steinhart CR, Nixon DG and Olsson RA. Intracoronary adenosine deaminase reduces canine myocardial reactive hyperemia. Circ Res 49: 1262–1267, 1981.
 876. Kroll K and Feigl EO. Adenosine is unimportant in controlling coronary blood flow in unstressed dog hearts. Am J Physiol (Heart Circ Physiol) 249: H1176–H1187, 1985.
 877. Doughty JM, Plane F and Langton PD. Charybdotoxin and apamin block EDHF in rat mesenteric artery if selectively applied to the endothelium. Am J Physiol (Heart Circ Physiol) 276: H1107–H1112, 1999.
 878. Lew MJ, Rivers RJ and Duling BR. Arteriolar smooth muscle responses are modulated by an intramural diffusion barrier. Am J Physiol (Heart Circ Physiol) 257: H10–H16, 1989.
 879. Komaru T, Kanatsuka H and Shirato K. Coronary microcirculation: physiology and pharmacology. Pharmacol Therapeut 86: 217–261, 2000.
 880. Laxson DD, Homans DC and Bache RJ. Inhibition of adenosine‐mediated coronary vasodilation exacerbates myocardial ischemia during exercise. Am J Physiol (Heart Circ Physiol) 265: H1471–H1477, 1993.
 881. Phair RD and Sparks HV. Adenosine content of skeletal muscle during active hyperemia and ischemic contraction. Am J Physiol (Heart Circ Physiol) 237: H1–H9, 1979.
 882. Laughlin MH, Klabunde RE, Delp MD and Armstrong RB. Effects of dipyridamole on muscle blood flow in exercising miniature swine. Am J Physiol (Heart Circ Physiol) 257: H1507–H1515, 1989.
 883. Terjung RL, Dudley GA and Meyer RA. Metabolic and circulatory limitations to muscular performance at the organ level. J Exp Biol 115: 307–318, 1985.
 884. Bryan PT and Marshall JM. Cellular mechanisms by which adenosine induces vasodilatation in rat skeletal muscle: significance for systemic hypoxia. J Physiol 514: 163–175, 1999.
 885. Marshall JM, Thomas T and Turner L. A link between adenosine, ATP‐sensitive K+ channels, potassium and muscle vasodilatation in the rat in systemic hypoxia. J Physiol 472: 1–9, 1993.
 886. Jackson WF. Arteriolar tone is determined by activity of ATP‐sensitive potassium channels. Am J Physiol (Heart Circ Physiol) 265: H1797–H1803, 1993.
 887. Hein TW and Kuo L. cAMP‐independent dilation of coronary arterioles to adenosine: role of nitric oxide, G proteins, and KATP channels. Circ Res 85: 634–642, 1999.
 888. Stepp DW, Kroll K and Feigl EO. K+ATP channels and adenosine are not necessary for coronary autoregulation. Am J Physiol (Heart Circ Physiol) 273: H1299–H1308, 1997.
 889. Hein TW, Yuan Z, Rosa RH, Jr. and Kuo L. Requisite roles of A2A receptors, nitric oxide, and KATP channels in retinal arteriolar dilation in response to adenosine. Invest Ophthalmol Visual Sci 46: 2113–2119, 2005.
 890. Saito Y, McKay M, Eraslan A and Hester RL. Functional hyperemia in striated muscle is reduced following blockade of ATP‐sensitive potassium channels. Am J Physiol (Heart Circ Physiol) 270: H1649–H1654, 1996.
 891. Persson MG, Öhlén A, Lindbom L, Hedqvist P and Gustafsson LE. Role of adenosine in functional hyperemia in skeletal muscle as indicated by pharmacological tools. Naunyn Schmiedeberg's Arch Pharmacol 343: 52–57, 1991.
 892. Maekawa K, Saito D, Obayashi N, Uchida S and Haraoka S. Role of endothelium‐derived nitric oxide and adenosine in functional myocardial hyperemia. Am J Physiol (Heart Circ Physiol) 267: H166–H173, 1994.
 893. Martin SE, Lenhard SD, Schmarkey LS, Offenbacher S and Odle BM. Adenosine regulates coronary blood flow during increased work and decreased supply. Am J Physiol (Heart Circ Physiol) 264: H1438–H1446, 1993.
 894. Kille JM and Klabunde RE. Adenosine as a mediator of postcontraction hyperemia in dog gracilis muscle. Am J Physiol (Heart Circ Physiol) 246: H274–H282, 1984.
 895. Poucher SM. The role of the A2A adenosine receptor subtype in functional hyperaemia in the hindlimb of anaesthetized cats. J Physiol 492: 495–503, 1996.
 896. Goonewardene IP and Karim F. Attenuation of exercise vasodilatation by adenosine deaminase in anaesthetized dogs. J Physiol 442: 65–79, 1991.
 897. Abood LG, Koketsu K and Miyamoto S. Outflux of various phosphates during membrane depolarization of excitable tissues. Am J Physiol 202: 469–474, 1962.
 898. Hilton SM, Hudlicka O and Marshall JM. Possible mediators of functional hyperaemia in skeletal muscle. J Physiol 282: 131–147, 1978.
 899. Hilton SM and Vrbova G. Inorganic phosphate ‐ a new candidate for mediator of functional vasodilatation in skeletal muscle. J Physiol 206: 29P–30P, 1970.
 900. Tominaga S, Suzuki T and Nakamura T. Evaluation of roles of potassium, inorganic phosphate, osmolality, pH, pCO2, pO2, and adenosine or AMP in exercise and reactive hyperemias in canine hindlimb muscles. Tohoku J Exp Med 109: 347–363, 1973.
 901. Barcroft H, Foley TH and McSwiney RR. Experiments on the liberation of phosphate from the muscles of the human forearm during vigorous exercise and on the action of sodium phosphate on forearm muscle blood vessels. J Physiol 213: 411–420, 1971.
 902. Hilton SM. Evidence for phosphate as a mediator of functional hyperaemia in skeletal muscles. Pflügers Arch Eur J Physiol 369: 151–159, 1977.
 903. Bohr DF and Goulet PL. Role of electrolytes in the contractile machinery of vascular smooth muscle. Am J Cardiol 8: 549–556, 1961.
 904. Mohrman DE and Sparks HV. Role of potassium ions in the vascular response to a brief tetanus. Circ Res 35: 384–390, 1974.
 905. Kjellmer I. The potassium ion as a vasodilator during muscular exercise. Acta Physiol Scand 63: 460–468, 1965.
 906. Sreter FA. Cell water, sodium, and potassium in stimulated red and white mammalian muscles. Am J Physiol 205: 1295–1298, 1963.
 907. Kjellmer I and Oldelram H. The effect of some physiological vasodilators on the vascular bed of skeletal muscle. Acta Physiol Scand 63: 94–102, 1965.
 908. Gebert G. Measurement of K+ and Na+ activity in the extracellular space of rabbit skeletal muscle during muscular work by means of glass microelectrodes. Pflügers Arch Eur J Physiol 331: 204–214, 1972.
 909. Juel C, Pilegaard H, Nielsen JJ and Bangsbo J. Interstitial K+ in human skeletal muscle during and after dynamic graded exercise determined by microdialysis. Am J Physiol (Regul Integrat Comparat Physiol) 278: R400–R406, 2000.
 910. Naik JS, Valic Z, Buckwalter JB and Clifford PS. Rapid vasodilation in response to a brief tetanic muscle contraction. J Appl Physiol 87: 1741–1746, 1999.
 911. Hazeyama Y and Sparks HV. A model of potassium ion efflux during exercise of skeletal muscle. Am J Physiol (Regul Integrat Comparat Physiol) 236: R83–R90, 1979.
 912. Sheehan RM and Renkin EM. Capillary, interstitial and cell membrane barriers to blood‐tissue transport of potassium and rubidium in mammalian skeletal muscle. Circ Res 30: 588–607, 1972.
 913. Edwards G, Dora KA, Gardener MJ, Garland CJ and Weston AH. K+ is an endothelium‐derived hyperpolarizing factor in rat arteries. Nature 396: 269–272, 1998.
 914. Lo A, Fuglevand AJ and Secomb TW. Theoretical simulation of K+‐based mechanisms for regulation of capillary perfusion in skeletal muscle. Am J Physiol (Heart Circ Physiol) 287: H833–H840, 2004.
 915. Duling BR. Effects of potassium ion on the microcirculation of the hamster. Circ Res 37: 325–332, 1975.
 916. Knochel JP and Schlein EM. On the mechanism of rhabdomyolysis in potassium depletion. J Clin Invest 51: 1750–1758, 1972.
 917. Hazeyama Y and Sparks HV. Exercise hyperemia in potassium‐depleted dogs. Am J Physiol (Heart Circ Physiol) 236: H480–H486, 1979.
 918. McCarron JG and Halpern W. Potassium dilates rat cerebral arteries by two independent mechanisms. Am J Physiol (Heart Circ Physiol) 259: H902–H908, 1990.
 919. Kunze DL. Rate‐dependent changes in extracellular potassium in the rabbit atrium. Circ Res 41: 122–127, 1977.
 920. Reis DJ and Ladecola C. Regulation by the brain of blood flow and metabolism: role of intrisic neuronal networks and circulating catecholamines. News Physiol Sci 17: 129–145, 1986.
 921. Villringer A and Dirnagl U. Coupling of brain activity and cerebral blood flow: basis of functional neuroimaging. Cerebrovasc Brain Metabol Rev 7: 240–276, 1995.
 922. Paulson OB and Newman EA. Does the release of potassium from astrocyte endfeet regulate cerebral blood flow? Science 237: 896–898, 1987.
 923. Edvinsson L. Neurogenic mechanisms in the cerebrovascular bed. Autonomic nerves, amine receptors and their effects on cerebral blood flow. Acta Physiol Scand Suppl 427: 1–35, 1975.
 924. Gebremedhin D, Ma YH, Falck JR, Roman RJ, VanRollins M and Harder DR. Mechanism of action of cerebral epoxyeicosatrienoic acids on cerebral arterial smooth muscle. Am J Physiol (Heart Circ Physiol) 263: H519–H525, 1992.
 925. Norton JM and Detar R. Potassium and isolated coronary vascular smooth muscle. Am J Physiol 222: 474–479, 1972.
 926. Friedman SM, Nakashima M. Palaty V and Walters BK. Vascular resistance and Na+‐K+ gradients in the perfused rat‐tail artery. Can J Physiol Pharmacol 51: 410–417, 1973.
 927. Doughty JM, Boyle JP and Langton PD. Blockade of chloride channels reveals relaxations of rat small mesenteric arteries to raised potassium. Br J Pharmacol 132: 293–301, 2001.
 928. Burns WR, Cohen KD and Jackson WF. K+‐induced dilation of hamster cremasteric arterioles involves both the Na+/K+‐ATPase and inward‐rectifier K+ channels. Microcirculation 11: 279–293, 2004.
 929. Quayle JM, McCarron JG, Asbury JR and Nelson MT. Single calcium channels in resistance‐sized cerebral arteries from rats. Am J Physiol (Heart Circ Physiol) 264: H470–H478, 1993.
 930. Gebert G, Nguyen‐Duong H, Schnizer W, Konold P, Hillenbrand F, Yabu H and Brecht K. The response of isolated arteries and veins to potassium, osmolarity and drugs. Arzneimittelforschung 23: 391–398, 1969.
 931. Lombard JH and Stekiel WJ. Responses of cremasteric arterioles of spontaneously hypertensive rats to changes in extracellular K+ concentration. Microcirculation 2: 355–362, 1995.
 932. Wunsch SA, Muller‐Delp J and Delp MD. Time course of vasodilatory responses in skeletal muscle arterioles: role in hyperemia at onset of exercise. Am J Physiol (Heart Circ Physiol) 279: H1715–H1723, 2000.
 933. Haddy FJ. Potassium effects on contraction in arterila smooth muscle mediated by Na+. K+‐ATPase. FASEB J 42: 239–245, 1983.
 934. Hermsmeyer K. Sodium pump hyperpolarization‐relaxation in rat caudal artery. FASEB J 42: 246–252, 1983.
 935. Nguyen TS, Winn HR and Janigro D. ATP‐sensitive potassium channels may participate in the coupling of neuronal activity and cerebrovascular tone. Am J Physiol (Heart Circ Physiol) 278: H878–H885, 2000.
 936. Horiuchi T, Dietrich HH, Hongo K and Dacey RG, Jr. Mechanism of extracellular K+‐induced local and conducted responses in cerebral penetrating arterioles. Stroke 33: 2692–2699, 2002.
 937. Murad F. Cyclic guanosine monophosphate as a mediator of vasodilation. J Clin Invest 78: 1–5, 1986.
 938. Plane F, Wiley KE, Jeremy JY, Cohen RA and Garland CJ. Evidence that different mechanisms underlie smooth muscle relaxation to nitric oxide and nitric oxide donors in the rabbit isolated carotid artery. Br J Pharmacol 123: 1351–1358, 1998.
 939. Trepakova ES, Cohen RA and Bolotina VM. Nitric oxide inhibits capacitative cation influx in human platelets by promoting sarcoplasmic/endoplasmic reticulum Ca2+‐ATP‐ase‐dependent refilling of Ca2+ stores. Circ Res 84: 201–209, 1999.
 940. Frandsenn U, Bangsbo J, Sander M, Hoffner L, Betak A, Saltin B and Hellsten Y. Exercise‐induced hyperaemia and leg oxygen uptake are not altered during effective inhibition of nitric oxide synthase with NG‐nitro‐L‐arginine methyl ester in humans. J Physiol 531: 257–264, 2001.
