Abstract
Bradykinin-induced activation of the pulmonary endothelium triggers nitric oxide production and other signals that cause vasorelaxation, including stimulation of large-conductance Ca2+-activated K+ (BKCa) channels in myocytes that hyperpolarize the plasma membrane and decrease intracellular Ca2+. Intrauterine chronic hypoxia (CH) may reduce vasorelaxation in the fetal-to-newborn transition and contribute to pulmonary hypertension of the newborn. Thus we examined the effects of maturation and CH on the role of BKCa channels during bradykinin-induced vasorelaxation by examining endothelial Ca2+ signals, wire myography, and Western immunoblots on pulmonary arteries isolated from near-term fetal (∼140 days gestation) and newborn, 10- to 20-day-old, sheep that lived in normoxia at 700 m or in CH at high altitude (3,801 m) for >100 days. CH enhanced bradykinin-induced relaxation of fetal vessels but decreased relaxation in newborns. Endothelial Ca2+ responses decreased with maturation but increased with CH. Bradykinin-dependent relaxation was sensitive to 100 μM nitro-l-arginine methyl ester or 10 μM 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, supporting roles for endothelial nitric oxide synthase and soluble guanylate cyclase activation. Indomethacin blocked relaxation in CH vessels, suggesting upregulation of PLA2 pathways. BKCa channel inhibition with 1 mM tetraethylammonium reduced bradykinin-induced vasorelaxation in the normoxic newborn and fetal CH vessels. Maturation reduced whole cell BKCa channel α1-subunit expression but increased β1-subunit expression. These results suggest that CH amplifies the contribution of BKCa channels to bradykinin-induced vasorelaxation in fetal sheep but stunts further development of this vasodilatory pathway in newborns. This involves complex changes in multiple components of the bradykinin-signaling axes.
Keywords: potassium channels, sheep, pulmonary artery, contractility, maturation, hypoxia
regulation of smooth muscle tone in pulmonary arteries during development is a delicate balance of vasoconstrictive and vasorelaxant pathways. Endothelial cells play a crucial role in determining the overall level of vasorelaxation (39, 67), and endothelium-dependent relaxation is partially mediated through bradykinin stimulation (31). Bradykinin is a potent vasodilator that is important in the fetal pulmonary circulation, as well as during inflammation, and its relationship to pulmonary hypertension has been explored (5, 31, 83).
Endothelial bradykinin receptor activation induces vasorelaxation through modulation of several different intracellular signaling pathways that are largely dependent on a rise of endothelial intracellular Ca2+ ([Ca2+]i) (67). The most widely studied pathway is bradykinin-induced activation of endothelial nitric oxide (NO) synthase (eNOS), an enzyme that generates NO (64). NO acts on nearby smooth muscle cells to cause downstream stimulation of soluble guanylate cyclase (sGC) pathways that leads to vasorelaxation (3, 45). Previous studies have shown that regulation of vessel relaxation through various receptor signaling systems is altered during pre- and postnatal development, as well as following prenatal chronic hypoxia, which imposes a significant stress on the fetus (9, 10, 35). Evidence suggests that acetylcholine (ACh)-dependent endothelium-mediated relaxation of the pulmonary vasculature is reduced in the fetus relative to the adult (57). Prenatal chronic hypoxia further suppresses ACh-induced relaxation in the fetus, but this is not due to changes in eNOS expression (91). The suppression of ACh-dependent relaxation maintains high pulmonary vascular resistance, which restricts blood flow, which, in turn, is important because the lung is not yet required for gas exchange. However, during the transition at birth from fetus to newborn, the pulmonary vessels dilate rapidly, increasing blood flow to the alveoli, and allow for proper gas exchange in the newborn lung.
Chronic hypoxia is a known risk factor in the development of pulmonary hypertension (67). It can significantly enhance vasoconstriction as well as reduce vasodilatory capacity. Subsequently, these effects elevate pulmonary pressure, which can result in pulmonary hypertension. The risk of pulmonary hypertension is especially prominent among newborns exposed to chronic hypoxia in utero due to pregnancy at high altitude, placental insufficiency, smoking, maternal anemia, or other causes (67). Persistent pulmonary hypertension of the newborn due to hypoxia or other etiologies is an incapacitating disease that can lead to failure of the ductus arteriosus to close, resulting in severe systemic hypoxia. Unfortunately, there are few treatment options and no cures (67). Numerous studies have indicated that endothelium-derived relaxing factors, especially those associated with NO, are crucial in the modifications associated with loss of vasodilatory capacity and development of pulmonary hypertension (1, 30, 48).
Our knowledge regarding the influence of prenatal chronic hypoxia on endothelium-dependent relaxation is limited. The available evidence indicates that there is enhanced eNOS expression (41, 59), reduced sGC (41), and CO-mediated relaxation (41, 59, 60) but enhanced large-conductance Ca2+-activated K+ (BKCa) channel function (42). In fetal lamb, eNOS expression is unchanged (91), but PKG function is enhanced and cGMP function is reduced (26). The diversity in the dysfunctions and compensations associated with high-altitude gestation have led us to design a series of studies to test how prenatal chronic hypoxia affects early postnatal bradykinin-induced vasorelaxation. We tested the specific hypothesis that prenatal chronic hypoxia impairs the normal development of relaxation through eNOS-dependent pathways. This hypothesis was tested in studies on arteries isolated from full-term fetal and newborn sheep housed at low or high altitude, allowing for direct comparative analyses.
METHODS
Experimental animals.
Experimental procedures were performed on sheep arteries, because the developmental progression of ovine lungs and the extent to which prenatal chronic hypoxia affects them are comparable to humans (67). The studies were performed within the regulations of the Animal Welfare Act, the National Institutes of Health Guide for the Care and Use of Laboratory Animals, “The Guiding Principles in the Care and Use of Animals” approved by the Council of the American Physiological Society, and the Animal Care and Use Committee of Loma Linda University (LLU). The tissue preparation, wire myography, and imaging experimental procedures and protocols are based on previously published methodology (10, 35, 37). Sheep were obtained from Nebeker Ranch (Lancaster, CA; 720 m altitude), and normoxic animals were brought to LLU [353 m altitude, 95 ± 5 Torr arterial Po2 (PaO2)] for experimental study. Animals in the chronic hypoxic experimental group were acclimatized to high altitude (3,801 m, 60 ± 5 Torr PaO2) at the Barcroft Laboratory, White Mountain Research Station (Bishop, CA) for ∼110 days (35, 37). The hypoxic ewes were then transported to LLU for experimental study. To maintain hypoxic conditions in pregnant sheep, a tracheal catheter was placed in the ewe shortly after the animal arrived at LLU. The tracheal catheter allowed N2 to reach the animal at a rate adjusted to maintain PaO2 at ∼60 Torr, equivalent to the PaO2 at the White Mountain Research Station (47). This PaO2 was maintained until the time of the experimental study. Ewes in the normoxic and chronic hypoxic groups were allowed to give birth naturally and transported to LLU Animal Care when the lambs were 8–17 days of age, as described previously (10). After arrival and until study, 1–2 days later, hypoxic animals were kept in a chamber supplied with 14–16% O2 that simulated the altitude at the Barcroft Laboratory. Both groups of lambs were studied between 10 and 20 days after birth.
For the fetal studies, within 1–5 days of arriving at LLU, pregnant sheep were euthanized with an overdose of Euthasol [pentobarbital sodium (100 mg/kg) and phenytoin sodium (10 mg/kg); Virbac, Ft. Worth, TX], the proprietary euthanasia solution. Lungs were removed from the fetuses and used immediately for contractility and imaging experiments. Care was taken during arterial isolation and wire mounting to ensure that the endothelium was not disrupted.
Tissue preparation.
As described elsewhere, fourth- to fifth-branch-order pulmonary arteries with internal diameters of ∼500–700 μm were isolated from full-term fetal (138–141 days) or newborn (10–21 days old) sheep from the different experimental groups (10, 35, 37). The parenchyma was removed carefully from the pulmonary arteries for contractility studies. The arteries were then cut into 5-mm-long rings in ice-cold phosphate-free balanced salt solution (BSS) of the following composition (in mM): 126 NaCl, 5 KCl, 10 HEPES, 1 MgCl2, 2 CaCl2, and 10 glucose, with pH adjusted to 7.4 with NaOH. Imaging studies and the contraction studies in which arteries were treated with the NO donor N-nitrosoproline (ProliNO), which releases NO in a 1:2 ProliNO-to-NO molar ratio with a dissociation half-life of ∼2 s at physiological pH and temperature (76), were performed in BSS. All other contraction studies were performed with a modified Krebs-Henseleit (K-H) solution containing (in mM) 120 NaCl, 4.8 KCl, 1.2 K2HPO4, 25 NaHCO3, 1.2 MgCl2, 2.5 CaCl2, and 10 glucose. To depolarize the arteries, NaCl was omitted from the BSS or K-H solution and replaced with equimolar KCl.
Contraction studies.
Wire-mounted pulmonary arterial rings were suspended in organ baths (Radnoti Glass Instruments, Monrovia, CA) containing 5 or 10 ml of modified K-H solution or BSS maintained at 37°C. Arteries in modified K-H solution were aerated with 95% O2-5% CO2 (pH 7.4), while unaerated HEPES-buffered BSS was used for the ProliNO studies to prevent sparge-induced loss of free NO into the atmosphere. Each ring was suspended between two tungsten wires passed through the lumen: one wire was anchored to the glass hook at the bottom of the organ chamber, while the other was connected to a tissue hook attached to a low-compliance force transducer (Radnoti Glass Instruments) that measured isometric force (10, 35, 37). The transducers were connected to an analog-to-digital data interface (Powerlab 16/30, A/D Instruments, Colorado Springs, CO; or MP100, Biopac, Goleta, CA) attached to a computer. Changes in tension were recorded using Chart 5.5 or 7.0 (AD Instruments) or AcqKnowledge 3.9 (Biopac), and the data were stored on digital media for later analysis.