 941. Radegran G and Saltin B. Nitric oxide in the regulation of vasomotor tone in human skeletal muscle. Am J Physiol (Heart Circ Physiol) 276: H1951–H1960, 1999.
 942. Friebel M, Klotz KF, Ley K, Gaehtgens P and Pries AR. Flow‐dependent regulation of arteriolar diameter in rat skeletal muscle in situ: Role of endothelium‐derived relaxing factor and prostanoids. J Physiol 483: 715–726, 1995.
 943. Hirai T, Visneski MD, Kearns KJ, Zelis R and Müsch TI. Effects of NO synthase inhibition on the muscular blood flow response to treadmill exercise in rats. J Appl Physiol 77: 1288–1293, 1994.
 944. Shoemaker JK, Halliwill JR, Hughson RL and Joyner MJ. Contributions of acetylcholine and nitric oxide to forearm blood flow at exercise onset and recovery. Am J Physiol (Heart Circ Physiol) 273: H2388–H2395, 1997.
 945. Busse R, Mülsch A, Fleming I and Hecker M. Mechanisms of nitric oxide release from the vascular endothelium. Circulation 87: V‐18–V‐25, 1993.
 946. Fisslthaler B, Dimmeier S, Hermann C, Busse R and Fleming I. Phosphorylation and activation of the endothelial nitric oxide synthase by fluid shear stress. Acta Physiol Scand 168: 81–88, 2000.
 947. 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.
 948. Fulton D, Gratton JP and Sessa WC. Post‐translational control of endothelial nitric oxide synthase: why isn't calcium/calmodulin enough? J Pharmacol Exp Therapeut 299: 818–824, 2001.
 949. Sessa WC. The nitric oxide synthase family of proteins. J Vasc Res 31: 131–143, 1994.
 950. Kobzik L, Reid MB, Bredt DS and Stamler JS. Nitric oxide in skeletal muscle. Nature 372: 546–548, 1994.
 951. Grange RW, Isotani E, Lau KS, Kamm KE, Huang PL and Stull JT. Nitric oxide contributes to vascular smooth muscle relaxation in contracting fast‐twitch muscles. Physiol Genom 5: 35–44, 2001.
 952. Silveira LR, Pereira‐Da‐Silva L, Juel C and Hellsten Y. Formation of hydrogen peroxide and nitric oxide in rat skeletal muscle cells during contractions. Free Rad Biol Med 35: 455–464, 2003.
 953. Lau KS, Grange RW, Chang WJ, Kamm KE, Sarelius I and Stull JT. Skeletal muscle contractions stimulate cGMP formation and attenuate vascular smooth muscle myosin phosphorylation via nitric oxide. FEBS Lett 431: 71–74, 1998.
 954. Stamler JS, Jia L, Eu JP, McMahon TJ, Demchenko IT, Bonaventura J, Gernert K and Piantadosi CA. Blood flow regulation by S‐nitrosohemoglobin in the physiological oxygen gradient. Science 276: 2034–2037, 1997.
 955. Gow AJ and Stamler JS. Reactions between nitric oxide and haemoglobin under physiological conditions. Nature 391: 169–173, 1998.
 956. Cosby K, Partovi KS, Crawford JH, Patel RP, Reiter CD, Martyr S, Yang BK, Waclawiw MA, Zalos G, Xu X, Huang KT, Shields H, Kim‐Shapiro DB, Schechter AN, Cannon III RO, and Gladwin MT. Nitrite reduction to nitric oxide by deoxyhemoglobin vasodilates the human circulation. Nat Med 9: 1498–1505, 2003.
 957. Ellsworth ML, Pittman RN and Ellis CG. Measurement of hemoglobin oxygen saturation in capillaries. Am J Physiol (Heart Circ Physiol) 252: H1031–H1040, 1987.
 958. Vaughn MW, Kuo L and Liao JC. Effective diffusion distance of nitric oxide in the microcirculation. Am J Physiol (Heart Circ Physiol) 274: H1705–H1714, 1998.
 959. Ellsworth ML. The red blood cell as an oxygen sensor: What is the evidence? Acta Physiol Scand 168: 551–559, 2000.
 960. Boegehold MA. Shear‐dependent release of venular nitric oxide: effect on arteriolar tone in rat striated muscle. Am J Physiol (Heart Circ Physiol) 271: H387–H395, 1996.
 961. Olearczyk JJ, Ellsworth ML, Stephenson AH, Lonigro AJ and Sprague RS. Nitric oxide inhibits ATP release from erythrocytes. J Pharmacol Exp Therapeut 309: 1079–1084, 2004.
 962. Dyke CK, Proctor DN, Dietz NM and Joyner MJ. Role of nitric oxide in exercise hyperaemia during prolonged rhythmic handgrip‐ping in humans. J Physiol 488: 259–265, 1995.
 963. Gilligan DM, Panza JA, Kilcoyne CM, Waclawiw MA, Casino PR and Quyyumi AA. Contribution of endothelium‐derived nitric oxide to exercise‐induced vasodilation. Circulation 90: 2853–2858, 1994.
 964. Hickner RC, Fisher JS, Ehsani AA and Kohrt WM. Role of nitric oxide in skeletal muscle blood flow at rest and during dynamic exercise in humans. Am J Physiol (Heart Circ Physiol) 273: H405–H410, 1997.
 965. Mitchell D and Tyml K. Nitric oxide release in rat skeletal muscle capillary. Am J Physiol (Heart Circ Physiol) 270: H1696–H1703, 1996.
 966. Lau KS, Grange RW, Isotani E, Sarelius IH, Kamm KE, Huang PL and Stull JT. nNOS and eNOS modulate cGMP formation and vascular response in contracting fast‐twitch skeletal muscle. Physiol Genom 2: 21–27, 2000.
 967. Cohen KD, Berg BR and Sarelius IH. Remote arteriolar dilations in response to muscle contraction under capillaries. Am J Physiol (Heart Circ Physiol) 278: H1916–H1923, 2000.
 968. Murrant CL and Sarelius IH. Multiple dilator pathways in skeletal muscle contraction‐induced arteriolar dilations. Am J Physiol (Regul Intergrat Comparat Physiol) 282: R969–R978, 2002.
 969. Budel S, Bartlett IS and Segal SS. Homocellular conduction along endothelium and smooth muscle of arterioles in hamster cheek pouch: unmasking an NO wave. Circ Res 93: 61–68, 2003.
 970. Dreier JP, Korner K, Gorner A, Lindauer U, Weih M, Villringer A and Dirnagl U. Nitric oxide modulates the CBF response to increased extracellular potassium. J Cerebr Blood Flow Metabol 15: 914–919, 1995.
 971. Yang G, Chen G, Ebner TJ and Iadecola C. Nitric oxide is the predominant mediator of cerebellar hyperemia during somatosensory activation in rats. Am J Physiol (Regul Integral Comparat Physiol) 277: R1760–R1770, 1999.
 972. Griffith OW and Stuehr DJ. Nitric oxide synthases: Properties and catalytic mechanism. Annu Rev Physiol 57: 707–736, 1995.
 973. Gen‐its RJ, Stein EA and Greene AS. Anesthesia alters NO‐mediated functional hyperemia. Brain Res 907: 20–26, 2001.
 974. Sahlin K. Intracellular pH and energy metabolism in skeletal muscle of man. With special reference to exercise. Acta Physiol Scand Suppl 455: 1–56, 1978.
 975. Geers C and Gros G. Carbon dioxide transport and carbonic anhydrase in blood and muscle. Physiol Rev 80: 681–715, 2000.
 976. Lindinger MI. Origins of [H+] changes in exercising skeletal muscle. Canadian J Appl Physiol 20: 357–368, 1995.
 977. Gaskell WH. On the tonicity of the heart and blood vessels. J Physiol (London) 3: 48–75, 1880.
 978. Connett RJ, Gayeski TE and Honig CR. Lactate accumulation in fully aerobic, working, dog gracilis muscle. Am J Physiol (Heart Circ Physiol) 246: H120–H128, 1984.
 979. Jorfeldt L, Juhlin‐Dannfelt A and Karlsson J. Lactate release in relation to tissue lactate in human skeletal muscle during exercise. J Appl Physiol 44: 350–352, 1978.
 980. Katz A and Sahlin K. Regulation of lactic acid production during exercise. J Appl Physiol 65: 509–518, 1988.
 981. Sahlin K, Harris RC, Nylind B and Hultman E. Lactate content and pH in muscle obtained after dynamic exercise. Pflugers Arch Eur J Physiol 367: 143–149, 1976.
 982. Achike FI and Ballard HJ. Influence of stimulation parameters on the release of adenosine, lactate and CO2 from contracting dog gracilis muscle. J Physiol 463: 107–121, 1993.
 983. Gollnick PD, Bayly WM and Hodgson DR. Exercise intensity, training, diet and lactate concentration in muscle and blood. Med Sci Sports Exer 18: 334–340, 1986.
 984. Lundholm L. The mechanism of the vasodilator effect of adrenaline. I. Effect on skeletal muscle vessels. Acta Physiol Scand 39: 1–52, 1956.
 985. Kurtz CM and Leake CD. The influence of the hydrion concentration on vascular tonicity II. With special reference to the dilating effect of lactic acid and urea. Am J Physiol 80: 107–113, 1927.
 986. McDowall R. The influence of acid base equilibrium on the activities of blood vessels. J Physiol 65: 25–31, 1928.
 987. Issekutz B, Jr., Lichtneckert I, Gaspar‐Nemeth Z and Hetenyi G, Jr. Tissue metabolism and peripheral circulation. II. Effect of iodoacetic acid on peripheral circulation. Arch Int Physiol 59: 116–124, 1951.
 988. Gasser R, Koppel H, Brussee H, Grisold M, Holzmann S and Klein W. EDRF does not mediate coronary vasodilation secondary to simulated ischemia: a study on KATP channels and Nω‐nitro‐L‐arginine on coronary perfusion pressure in isolated Langendorff‐perfused guinea‐pig hearts. Cardiovasc Drug Therap 12: 279–284, 1998.
 989. Paajanen V and Vornanen M. The induction of an ATP‐sensitive K+ current in cardiac myocytes of air‐ and water‐breathing vertebrates. Pflügers Arch Eur J Physiol 444: 760–770, 2002.
 990. Xie LH, Takano M and Noma A. The inhibitory effect of propranolol on ATP‐sensitive potassium channels in neonatal rat heart. Br J Pharmacol 123: 599–604, 1998.
 991. Ido Y, Chang K, Woolsey TA and Williamson JR. NADH: sensor of blood flow need in brain, muscle, and other tissues. FASEB J 15: 1419–1421, 2001.
 992. Chen YL, Wolin MS and Messina EJ. Evidence for cGMP mediation of skeletal muscle arteriolar dilation to lactate. J Appl Physiol 81: 349–354, 1996.
 993. Bangsbo J, Johansen L, Graham T and Saltin B. Lactate and H+ effluxes from human skeletal muscles during intense, dynamic exercise. J Physiol 462: 115–133, 1993.
 994. MacLean DA, Imadojemu VA and Sinoway LI. Interstitial pH, K+. lactate, and phosphate determined with MSNA during exercise in humans. Am J Physiol (Regul Intergrat Comparat Physiol) 278: R563–R571, 2000.
 995. Lott ME, Hogeman CS, Vickery L, Kunselman AR, Sinoway LI and MacLean DA. Effects of dynamic exercise on mean blood velocity and muscle interstitial metabolite responses in humans. Am J Physiol (Heart Circ Physiol) 281: H1734–H1741, 2001.
 996. Boschmann M, Rosenbaum M, Leibel RL and Segal KR. Metabolic and hemodynamic responses to exercise in subcutaneous adipose tissue and skeletal muscle. Int J Sports Med 23: 537–543, 2002.
 997. MacLean DA, Bangsbo J and Saltin B. Muscle interstitial glucose and lactate levels during dynamic exercise in humans determined by microdialysis. J Appl Physiol 87: 1483–1490, 1999.
 998. Ashina M, Stallknecht B, Bendtsen L, Pedersen JF, Galbo H, Dalgaard P and Olesen J. In vivo evidence of altered skeletal muscle blood flow in chronic tension‐type headache. Brain 125: 320–326, 2002.
 999. MacLean DA, LaNoue KF, Gray KS and Sinoway LI. Effects of hindlimb contraction on pressor and muscle interstitial metabolite responses in the cat. J Appl Physiol 85: 1583–1592, 1998.
 1000. Barcroft H. Blood flow and metabolism in skeletal muscle. In: Circulation in Skeletal Muscle, ed. Hudlicka O. Oxford, UK: Pergamon, 1968, pp. 121–134.
 1001. Barcroft H, Greenwood B, McArdle B, McSwiney RR, Semple SJG. Whelan RF and Youlten LJF. The effect of exercise on forearm blood flow and on venous blood pH, PCO2 and lactate in a subject with phosphorylase deficiency in skeletal muscle (McArdle's syndrome). J Physiol 189: 44P–46P, 1967.
 1002. McArdle B. Myopathy due to a defect in muscle glycogen breakdown. Clin Sci 10: 13–33, 1951.
 1003. Molnar JI, Scott JB, Frohlich ED and Haddy FJ. Local effects of various anions and H+ on dog limb and coronary vascular resistances. Am J Physiol 203: 125–132, 1962.
 1004. McKinnon W, Aaronson PI, Knock G, Graves J and Poston L. Mechanism of lactate‐induced relaxation of isolated rat mesenteric resistance arteries. J Physiol 490: 783–792, 1996.
 1005. Omar HA, Figueroa R, Omar RA and Wolin MS. Properties of an endogenous arachidonic acid‐elicited relaxing mechanism in human placental vessels. Am J Obstetr Gyn 167: 1064–1070, 1992.
 1006. Omar HA, Mohazzab‐H KM, Mortelliti MP and Wolin MS. O2‐dependent modulation of calf pulmonary artery tone by lactate: potential role of H2O2 and cGMP. Am J Physiol (Lung Respir Physiol) 264: L141–L145, 1993.
 1007. Mori K, Nakaya Y, Sakamoto S, Hayabuchi Y, Matsuoka S and Kuroda Y. Lactate‐induced vascular relaxation in porcine coronary arteries is mediated by Ca2+‐activated K+ channels. J Mol Cell Cardiol 30: 349–356, 1998.
 1008. Frøbert O, Mikkelsen EO, Bagger JP and Gravholt CH. Measurement of interstitial lactate during hypoxia‐induced dilatation in isolated pressurised porcine coronary arteries. J Physiol 539: 277–284, 2002.
 1009. Hein TW, Xu W and Kuo L. Dilation of retinal arterioles in response to lactate: role of nitric oxide, guanylyl cyclase, and ATP‐sensitive potassium channels. Invest Ophthalmol Visual Sci 47: 693–699, 2006.
 1010. Fukao M, Mason HS, Britton FC, Kenyon JL, Horowitz B and Keef KD. Cyclic GMP‐dependent protein kinase activates cloned BKCa channels expressed in mammalian cells by direct phosphorylation at serine 1072. J Biol Chem 274: 10927–10935, 1999.
 1011. Stainsby WN, Brechue WF, O'Drobinak DM and Barclay JK. Oxidation/reduction state of cytochrome oxidase during repetitive contractions. J Appl Physiol 67: 2158–2162, 1989.
 1012. Jöbsis FF and Stainsby WN. Oxadation of NADH during contractions of circulated mammalian skeletal muscle. Respir Physiol 4: 292–300, 1968.
 1013. Case RB and Greenberg H. The response of canine coronary vascular resistance to local alterations in coronary arterial PCO2. Circ Res 39: 558–566, 1976.
 1014. Daugherty RM, Jr., Scott JB, Dabney JM and Haddy FJ. Local effects of O2 and CO2 on limb, renal, and coronary vascular resistances. Am JPhysiol 213: 1102–1110, 1967.
 1015. Dietzel W, Samuelson SA, Guenter CA, Massion WH and Hinshaw LB. The effect of changes in arterial pCO2 on isogravimetric capillary pressure and vascular resistances. Proc Soc Exp Biol Med 131: 845–850, 1969.
 1016. Fujishima M, Scheinberg P, Busto R and Reinmuth OM. The relation between cerebral oxygen consumption and cerebral vascular reactivity to carbon dioxide. Stroke 2: 251–257, 1971.
 1017. Kittle CF, Aoki H and Brown EB. The role of pH and CO2 in the distribution of blood flow. Surgery 57: 139–154, 1965.
 1018. Kontos HA, Raper AJ and Patterson JL. Analysis of vasoactivity of local pH. pCO2 and bicarbonate on pial vessels. Stroke 8: 358–360, 1977.
 1019. Kontos HA, Richardson DW and Patterson JL, Jr. Effects of hypercapnia on human forearm blood vessels. Am J Physiol 212: 1070–1080, 1967.
 1020. Ledingham IM, McBride TI, Parratt JR and Vince JP. The effect of hypercapnia on myocardial blood flow and metabolism. J Physiol 210: 87–105, 1970.
 1021. Radawski D, Dabney JM, Daugherty RM Jr., Haddy FJ and Scott JB. Local effects of CO2 on vascular resistances and weight of the dog forelimb. Am J Physiol 222: 439–443, 1972.
 1022. Wang X, Wu J, Li L, Chen F, Wang R and Jiang C. Hypercapnic acidosis activates KATP channels in vascular smooth muscles. Circ Res 92: 1225–1232, 2003.
 1023. Wexels JC. Myocardial oxygen supply during hypocapnia and hypercapnia in the dog. Can J Physiol Pharmacol 64: 1376–1380, 1986.
 1024. Tarnow J, Bruckner JB, Eberlein HJ, Gethmann JW, Hess W, Patschke D and Wilde J. Blood pH and PaCO2 as chemical factors in myocardial blood flow control. Basic Res Cardiol 70: 685–696, 1975.
 1025. Rowe GG, Castillo CA and Crumpton CW. Effects of hyperventilation on systemic and coronary hemodynamics. Am Heart J 63: 67–77, 1962.
 1026. Vance JP, Brown DM and Smith G. The effects of hypocapnia on myocardial blood flow and metabolism. Br J Anaesth 45: 455–463, 1973.
 1027. Tenney SM. Sympatho‐adrenal stimulation by carbon dioxide and the inhibitory effect of carbonic acid on epinephrine response. Am J Physiol 187: 341–346, 1956.
 1028. Mithoefer JC, Kazemi H, Holford FD and Friedman I. Myocardial potassium exchange during respiratory acidosis: the interaction of carbon dioxide and sympathoadrenal discharge. Respir Physiol 5: 91–107, 1968.
 1029. Foëx P and Fordham RM. Intrinsic myocardial recovery from the negative inotropic effects of acute hypercapnia. Cardiovasc Res 6: 257–262, 1972.
 1030. Gurevicius J, Salem MR, Metwally AA, Silver JM and Crystal GJ. Contribution of nitric oxide to coronary vasodilation during hypercapnic acidosis Am J Physiol (Heart Circ Physiol) 268: H39–H47, 1995.
 1031. Wexels JC, Myhre ES and Mjøs OD. Effects of carbon dioxide and pH on myocardial blood‐flow and metabolism in the dog. Clin Physiol 5: 575–588, 1985.
 1032. Richardson DW, Wasserman AJ and Patterson JL, Jr. General and regional circulatory responses to change in blood pH and carbon dioxide tension. J Clin Invest 40: 31–43, 1961.
 1033. Kontos HA and Patterson JL, Jr. Carbon dioxide as a major factor in the production of reactive hyperaemia in the human forearm. Clin Sci 27: 143–154, 1964.
 1034. Kontos HA. Regulation of the cerebral circulation. Annu Rev Physiol 43: 397–407, 1981.
 1035. Duling BR. Changes in microvascular diameter and oxygen tension induced by carbon dioxide. Circ Res 32: 370–376, 1973.
 1036. McGillivray‐Anderson KM and Faber JE. Effect of acidosis on contraction of microvascular smooth muscle by α1‐ and α2‐adrenoceptors: implications for neural and metabolic regulation. Circ Res 66: 1643–1657, 1990.
 1037. Tateishi J and Faber JE. Inhibition of arteriole α2‐ but not α1‐adrenoceptor constriction by acidosis and hypoxia in vitro. Am J Physiol (Heart Circ Physiol) 268: H2068–H2076, 1995.
 1038. Broten TP, Romson JL, Fullerton DA, Van Winkle DM and Feigl EO. Synergistic action of myocardial oxygen and carbon dioxide in controlling coronary blood flow. Circ Res 68: 531–542, 1991.
 1039. Broten TP and Feigl EO. Role of myocardial oxygen and carbon dioxide in coronary autoregulation. Am J Physiol (Heart Circ Physiol) 262: H1231–H1237, 1992.
 1040. Case RB, Felix A, Wachter M, Kyriakidis G and Castellana F. Relative effect of CO2 on canine coronary vascular resistance. Circ Res: 42: 410–418, 1978.
 1041. Carr P, Graves JE and Poston L. Carbon dioxide induced vasorelaxation in rat mesenteric small arteries precontracted with noradrenaline is endothelium dependent and mediated by nitric oxide. Pflügers Arch Eur J Physiol 423: 343–345, 1993.
 1042. Hsu P, Albuquerque ML and Leffler CW. Mechanisms of hypercapnia‐stimulated PG production in piglet cerebral microvascular endothelial cells. Am J Physiol (Heart Circ Physiol) 268: H591–H603, 1995.
 1043. Mo FM and Ballard HJ. Adenosine output from dog gracilis muscle during systemic hypercapnia and/or amiloride‐SITS infusion. Am J Physiol (Heart Circ Physiol) 267: H1243–H1249, 1994.
 1044. Mustafa SJ and Mansour MM. Effect of perfusate pH on coronary flow and adenosine release in isolated rabbit heart. Proc Soc Exp Biol Med 176: 22–26, 1984.
 1045. Phillis JW, Song D and O'Regan MH. The role of adenosine in rat coronary flow regulation during respiratory and metabolic acidosis. Eur J Pharmacol 356: 199–206, 1998.
 1046. Belloni FL, Bruttig SP, Rubio R and Berne RM. Uptake and release of adenosine by cultured rat aortic smooth muscle. Microvasc Res 32: 200–210, 1986.
 1047. Nagi MM and Ward ME. Modulation of myogenic responsiveness by CO2 in rat diaphragmatic arterioles: Role of the endothelium. Am J Physiol (Heart Circ Physiol) 272: H1419–H1425, 1997.
 1048. Bardenheuer H, Whelton B and Sparks HV, Jr. Adenosine release by the isolated guinea pig heart in response to isoproterenol, acetylchloine, and acidosis: the minimal role of vascular endothelium. Circ Res 61: 594–600, 1987.
 1049. Deal CP, Jr. and Green HD. Effects of pH on blood flow and peripheral resistance in muscular and cutaneous vascular beds in the hind limb of the pentobarbitalized dog. Circ Res 2: 148–154, 1954.
 1050. Lobov GI and Kubyshkina NA. Effect of acidosis on contractile function of mesenterial lymphatic vessels in bulls. Bull Exp Biol Med 132: 622–624, 2001.
 1051. Sweeney M, Beddy D, Honner V, Sinnott B, O'Regan RG and McLoughlin P. Effects of changes in pH and CO2 on pulmonary arterial wall tension are not endothelium dependent. J Appl Physiol 85: 2040–2046, 1998.
 1052. Hermansen L and Osnes JB. Blood and muscle pH after maximal exercise in man. J Appl Physiol 32: 304–308, 1972.
 1053. Mainwood GW and Renaud JM. The effect of acid‐base balance on fatigue of skeletal muscle. Can J Physiol Pharmacol 63: 403–416, 1985.
 1054. Street D, Bangsbo J and Juel C. Interstitial pH in human skeletal muscle during and after dynamic graded exercise. J Physiol 537: 993–998, 2001.
 1055. Ballard HJ. The influence of lactic acid on adenosine release from skeletal muscle in anaesthetized dogs. J Physiol 433: 95–108, 1991.
 1056. Ishizaka H and Kuo L. Acidosis‐induced coronary arteriolar dilation is mediated by the ATP‐sensitive potassium channels in vascular smooth muscle. Circ Res 78: 50–57, 1996.
 1057. Radawski DP, Hoppe W and Haddy FJ. Role of vasoactive substances in active hyperemia in skeletal muscle. Proc Soc Exp Biol Med 148: 270–276, 1975.
 1058. Harder DR. Effect of H+ and elevated PCO2 on membrane electrical properties of rat cerebral arteries. Pflügers Arch Eur J Physiol 394: 182–185, 1982.
 1059. Dietrich HH and Dacey RG, Jr. Effects of extravascular acidification and extravascular alkalinization on constriction and depolarization in rat cerebral arterioles in vitro. J Neurosurg 81: 437–142, 1994.
 1060. Siegel G, Emden J, Wenzel K, Mironneau J and Stock G. Potassium channel activation in vascular smooth muscle. Adv Exp Med Biol 311: 53–72, 1992.
 1061. West GA, Leppla DC and Simard JM. Effects of external pH on ionic currents in smooth muscle cells from the basilar artery of the guinea pig. Circ Res 71: 201–209, 1992.
 1062. Weirich J, Dumont L and Fleckenstein‐Grun G. Contribution of store‐operated Ca2+ entry to pH0‐dependent changes in vascular tone of porcine coronary smooth muscle. Cell Calc 35: 9–20, 2004.
 1063. Loutzenhiser R, Matsumoto Y, Okawa W and Epstein M. H+‐induced vasodilation of rat aorta is mediated by alterations in intracellular calcium sequestration. Circ Res 67: 426–439, 1990.
 1064. Peng HL, Ivarsen A, Nilsson H and Aalkjaer C. On the cellular mechanism for the effect of acidosis on vascular tone. Acta Physiol Scand 164: 517–525, 1998.
 1065. Nakahata K, Kinoshita H, Hirano Y, Kimoto Y, Iranami H and Hatano Y. Mild hypercapnia induces vasodilation via adenosine triphosphate‐sensitive K+ channels in parenchymal microvessels of the rat cerebral cortex. Anesthesiology 99: 1333–1339, 2003.
 1066. Kinoshita H and Katusic ZS. Role of potassium channels in relaxations of isolated canine basilar arteries to acidosis. Stroke 28: 433–437, 1997.
 1067. Shioiri H, Komaru T, Sato K, Takahashi K, Takeda S, Kanatsuka H, Watanabe J and Shirato K. Impact of hypercholesterolemia on acidosis‐induced coronary microvascular dilation. Basic Res Cardiol 98: 76–83, 2003.
 1068. Phillis JW, Song D and O'Regan MH. Mechanisms involved in coronary artery dilatation during respiratory acidosis in the isolated perfused rat heart. Basic Res Cardiol 95: 93–97, 2000.