At the beginning of each experiment, vessels were equilibrated without tension for ≥30 min. Tension of ∼0.75 g was then applied to each vessel and vessel tension was allowed to stabilize, as previously described (10, 35, 37). To allow for comparative evaluation of smooth muscle contraction and relaxation, isolated pulmonary arterial rings bathed in modified K-H solution were stimulated with 125 mM KCl (high-K) to cause membrane depolarization and activate L-type Ca2+ channels (35). In some experiments, the tension was normalized to a control response obtained with high K. To evaluate dose-response relaxation characteristics, bradykinin was used to relax the arteries following precontraction with 10 μM phenylephrine (PE). In these experiments, the tension was normalized to the maximum 10 μM PE-precontracted tension (%T10 μM PE).
Confocal microscopy studies.
[Ca2+]i was measured in pulmonary arterial endothelial cells in situ with the Ca2+-sensitive dye fluo 4-AM (Invitrogen, Carlsbad, CA) using a laser scanning confocal imaging workstation (model 710 NLO, Zeiss, Thornwood, NY) with an inverted microscope (Zeiss Axio Observer) according to procedures based on those previously described (35, 37). Fluo 4-AM was dissolved in DMSO and added from a 1 mM stock solution to the arterial suspension at a final fluo 4 concentration of 10 μM, along with 0.1% Pluronic F-127, for 1 h at room temperature in darkness in BSS. Arterial segments were then washed with BSS for 30 min to allow for dye esterification and cut into linear strips. The arterial segments were pinned to Sylgard (Ellsworth Adhesives, Germantown, WI) and placed in an open-bath imaging chamber (Warner Instruments, Hamden, CT) mounted on the confocal imaging stage. Cells were illuminated at 488 nm with a krypton-argon laser, and the emitted light was collected using a photomultiplier tube with a band-limited spectral grating in the range 493–622 nm, with full-frame images obtained every 700 ms. To ensure that the endothelial [Ca2+]i was recorded, the pinhole was adjusted to provide an imaging depth of 5.4 μm, and the sample was focused above the internal elastic lamina layer, which has significant autofluorescence when excited at 488 nm in this preparation. This depth is substantially deeper than an individual endothelial cell, on the basis of morphological examination of fixed and live preparations (data not shown), and accounts for sample ruffling, thus allowing for the examination of many more endothelial cells than otherwise would have been achieved. Images were acquired at a 12-bit sampling depth with use of a water-immersion ×40 Plan-Apochromat, 1.0 numerical aperture objective mounted on an InverterScope objective inverter (LSM Tech, Wellsville, PA), which allowed for imaging in an upright configuration. To prevent arterial movement during recordings, arteries were pretreated for 1 h with a cocktail including 10 μM Y27632 to inhibit Rho kinase, 10 μM cytochalasin D to inhibit actin polymerization, and 10 μM W-7 to inhibit myosin light chain kinase (10, 43, 56, 88). The concentrations of these drugs were chosen on the basis of the outcomes of wire myography studies where the influence of the drugs on vessel reactivity were examined individually. Regions of interest were detected automatically post hoc using the LC Pro plug-in for ImageJ (22). For presentation purposes, the fractional fluorescence intensity was automatically calculated using LC Pro.
Background was subtracted from measurements as follows: F/F0 = F − baseline/F0 − baseline, where baseline is the intensity from a region of interest with no cells, F is the fluorescence intensity for the region of interest, and F0 is the fluorescence intensity during a period when there was no Ca2+ activity as determined automatically. False positives were excised from the final data set by their relationship to the timing of the application of bradykinin and by visual analysis of the data.
Western immunoblot studies.
To generate whole cell lysates, pulmonary arteries from fetal and newborn sheep were harvested, cleaned, and rapid-frozen in liquid nitrogen and stored at −80°C. Tissues were then homogenized using glass-on-glass in a RIPA extraction buffer containing 10 mM DTT and a protease inhibitor cocktail (catalog no. M1745, Sigma-Aldrich, St. Louis, MO). Samples were centrifuged at 5,000 g for 20 min, and supernatants were collected and analyzed using SDS-PAGE along with reference control samples from pregnant adult sheep middle cerebral arteries. Separated proteins were transferred to nitrocellulose membranes at 200 mA for 90 min in Towbin buffer (25 mM Tris, 192 mM glycine, and 20% methanol). Membranes were blocked using 5% milk in Tris-buffered saline at pH 7.45 for 1 h at room temperature with continuous shaking. Primary antibodies were incubated for 12 h at 4°C using the following dilutions: 300:1 for BK-α (catalog no. APC-021, Alomone, Jerusalem, Israel) and 300:1 for BK-β1 (catalog no. APC-036, Alomone) (44). For visualization, membranes were incubated for 90 min with a secondary antibody conjugated to DyLight 800 (catalog no. 46422, Pierce Chemical, Rockford, IL). Subsequently, membranes were stripped and reprobed using antibodies against β-actin [monoclonal anti-β-actin produced in mouse, clone AC-74 (catalog no. A2228, Sigma-Aldrich)] as a loading control. Anti-β-actin was diluted 1:5,000 and incubated for 90 min in 5% milk in Tris-buffered saline with 0.1% Tween 20. Membranes were imaged on an infrared imaging system (Odyssey, LI-COR), and individual protein bands were quantified using Image Studio software (LI-COR). Protein abundance was normalized using β-actin as a loading control and expressed as relative abundance compared with an arbitrary reference standard (pooled whole cell lysate obtained from pregnant adult sheep middle cerebral arteries).
Chemicals and drugs.
Most reagents and chemicals were purchased from Sigma-Aldrich; bradykinin, Y-27632, and W-7 were purchased from Tocris (Minneapolis, MN).
Statistical methods and sampling.
All time-series recordings were graphed with IGOR Pro 6.0 (Wavemetrics, Lake Oswego, OR), and summarized data are presented as means ± SE. Statistical analyses were made using Prism 5.0 (GraphPad, La Jolla, CA). Data were evaluated for normality prior to comparative statistical analysis. Dose-response curves were fitted in Prism 5.0 using the Hill equation with nonlinear curve fit analysis (10, 35). A total of 605 arterial segments from 61 sheep were tested for contractility. The Ca2+ imaging studies were performed on tissues from six fetal normoxic, four fetal hypoxic, three newborn normoxic, and four newborn hypoxic sheep. Western blot analysis was performed on tissues from nine fetal normoxic sheep and six animals in each of the other three groups. P < 0.05 was accepted as statistically significant, unless otherwise noted.
RESULTS
Endothelial cells of fetal sheep pulmonary arteries.
The first set of studies imaged the endothelium of intact, isolated, ovine pulmonary arteries to demonstrate that our arterial isolation techniques do not disrupt the endothelial layer (Fig. 1). The images of Fig. 1 are important, as Figs. 2–10 show the findings of studies designed to evaluate functional responses of the endothelium in pulmonary arteries from fetal and newborn sheep housed at low or high altitude. Representative images in Fig. 1 illustrate that the endothelial cells have an ellipsoid shape, which is presumed to be along the direction of blood flow, and that the cells are closely associated with one another. No quantitative measurements of endothelial cell shape or structure were performed in the present studies.
Prenatal chronic hypoxia and bradykinin-mediated relaxation.
The first series of functional studies was designed to determine the extent to which bradykinin relaxes pulmonary arteries and the extent to which this is altered by prenatal chronic hypoxia. These studies were based on the general premise that prenatal chronic hypoxia impedes relaxation (20, 21, 87). This proposition was investigated by examining dose-dependent bradykinin-mediated relaxation of 10 μM PE-precontracted vessels. Dose-response curves of the data from these studies and summary results are shown in Fig. 2. Figure 2, A and B, shows that prenatal chronic hypoxia enhances the bradykinin-induced vasorelaxant response in arteries from the fetus but that relaxation is blunted in vessels from the chronic hypoxic newborn. Figure 2C and Table 1 illustrate that the potency is enhanced by postnatal maturity but unaffected by high-altitude chronic hypoxia. Figure 2D and Table 2 show that continued life at high altitude impairs the normal developmental increase in the maximum response to bradykinin, in that relaxation is equivalent to that of vessels from the hypoxic fetus.
Table 1.
Normoxia |
Hypoxia |
|||
---|---|---|---|---|
Fetus | Newborn | Fetus | Newborn | |
Control | −6.86 ± 0.21 (14) | −7.89 ± 0.16* (6) | −6.95 ± 0.15 (14) | −7.91 ± 0.12* (16) |
l-NAME | −6.93 ± 1.09 (5) | −8.43 ± 0.26 (4) | −6.83 ± 0.53 (6) | −7.61 ± 0.43 (7) |
Indo | −6.38 ± 0.78 (5) | −7.89 ± 0.15 (4) | −6.87 ± 0.74 (3) | −7.04 ± 0.38 (4) |
ODQ | −6.89 ± 0.84 (4) | −7.89 ± 0.36 (3) | −6.51 ± 0.51 (3) | −7.86 ± 0.67 (4) |
TEA | −6.44 ± 0.90 (5) | −7.65 ± 0.24 (4) | −7.05 ± 0.32 (3) | −7.68 ± 0.23 (4) |
Values (means ± SE) are shown as log concentration for the IC50 for bradykinin-induced relaxation for data presented graphically in Figs. 2, 5–7, and 9; number of animals is shown in parentheses. l-NAME, NG-nitro-l-arginine methyl ester; Indo, indomethacin; ODQ, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one; TEA, tetraethylammonium. Statistical significance as shown using a 95% confidence interval based on the log (inhibitor) vs. response curve fit to the data:
significant leftward shift compared with Fetus on the basis of control traces in Fig. 2.