 1069. Hattori K, Tsuchida S, Tsukahara H, Mayumi M, Tanaka T, Zhang L, Taniguchi T and Muramatsu I. Augmentation of NO‐mediated vasodilation in metabolic acidosis. Life Sci 71: 1439–1447, 2002.
 1070. Lindauer U, Kunz A, Schuh‐Hofer S, Vogt J, Dreier JP and Dirnagl U. Nitric oxide from perivascular nerves modulates cerebral arterial pH reactivity. Am J Physiol (Heart Circ Physiol) 281: H1353–H1363, 2001.
 1071. Cheng B, Essackjee HC and Ballard HJ. Evidence for control of adenosine metabolism in rat oxidative skeletal muscle by changes in pH. J Physiol 522: 467–477, 2000.
 1072. Phillis JW, O'Regan MH and Song D. Further evidence for the role of adenosine in hypercapnia/acidosis‐evoked coronary flow regulation. Gen Pharmacol 33: 431–437, 1999.
 1073. Kingwell BA. Nitric oxide as a metabolic regulator during exercise: effects of training in health and disease. Clin Exp Pharmacol Physiol 27: 239–250, 2000.
 1074. Bredt DS. Endogenous nitric oxide synthesis: biological functions and pathophysiology. Free Rad Res 31: 577–596, 1999.
 1075. Patwell DM, McArdle A, Morgan JE, Patridge TA and Jackson MJ. Release of reactive oxygen and nitrogen species from contracting skeletal muscle cells. Free Rad Biol Med 37: 1064–1072, 2004.
 1076. Lundvall J. Tissue hyperosmolality as a mediator of vasodilatation and transcapillary fluid flux in exercising skeletal muscle. Acta Physiol Scand Suppl 379: 1–142, 1972.
 1077. Gray SD. Effect of hypertonicity on vascular dimensions in skeletal muscle. Microvasc Res 3: 117–124, 1971.
 1078. Duling BR and Staples E. Microvascular effects of hypertonic solutions in the hamster. Microvasc Res 11: 51–56, 1976.
 1079. Sparks HV, Jr. and Belloni FL. The peripheral circulation: local regulation. Ann Rev Physiol 40: 67–92, 1978.
 1080. Haljamae H, Johansson B, Jonsson O and Rockert H. The distribution of sodium, potassium and chloride in the smooth muscle of the rat portal vein. Acta Physiol Scand 78: 255–268, 1970.
 1081. Scott JB and Radawski D. Role of hyperosmolarity in the genesis of active and reactive hyperemia. Circ Res 28 (Suppl. 1): 26–32, 1971.
 1082. Stainsby WN and Barclay JK. Effect of infusions of osmotically active substances on muscle blood flow and systemic blood pressure. Circ Res 28 (Suppl. 11: 33–38, 1971.
 1083. Mellander S and Lundvall J. Role of tissue hyperosmolality in exercise hyperemia. Circ Res 28 (Suppl. 1): 39–45, 1971.
 1084. Wahl M, Kuschinsky W, Bosse O and Thurau K. Dependency of pial arterial and arteriolar diameter on perivascular osmolarity in the cat. A microapplication study. Circ Res 32: 162–169, 1973.
 1085. Ishizaka H and Kuo L. Endothelial ATP‐sensitive potassium channels mediate coronary microvascular dilation to hyperosmolarity. Am J Physiol (Heart Circ Physiol) 273: H104–H112, 1997.
 1086. Massett MP, Koller A and Kaley G. Effect of MAP kinase kinase (MEK) inhibitor PD98059 on constrictor responses in skeletal muscle arterioles. FASEB J 14: A29, 2000.
 1087. Hallback DA, Jodal M, Sjoqvist A and Lundgren O. Villous tissue osmolality and intestinal transport of water and electrolytes. Acta Physiol Scand 107: 115–126, 1979.
 1088. Bohlen HG. Na+ induced intestinal interstitial hyperosmolality and vascular responses during absorptive hyperemia. Am J Physiol (Heart Circ Physiol) 242: H785–H789, 1982.
 1089. Bohlen HG and Lash JM. Intestinal absorption of sodium and nitric oxide‐dependent vasodilation interact to dominate resting vascular resistance. Circ Res 78: 231–237, 1996.
 1090. Bohlen HG. Integration of intestinal structure, function, and microvascular regulation. Microcirculation 5: 27–37, 1998.
 1091. Bohlen HG and Lash JM. Intestinal lymphatic vessels release endothelial‐dependent vasodilators. Am J Physiol (Heart Circ Physiol) 262: H813–H818, 1992.
 1092. Steenbergen JM and Bohlen HG. Sodium hyperosmolarity of intestinal lymph causes arteriolar vasodilation in part mediated by EDRF. Am J Physiol (Heart Circ Physiol) 265: H323–H328, 1993.
 1093. Steenbergen JF and Fay FS. The quantal nature of calcium release to caffeine in single smooth muscle cells results from activation of the sarcoplasmic reticulum Ca2+‐ATPase. J Biol Chem 271: 1821–1824, 1996.
 1094. Bohlen HG and Nase GP. Dependence of intestinal arteriolar regulation on flow‐mediated nitric oxide formation. Am J Physiol (Heart Circ Physiol) 279: H2249–H2258, 2000.
 1095. Proctor KG. Contribution of hyperosmolality to glucose‐induced intestinal hyperemia. Am J Physiol (Gastrointest Liver Physiol) 248: G521–G525, 1985.
 1096. Johansson B and Jonsson O. Cell volume as a factor influencing electrical and mechanical activity of vascular smooth muscle. Acta Physiol Scand 72: 456–468, 1968.
 1097. Keef KD and Ross G. Relaxation induced by KC1, NaCl and sucrose in rabbit cornary arteries. Pflügers Arch Eur J Physiol 409: 308–313, 1987.
 1098. Yamaguchi M, Tomiyama Y, Katayama T, Kitahata H and Oshita S. Involvement of adenosine triphosphate‐sensitive potassium channels in the response of membrane potential to hyperosmolality in cultured human aorta endothelial cells. Anesth Analg 100: 419–426, 2005.
 1099. Nilius B and Droogmans G. Amazing chloride channels: an overview. Acta Physiol Scand 177: 119–147, 2003.
 1100. Daut J, Dischner A and Mehrke G. Bradykinin induces a transient hyperpolarization of cultured guinea‐pig coronary endothelial cells. J Physiol 410: 48 P, 1989.
 1101. Cannell MB and Sage SO. Bradykinin‐evoked changes in cytosolic calcium and membrane currents in cultured bovine pulmonary artery endothelial cells. J Physiol 419: 555–568, 1989.
 1102. Sharma NR and Davis MJ. Substance P‐induced calcium entry in endothelial cells is secondary to depletion of intracellular stores. Am J Physiol (Heart Circ Physiol) 268: H962–H973, 1995.
 1103. Nilius B, Vriens J, Prenen J, Droogmans G and Voets T. TRPV4 calcium entry channel: a paradigm for gating diversity. Am J Physiol (Cell Physiol) 286: C195–C205, 2004.
 1104. Nilius B and Sage SO. TRP channels: novel gating properties and physiological functions. J Physiol 567: 33–34, 2005.
 1105. Messina EJ, Weiner R and Kaley G. Prostaglandins and local circulatory control. Fed Proc 35: 2367–2375, 1976.
 1106. Vanhoutte PM and Mombouli JV. Vascular endothelium: vasoactive mediators. Progr Cardiovasc Dis 39: 229–238, 1996.
 1107. Karamouzis M, Karamouzis I, Vamvakoudis E, Ampatzidis G, Christoulas K, Angelopoulou N and Mandroukas K. The response of muscle interstitial prostaglandin E2(PGE2), prostacyclin I2(PGI2) and thromboxane A2(TXA2) levels during incremental dynamic exercise in humans determined by in vivo microdialysis. Prostaglandins, Leukotrienes and Essential Fatty Acid 64: 259–263, 2001.
 1108. Frandsen U, Bangsbo J, Langberg H, Saltin B and Hellsten Y. Inhibition of nitric oxide synthesis by systemic NG‐monomethyl‐L‐arginine administration in humans: effects on interstitial adenosine, prostacyclin and potassium concentrations in resting and contracting skeletal muscle. J Vasc Res 37: 297–302, 2000.
 1109. Symons JD, Theodossy SJ, Longhurst JC and Stebbins CL. Intramuscular accumulation of prostaglandins during static contraction of the cat triceps surae. J Appl Physiol 71: 1837–1842, 1991.
 1110. Wilson JR and Kapoor SC. Contribution of prostaglandins to exercise‐induced vasodilation in humans. Am J Physiol (Heart Circ Physiol) 265: H171–H175, 1993.
 1111. Young EW and Sparks HV. Prostaglandins and exercise hyperemia of dog skeletal muscle. Am J Physiol (Heart Circ Physiol) 238: H191–H195, 1980.
 1112. Herbaczynska‐Cedro K, Staszewska‐Barczak J and Janczewska H. Muscular work and the release of prostaglandin‐like substances. Cardiovasc Res 10: 413–420, 1976.
 1113. Weiner R, Messina EJ, Rodenburh J and Kaley K. Indomethacin reduces skeletal muscle vasodilatation induced by exercise and ishemia. Artery 3: 52–58, 1977.
 1114. Duffy SJ, Castle SF, Harper RW and Meredith IT. Contribution of vasodilator prostanoids and nitric oxide to resting flow, metabolic vasodilation, and flow‐mediated dilation in human coronary circulation. Circulation 100: 1951–1957, 1999.
 1115. Kilbom A and Wennmalm A. Endogenous prostaglandins as local regulators of blood flow in man: effect of indomethacin on reactive and functional hyperaemia. J Physiol 257: 109–121, 1976.
 1116. Nuttle LC, Ligon AL, Farrell KR and Hester RL. Inhibition of phospholipase A2 attenuates functional hyperemia in the hamster cremaster muscle. Am J Physiol (Heart Circ Physiol) 276: H1289–H1294, 1999.
 1117. Dai XZ and Bache RJ. Effect of indomethacin on coronary blood flow during graded treadmill exercise in the dog. Am J Physiol (Heart Circ Physiol) 247: H452–H458, 1984.
 1118. Young EW and Sparks HV. Prostaglandin E release from dog skeletal muscle during restricted flow exercise. Am J Physiol (Heart Circ Physiol) 236: H596–H599, 1979.
 1119. Beaty III O, and Donald DE. Contribution of prostaglandins to muscle blood flow in anesthetized dogs at rest, during exercise, and followirg inflow occlusion. Circ Res 44: 67–75, 1979.
 1120. Morganroth ML, Young EW and Sparks HV. Prostaglandin and histaminergic mediation of prolonged vasodilation after exercise. Am J Physiol (Heart Circ Physiol) 233: H27–H33, 1977.
 1121. Nowak J and Wennmalm A. A study on the role of endogenous prostaglandins in the development of exercise‐induced and postocclusive hyperemia in human limbs. Acta Physiol Scand 106: 365–369, 1979.
 1122. Jackson WF. Ion channels and vascular tone. Hypertension 35: 173–178, 2000.
 1123. Jackson WF. Lipoxygenase inhibitors block O2 responses of hamster cheek pouch arterioles. Am J Physiol (Heart Circ Physiol) 255: H711–H716, 1988.
 1124. Jackson WF. Prostaglandins do not mediate arteriolar oxygen reactivity. Am J Physiol (Heart Circ Physiol) 250: H1102–H1108, 1986.
 1125. Jackson WF. Arteriolar oxygen reactivity is inhibited by leukotriene antagonists. Am J Physiol (Heart Circ Physiol) 257: H1565–H1572, 1989.
 1126. Frisbee JC, Krishna UM, Falck JR and Lombard JH. Role of prostanoids and 20‐HETE in mediating oxygen‐induced constriction of skeletal muscle resistance arteries. Microvasc Res 62: 271–283, 2001.
 1127. Harder DR, Roman RJ and Gebremedhin D. Molecular mechanisms controlling nutritive blood flow: role of cytochrome P450 enzymes. Acta Physiol Scand 168: 543–549, 2000.
 1128. Li PL, Chen CL, Bortell R and Campbell WB. 11, 12‐epoxyeicosatrienoic acid stimulates endogenous mono‐ADP‐ribosylation in bovine coronary arterial smooth muscle. Circ Res 85: 349–356, 1999.
 1129. Oltman CL, Weintraub NL, VanRollins M and Dellsperger KC. Epoxyeicosatrienoic acids and dihydroxyeicosatrienoic acids are potent vasodilators in the canine coronary microcirculation. Circ Res 83: 932–939, 1998.
 1130. Fisslthaler B, Popp R, Kiss L, Potente M, Harder DR, Fleming I and Busse R. Cytochrome P450 2C is an EDHF synthase in coronary arteries. Nature 401: 493–497, 1999.
 1131. Hillig T, Krustrup P, Fleming I, Osada T, Saltin B and Hellsten Y. Cytochrome P450 2C9 plays an important role in the regulation of exercise‐induced skeletal muscle blood flow and oxygen uptake in humans. J Physiol 546: 307–314, 2003.
 1132. Peng X, Carhuapoma JR, Bhardwaj A, Alkayed NJ, Falck JR, Harder DR, Traystman RJ and Koehler RC. Suppression of cortical functional hyperemia to vibrissal stimulation in the rat by epoxygenase inhibitors. Am J Physiol (Heart Circ Physiol) 283: H2029–H2037, 2002.