Table 2.
Normoxia |
Hypoxia |
|||
---|---|---|---|---|
Fetus | Newborn | Fetus | Newborn | |
Control | 83 ± 2 (14) | 11 ± 4* (6) | 47 ± 3‡ (14) | 47 ± 2‡ (16) |
l-NAME | 91 ± 5 (5) | 42 ± 4† (4) | 79 ± 5† (6) | 76 ± 3†‡ (7) |
Indo | 85 ± 10 (5) | 19 ± 5 (4) | 76 ± 12† (3) | 66 ± 7†‡ (4) |
ODQ | 91 ± 5 (4) | 44 ± 6† (3) | 79 ± 8† (3) | 73 ± 5†‡ (4) |
TEA | 78 ± 14 (5) | 36 ± 5† (4) | 73 ± 5† (3) | 40 ± 5 (4) |
Values (means ± SE) are shown as percentage of tension normalized to control response obtained in high-K solution for phenylephrine-induced contraction for data presented graphically in Figs. 2, 5–7, and 9; number of animals is shown in parentheses. Statistical significance as shown using a 95% confidence interval based on the log (inhibitor) vs. response curve fit to the data:
significant increase in maximum response compared with Fetus,
significant depression in maximum response compared with Control,
significant difference in maximum response compared with Normoxia counterpart.
Prenatal chronic hypoxia and endothelial Ca2+ signals.
Bradykinin-induced relaxation is well regarded as being related to Ca2+-dependent signaling in endothelial cells (78). Because of the influence of development and chronic hypoxia on bradykinin-induced relaxation of pulmonary vessels presented in Fig. 2, we examined the potential that prenatal chronic hypoxia enhances bradykinin-induced Ca2+ signals in fetal endothelial cells but restricts these signals in the newborn. Figure 3 shows Ca2+ responses in fetal endothelial cells recorded before and during administration of 1 μM bradykinin in the presence of extracellular Ca2+. Figure 3A shows in situ fluorescence images of endothelial cells in the arterial wall isolated from a prenatal chronic hypoxic fetal sheep for various times, as shown for the fluorescence intensity trace in Fig. 3B. Bradykinin rapidly increased cytosolic Ca2+ in the pulmonary arterial endothelial cells, and these signals decayed slowly back to baseline. Qualitatively, the increase in Ca2+ was similar for all experimental groups examined; thus other representative images are not provided.
The magnitude and kinetics of these fluorescence Ca2+ signals were then quantified. Figure 4 provides summaries of the quantitative analysis for various aspects of the Ca2+ responses. These data demonstrate that the amplitude (Fig. 4A) increases modestly after birth in arterial endothelial cells from control animals. However, prenatal chronic hypoxia enhances the amplitude of the Ca2+ signal in the fetus, but not the newborn. The area under the curve (Fig. 4B), event duration (Fig. 4C), and decay (Fig. 4D) decreased following birth but increased somewhat by chronic hypoxia in the newborn. The duration of Ca2+ rise (attack, Fig. 4E) became shorter after birth and following prenatal chronic hypoxia in cells from the fetus. Compared with the vasorelaxant influences of bradykinin, the data suggest that postnatal maturation enhances Ca2+ sensitivity, while chronic hypoxia augments Ca2+ signals, in neonatal endothelial cells but suppresses postnatal sensitivity.
Prenatal chronic hypoxia and eNOS-dependent relaxation.
Given the uncoupling between endothelial Ca2+ signals and vasorelaxation following prenatal chronic hypoxia and postnatal maturation, the next series of studies tested the hypothesis that altered bradykinin-induced eNOS activation mediates this detachment. To evaluate this process, NO synthesis was inhibited with 100 μM NG-nitro-l-arginine methyl ester (l-NAME) (71). Figure 5 shows that l-NAME successfully reduced bradykinin-induced vasorelaxation in 10 μM PE-precontracted vessels from the hypoxic fetus as well as from normoxic and hypoxic newborns. In addition to the increased sensitivity of fetal vessels to l-NAME following hypoxia, hypoxic fetal vessels displayed greater reductions in overall vasorelaxation. The summary data in Table 1 show that the potency of bradykinin-induced relaxation was unaffected by l-NAME. Table 2 demonstrates that the maximum response was not affected by eNOS inhibition in normoxic fetus but was significantly reduced in the normoxic and hypoxic newborn and the hypoxic fetus. Before birth, prenatal chronic hypoxia increased the role for eNOS, but the change in maximum response was virtually identical to that of hypoxic newborns. Overall, these findings suggest that prenatal chronic hypoxia accelerates development of eNOS-dependent vasorelaxation in the fetus, possibly involving sGC, PKG, or other downstream components of the pathway, but this acceleration restrains the normal development of pulmonary vascular relaxation to bradykinin.
Prenatal chronic hypoxia and prostacyclin-dependent vasorelaxation.
Prostacyclin is released by the endothelium and is another significant signaling arm activated by endothelial bradykinin stimulation (11). To test the role of prostacyclin in bradykinin-induced vasorelaxation, 10 μM indomethacin (Indo) was added to inhibit cyclooxygenase (COX)-dependent prostacyclin production. At this concentration, Indo inhibits COX-1 and -2 activity (61, 66). Figure 6 shows dose-dependent relaxation of 10 μM PE-precontracted vessels to bradykinin in the presence of 10 μM indomethacin. As illustrated in Fig. 6, A and B, COX inhibition attenuated the maximal vasorelaxation response in arteries from fetuses following prenatal chronic hypoxia, with no effect on arteries from normoxic fetuses. Figure 6C shows that the relaxation to bradykinin in normoxic newborn arteries was largely unaffected by COX inhibition, although PE vasoreactivity was enhanced. This compares with the data shown in Fig. 6D for hypoxic newborns, which were far more sensitive to Indo. Table 1 shows that COX inhibition failed to alter potency in all groups, but Table 2 shows that normoxic arteries were resistant to Indo, while prenatal chronic hypoxia unmasked prostacyclin-dependent relaxation in arteries from the fetus and the newborn.
Prenatal chronic hypoxia and sGC relaxation.
sGC is activated by NO in smooth muscle downstream of eNOS stimulation (4). Such activation leads to cGMP formation with subsequent vasorelaxation. To establish a more complete picture of the impact of prenatal chronic hypoxia on the role of sGC, we inhibited this enzyme with 10 μM 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) prior to endothelial stimulation with bradykinin (29). Figure 7 shows the dose-dependent vasorelaxation to bradykinin in the presence and absence of ODQ. Figure 7, B–D, shows that sGC inhibition suppresses bradykinin-mediated relaxation in the normoxic and hypoxic newborn and the hypoxic fetus, while the meager relaxation in the normoxic fetus was relatively unaffected (Fig. 7A). As summarized in Table 1, ODQ treatment did not affect potency in arteries from fetal or newborn normoxic or hypoxic sheep. However, Table 2 shows that the maximum response was reduced in all groups, except normoxic fetus.
The results of the l-NAME and ODQ studies illustrate that eNOS and sGC are important to bradykinin-dependent relaxation. It is possible that chronic hypoxia accelerates their development in utero but impedes normal development of these or other pathway components after birth. Nonetheless, previous evidence indicates that eNOS expression is unaffected by prenatal chronic hypoxia in our fetal sheep (91). Because of these findings, we performed a series of experiments to examine the direct impact of NO on vessel reactivity. In particular, we evaluated the potential for NO to directly stimulate sGC activity by addition of the NO donor ProliNO (76). Figure 8 shows results from 10 μM 5-HT-preconstricted vessels relaxed with ProliNO in the presence and absence of the sGC inhibitor ODQ, where 5-HT was used because it induces robust contraction (35). Overall, ProliNO effectively relaxed 5-HT-precontracted vessels from normoxic and prenatal chronic hypoxic fetal and newborn sheep (Fig. 8A). sGC inhibition effectively reduced the potency in all groups and reduced the maximum response of ProliNO-induced relaxation in the hypoxic fetus.
BKCa channels and bradykinin relaxation.
K+ channels are critical for establishing myocyte resting membrane potential and are frequent targets of endogenous and therapeutic vasodilators (52). One primary target of NO-dependent signaling is the BKCa channel (79). The BKCa channel is stimulated by ryanodine receptor-dependent Ca2+ sparks and independently modulated by PKA and PKG downstream of sGC and cGMP (3, 74, 77, 79). To establish the role of BKCa channels in bradykinin-mediated vasorelaxation, arteries were treated with tetraethylammonium (TEA) at 1 mM, a concentration that blocks roughly one-half of the current due to BKCa channels (44, 46). Figure 9 shows the dose-dependent relaxation of PE-precontracted vessels to bradykinin in the presence and absence of 1 mM TEA. BKCa channel inhibition with TEA had no significant effect on bradykinin-induced vasorelaxation in normoxic fetal and hypoxic newborn vessels, as shown in Fig. 9, A and D, respectively. However, Fig. 9B shows that 1 mM TEA impaired vasorelaxation mediated by 1 μM bradykinin in arteries from prenatal chronic hypoxic fetuses, while inhibition of the BKCa channel had no effect on bradykinin-induced relaxation in arteries from hypoxic newborns (Fig. 9D). BKCa channel inhibition weakened bradykinin-induced relaxation in the normoxic newborn (Fig. 9C), indicating a substantial developmental increase in the role of the BKCa channel in bradykinin relaxation. Table 1 shows that 1 mM TEA failed to affect the potency of bradykinin-induced relaxation in any of the groups. Table 2 shows that BKCa channel inhibition reduced the maximum response in the prenatal chronic hypoxic fetus and the normoxic newborn. Although chronic prenatal hypoxia promoted BKCa-related relaxation in the fetus, it suppressed the normal development of BKCa-mediated relaxation in the newborn.