 1133. Peng X, Zhang C, Alkayed NJ, Harder DR and Koehler RC. Dependency of cortical functional hyperemia to forepaw stimulation on epoxygenase and nitric oxide synthase activities in rats. J Cerebr Blood Flow Metabol 24: 509–517, 2004.
 1134. Bhardwaj A, Northington FJ, Carhuapoma JR, Falck JR, Harder DR, Traystman RJ and Koehler RC. P‐450 epoxygenase and NO synthase inhibitors reduce cerebral blood flow response to N‐methyl‐D‐aspartate. Am J Physiol (Heart Circ Physiol) 279: H1616–H1624, 2000.
 1135. Nitenberg A, Ledoux S, Valensi P, Sachs R and Antony I. Coronary microvascular adaptation to myocardial metabolic demand can be restored by inhibition of iron‐catalyzed formation of oxygen free radicals in type 2 diabetic patients. Diabetes 51: 813–818, 2002.
 1136. Harrison DG. Cellular and molecular mechanisms of endothelial cell dysfunction. J Clin Invest 100: 2153–2157, 1997.
 1137. Katusic ZS. Superoxide anion and endothelial regulation of arterial tone. Free Rad Biol Med 20: 443–448, 1996.
 1138. Beckman JS and Koppenol WH. Nitric oxide, superoxide, and peroxynitrite: the good, the bad, and ugly. Am J Physiol 271: C1424–C1437, 1996.
 1139. Wei EP, Kontos HA and Beckman JS. Mechanisms of cerebral vasodilation by superoxide, hydrogen peroxide, and peroxynitrite. Am J Physiol (Heart Circ Physiol) 271: H1262–H1266, 1996.
 1140. Didion SP, Ryan MJ, Baumbach GL, Sigmund CD and Faraci FM. Superoxide contributes to vascular dysfunction in mice that express human renin and angiotensinogen. Am J Physiol (Heart Circ Physiol) 283: H1569–H1576, 2002.
 1141. Yada T, Shimokawa H, Hiramatsu O, Kajita T, Shigeto F, Goto M, Ogasawara Y and Kajiya F. Hydrogen peroxide, an endogenous endothelium‐derived hyperpolarizing factor, plays an important role in coronary autoregulation in vivo. Circulation 107: 1040–1045, 2003.
 1142. Shimokawa H and Matoba T. Hydrogen peroxide as an endothelium‐derived hyperpolarizing factor. Pharmacol Res 49: 543–549, 2004.
 1143. Bény J‐L and Von der Weid PY. Hydrogen peroxide: An endogenous smooth muscle cell hyperpolarizing factor. Biochem Biophys Res Commun 176: 378–384, 1991.
 1144. Sobey CG, Heistad DD and Faraci FM. Potassium channels mediate dilatation of cerebral arterioles in response to arachidonate. Am J Physiol (Heart Circ Physiol) 275: H1606–H1612, 1998.
 1145. Pelaez NJ, Braun TR, Paul RJ, Meiss RA and Packer CS. H2O2 mediates Ca2+‐ and MLC20 phosphorylation‐independent contraction in intact and permeabilized vascular muscle. Am J Physiol (Heart Circ Physiol) 279: H1185–H1193, 2000.
 1146. Langberg H, Bjorn C, Boushel R, Hellsten Y and Kjaer M. Exercise‐induced increase in interstitial bradykinin and adenosine concentrations in skeletal muscle and peritendinous tissue in humans. J Physiol 542: 977–983, 2002.
 1147. Sarelius IH, Cohen KD and Murrant CL. Role for capillaries in coupling blood flow with metabolism. Clin Exp Pharmacol Physiol 27: 826–829, 2000.
 1148. Boushel R. Metabolic control of muscle blood flow during exercise in humans. Canadian J Appl Physiol 28: 754–773, 2003.
 1149. Degenring FH. The effects of acidosis and alkalosis on coronary flow and cardiac nucleotide metabolism. Basic Res Cardiol 71: 287–290, 1976.
 1150. Raberger G, Schütz W and Kraupp O. Coronary reactive hyperemia and coronary dilator action of adenosine during normal respiration and hypercapnic acidosis in the dog. Clin Exp Pharmacol Physiol 2: 373–382, 1975.
 1151. Frisbee JC. Regulation of in situ skeletal muscle arteriolar tone: interactions between two parameters. Microcirculation 9: 443–462, 2002.
 1152. Bohlen HG. The microcirculation in hypertension. J Hypertens 7 (Suppl. 4): S117–S124, 1989.
 1153. Stacey DL, Joyner WL and Gilmore JP. Arteriolar reactivity to pressure stimuli in hamsters with renal hypertension. Hypertension 10: 82–92, 1987.
 1154. Falcone JC, Granger HJ and Meininger GA. Enhanced myogenic activation in skeletal muscle arterioles from spontaneously hypertensive rats. Am J Physiol (Heart Circ Physiol) 265: H1847–H1855, 1993.
 1155. Meininger GA, Routh LK and Granger HJ. Autoregulation and vasoconstriction in the intestine during acute renal hypertension. Hypertension 7: 364–373, 1985.
 1156. Schretzenmayr A. Uber Kreislaufregulatorische Vorgange an den groben Arterien bei der Muskelarbeit. Pflügers Arch Eur J Physiol 232: 743–748, 1933.
 1157. Smiesko V, Lang DJ and Johnson PC. Dilator response of rat mesenteric arcading arterioles to increased blood flow velocity. Am J Physiol (Heart Circ Physiol) 257: H1958–H1965, 1989.
 1158. Falcone JC, Kuo L and Meininger GA. Endothelial cell calcium increases during flow‐induced dilation in isolated arterioles. Am J Physiol (Heart Circ Physiol) 264: H653–H659, 1993.
 1159. Koller A and Kaley G. Endothelium regulates skeletal muscle microcirculation by a blood flow velocity‐sensing mechanism. Am J Physiol (Heart Circ Physiol) 258: H916–H920, 1990.
 1160. Griffith TM, Edwards DH, Davies RL and Henderson AH. The role of EDRF in flow distribution: a micorangiographic study of the rabbit isolated ear. Microvasc Res 37: 162–177, 1989.
 1161. Fujii K, Heistad DD and Faraci FM. Flow‐mediated dilatation of the basilar artery in vivo. Circ Res 69: 697–705, 1991.
 1162. Viswanathan M, Rivera O and Short BL. Heat shock protein 90 is involved in pulsatile flow‐induced dilation of rat middle cerebral artery. J Vasc Res 36: 524–527, 1999.
 1163. Miura H, Wachtel RE, Liu Y, Loberiza FR, Jr., Saito T, Miura M and Gutterman DD. Flow‐induced dilation of human coronary arterioles: important role of Ca2+‐activated K+ channels. Circulation 103: 1992–1998, 2001.
 1164. Hull SS, Jr., Kaiser L, Jaffe MD and Sparks HV, Jr. Endothelium‐dependent flow‐induced dilation of canine femoral and saphenous arteries. Blood Vessels 23: 183–198, 1986.
 1165. Lie M, Sejersted OM and Kiil F. Local regulation of vascular cross section during changes in femoral arterial blood flow in dogs. Circ Res 27: 727–737, 1970.
 1166. Melkumyants AM and Balashov SA. Effect of blood viscosity on arterial flow induced dilator response. Cardiovasc Res 24: 165–168, 1990.
 1167. Pohl U, Herlan K, Huang A and Bassenge E. EDRF‐mediated shear‐induced dilation opposes myogenic vasoconstriction in small rabbit arteries. Am J Physiol (Heart Circ Physiol) 261: H2016–H2023, 1991.
 1168. Drexler H, Zeiher AM, Wollschlager H, Meinertz T, Just H and Bonzel T. Row‐dependent coronary artery dilatation in humans. Circulation 80: 466–474, 1989.
 1169. Sinoway LI, Hendrickson C, Davidson WR, Jr., Prophet S and Zelis R. Characteristics of flow‐mediated brachial artery vasodilation in human subjects. Circ Res 64: 32–42, 1989.
 1170. Miura H, Liu Y and Gutterman DD. Human coronary arteriolar dilation to bradykinin depends on membrane hyperpolarization: contribution of nitric oxide and Ca2+‐activated K+ channels. Circulation 99: 3132–3138, 1999.
 1171. Bevan JA, Garcia‐Roldan JL and Joyce EH. Resistance artery tone is influenced independently by pressure and by flow. Blood Vessels 27: 202–207, 1990.
 1172. Bryan RM, Jr., Marrelli SP, Steenberg ML, Schildmeyer LA and Johnson TD. The effects of luminal shear stress on cerebral arteries and arterioles. Am J Physiol (Heart Circ Physiol) 280: H2011–H2022, 2001.
 1173. Rubanyi GM, Romero JC and Vanhoutte PM. Flow‐induced release of endothelium‐derived relaxing factor. Am J Physiol (Heart Circ Physiol) 250: H1145–H1149, 1986.
 1174. Kuo L, Chilian WM and Davis MJ. Interaction of pressure‐and flow‐induced responses in porcine coronary resistance vessels. Am J Physiol (Heart Circ Physiol) 261: H1706–H1715, 1991.
 1175. Holtz J, Forstermann U, Pohl U, Giesler M and Bassenge E. Flow‐dependent, endothelium‐mediated dilation of epicardial coronary arteries in conscious dogs: effects of cyclooxygenase inhibition. Journal of Cardiovascular Pharmacology 6: 1161–1169, 1984.
 1176. Koller A and Huang A. Impaired nitric oxide‐mediated flow‐induced dilation in arterioles of spontaneously hypertensive rats. Circ Res 74: 416–421, 1994.
 1177. Koller A, Sun D, Huang A and Kaley G. Corelease of nitric oxide and prostaglandins mediates flow‐dependent dilation of rat gracilis muscle arterioles. Am J Physiol (Heart Circ Physiol) 267: H326–H332, 1994.
 1178. Bergaya S, Meneton P, Bloch‐Faure M, Mathieu E, Alhenc‐Gelas F, Levy BI and Boulanger CM. Decreased flow‐dependent dilation in carotid arteries of tissue kallikrein‐knockout mice. Circ Res 88: 593–599, 2001.
 1179. Matrougui K, Loufrani L, Heymes C, Levy BI and Henrion D. Activation of AT2 receptors by endogenous angiotensin II is involved in flow‐induced dilation in rat resistance arteries. Hypertension 34: 659–665, 1999.
 1180. Miura H, Bosnjak JJ, Ning G, Saito T, Miura M and Gutterman DD. Role for hydrogen peroxide in flow‐induced dilation of human coronary arterioles. Circ Res 92: 31–40, 2003.
 1181. Laurindo FRM, Pedro MA, Barbeiro HV, Pileggi F, Carvalho MHC, Augusto O and Luz PL. Vascular free radical release: Ex vivo and in vivo evidence for a flow‐dependent endothelial mechanism. Circ Res 74: 700–709, 1994.
 1182. Ingebrigtsen R, Lie M, Hol R, Leraand D and Fonstelien E. Dilation of the ileo‐femoral artery following the opening of an experimental artero‐venous fistula in the dog. Scand J Clin Lab Invest 31: 255–263, 1973.
 1183. Smiesko V, Kozik J and Dolezel S. Role of endothelium in the control of arterial diameter by blood flow. Blood Vessels 22: 247–251, 1985.
 1184. Smiesko V, Khayutin VM, Kozik J and Rogoza AN. Flow‐induced dilation of the dog gracilis muscle artery. Physiol Bohemoslovaca 36: 289–300, 1987.
 1185. Chu A, Chambers DE, Lin C, Kuehl WD, Palmer RMJ, Moncada S and Cobb FR. Effects of inhibition of nitric oxide formation on basal vasomotion and endothelium‐dependent responses of the coronary arteries of awake dogs. J Clin Invest 87: 1964–1968, 1991.
 1186. Kuo L, Davis MJ and Chilian WM. Longitudinal gradients for endothelium‐dependent vascular responses in the coronary microcirculation. Circulation 92: 518–525, 1995.
 1187. Rodbard S. Vascular caliber. Cardiology 60: 4–49, 1975.
 1188. Tesfamariam B and Cohen RA. Inhibition of adrenergic vasoconstriction by endothelial cell shear stress. Circ Res 63: 720–725, 1988.
 1189. Griffith TM, Edwards DH, Davies RL, Harrison TJ and Evans KT. EDRF coordinates the behaviour of vascular resistance vessels. Nature 329: 442–445, 1987.
 1190. Quick CM, Hashimoto T and Young WL. Lack of flow regulation may explain the development of arteriovenous malformations. Neurol Res 23: 641–644, 2001.
 1191. Koller A, Sun D and Kaley G. Role of shear stress and endothelial prostaglandins in flow‐ and viscosity‐induced dilation of arterioles in vitro. Circ Res 12: 1276–1284, 1993.
 1192. Stepp DW, Nishikawa Y and Chilian WM. Regulation of shear stress in the canine coronary microcirculation. Circulation 100: 1555–1561, 1999.
 1193. Liao JC and Kuo L. Interaction between adenosine and flow‐induced dilation in coronary microvascular network. Am J Physiol (Heart Circ Physiol) 272: H1571–H1581, 1997.
 1194. Hester RL and Duling BR. Red cell velocity during functional hyperemia: implications for rheology and oxygen transport. Am J Physiol (Heart Circ Physiol) 255: H236–H244, 1988.