BKCa channel protein expression.
It has been well established that the BKCa channel is a crucial regulator of myogenic tone and blood pressure and is especially important to O2-induced pulmonary arterial relaxation at birth (12, 70, 86). The BKCa channel is made up of four pore-forming α-subunits to which accessory β-subunits can attach (51, 63). Four classes of β-subunits have been identified, although there may be more (62, 84). These accessory β-subunits can regulate voltage and Ca2+ sensitivity to specialize BKCa channel-opening behavior, with the β1-subunit being the most common β-subunit in vascular smooth muscle cells (13, 63). In its absence, mouse arterial tone and blood pressure have been shown to increase due to a reduction in BKCa channel Ca2+ sensitivity and coupling to Ca2+ sparks (69). Furthermore, other studies have shown that the activity of the BKCa channel is developmentally regulated and is differentially affected by Po2 (73). To determine whether the increase in relaxation in fetal arteries following prenatal chronic hypoxia was mediated by a change in BKCa channel expression, Western immunoblot analysis was conducted for the α- and β1-subunits. Figure 10 shows the results of this analysis. As shown in Fig. 10B, maturation of the fetus to the newborn significantly decreased α-subunit expression under normoxic conditions. However, α-subunit expression did not decrease further following prenatal chronic hypoxia. Figure 10, C and D, shows that maturation significantly increased β1-subunit expression, as well as the ratio of β1- to α-subunit expression, regardless of oxygenation status.
DISCUSSION
These studies are the first to examine the influence of prenatal chronic hypoxia on bradykinin-mediated pulmonary arterial relaxation of the fetus and the newborn and to elucidate the roles of important underlying signaling pathways. A schematic summary of these findings and the relationship to our previous work is presented in Fig. 11 (37). In the pulmonary arterial endothelium, we examined the importance of Ca2+ elevations, eNOS, and COX signaling. In smooth muscle, we evaluated subsequent activation of sGC and downstream influences on BKCa channels. Our studies demonstrate that prenatal chronic hypoxia accentuates relaxation in the fetal pulmonary vasculature through enhanced pathway development, but this elevation leads to impediments following birth that ultimately impair pulmonary vascular relaxation in the newborn.
Prenatal chronic hypoxia enhanced bradykinin-mediated pulmonary arterial relaxation in the fetus. This was a surprise, because chronic hypoxia generally increases pulmonary arterial pressure and depresses pulmonary arterial vasodilatory capacity, as was observed in the newborn (Fig. 2D) and in previous studies (40). The findings suggest that fetal sheep adapt in utero to compensate for eventual birth in a high-altitude, low-O2 environment. On the basis of this line of reasoning, the resultant increase in blood flow at birth in high-altitude-adapted fetuses with heightened bradykinin-mediated arterial relaxation would enhance O2 diffusion from the alveoli into the bloodstream. O2 uptake would be improved when the newborn breathes, allowing the animal to thrive during the fetal-to-newborn transition.
This theoretical cause-and-effect relationship was not seen in high-altitude newborn sheep 2 wk after birth. Rather, the normal course of the development of pulmonary vasorelaxation was restricted (Fig. 2D). This loss could potentially contribute to the elevation in pulmonary pressures and increases in pulmonary vascular resistance that we and others have documented in lambs born at high altitude (10, 40). Furthermore, it is possible that the prenatal acceleration of vasorelaxation responses to bradykinin is redistributing blood flow from developing vital organs to the lung, resulting in blood flow distribution issues. Such an alteration of distribution following chronic hypoxia was previously reported in studies on our fetal sheep (47). These studies showed that blood flow to the fetal brain and heart was maintained, despite a significant decline in cardiac output and reduction of flow throughout the body. This preservation of perfusion is indicative of the autoregulatory nature of blood flow to the brain and heart. If chronic prenatal hypoxia also decreases fetal pulmonary vascular resistance and reallocates blood flow to the lung, these effects could, jointly, place additional stress on other vital organ systems that potentially compromises their function and growth.
The improvement in bradykinin potency following birth could be due to various modifications in the bradykinin signaling pathway. On the basis of classical pharmacological principles, there could be changes in receptor density, ligand affinity, or receptor coupling. Changes in bradykinin receptor expression likely underlie the enhancements and reductions in the maximal relaxation response, but not the effects on potency. The modifications in potency more likely reflect improved receptor affinity or coupling of the receptor to intracellular signaling pathways. Receptor glycosylation can influence receptor occupancy and ligand binding affinity (53). Another possibility is that changes in expression of the angiotensin (AT) type 2 (AT2) receptor can accentuate the potency of bradykinin (7, 92). Indeed, in the mouse lung, AT2 receptor mRNA expression increases following antenatal hypoxia, while AT1 receptor expression remains stable (34). This finding mirrors the protein expression changes with ontogeny, where AT2 receptor expression increases from the neonatal mouse to the adult, while AT1 receptor expression is steady (25). Another alternative that influences receptor activation is that there could be alterations in tyrosine or serine/threonine phosphorylation, which are important to bradykinin receptor desensitization (53). Ontogeny also could lead to modifications in cell signaling pathways downstream of bradykinin receptor activation. The reduction in Ca2+ signaling supports this premise and illustrates that ontogeny enhances Ca2+ sensitization of eNOS. However, there also may be changes in MAPK, JAK/STAT, or other signaling systems, such as changes in receptor acylation, which influences G protein receptor coupling (53). Although it is quite plausible that one or more of these options are involved in the changes in bradykinin potency, it was beyond the scope of the present studies to evaluate this in depth. Nonetheless, such changes are important, as they likely facilitate vessel relaxation and are, therefore, important during the transition to air-breathing.
NO signaling is well regarded as being vital to pulmonary vasorelaxation (17, 38, 64, 67). Thus the finding that eNOS was important to bradykinin-induced vasorelaxation was expected. Although the data are not wholly conclusive, the prospect that eNOS inhibition has a greater effect on the arteries of fetal prenatal chronic hypoxic sheep than on the arteries of normoxic fetal sheep is intriguing. These enhancements in eNOS-dependent vasorelaxation are potentially related to the increased endothelial Ca2+ signaling observed in prenatal chronic hypoxic vessels and concomitant enhancement in Ca2+-dependent activation of eNOS (36, 64).
The discovery that bradykinin Ca2+ signals in the endothelium were suppressed following birth, although relaxation was enhanced, is provocative. These data suggest that, after birth, eNOS is more readily activated by Ca2+. The improvement of Ca2+-eNOS coupling can be due to a number of processes (17, 36, 82). Potentially, amino acid residues that sensitize eNOS to calmodulin, as well as Ca2+, are phosphorylated (36). Binding partners may have greater interaction, or there may be changes in the abundances of cofactors such as l-arginine, flavin adenine dinucleotide, flavin mononucleotide, and tetrahydrobiopterin that facilitate NO production directly or indirectly (23, 64, 82). These changes could also include alterations in spatial-temporal coupling based on the characteristics of the Ca2+ signals and eNOS cellular distribution (22, 64). Indeed, there was a faster rate of Ca2+ rise following chronic hypoxia that paralleled the changes due to ontogeny. In addition, after birth, the decay of Ca2+ was more rapid, an effect that was blunted by hypoxia. These findings, coupled with the multifaceted changes in the Ca2+ spike amplitude and area under the curve, suggest that there are complex changes in release, influx, and sequestration, as well as extrusion. This includes modifications in the expression or activation of inositol trisphosphate receptors or Ca2+-permeable ion channels at the plasma membrane, extrusion of Ca2+ from the cytosol by plasma membrane or endoplasmic reticulum Ca2+-ATPases, or even storage of Ca2+ in the endoplasmic reticulum (32, 33). Resolution of the developmental mechanisms that govern increased bradykinin-mediated relaxation through eNOS-Ca2+ coupling and the influence of chronic hypoxia on the developmental process are important but require additional experimentation.
The finding that sGC and eNOS antagonism caused equivalent inhibition of bradykinin-mediated relaxation provides additional evidence that a substantial portion of the bradykinin relaxation is through coupling of eNOS and sGC. Furthermore, the inhibitory studies suggest that the fidelity of coupling between eNOS and sGC is not modified by maturation or chronic hypoxia. Nonetheless, ProliNO greatly intensified and normalized the extent of relaxation across all groups (Fig. 8). Interestingly, sGC inhibition shifted the potency of ProliNO as opposed to reducing the maximal relaxation to bradykinin shown in Fig. 7. Although such differences in how sGC inhibition influences bradykinin vs. ProliNO relaxation are unlikely, they may reflect nonspecific effects of ODQ that attenuate the bradykinin-induced vasodilation pathway at points upstream of sGC activation (6, 29, 80, 85). The differences in the actions of ODQ also suggest that while a major portion of bradykinin-induced relaxation is through cGMP generation, its mechanism of action may also involve NO degradation through phosphodiesterases or NO-independent pathways (6, 85). The ProliNO data further illustrate that bradykinin does not maximally activate relaxation pathways and that there is substantial NO-sensitive and sGC-insensitive reserve. This sGC-insensitive reserve likely includes protein nitrosylation, which is a key mediator of NO-dependent signaling (55).