 1195. Chilian WM, Layne SM, Klausner EC, Eastham CL and Marcus ML. Redistribution of coronary microvascular resistance produced by dipyridamole. Am J Physiol (Heart Circ Physiol) 256: H383–H390, 1989.
 1196. Jones CJH, Kuo L, Davis MJ and Chilian WM. Regulation of coronary blood flow: coordination of heterogeneous control mechanisms in vascular microdomains. Cardiovasc Res 29: 585–596, 1995.
 1197. Marcus ML, Chilian WM, Kanatsuka H, Dellsperger KC, Eastham CL and Lamping KG. Understanding the coronary circulation through studies at the microvascular level. Circulation 82: 1–7, 1990.
 1198. Kanatsuka H, Lamping KG, Eastham CL and Marcus ML. Heterogeneous changes in epimyocardial microvascular size during graded coronary stenosis. Circ Res 66: 389–396, 1990.
 1199. Jones CJH, Kuo L, Davis MJ and Chilian WM. The role of nitric oxide in the coronary microvascular responses to adenosine and increased metabolic demand. Circulation 91: 1807–1813, 1995.
 1200. Joyner MJ and Halliwill JR. Neurogenic vasodilation in human skeletal muscle: possible role in contraction‐induced hyperaemia. Acta Physiol Scand 168: 481–488, 2000.
 1201. Thomas GD and Segal SS. Neural control of muscle blood flow during exercise. J Appl Physiol 97: 731–738, 2004.
 1202. Hansen J, Sander M, Hald CF, Victor RG and Thomas GD. Metabolic modulation of sympathetic vasoconstriction in human skeletal muscle: role of tissue hypoxia. J Physiol 527: 387–396, 2000.
 1203. Martin III WH, Tolley TK and Saffitz JE. Autoradiographic delineation of skeletal muscle α1‐adrenergic receptor distribution. Am J Physiol (Heart Circ Physiol) 259: H1402–H1408, 1990.
 1204. Struijker‐Boudier HA. Messing MW and van Essen H. Alpha‐adrenergic reactivity of the microcirculation in conscious spontaneously hypertensive rats. Mol Cell Biochem 157: 239–244, 1996.
 1205. Faber JE. In situ analysis of alpha‐adrenoceptors on arteriolar and venular smooth muscle in rat skeletal muscle microcirculation. Circ Res 62: 37–50, 1988.
 1206. Ohyanagi M, Faber JE and Nishigaki K. Differential activation of α1‐ and α2‐adrenoceptors on microvascular smooth muscle during sympathetic nerve stimulation. Circ Res 68: 232–244, 1991.
 1207. Leech CJ and Faber JE. Different α‐adrenoceptor subtypes mediate constriction of arterioles and venules. Am J Physiol (Heart Circ Physiol) 270: H710–H722, 1996.
 1208. Heusch G, Deussen A, Schipke J and Thamer V. α1 and α2‐adrenoceptor‐mediated vasoconstriction of large and small canine coronary arteries in vivo. J Cardiovasc Pharmacol 6: 961–968, 1984.
 1209. Chilian WM. Functional distribution of α1‐ and α2‐adrenergic receptors in the coronary microcirculation. Circulation 84: 2108–2122, 1991.
 1210. Jones CJH, DeFily DV, Patterson JL and Chilian WM. Endothelium‐dependent relaxation competes with α1‐ and α2‐adrenergic constriction in the canine epicardial coronary microcirculation. Circulation 87: 1264–1274, 1993.
 1211. O'Donnell SR and Wanstall JC. The classification of beta‐adrenoceptors in isolated ring preparations of canine coronary arteries. Br J Pharmacol 81: 637–644, 1984.
 1212. Berkenboom G, Fontaine J, Desmet J‐M and Degre S. Comparison of the effect of beta adrenergic antagonists with different ancillary properties on isolated canine and human coronary arteries. Cardiovasc Res 21: 299–304, 1987.
 1213. Drew GM and Levy GP. Characterization of the coronary vascular‐adrenoceptor in the pig. Br J Pharmacol 46: 348–350, 1972.
 1214. Baron GD, Speden RN and Bohr DF. Beta‐adrenergic receptors in coronary and skeletal muscle arteries. Am J Physiol 223: 878–881, 1972.
 1215. Hein TW, Zhang C, Wang W and Kuo L. Heterogeneous β2‐adrenoceptor expression and dilation in coronary arterioles across the left ventricular wall. Circulation 110: 2708–2712, 2004.
 1216. Murphree SS and Saffitz JE. Delineation of the distribution of beta‐adrenergic receptor subtypes in canine myocardium. Circ Res 63: 117–125, 1988.
 1217. Sun D, Huang A, Mital S, Kichuk MR, Marboe CC, Addonizio LJ, Michler RE, Koller A, Hintze TH and Kaley G. Norepinephrine elicits β2‐receptor‐mediated dilation of isolated human coronary arterioles. Circulation 106: 550–555, 2002.
 1218. Curro FA and Greenberg S. Characteristics of postsynaptic α1 and α2 adrenergic receptors in canine vascular smooth muscle. Can J Physiol Pharmacol 61: 893–904, 1983.
 1219. Medgett IC, Hicks PE and Langer SZ. Effect of acidosis on α1 and α2‐adrenoceptor‐mediated vasoconstrictor responses in isolated arteries. Eur J Pharmacol 135: 443–447, 1987.
 1220. Chen LQ and Shepherd AP. Role of H+ and α2‐receptors in escape from sympathetic vasoconstriction. Am J Physiol (Heart Circ Physiol) 261: H868–H873, 1991.
 1221. Gorman MW, Tune JD, Richmond KN and Feigl EO. Quantitative analysis of feedforward sympathetic coronary vasodilation in exercising dogs. J Appl Physiol 89: 1903–1911, 2000.
 1222. Björnberg J, Maspers M and Mellander S. Metabolic control of large‐bore arterial resistance vessels, arterioles, and veins in cat skeletal muscle during exercise. Acta Physiol Scand 135: 83–94, 1989.
 1223. Ekelund U and Mellander S. Role of endothelium‐derived nitric oxide in the regulation of tonus in large‐bore arterial resistance vessels, arterioles and veins in cat skeletal muscle. Acta Physiol Scand 140: 301–309, 1990.
 1224. Granger HJ. Coordinated microvascular reactions mediated by series‐coupling of metabolic, myogenic and flow‐sensitive resistance elements. FASEB J 3: A1387, 1989.
 1225. Comelissen AJ, Dankelman J, VanBavel E and Spaan JA. Balance between myogenic, flow‐dependent, and metabolic flow control in coronary arterial tree: a model study. Am J Physiol (Heart Circ Physiol) 282: H2224–H2237, 2002.
 1226. Krogh A. Studies on the capillariometer mechanism: I. The reaction to stimuli and the innervation of the blood vessels in the tongue of the frog. J Physiol 53: 399–419, 1920.
 1227. Fleisch A. Les reflexes nutritifs ascendants producteurs de dilatation arterielle. Arch Int Physiol 41: 141, 1935.
 1228. Hilton SM. A peripheral arterial conducting mechanism underlying dilatation of the femoral artery and concerned in functional vasodilatation in skeletal muscle. J Physiol 149: 93–111, 1959.
 1229. Segal SS and Duling BR. Arteriolar dilation propagates in an electronic fashion. Proc Microcircul Soc 1158‐1150, 1986.
 1230. Segal SS and Duling BR. Propagation of vasodilation in resistance vessels of the hamster: development and review of a working hypothesis. Circ Res 61: II‐20–II‐25, 1987.
 1231. Emerson GG and Segal SS. Endothelial cell pathway for conduction of hyperpolarization and vasodilation along hamster feed artery. Circ Res 86: 94–100, 2000.
 1232. Segal SS and Jacobs TL. Role for endothelial cell conduction in ascending vasodilatation and exercise hyperaemia in hamster skeletal muscle. J Physiol 536: 937–946, 2001.
 1233. Takano H, Dora KA and Garland CJ. Spreading vasodilatation in resistance arteries. J Smooth Muscle Res 41: 303–311, 2005.
 1234. Hill MA, Davis MJ and Meininger GA. Cyclooxygenase inhibition potentiates myogenic activity in skeletal muscle arterioles. Am J Physiol (Heart Circ Physiol) 258: H127–H133, 1990.
 1235. Hoepfl B, Rodenwaldt B, Pohl U and de Wit C. EDHF. but not NO or prostaglandins, is critical to evoke a conducted dilation upon ACh in hamster arterioles. Am J Physiol (Heart Circ Physiol) 283: H996–H1004, 2002.
 1236. Rivers RJ, Hein TW, Zhang C and Kuo L. Activation of barium‐sensitive inward rectifier potassium channels mediates remote dilation of coronary arterioles. Circulation 104: 1749–1753, 2001.
 1237. Lin Y and Duling BR. Vulnerability of conducted vasomotor response to ischemia. Am J Physiol (Heart Circ Physiol) 267: H2363–H2370, 1994.
 1238. Kurjiaka DT, Bender SB, Nye DD, Wiehler WB and Welsh DG. Hypertension attenuates cell‐to‐cell communication in hamster retractor muscle feed arteries. Am J Physiol (Heart Circ Physiol) 288: H861–H870, 2005.
 1239. Goto K, Rummery NM, Grayson TH and Hill CE. Attenuation of conducted vasodilatation in rat mesenteric arteries during hypertension: role of inwardly rectifying potassium channels. J Physiol 561: 215–231, 2004.
 1240. Haug SJ, Welsh DG and Segal SS. Sympathetic nerves inhibit conducted vasodilatation along feed arteries during passive stretch of hamster skeletal muscle. J Physiol 552: 273–282, 2003.
 1241. Lutz BR and Fultion GP. The neuromotor mechanism of the small blood vessels in membranes of the frog (Rana Pipiens) and the hamster (Mesocricetus Auratus) with reference to the normal and pathological conditions of blood flow. Exp Med Surg 8: 258–287, 1950.
 1242. Emerson GG, Neild TO and Segal SS. Conduction of hyperpolarization along hamster feed arteries: augmentation by acetylcholine. Am J Physiol (Heart Circ Physiol) 283: H102–H109, 2002.
 1243. Prosser CL. Conduction in nonstriated muscles. Physiol Rev Suppl 5: 193–212, 1962.
 1244. Hirst GDS and Neild TO. An analysis of excitatory junctional potentials recorded from arterioles. J Physiol 280: 87–104, 1978.
 1245. Vonderlage M. Spread of contraction in rabbit ear artery preparations in response to stimualation by norepinephrine. Circ Res 59: 600–608, 1981.
 1246. Emerson GG and Segal SS. Electrical activation of endothelium evokes vasodilation and hyperpolarization along hamster feed arteries. Am J Physiol (Heart Circ Physiol) 280: H160–H167, 2001.
 1247. Koller A and Kaley G. Shear stress‐induced dilation of arterioles. Am J Physiol (Heart Circ Physiol) 274: H382–H383, 1998.
 1248. Segal SS, Welsh DG and Kurjiaka DT. Spread of vasodilatation and vasoconstriction along feed arteries and arterioles of hamster skeletal muscle. J Physiol 516: 283–291, 1999.
 1249. Spray DC. Molecular physiology of gap junction channels. Clin Exp Pharmacol Physiol 23: 1038–1040, 1996.
 1250. Goodenough DA, Goliger JA and Paul DL. Connexins, connexons and intercellular communication. Annu Rev Biochem 65: 475–502, 1996.
 1251. Little TL, Beyer EC and Duling BR. Connexin 43 and connexin 40 gap junctional proteins are present in arteriolar smooth muscle and endothelium in vivo. Am J Physiol (Heart Circ Physiol) 268: H729–H739, 1995.
 1252. Mather S, Dora KA, Sandow SL, Winter P and Garland CJ. Rapid endothelial cell‐selective loading of connexin 40 antibody blocks endothelium‐derived hyperpolarizing factor dilation in rat small mesenteric arteries. Circ Res 97: 399–407, 2005.
 1253. Bartlett IS and Segal SS. Resolution of smooth muscle and endothelial pathways for conduction along hamster cheek pouch arterioles. Am J Physiol (Heart Circ Physiol) 278: H604–H612, 2000.
 1254. Figueroa X, Chen CC, Campbell KP, Damon DN, Day KH, Ramos S and Duling BR. Are voltage‐dependent ion channels involved in the endothelial cell control of vasomotor tone? Am J Physiol (Heart Circ Physiol) 293 (3): H1371–H1383, 2007.
 1255. de Wit C, Roos F, Bolz SS, Kirchhoff S, Krüger O, Willecke K and Pohl U. Impaired conduction of vasodilation along arterioles in connexin40‐deficient mice. Circ Res 86: 649–655, 2000.
 1256. Figueroa XF, Paul DL, Simon AM, Goodenough DA, Day KH, Damon DN and Duling BR. Central role of connexin40 in the propagation of electrically activated vasodilation in mouse cremasteric arterioles in vivo. Circ Res 92: 793–800, 2003.
 1257. Welsh DG and Segal SS. Endothelial and smooth muscle cell conduction in arterioles controlling blood flow. Am J Physiol 274: H178–H186, 1998.
 1258. Xia J and Duling BR. Electromechanical coupling and the conducted vasomotor response. Am J Physiol 269: H2022–H2030, 1995.
 1259. Xia J, Little TL and Duling BR. Cellular pathways of the conducted electrical response in arterioles of hamster cheek pouch in vitro. Am J Physiol (Heart Circ Physiol) 269: H2031–H2038, 1995.