Although prostacyclin signaling pathways were not fully evaluated, our indomethacin studies were revealing. The lack of a role for COX-dependent vasorelaxation in the fetal normoxic studies was expected and follows from earlier studies that illustrate that prostacyclins have little effect on pulmonary vascular tone until after birth (54). The failure of indomethacin to substantially reduce bradykinin-dependent relaxation in newborns was interesting and compares with previous studies. In studies performed in newborn sheep as well as goats, direct stimulation with prostacyclin agonists and precursors elicits pulmonary vascular vasorelaxation (15, 28). One explanation is that bradykinin does not fully activate resident prostacyclin synthesis pathways. The finding that chronic hypoxia augmented indomethacin-sensitive bradykinin-induced relaxation was also unexpected, as previous work in piglets showed that prostacyclin production is suppressed (20, 21). Resolving whether greater indomethacin sensitivity is due to improved activation of dormant prostacyclin synthesis pathways or whether there are phenotypic changes that increase the expression of key synthesis enzymes or pathway elements in the arterial myocytes that govern prostacyclin-dependent vasorelaxation is worthy of future investigation (20, 21, 28).
The suppression of the NOS-independent component to bradykinin-induced vasodilation following hypoxia in the newborn is intriguing. Restriction of prostacyclin-dependent signaling is unlikely, because our indomethacin data suggest that PLA2-dependent mechanisms are upregulated following chronic hypoxia. However, chronic hypoxia may suppress the function of other NO synthase-independent pathways important to vasodilation not described in this report, including activation of stored forms of NO (16) and epoxyeicosatrienoic acids (14), as well as myoendothelial gap junctions (24, 50). Resolution of which, if either, of these or other potential ionic or nonionic pathways are suppressed by chronic hypoxia requires additional studies.
Bradykinin-induced relaxation in porcine pulmonary resistance arteries is dependent on the activity of multiple K+ channels (5). In particular, BKCa channels in vascular smooth muscle are important to fetal bradykinin-induced pulmonary arterial relaxation. Previous work has also shown that BKCa channels are important to O2-dependent relaxation in fetal ovine pulmonary arteries (65, 70). Our findings are consistent with these reports and suggest that prenatal chronic hypoxia augments the role for BKCa channels in fetal sheep (Fig. 9B), which may provide a distinct advantage for the lamb during birth. Nonetheless, whatever temporary advantage this increased role for BKCa channels may provide during the fetal-to-newborn transition is followed quickly by impaired development of normal BKCa channel-related vasorelaxation 2 wk after birth (Fig. 9D).
Impediments in BKCa channel function are often caused by chronic hypoxia-induced channel dysfunction or changes in subunit expression. In the present study we inhibited BKCa channel activity in myography studies and conducted Western immunoblots to investigate the possibility that BKCa channel expression parallels the changes in BKCa channel-mediated relaxation (Fig. 9). Such a correlation would indicate that the augmented role for BKCa channels in the fetus and the attenuation of normal BKCa relaxation in the newborn are brought on by hypoxia-induced changes in channel expression, as we observed in the uterine arteries of pregnant ewes (44). However, the tension and expression studies did not agree with one another. The lack of change in α- or β1-subunit expression following chronic hypoxia in the fetus or newborn (Fig. 10) was surprising. We expected to see increases in α- and β1-subunit expression in the prenatal chronic hypoxic fetus to explain the increase in vasorelaxation (Fig. 9B). Indeed, previous studies have shown such a correlation between BKCa channel function and expression, where BKCa channels in cerebral arteries and aorta of spontaneously hypertensive rat arteries undergo a decrease in Ca2+ sensitivity due to a disproportionate expression of α- and β1-subunits, either by downregulation of the β1-subunit or upregulation of the α-subunit (2, 58). Furthermore, a study of fetal and adult cultured ovine pulmonary arterial smooth muscle cells showed that short-term hypoxia increased expression of the α- and β1-subunits (72). The discrepancies between our results and those of previous studies point to a number of possible explanations. The implication is that sustained, intrauterine low-O2 conditions have a unique effect on the function and expression of the BKCa channel compared with the effects seen following induced, short-term hypoxia. However, we have yet to explore activation of whole cell or transient BKCa currents by membrane voltage or the coupling to Ca2+ sparks, which are vital to how BKCa channel-induced membrane hyperpolarization induces vessel relaxation (12, 37).
The increase in BKCa channel activity following prenatal chronic hypoxia in the fetus may also be explained by changes in expression or posttranslational modifications that increase BKCa channel function. Studies also illustrate that single-nucleotide polymorphisms within the α- or β1-subunit can confer a gain or loss of function (8, 19). It follows that “gain-of-function” alternative splice variants of the α- and β1-subunits could be activated during hypoxic stress, thereby conferring an increase in Ca2+ sensitivity and vasorelaxation. The location of the subunits is also vital to channel function. For example, in addition to the plasma membrane, BKCa channels are also expressed in the mitochondrial membrane of neurons, and this can influence cell excitability (75, 90). Unfortunately, our Western immunoblot analysis only measured whole cell protein content and did not provide information regarding subunit cellular location. Thus it is possible that chronic prenatal hypoxia could modify channel targeting and the proportion of the channels retained in the sarcoplasmic-endoplasmic reticulum vs. those channels transported to the mitochondrial and plasma membranes. An alternative explanation for our findings is that chronic prenatal hypoxia induces changes in the expression of BKCa channel γ-subunits, which interact with β1-subunits. In a key study performed in rat cerebral arterial myocytes, the γ-subunit was found to be important to vasorelaxation, where it facilitated the coupling of BKCa channels to Ca2+ sparks by shifting voltage dependence and Ca2+ sensitivity, assisting in membrane hyperpolarization and concomitant vasorelaxation (18). Thus one potential explanation for the mismatch between data from tension studies and α- and β1-subunit expression data could be an upregulation of the γ-subunit, which improves BKCa channel function. Ultimately, however, resolution of this and other potential explanations requires further investigation.
Although the data suggest that BKCa channel-dependent dilation of pulmonary arteries is mediated through the NO-cGMP-PKG signaling axis, PKG also causes vasodilation through other mechanisms. In addition to activating K+ channels, hyperpolarizing the plasma membrane, and reducing voltage-dependent Ca2+ influx, PKG also functionally decreases the sensitivity of contraction to increases in the cytosolic Ca2+ concentration. In particular, PKG can dilate vessels by promoting dephosphorylation of the myosin light chain kinase. This is achieved by phosphorylation and activation of the myosin light chain phosphatase (26, 27, 81). Furthermore, PKG inhibits RhoA activity, which is a key modulator of the Rho kinase pathway (27, 49). This is directly relevant to the current studies, as we previously showed that Rho kinase accounts for roughly half of the arterial tension due to serotonin or membrane depolarization in pulmonary arteries of sheep (10, 68). Future studies are therefore needed to discriminate the interaction between PKG and various pathways.
Perspective.
The present series of studies illustrate that bradykinin-dependent vasorelaxation is mediated through a combination of eNOS-sensitive and -insensitive pathways. From a methodological standpoint, our results demonstrate the importance of conducting whole animal studies, in addition to in vitro experimentation. Sustained stress, such as prenatal chronic hypoxia, can result in complex changes via the interaction of numerous systems not represented by inducing short-term stress in isolated cells. Terrestrial prenatal chronic hypoxia due to living at moderately high altitude influenced the development of eNOS-dependent pathways, causing adaptations that improve pulmonary blood flow and blood oxygenation in the fetus and newborn infant. Presumably, these adaptations allow newborn lambs to survive birth in the rarified environment. Unfortunately, the increased blood flow that results from these adaptations may lessen blood flow necessary for the proper development of other vital organs. On the basis of our data, prenatal chronic hypoxia blunts normal development of bradykinin-mediated vasodilatory pathways, resulting in lessened responsiveness. In turn, this could exacerbate pulmonary pressures and reduce the ability of newborn hypoxic lambs to thrive. On the whole, our results demonstrate that prenatal chronic hypoxia takes a significant toll on proper pre- and postnatal development of the bradykinin-activated, eNOS-BKCa channel vasodilatory pathway in sheep. The dramatic effects of hypoxia observed here provide valuable insights into the mechanisms contributing to pulmonary hypertension in the newborn and demonstrate the potential for targeting the eNOS-BKCa channel signaling axis in afflicted newborns.
GRANTS
This material is based on work supported by National Science Foundation Grant MRI 0923559 and National Institutes of Health Grants HD-069746 (S. M. Wilson), HL-085887 (M. S. Taylor), R01 NS-076945 (W. J. Pearce), HL-095973 (A. B. Blood), and P01 HD-031226 and R01 HD-003807 (L. D. Longo). Additional funding for research reported in this publication was provided by National Institute of General Medical Sciences Grant 2R25 GM-060507. C. Blum-Johnston was a summer research fellow in the Apprenticeship Bridge to College and Undergraduate Training Programs through the Initiative for Maximizing Student Development Program in the Center for Health Disparities and Molecular Medicine. Additional support for the Advanced Imaging and Microscopy Core was provided by the Loma Linda University School of Medicine.
DISCLAIMERS
The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health or National Science Foundation.
DISCLOSURES
No conflicts of interest, financial or otherwise, are declared by the authors.
AUTHOR CONTRIBUTIONS
C.B.-J., R.B.T., C.W., M.R., A.R.B., Q.B., R.W., and S.M.W. analyzed the data; C.B.-J., R.B.T., C.W., M.R., A.R.B., Q.B., R.W., A.B.B., M.F., M.S.T., L.D.L., W.J.P., and S.M.W. interpreted the results of the experiments; C.B.-J., R.B.T., C.W., M.R., A.R.B., Q.B., and S.M.W. prepared the figures; C.B.-J., R.B.T., C.W., L.D.L., W.J.P., and S.M.W. drafted the manuscript; C.B.-J., R.B.T., C.W., M.R., A.R.B., Q.B., R.W., A.B.B., M.F., M.S.T., L.D.L., W.J.P., and S.M.W. edited and revised the manuscript; C.B.-J., R.B.T., C.W., M.R., A.R.B., Q.B., R.W., A.B.B., M.F., M.S.T., L.D.L., W.J.P., and S.M.W. approved the final version of the manuscript; R.B.T., A.B.B., M.F., M.S.T., L.D.L., W.J.P., and S.M.W. developed the concept and designed the research; R.B.T., M.R., A.R.B., Q.B., R.W., and S.M.W. performed the experiments.
ACKNOWLEDGMENTS
We thank Kurt Vrancken and Dr. Taiming Liu for technical assistance with the ProliNO studies. All imaging was performed in the Loma Linda University Advanced Imaging and Microscopy Core Facility.
REFERENCES
- 1.Adnot S, Raffestin B, Eddahibi S, Braquet P, Chabrier PE. Loss of endothelium-dependent relaxant activity in the pulmonary circulation of rats exposed to chronic hypoxia. J Clin Invest 87: 155–162, 1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Amberg GC, Santana LF. Downregulation of the BK channel β1-subunit in genetic hypertension. Circ Res 93: 965–971, 2003. [DOI] [PubMed] [Google Scholar]
- 3.Archer SL, Huang JM, Hampl V, Nelson DP, Shultz PJ, Weir EK. Nitric oxide and cGMP cause vasorelaxation by activation of a charybdotoxin-sensitive K channel by cGMP-dependent protein kinase. Proc Natl Acad Sci USA 91: 7583–7587, 1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Arnold WP, Mittal CK, Katsuki S, Murad F. Nitric oxide activates guanylate cyclase and increases guanosine 3′:5′-cyclic monophosphate levels in various tissue preparations. Proc Natl Acad Sci USA 74: 3203–3207, 1977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Aschner JL, Smith TK, Kovacs N, Pinheiro JM, Fuloria M. Mechanisms of bradykinin-mediated dilation in newborn piglet pulmonary conducting and resistance vessels. Am J Physiol Lung Cell Mol Physiol 283: L373–L382, 2002. [DOI] [PubMed] [Google Scholar]
- 6.Batenburg WW, Garrelds IM, van Kats JP, Saxena PR, Danser AH. Mediators of bradykinin-induced vasorelaxation in human coronary microarteries. Hypertension 43: 488–492, 2004. [DOI] [PubMed] [Google Scholar]
- 7.Batenburg WW, Tom B, Schuijt MP, Danser AH. Angiotensin II type 2 receptor-mediated vasodilation. Focus on bradykinin, NO and endothelium-derived hyperpolarizing factor(s). Vasc Pharmacol 42: 109–118, 2005. [DOI] [PubMed] [Google Scholar]
- 8.Beitelshees AL, Gong Y, Wang D, Schork NJ, Cooper-Dehoff RM, Langaee TY, Shriver MD, Sadee W, Knot HJ, Pepine CJ, Johnson JA, INVEST Investigators. KCNMB1 genotype influences response to verapamil SR and adverse outcomes in the INternational VErapamil SR/Trandolapril STudy (INVEST). Pharmacogenet Genomics 17: 719–729, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Bixby CE, Ibe BO, Abdallah MF, Zhou W, Hislop AA, Longo LD, Raj JU. Role of platelet-activating factor in pulmonary vascular remodeling associated with chronic high altitude hypoxia in ovine fetal lambs. Am J Physiol Lung Cell Mol Physiol 293: L1475–L1482, 2007. [DOI] [PubMed] [Google Scholar]
- 10.Blood AB, Terry MH, Merritt TA, Papamatheakis DG, Blood Q, Ross JM, Power GG, Longo LD, Wilson SM. Effect of chronic perinatal hypoxia on the role of rho-kinase in pulmonary artery contraction in newborn lambs. Am J Physiol Regul Integr Comp Physiol 304: R136–R146, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Bradbury DA, Newton R, Zhu YM, Stocks J, Corbett L, Holland ED, Pang LH, Knox AJ. Effect of bradykinin, TGF-β1, IL-1β, and hypoxia on COX-2 expression in pulmonary artery smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 283: L717–L725, 2002. [DOI] [PubMed] [Google Scholar]
- 12.Brayden JE, Nelson MT. Regulation of arterial tone by activation of calcium-dependent potassium channels. Science 256: 532–535, 1992. [DOI] [PubMed] [Google Scholar]
- 13.Brenner R, Perez GJ, Bonev AD, Eckman DM, Kosek JC, Wiler SW, Patterson AJ, Nelson MT, Aldrich RW. Vasoregulation by the β1-subunit of the calcium-activated potassium channel. Nature 407: 870–876, 2000. [DOI] [PubMed] [Google Scholar]
- 14.Campbell WB, Fleming I. Epoxyeicosatrienoic acids and endothelium-dependent responses. Pflügers Arch 459: 881–895, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Cassin S, Winikor I, Tod M, Philips J, Frisinger J, Jordan J, Gibbs C. Effects of prostacyclin on the fetal pulmonary circulation. Pediatr Pharmacol 1: 197–207, 1981. [PubMed] [Google Scholar]
- 16.Danser AH, de Vries R, Schoemaker RG, Saxena PR. Bradykinin-induced release of nitric oxide by the isolated perfused rat heart: importance of preformed pools of nitric oxide-containing factors. J Hypertens 16: 239–244, 1998. [DOI] [PubMed] [Google Scholar]
- 17.Dudzinski DM, Michel T. Life history of eNOS: partners and pathways. Cardiovasc Res 75: 247–260, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Evanson KW, Bannister JP, Leo MD, Jaggar JH. LRRC26 is a functional BK channel auxiliary γ-subunit in arterial smooth muscle cells. Circ Res 115: 423–431, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Fernandez-Fernandez JM, Tomas M, Vazquez E, Orio P, Latorre R, Senti M, Marrugat J, Valverde MA. Gain-of-function mutation in the KCNMB1 potassium channel subunit is associated with low prevalence of diastolic hypertension. J Clin Invest 113: 1032–1039, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Fike CD, Kaplowitz MR, Pfister SL. Arachidonic acid metabolites and an early stage of pulmonary hypertension in chronically hypoxic newborn pigs. Am J Physiol Lung Cell Mol Physiol 284: L316–L323, 2003. [DOI] [PubMed] [Google Scholar]
- 21.Fike CD, Kaplowitz MR, Zhang Y, Pfister SL. Cyclooxygenase-2 and an early stage of chronic hypoxia-induced pulmonary hypertension in newborn pigs. J Appl Physiol 98: 1111–1118; discussion 1091, 2005. [DOI] [PubMed] [Google Scholar]
- 22.Francis M, Qian X, Charbel C, Ledoux J, Parker JC, Taylor MS. Automated region of interest analysis of dynamic Ca2+ signals in image sequences. Am J Physiol Cell Physiol 303: C236–C243, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Fulton D, Fontana J, Sowa G, Gratton JP, Lin M, Li KX, Michell B, Kemp BE, Rodman D, Sessa WC. Localization of endothelial nitric-oxide synthase phosphorylated on serine 1179 and nitric oxide in Golgi and plasma membrane defines the existence of two pools of active enzyme. J Biol Chem 277: 4277–4284, 2002. [DOI] [PubMed] [Google Scholar]
- 24.Gairhe S, Bauer NN, Gebb SA, McMurtry IF. Myoendothelial gap junctional signaling induces differentiation of pulmonary arterial smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 301: L527–L535, 2011. [DOI] [PubMed] [Google Scholar]
- 25.Gao J, Chao J, Parbhu KJ, Yu L, Xiao L, Gao F, Gao L. Ontogeny of angiotensin type 2 and type 1 receptor expression in mice. J Renin Angiotensin Aldosterone Syst 13: 341–352, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Gao Y, Portugal AD, Liu J, Negash S, Zhou W, Tian J, Xiang R, Longo LD, Raj JU. Preservation of cGMP-induced relaxation of pulmonary veins of fetal lambs exposed to chronic high altitude hypoxia: role of PKG and Rho kinase. Am J Physiol Lung Cell Mol Physiol 295: L889–L896, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Gao Y, Portugal AD, Negash S, Zhou W, Longo LD, Raj UJ. Role of Rho kinases in PKG-mediated relaxation of pulmonary arteries of fetal lambs exposed to chronic high altitude hypoxia. Am J Physiol Lung Cell Mol Physiol 292: L678–L684, 2007. [DOI] [PubMed] [Google Scholar]