 1260. Bény J‐L and Chabaud F. Kinins and endothelium‐dependent hyperpolarization in porcine coronary arteries. In: Endothelium‐Derived Hyperpolarizing Factor, ed. Vanhoutte PM. Amsterdam: Harwood Academic, 1996, pp. 41–49.
 1261. de Wit C. Connexins pave the way for vascular communication. News Physiol Sci 19: 148–153, 2004.
 1262. Tran CH and Welsh DG. The mechanistic basis of cell‐cell communication in resistance arterioles. Am J Physiol (Heart Circ Physiol) in preparation 2008.
 1263. Bény J‐L and Connat JL. An electron‐microscopic study of smooth muscle cell dye coupling in the pig coronary arteries: Role of gap junctions. Circ Res 70: 49–55, 1992.
 1264. Yamamoto Y, Klemm MF, Edwards FR and Suzuki H. Intercellular electrical communication among smooth muscle and endothelial cells in guinea‐pig mesenteric arterioles. J Physiol 535: 181–195, 2001.
 1265. Bény J‐L. Zhu PL and Haefliger IO. Lack of bradykinin‐induced smooth muscle cell hyperpolarization despite heterocellular dye coupling and endothelial cell hyperpolarization in porcine ciliary artery. J Vasc Res 34: 344–350, 1997.
 1266. Kumer SC, Damon DN and Duling BR. Patterns of conducted vasomotor response in the mouse. Microvasc Res 59: 310–315, 2000.
 1267. Diep HK, Vigmond EJ, Segal SS and Welsh DG. Defining electrical communication in skeletal muscle resistance arteries: A computational approach. J Physiol 568: 267–281, 2005.
 1268. Bény J‐L. Electrical coupling between smooth muscle cells and endothelial cells in pig coronary arteries. Pflügers Arch Eur J Physiol 433: 364–367, 1997.
 1269. Segal SS and Neild TO. Conducted depolarization in arteriole networks of the guinea‐pig small intestine: Effect of branching on signal dissipation. J Physiol 496: 229–244, 1996.
 1270. Takano H, Dora KA, Spitaler MM and Garland CJ. Spreading dilatation in rat mesenteric arteries associated with calcium‐independent endothelial cell hyperpolarization. J Physiol 556: 887–903, 2004.
 1271. Jantzi MC, Brett SE, Jackson WF, Corteling R, Vigmond EJ and Welsh DG. Inward rectifying potassium channels facilitate cell‐to‐cell communication in hamster retractor muscle feed arteries. Am J Physiol (Heart Circ Physiol) 291: H1319–H1328, 2006.
 1272. Sharma NR and Davis MJ. Mechanism of substance P‐induced hyperpolarization of porcine coronary artery endothelial cells. Am J Physiol (Heart Circ Physiol) 266: H156–H164, 1994.
 1273. Schilling WP, Cabello OA and Rajan L. Depletion of the inositol 1,4,5‐trisphosphate‐sensitive intracellular Ca2+ store in vascular endothelial cells activates the agonist‐sensitive Ca2+‐influx pathway. Biochem J 284: 521–530, 1992.
 1274. Crane GJ, Gallagher N, Dora KA and Garland CJ. Small‐ and intermediate‐conductance calcium‐activated K+ channels provide different facets of endothelium‐dependent hyperpolarization in rat mesenteric artery. J Physiol 553: 183–189, 2003.
 1275. Vargas FF, Caviedes PF and Grant DS. Electrophysiological characteristics of cultured human umbilical vein endothelial cells. Microvasc Res 47: 153–165, 1994.
 1276. Bossu JL, Feltz A, Rodeau JL and Tanzi F. Voltage‐dependent transient calcium currents in freshly dissociated capillary endothelial cells. FEBS Lett 255: 377–380, 1989.
 1277. Bossu JL, Elhamdani A, Feltz A, Tanzi F, Aunis D and Thierse D. Voltage‐gated Ca2+ entry in isolated bovine capillary endothelial cells: instance of a new type of BAY K 8644‐sensitive channel. Pflügers Arch Eur J Physiol 420: 200–207, 1992.
 1278. Wu S, Haynes J, Jr., Taylor JT, Obiako BO, Stubbs JR, Li M and Stevens T. Cav3.1 (alpha1G) T‐type Ca2+ channels mediate vaso‐occlusion of sickled erythrocytes in lung microcirculation. Circ Res 93: 346–353, 2003.
 1279. Duza T and Sarelius IH. Localized transient increases in endothelial cell Ca2+ in arterioles in situ: implications for coordination of vascular function. Am J Physiol (Heart Circ Physiol) 286: H2322–H2331, 2004.
 1280. Domeier TL and Segal SS. Electromechanical and pharmacomechanical signalling pathways for conducted vasodilatation along endothelium of hamster feed arteries. J Physiol 579: 175–186, 2007.
 1281. Uhrenholt TR, Domeier TL and Segal SS. Propagation of calcium waves along endothelium of hamster feed arteries. Am J Physiol (Heart Circul Physiol) 292: H1634–H1640, 2007.
 1282. Tallini YN, Brekke JF, Shui B, Doran R, Hwang SM, Nakai J, Salama G, Segal SS and Kotlikoff MI. Propagated endothelial Ca2+ waves and arteriolar dilation in vivo: measurements in Cx40BAC GCaMP2 transgenic mice. Circ Res 101: 1300–1309, 2007.
 1283. Girard S and Clapham D. Acceleration of intracellular calcium waves in Xenopus oocytes by calcium influx. Science 260: 229–232, 1993.
 1284. Domenighetti AA, Bény J‐L, Chabaud F and Frieden M. An intercellular regenerative calcium wave in porcine coronary artery endothelial cells in primary culture. J Physiol 513: 103–116, 1998.
 1285. Clapham DE. Calcium signaling. Cell 80: 259–268, 1995.
 1286. Clapham DE. Intracellular calcium: Replenishing the stores. Nature 375: 634–635, 1995.
 1287. Segal SS. Microvascular recruitment in hamster striated muscle: a role for conducted vasodilation. Am J Physiol (Heart Circ Physiol): H181–H189, 1991.
 1288. Segal SS. Regulation of blood flow in the microcirculation. Microcirculation 12: 33–45, 2005.
 1289. Johnson PC. Myogenic nature of increase in intestinal vascular resistance with venous pressure elevation. Circ Res 7: 992–999, 1959.
 1290. Johnson PC, Hanson KM and Thulesius O. Pre‐ and postcapillary resistance in the dog forelimb. Am J Physiol 210: 873–876, 1966.
 1291. Silver IA. Cellular microenvironment in relation to local blood flow. Ciba Found Symp 49‐67, 1978.
 1292. Johnson PC. Effect of venous pressure on mean capillary pressure and vascular resistance in the intestine. Circ Res 16: 294–300, 1965.
 1293. Hanson KM and Johnson PC. Evidence for local arteriovenous reflex in intestine. J Appl Physiol 17: 509–513, 1962.
 1294. Nurkiewicz TR and Boegehold MA. Limitation of arteriolar myogenic activity by local nitric oxide: Segment‐specific effect of dietary salt. Am J Physiol (Heart Circ Physiol) 277: H1946–H1955, 1999.
 1295. Davis MJ, Gilmore JP and Joyner WL. Responses of pulmonary allograft and cheek pouch arterioles in the hamster to alterations in extravascular pressure in different oxygen environments. Circ Res 49: 133–140, 1981.
 1296. Griffith TM and Edwards DH. Myogenic autoregulation of flow may be inversely related to endothelium‐derived relaxing factor activity. Am J Physiol (Heart Circ Physiol) 258: H1171–H1180, 1990.
 1297. Bevan JA and Joyce EH. Flow‐induced resistance artery tone: Balance between constrictor and dilator mechanisms. Am J Physiol (Heart Circ Physiol) 258: H663–H668, 1990.
 1298. Garcia‐Roldan JL and Bevan J. Flow‐induced constriction and dilation of cerebral resistance arteries. Circ Res 66: 1445–1448, 1990.
 1299. New DI, Chesser AM, Thuraisingham RC and Yaqoob MM. Cerebral artery responses to pressure and flow in uremic hypertensive and spontaneously hypertensive rats. Am J Physiol (Heart Circ Physiol) 284: H1212–H1216, 2003.
 1300. Madden JA and Christman NJ. Integrin signaling, free radicals, and tyrosine kinase mediate flow constriction in isolated cerebral arteries. Am J Physiol (Heart Circ Physiol) 277: H2264–H2271, 1999.
 1301. Shimoda LA, Norins NA and Madden JA. Responses to pulsatile flow in piglet isolated cerebral arteries. Pediatr Res 43: 514–520, 1998.
 1302. Juncos LA. Garvin J, Carretero OA and Ito S. Flow modulates myogenic responses in isolated microperfused rabbit afferent arterioles via endothelium‐derived nitric oxide. J Clin Invest 95: 2741–2748, 1995.
 1303. Pohl U, Lamontagne D, Bassenge E and Busse R. Attenuation of coronary autoregulation in the isolated rabbit heart by endothelium derived nitric oxide. Cardiovasc Res 28: 414–419, 1994.
 1304. Ekelund U, Bjornberg J, Grande PO, Albert U and Mellander S. Myogenic vascular regulation in skeletal muscle in vivo is not dependent of endothelium‐derived nitric oxide. Acta Physiol Stand 144: 199–207, 1992.
 1305. de Wit C, Jahrbeck B, Schafer C. Bolz SS and Pohl U. Nitric oxide opposes myogenic pressure responses predominantly in large arterioles in vivo. Hypertension 31: 787–794, 1998.
 1306. Faber JE and Meininger GA. Selective interaction of α‐adrenoceptors with myogenic regulation of microvascular smooth muscle. Am J Physiol (Heart Circ Physiol) 259: H1126–H1133, 1990.
 1307. Ikeoka K, Nishigaki K, Ohyanagi M and Faber JE. In vitro analysis of α‐adrenoceptor interactions with the myogenic response in resistance vessels. J Vasc Res 29: 313–321, 1992.
 1308. Liu J, Hill MA and Meininger GA. Mechanisms of myogenic enhancement by norepinephrine. Am J Physiol (Heart Circ Physiol) 266: H440–H446, 1994.
 1309. Nilsson H, Ljung B, Sjoblom N and Wallin BG. The influence of the sympathetic impulse pattern on contractile responses of rat mesenteric arteries and veins. Acta Physiol Scand 123: 303–309, 1985.
 1310. Porsa E, Harper MJ and Herlihy JT. Effects of preload and eicosanoid synthesis inhibition on rat aortic smooth muscle sensitivity. Prostaglan Leukotrienes Essen Fatty Acid 38: 61–65, 1989.
 1311. Tallarida RJ, Sevy RW, Harakal C, Bendrick J and Faust R. The effect of preload on the dissociation constant of norepinephrine in isolated strips of rabbit thoracic aorta. Arch Int Pharmacodyn Therap 210: 67–74, 1974.
 1312. Dunn WR, Wellman GC and Bevan JA. Enhanced resistance artery sensitivity to agonists under isobaric compared with isometric conditions. Am J Physiol (Heart Circ Physiol) 266: H147–H155, 1994.
 1313. Henriksen O, Amtorp O, Faris I and Agerskov K. Evidence for a local sympathetic venoarteriolar “reflex” in the dog hindleg. Circ Res 52: 534–542, 1983.
 1314. Hassan AA and Tooke JE. Mechanism of the postural vasoconstrictor response in the human foot. Clin Sci (London) 75: 379–387, 1988.
 1315. Ping P and Johnson PC. Role of myogenic response in enhancing autoregulation of flow during sympathetic nerve stimulation. Am J Physiol (Heart Circ Physiol) 263: H1177–H1184, 1992.
 1316. Ping P and Johnson PC. Mechanism of enhanced myogenic response in arterioles during sympathetic nerve stimulation. Am J Physiol (Heart Circ Physiol) 263: H1185–H1189, 1992.
 1317. Kurjiaka DT and Segal SS. Autoregulation during pressor response elevates wall shear rate in arterioles. J Appl Physiol 80: 598–604, 1996.
 1318. Mellander S. Comparative studies on the adrenergic neuro‐hormonal control of resistance and capancitance blood vessels in the cat. Acta Physiol Scand 50: 3–86, 1960.
 1319. Boegehold MA and Johnson PC. Response of arteriolar network of skeletal muscle to sympathetic nerve stimulation. Am J Physiol (Heart Circ Physiol) 254: H919–H928, 1988.
 1320. Hillman J. Further studies on beta‐adrenergic control of transcapillary fluid aborption from skeletal muscle to blood during hemorrhage. Acta Physiol Scand 112: 281–286, 1981.
 1321. Hillman J. Beta 2‐adrenergic control of transcapillary fluid absorption and plasma volume in hemorrhage. Acta Physiol Scand 516: 1–62, 1983.
 1322. Björnberg J. Forces involved in transcapillary fluid movement in exercising cat skeletal muscle. Acta Physiol Scand 140: 221–236, 1990.
 1323. Cryer HM, Kaebnick H, Harris PD and Flint LM. Effects of tissue acidosis on skeletal muscle microcirculatory responses to hemorrhagic shock in unanesthetized rats. J Surg Res 39: 59–67, 1985.
 1324. Arslan E, Sierko E, Waters JH and Siemionow M. Microcirculatory hemodynamics after acute blood loss followed by fresh and banked blood transfusion. Am J Surg 190: 456–462, 2005.
 1325. Amundson B, Jennische E and Haljamae H. Correlative analysis of microcirculatory and cellular metabolic events in skeletal muscle during hemorrhagic shock. Acta Physiol Scand 108: 147–158, 1980.