- 28.Gao Y, Zhou H, Ibe BO, Raj JU. Prostaglandins E2 and I2 cause greater relaxations in pulmonary veins than in arteries of newborn lambs. J Appl Physiol 81: 2534–2539, 1996. [DOI] [PubMed] [Google Scholar]
- 29.Garthwaite J, Southam E, Boulton CL, Nielsen EB, Schmidt K, Mayer B. Potent and selective inhibition of nitric oxide-sensitive guanylyl cyclase by 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one. Mol Pharmacol 48: 184–188, 1995. [PubMed] [Google Scholar]
- 30.Giaid A, Saleh D. Reduced expression of endothelial nitric oxide synthase in the lungs of patients with pulmonary hypertension. N Engl J Med 333: 214–221, 1995. [DOI] [PubMed] [Google Scholar]
- 31.Glasgow RE, Buga GM, Ignarro LJ, Chaudhuri G, Heymann MA. Endothelium-derived relaxing factor as a mediator of bradykinin-induced perinatal pulmonary vasodilatation in fetal sheep. Reprod Fertil Dev 9: 213–216, 1997. [DOI] [PubMed] [Google Scholar]
- 32.Goyal R, Angermann JE, Ostrovskaya O, Buchholz JN, Smith GD, Wilson SM. Enhanced capacitative calcium entry and sarcoplasmic-reticulum calcium storage capacity with advanced age in murine mesenteric arterial smooth muscle cells. Exp Gerontol 44: 201–207, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Goyal R, Creel KD, Chavis E, Smith GD, Longo LD, Wilson SM. Maturation of intracellular calcium homeostasis in sheep pulmonary arterial smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 295: L905–L914, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Goyal R, Leitzke A, Goyal D, Gheorghe CP, Longo LD. Antenatal maternal hypoxic stress: adaptations in fetal lung renin-angiotensin system. Reprod Sci 18: 180–189, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Goyal R, Papamatheakis DG, Loftin M, Vrancken K, Dawson AS, Osman NJ, Blood AB, Pearce WJ, Longo LD, Wilson SM. Long-term maternal hypoxia: the role of extracellular Ca2+ entry during serotonin-mediated contractility in fetal ovine pulmonary arteries. Reprod Sci 18: 948–962, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Greif DM, Sacks DB, Michel T. Calmodulin phosphorylation and modulation of endothelial nitric oxide synthase catalysis. Proc Natl Acad Sci USA 101: 1165–1170, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Hadley SR, Blood Q, Rubalcava M, Waskel E, Lumbard B, Le P, Longo LD, Buchholz JN, Wilson SM. Maternal high-altitude hypoxia and suppression of ryanodine receptor-mediated Ca2+ sparks in fetal sheep pulmonary arterial myocytes. Am J Physiol Lung Cell Mol Physiol 303: L799–L813, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Hampl V, Herget J. Role of nitric oxide in the pathogenesis of chronic pulmonary hypertension. Physiol Rev 80: 1337–1372, 2000. [DOI] [PubMed] [Google Scholar]
- 39.Haworth SG. Pulmonary endothelium in the perinatal period. Pharmacol Rep 58 Suppl: 153–164, 2006. [PubMed] [Google Scholar]
- 40.Herrera EA, Pulgar VM, Riquelme RA, Sanhueza EM, Reyes RV, Ebensperger G, Parer JT, Valdez EA, Giussani DA, Blanco CE, Hanson MA, Llanos AJ. High-altitude chronic hypoxia during gestation and after birth modifies cardiovascular responses in newborn sheep. Am J Physiol Regul Integr Comp Physiol 292: R2234–R2240, 2007. [DOI] [PubMed] [Google Scholar]
- 41.Herrera EA, Reyes RV, Giussani DA, Riquelme RA, Sanhueza EM, Ebensperger G, Casanello P, Mendez N, Ebensperger R, Sepulveda-Kattan E, Pulgar VM, Cabello G, Blanco CE, Hanson MA, Parer JT, Llanos AJ. Carbon monoxide: a novel pulmonary artery vasodilator in neonatal llamas of the Andean altiplano. Cardiovasc Res 77: 197–201, 2008. [DOI] [PubMed] [Google Scholar]
- 42.Herrera EA, Riquelme RA, Ebensperger G, Reyes RV, Ulloa CE, Cabello G, Krause BJ, Parer JT, Giussani DA, Llanos AJ. Long-term exposure to high-altitude chronic hypoxia during gestation induces neonatal pulmonary hypertension at sea level. Am J Physiol Regul Integr Comp Physiol 299: R1676–R1684, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Hidaka H, Asano M, Iwadare S, Matsumoto I, Totsuka T, Aoki N. A novel vascular relaxing agent, N-(6-aminohexyl)-5-chloro-1-naphthalensulfonamide, which affects vascular smooth muscle actomyosin. J Pharmacol Exp Ther 207: 8–15, 1978. [PubMed] [Google Scholar]
- 44.Hu XQ, Xiao D, Zhu R, Huang X, Yang S, Wilson SM, Zhang L. Chronic hypoxia suppresses pregnancy-induced upregulation of large-conductance Ca2+-activated K+ channel activity in uterine arteries. Hypertension 60: 214–222, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Ignarro LJ, Harbison RG, Wood KS, Kadowitz PJ. Activation of purified soluble guanylate cyclase by endothelium-derived relaxing factor from intrapulmonary artery and vein: stimulation by acetylcholine, bradykinin and arachidonic acid. J Pharmacol Exp Ther 237: 893–900, 1986. [PubMed] [Google Scholar]
- 46.Iwatsuki N, Petersen OH. Action of tetraethylammonium on calcium-activated potassium channels in pig pancreatic acinar cells studied by patch-clamp single-channel and whole-cell current recording. J Membr Biol 86: 139–144, 1985. [DOI] [PubMed] [Google Scholar]
- 47.Kamitomo M, Alonso JG, Okai T, Longo LD, Gilbert RD. Effects of long-term, high-altitude hypoxemia on ovine fetal cardiac output and blood flow distribution. Am J Obstet Gynecol 169: 701–707, 1993. [DOI] [PubMed] [Google Scholar]
- 48.Kaneko FT, Arroliga AC, Dweik RA, Comhair SA, Laskowski D, Oppedisano R, Thomassen MJ, Erzurum SC. Biochemical reaction products of nitric oxide as quantitative markers of primary pulmonary hypertension. Am J Respir Crit Care Med 158: 917–923, 1998. [DOI] [PubMed] [Google Scholar]
- 49.Kato M, Blanton R, Wang GR, Judson TJ, Abe Y, Myoishi M, Karas RH, Mendelsohn ME. Direct binding and regulation of RhoA protein by cyclic GMP-dependent protein kinase Iα. J Biol Chem 287: 41342–41351, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kerr PM, Wei R, Tam R, Sandow SL, Murphy TV, Ondrusova K, Lunn SE, Tran CH, Welsh DG, Plane F. Activation of endothelial IK channels underlies NO-dependent myoendothelial feedback. Vascul Pharmacol 74: 130–138, 2015. [DOI] [PubMed] [Google Scholar]
- 51.Knaus HG, Folander K, Garcia-Calvo M, Garcia ML, Kaczorowski GJ, Smith M, Swanson R. Primary sequence and immunological characterization of β-subunit of high conductance Ca2+-activated K+ channel from smooth muscle. J Biol Chem 269: 17274–17278, 1994. [PubMed] [Google Scholar]
- 52.Ko EA, Han J, Jung ID, Park WS. Physiological roles of K+ channels in vascular smooth muscle cells. J Smooth Muscle Res 44: 65–81, 2008. [DOI] [PubMed] [Google Scholar]
- 53.Leeb-Lundberg LM, Marceau F, Muller-Esterl W, Pettibone DJ, Zuraw BL. International union of pharmacology. XLV. Classification of the kinin receptor family: from molecular mechanisms to pathophysiological consequences. Pharmacol Rev 57: 27–77, 2005. [DOI] [PubMed] [Google Scholar]
- 54.Leffler CW, Hessler JR, Green RS. The onset of breathing at birth stimulates pulmonary vascular prostacyclin synthesis. Pediatr Res 18: 938–942, 1984. [DOI] [PubMed] [Google Scholar]
- 55.Lima B, Forrester MT, Hess DT, Stamler JS. S-nitrosylation in cardiovascular signaling. Circ Res 106: 633–646, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Lin DC, Lin S. Actin polymerization induced by a motility-related high-affinity cytochalasin binding complex from human erythrocyte membrane. Proc Natl Acad Sci USA 76: 2345–2349, 1979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Liu SF, Hislop AA, Haworth SG, Barnes PJ. Developmental changes in endothelium-dependent pulmonary vasodilatation in pigs. Br J Pharmacol 106: 324–330, 1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Liu Y, Pleyte K, Knaus HG, Rusch NJ. Increased expression of Ca2+-sensitive K+ channels in aorta of hypertensive rats. Hypertension 30: 1403–1409, 1997. [DOI] [PubMed] [Google Scholar]
- 59.Llanos AJ, Ebensperger G, Herrera EA, Reyes RV, Cabello G, Diaz M, Giussani DA, Parer JT. The heme oxygenase-carbon monoxide system in the regulation of cardiorespiratory function at high altitude. Respir Physiol Neurobiol 184: 186–191, 2012. [DOI] [PubMed] [Google Scholar]
- 60.Llanos AJ, Ebensperger G, Herrera EA, Reyes RV, Pulgar VM, Seron-Ferre M, Diaz M, Parer JT, Giussani DA, Moraga FA, Riquelme RA. Fetal and postnatal pulmonary circulation in the Alto Andino. Placenta 32 Suppl 2: S100–S103, 2011. [DOI] [PubMed] [Google Scholar]
- 61.Llorens O, Perez JJ, Palomer A, Mauleon D. Differential binding mode of diverse cyclooxygenase inhibitors. J Mol Graph Model 20: 359–371, 2002. [DOI] [PubMed] [Google Scholar]
- 62.Lorca RA, Prabagaran M, England SK. Functional insights into modulation of BKCa channel activity to alter myometrial contractility. Front Physiol 5: 289, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Meera P, Wallner M, Jiang Z, Toro L. A calcium switch for the functional coupling between α (hslo)- and β-subunits (KV,Caβ) of maxi K channels. FEBS Lett 382: 84–88, 1996. [DOI] [PubMed] [Google Scholar]
- 64.Mutchler SM, Straub AC. Compartmentalized nitric oxide signaling in the resistance vasculature. Nitric Oxide Biol Chem 49: 8–15, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Olschewski A, Hong Z, Linden BC, Porter VA, Weir EK, Cornfield DN. Contribution of the KCa channel to membrane potential and O2 sensitivity is decreased in an ovine PPHN model. Am J Physiol Lung Cell Mol Physiol 283: L1103–L1109, 2002. [DOI] [PubMed] [Google Scholar]
- 66.Palomer A, Cabre F, Pascual J, Campos J, Trujillo MA, Entrena A, Gallo MA, Garcia L, Mauleon D, Espinosa A. Identification of novel cyclooxygenase-2 selective inhibitors using pharmacophore models. J Med Chem 45: 1402–1411, 2002. [DOI] [PubMed] [Google Scholar]
- 67.Papamatheakis DG, Blood AB, Kim JH, Wilson SM. Antenatal hypoxia and pulmonary vascular function and remodeling. Curr Vasc Pharmacol 11: 616–640, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Papamatheakis DG, Patel JJ, Blood Q, Merritt TT, Longo LD, Wilson SM. Depolarization-dependent contraction increase after birth and preservation following long-term hypoxia in sheep pulmonary arteries. Pulm Circ 2: 41–53, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Pluger S, Faulhaber J, Furstenau M, Lohn M, Waldschutz R, Gollasch M, Haller H, Luft FC, Ehmke H, Pongs O. Mice with disrupted BK channel β1-subunit gene feature abnormal Ca2+ spark/STOC coupling and elevated blood pressure. Circ Res 87: E53–E60, 2000. [DOI] [PubMed] [Google Scholar]
- 70.Porter VA, Rhodes MT, Reeve HL, Cornfield DN. Oxygen-induced fetal pulmonary vasodilation is mediated by intracellular calcium activation of KCa channels. Am J Physiol Lung Cell Mol Physiol 281: L1379–L1385, 2001. [DOI] [PubMed] [Google Scholar]
- 71.Rees DD, Palmer RM, Schulz R, Hodson HF, Moncada S. Characterization of three inhibitors of endothelial nitric oxide synthase in vitro and in vivo. Br J Pharmacol 101: 746–752, 1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Resnik E, Herron J, Fu R, Ivy DD, Cornfield DN. Oxygen tension modulates the expression of pulmonary vascular BKCa channel α- and β-subunits. Am J Physiol Lung Cell Mol Physiol 290: L761–L768, 2006. [DOI] [PubMed] [Google Scholar]
- 73.Rhodes MT, Porter VA, Saqueton CB, Herron JM, Resnik ER, Cornfield DN. Pulmonary vascular response to normoxia and KCa channel activity is developmentally regulated. Am J Physiol Lung Cell Mol Physiol 280: L1250–L1257, 2001. [DOI] [PubMed] [Google Scholar]
- 74.Robertson BE, Schubert R, Hescheler J, 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. [DOI] [PubMed] [Google Scholar]
- 75.Roth M, Rupp M, Hofmann S, Mittal M, Fuchs B, Sommer N, Parajuli N, Quanz K, Schubert D, Dony E, Schermuly RT, Ghofrani HA, Sausbier U, Rutschmann K, Wilhelm S, Seeger W, Ruth P, Grimminger F, Sausbier M, Weissmann N. Heme oxygenase-2 and large-conductance Ca2+-activated K+ channels: lung vascular effects of hypoxia. Am J Respir Crit Care Med 180: 353–364, 2009. [DOI] [PubMed] [Google Scholar]
- 76.Saavedra JE, Southan GJ, Davies KM, Lundell A, Markou C, Hanson SR, Adrie C, Hurford WE, Zapol WM, Keefer LK. Localizing antithrombotic and vasodilatory activity with a novel, ultrafast nitric oxide donor. J Med Chem 39: 4361–4365, 1996. [DOI] [PubMed] [Google Scholar]
- 77.Sadoshima J, Akaike N, Kanaide H, Nakamura M. Cyclic AMP modulates Ca-activated K channel in cultured smooth muscle cells of rat aortas. Am J Physiol Heart Circ Physiol 255: H754–H759, 1988. [DOI] [PubMed] [Google Scholar]
- 78.Sage SO, Adams DJ, van Breemen C. Synchronized oscillations in cytoplasmic free calcium concentration in confluent bradykinin-stimulated bovine pulmonary artery endothelial cell monolayers. J Biol Chem 264: 6–9, 1989. [PubMed] [Google Scholar]
- 79.Saqueton CB, Miller RB, Porter VA, Milla CE, Cornfield DN. NO causes perinatal pulmonary vasodilation through K+ channel activation and intracellular Ca2+ release. Am J Physiol Lung Cell Mol Physiol 276: L925–L932, 1999. [DOI] [PubMed] [Google Scholar]
- 80.Sukumaran SV, Singh TU, Parida S, Narasimha Reddy CE, Thangamalai R, Kandasamy K, Singh V, Mishra SK. TRPV4 channel activation leads to endothelium-dependent relaxation mediated by nitric oxide and endothelium-derived hyperpolarizing factor in rat pulmonary artery. Pharmacol Res 78: 18–27, 2013. [DOI] [PubMed] [Google Scholar]
- 81.Surks HK, Mochizuki N, Kasai Y, Georgescu SP, Tang KM, Ito M, Lincoln TM, Mendelsohn ME. Regulation of myosin phosphatase by a specific interaction with cGMP-dependent protein kinase Iα. Science 286: 1583–1587, 1999. [DOI] [PubMed] [Google Scholar]
- 82.Takahashi S, Mendelsohn ME. Synergistic activation of endothelial nitric-oxide synthase (eNOS) by HSP90 and Akt: calcium-independent eNOS activation involves formation of an HSP90-Akt-CaM-bound eNOS complex. J Biol Chem 278: 30821–30827, 2003. [DOI] [PubMed] [Google Scholar]
- 83.Taraseviciene-Stewart L, Gera L, Hirth P, Voelkel NF, Tuder RM, Stewart JM. A bradykinin antagonist and a caspase inhibitor prevent severe pulmonary hypertension in a rat model. Can J Physiol Pharmacol 80: 269–274, 2002. [DOI] [PubMed] [Google Scholar]
- 84.Toro L, Li M, Zhang Z, Singh H, Wu Y, Stefani E. MaxiK channel and cell signalling. Pflügers Arch 466: 875–886, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Tracey A, Bunton D, Irvine J, MacDonald A, Shaw AM. Relaxation to bradykinin in bovine pulmonary supernumerary arteries can be mediated by both a nitric oxide-dependent and -independent mechanism. Br J Pharmacol 137: 538–544, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Tristani-Firouzi M, Martin EB, Tolarova S, Weir EK, Archer SL, Cornfield DN. Ventilation-induced pulmonary vasodilation at birth is modulated by potassium channel activity. Am J Physiol Heart Circ Physiol 271: H2353–H2359, 1996. [DOI] [PubMed] [Google Scholar]
- 87.Tulloh RM, Hislop AA, Boels PJ, Deutsch J, Haworth SG. Chronic hypoxia inhibits postnatal maturation of porcine intrapulmonary artery relaxation. Am J Physiol Heart Circ Physiol 272: H2436–H2445, 1997. [DOI] [PubMed] [Google Scholar]
- 88.Wang J, Weigand L, Foxson J, Shimoda LA, Sylvester JT. Ca2+ signaling in hypoxic pulmonary vasoconstriction: effects of myosin light chain and Rho kinase antagonists. Am J Physiol Lung Cell Mol Physiol 293: L674–L685, 2007. [DOI] [PubMed] [Google Scholar]
- 89.Xia Y, Fu Z, Hu J, Huang C, Paudel O, Cai S, Liedtke W, Sham JS. TRPV4 channel contributes to serotonin-induced pulmonary vasoconstriction and the enhanced vascular reactivity in chronic hypoxic pulmonary hypertension. Am J Physiol Cell Physiol 305: C704–C715, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Xu W, Liu Y, Wang S, McDonald T, Van Eyk JE, Sidor A, O'Rourke B. Cytoprotective role of Ca2+- activated K+ channels in the cardiac inner mitochondrial membrane. Science 298: 1029–1033, 2002. [DOI] [PubMed] [Google Scholar]
- 91.Xue Q, Ducsay CA, Longo LD, Zhang L. Effect of long-term high-altitude hypoxia on fetal pulmonary vascular contractility. J Appl Physiol 104: 1786–1792, 2008. [DOI] [PubMed] [Google Scholar]
- 92.Yayama K, Okamoto H. Angiotensin II-induced vasodilation via type 2 receptor: role of bradykinin and nitric oxide. Int Immunopharmacol 8: 312–318, 2008. [DOI] [PubMed] [Google Scholar]