 1326. Luchette FA, Jenkins WA, Friend LA, Su C, Fischer JE and James JH. Hypoxia is not the sole cause of lactate production during shock. J Trauma 52: 415–419, 2002.
 1327. Lemieux MD, Smith RN and Couch NP. Electrometric surface pH of skeletal muscle in hypovolemia. Am J Surg 117: 627–631, 1969.
 1328. Taylor JH, Beilman GJ, Conroy MJ, Mulier KE, Myers D, Gruessner A and Hammer BE. Tissue energetics as measured by nuclear magnetic resonance spectroscopy during hemorrhagic shock. Shock 21: 58–64, 2004.
 1329. Lundgren O, Lundwall J and Mellander S. Range of sympathetic discharge and reflex vascular adjustments in skeletal muscle during hemorrhagic hypotension. Acta Physiol Scand 62: 380–390, 1964.
 1330. Zhang R, Zuckerman JH, Iwasaki K, Wilson TE, Crandall CG and Levine BD. Autonomic neural control of dynamic cerebral autoregulation in humans. Circulation 106: 1814–1820, 2002.
 1331. Ide K, Boushel R, Sorensen HM, Fernandes A, Cai Y, Pott F and Secher NH. Middle cerebral artery blood velocity during exercise with beta‐1 adrenergic and unilateral stellate ganglion blockade in humans. Acta Physiol Scand 170: 33–38, 2000.
 1332. Jones CE, Farrell TA and Ator R. Evidence that coronary alpha‐adrenergic lone limits myocardial blood flow and oxygenation in acute hemorrhagic hypotension. Circul Shock 11: 329–340, 1983.
 1333. Birinyi F, Hackel DB and Mikat E. Effects of alpha‐adrenergic blockade on coronary blood flow of dogs in hemorrhagic shock. Circul Shock 4: 297–303, 1977.
 1334. Adachi T, Hori S, Miyazaki K, Nakagawa M, Inoue S, Ohnishi Y, Nakazawa H, Aikawa N and Ogawa S. Inhibition of nitric oxide synthesis aggravates myocardial ischemia in hemorrhagic shock in constant pressure model. Shock 9: 204–209, 1998.
 1335. Bond RF, Manning ES, Gonzalez NM, Gonzalez RR, Jr. and Becker VE. Myocardial and skeletal muscle responses to hemorrhage and shock during adrenergic blockade. Am J Physiol 225: 247–257, 1973.
 1336. Tateishi J and Faber JE. ATP‐sensitive K+ channels mediate α2D‐adrenergic receptor contraction of arteriolar smooth muscle and reversal of contraction by hypoxia. Circ Res 76: 53–63, 1995.
 1337. Loutzenhiser RD and Parker MJ. Hypoxia inhibits myogenic reactivity of renal afferent arterioles by activating ATP‐sensitive K+ channels. Circ Res 74: 861–869, 1994.
 1338. Gschwend S, Henning RH, de Zeeuw D and Buikema H. Coronary myogenic constriction antagonizes EDHF‐mediated dilation: role of KCa channels. Hypertension 41: 912–918, 2003.
 1339. Farouque HM, Worthley SG and Meredith IT. Effect of ATP‐sensitive potassium channel inhibition on coronary metabolic vasodilation in humans. Arterioscl Thromb Vasc Biol 24: 905–910, 2004.
 1340. Imamura Y, Tomoike H, Narishige T, Takahashi T, Kasuya H and Takeshita A. Glibenclamide decreases basal coronary blood flow in anesthetized dogs. Am J Physiol (Heart Circ Physiol) 263: H399–H404, 1992.
 1341. Mori H, Chujo M, Tanaka E, Yamakawa A, Shinozaki Y, Mohamed MU and Nakazawa H. Modulation of adrenergic coronary vasoconstriction via ATP‐sensitive potassium channel. Am J Physiol (Heart Circ Physiol) 268: H1077–H1085, 1995.
 1342. Samaha FF, Heineman FW, Ince C, Fleming J and Balaban RS. ATP‐sensitive potassium channel is essential to maintain basal coronary vascular tone in vivo. Am J Physiol (Cell Physiol) 262: C1220–C1227, 1992.
 1343. Zhang C, Hein TW and Kuo L. Transmural difference in coronary arteriolar dilation to adenosine: Effect of luminal pressure and KATP channels. Am J Physiol (Heart Circ Physiol) 279: H2612–H2619, 2000.
 1344. Faraci FM and Heistad DD. Regulation of the cerebral circulation: Role of endothelium and potassium channels. Physiological Rev 78: 53–97, 1998.
 1345. Toyoda K, Fujii K, Ibayashi S, Kitazono T, Nagao T and Fujishima M. Role of ATP‐sensitive potassium channels in brain stem circulation during hypotension. Am J Physiol (Heart Circ Physiol) 213: H1342–H1346, 1997.
 1346. Rosenblum WI. ATP‐sensitive potassium channels in the cerebral circulation. Stroke 34: 1547–1552, 2003.
 1347. Mironneau J and Macrez‐Lepretre N. Modulation of Ca2+ channels by α1A‐ and α2A‐adrenoceptors in vascular myocytes: involvement of different transduction pathways. Cell Signal 7: 471–479, 1995.
 1348. Adams DJ and Hill MA. Potassium channels and membrane potential in the modulation of intracellular calcium in vascular endothelial cells. J Cardiovasc Electrophysiol 15: 598–610, 2004.
 1349. Keller DM, Ogoh S, Greene S, Olivencia‐Yurvati A and Raven PB. Inhibition of KATP channel activity augments baroreflex‐mediated vasoconstriction in exercising human skeletal muscle. J Physiol 561: 273–282, 2004.
 1350. Thomas GD, Hansen J and Victor RG. ATP‐sensitive potassium channels mediate contraction‐induced attenuation of sympathetic vasoconstriction in rat skeletal muscle. J Clin Invest 99: 2602–2609, 1997.
 1351. Strandell T and Shepherd JT. The effect in humans of increased sympathetic activity on the blood flow to active muscles. Acta Med Scand Suppl 472: 146–167, 1967.
 1352. Remensnyder JP, Mitchell JH and Sarnoff SJ. Functional sympatholysis during muscular activity. Circ Res 11: 370–380, 1962.
 1353. Rowlands DJ and Donald DE. Sympathetic vasoconstrictive responses during exercise‐ or drug‐induced vasodilatation. A time‐dependent response. Circ Res 23: 45–60, 1968.
 1354. Burcher E and Garlick D. Antagonism of vasoconstrictor responses by exercise in the gracilis muscle of the dog. J Pharmacol Exp Therapeut 187: 78–85, 1973.
 1355. Anderson KM and Faber JE. Differential sensitivity of arteriolar alpha1‐ and alpha2‐adrenoceptor constriction to metabolic inhibition during rat skeletal muscle contraction. Circ Res 69: 174–184, 1991.
 1356. McGillivray‐Anderson KM and Faber JE. Effect of reduced blood flow on alpha1‐ and alpha2‐adrenoceptor constriction of rat skeletal muscle microvessels. Circ Res 69: 165–173, 1991.
 1357. Muldowney SM and Faber JE. Preservation of venular but not arteriolar smooth muscle alpha‐adrenoceptor sensitivity during redused blood flow. Circ Res 69: 1215–1225, 1991.
 1358. Thomas GD, Hansen J and Victor RG. Inhibition of α2‐adrenergic vasoconstriction during contraction of glycolytic, not oxidative, rat hindlimb muscle. Am J Physiol (Heart Circ Physiol) 266: H920–H929, 1994.
 1359. DeFily DV, Patterson JL and Chilian WM. Endogenous adenosine modulates α2‐ but not α1‐adrenergic constriction of coronary arterioles. Am J Physiol (Heart Circ Physiol) 268: H2487–H2494, 1995.
 1360. Nishigaki K, Faber JE and Ohyanagi M. Interactions between alpha‐adrenoceptors and adenosine receptors on microvascular smooth muscle. Am J Physiol (Heart Circ Physiol) 260: H1655–H1666, 1991.
 1361. Hansen J, Thomas GD, Harris SA, Parsons WJ and Victor RG. Differential sympathetic neural control of oxygenation in resting and exercising human skeletal muscle. J Clin Invest 98: 584–596, 1996.
 1362. Hutcheson IR and Griffith TM. Heterogeneous populations of K+ channels mediate EDRF release to flow but not agonists in rabbit aorta. Am J Physiol (Heart Circ Physiol) 266: H590–H596, 1994.
 1363. Cooke JP, Rossitch E, Jr., Andon NA, Loscaizo J and Dzau VJ. Flow activates an endothelial potassium channel to release an endogenous nitrovasodilator. J Clin Invest 88: 1663–1671, 1991.
 1364. Hein TW, Liao JC and Kuo L. oxLDL specifically impairs endothelium‐dependent, NO‐mediated dilation of coronary arterioles. Am J Physiol (Heart Circ Physiol) 278: H175–H183, 2000.
 1365. Sun D, Huang A, Koller A and Kaley G. Endothelial KCa channels mediate flow‐dependent dilation of arterioles of skeletal muscle and mesentery. Microvasc Res 61: 179–186, 2001.
 1366. Lückhoff A and Busse R. Activators of potassium channels enhance calcium influx into endothelial cells as a consequence of potassium currents. Naunyn‐Schmiedebergs Arch Pharmacol 342: 94–99, 1990.
 1367. Schnitzler MM, Derst C, Daut J and Preisig‐Muller R. ATP‐sensitive potassium channels in capillaries isolated from guinea‐pig heart. J Physiol 525: 307–317, 2000.
 1368. Langheinrich U, Mederos y Schnitzler M and Daut J. Ca2+‐transients induced by K+ channel openers in isolated coronary capillaries. Pflügers Arch Eur J Physiol 435: 435–438, 1998.
 1369. Frisbee JC and Lombard JH. Elevated oxygen tension inhibits flow‐induced dilation of skeletal muscle arterioles. Microvasc Res 58: 99–107, 1999.
 1370. Cábel M, Smiesko V and Johnson PC. Attenuation of blood flow‐induced dilation in arterioles after muscle contraction. Am J Physiol (Heart Circ Physiol) 266: H2114–H2121, 1994.
 1371. Keung EC and Li Q. Lactate activates ATP‐sensitive potassium channels in guinea pig ventricular myocytes. J Clin Invest 88: 1772–1777, 1991.
 1372. Bouchard JF, Dumont E and Lamontagne D. Evidence that prostaglandins I2, E2, and D2 may activate ATP sensitive potassium channels in the isolated rat heart. Cardiovasc Res 28: 901–905, 1994.
 1373. Randall MD. The involvement of ATP‐sensitive potassium channels and adenosine in the regulation of coronary flow in the isolated perfused rat heart. Br J Pharmacol 116: 3068–3074, 1995.
 1374. Goto K, Fujii K, Abe I and Fujishima M. Sympathetic control of arterial membrane potential by ATP‐sensitive K+‐channels. Hypertension 35: 379–384, 2000.
 1375. Kitazono T, Heistad DD and Faraci FM. Role of ATP‐sensitive K+ channels in CGRP‐induced dilatation of basilar artery in vivo. Am J Physiol (Heart Circ Physiol) 265: H581–H585, 1993.
 1376. Narishige T, Egashira K, Akatsuka Y, Imamura Y, Takahashi T, Kasuya H and Takeshita A. Glibenclamide prevents coronary vasodilation induced by β1‐adrenoceptor stimulation in dogs. Am J Physiol (Heart Circ Physiol) 266: H84–H92, 1994.
 1377. Chang HY. The involvement of ATP‐sensitive potassium channels in β2‐adrenoceptor agonist‐induced vasodilatation on rat diaphragmatic microcirculation. Br J Pharmacol 121: 1024–1030, 1997.
 1378. Borst HG, McGregor M, Whittenberger JL and Berglund E. Influence of pulmonary arterial and left atrial pressures on pulmonary vascular resistance. Circ Res 4: 393–399, 1956.
 1379. Liu Y, Harder DR and Lombard JH. Myogenic activation of canine small renal arteries after nonchemical removal of the endothelium. Am J Physiol (Heart Circ Physiol) 267: H302–H307, 1994.
 1380. Davis MJ and Meininger GA. The myogenic response in microvascular networks. In: Mechanotransduction by the Vascular Wall, ed. Rubanyi G, Mt. Kisco, New York: Futura Publishing Company, 1993, pp. 37–60.
 1381. McSherry IN, Sandow SL, Campbell WB, Falck JR, Hill MA and Dora KA. A role for heterocellular coupling and EETs in dilation of rat cremaster arterioles. Microcirculation 13: 119–130, 2006.
 1382. Hill MA, Davis MJ, Meininger GA, Potocnik SJ and Murphy TV. Arteriolar myogenic signalling mechanisms: Implications for local vascular function. Clin Hemorheol Microcircul 34: 67–79, 2006.
 1383. Kuo L, Davis MJ and Chilian WM. Endothelial modulation of arteriolar tone. News Physiol Sci 7: 5–9, 1992.

Related Articles:

Microvascular Consequences of Obesity and Diabetes

Contact Editor

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

* Required Field

How to Cite

Michael J Davis, Michael A Hill, Lih Kuo. Local Regulation of Microvascular Perfusion. Compr Physiol 2011, Supplement 9: Handbook of Physiology, The Cardiovascular System, Microcirculation: 161-284. First published in print 2008. doi: 10.1002/cphy.cp020406