Skip to main content
American Journal of Physiology - Lung Cellular and Molecular Physiology logoLink to American Journal of Physiology - Lung Cellular and Molecular Physiology
. 2008 Apr 18;295(1):L104–L113. doi: 10.1152/ajplung.00058.2008

Differences in STIM1 and TRPC expression in proximal and distal pulmonary arterial smooth muscle are associated with differences in Ca2+ responses to hypoxia

Wenju Lu 1,*, Jian Wang 1,*, Larissa A Shimoda 1, J T Sylvester 1
PMCID: PMC2494791  PMID: 18424621

Abstract

Hypoxic pulmonary vasoconstriction (HPV) requires Ca2+ influx through store-operated Ca2+ channels (SOCC) in pulmonary arterial smooth muscle cells (PASMC) and is greater in distal than proximal pulmonary arteries (PA). SOCC may be composed of canonical transient receptor potential (TRPC) proteins and activated by stromal interacting molecule 1 (STIM1). To assess the possibility that HPV is greater in distal PA because store-operated Ca2+ entry (SOCE) is greater in distal PASMC, we measured intracellular Ca2+ concentration ([Ca2+]i) and SOCE in primary cultures of PASMC using fluorescent microscopy and the Ca2+-sensitive dye fura 2. Both hypoxia (4% O2) and KCl (60 mM) increased [Ca2+]i. Responses to hypoxia, but not KCl, were greater in distal cells. We measured SOCE in PASMC perfused with Ca2+-free solutions containing cyclopiazonic acid to deplete Ca2+ stores in sarcoplasmic reticulum and nifedipine to prevent Ca2+ entry through L-type voltage-operated Ca2+ channels. Under these conditions, the increase in [Ca2+]i caused by restoration of extracellular Ca2+ and the decrease in fura 2 fluorescence caused by Mn2+ were greater in distal PASMC, indicating greater SOCE. Moreover, the increase in SOCE caused by hypoxia was also greater in distal cells. Real-time quantitative polymerase chain reaction analysis of PASMC and freshly isolated deendothelialized PA tissue demonstrated expression of STIM1 and five of seven known TRPC isoforms (TRPC1 > TRPC6 > TRPC4 ≫ TRPC3 ≈ TRPC5). For both protein, as measured by Western blotting, and mRNA, expression of STIM1, TRPC1, TRPC6, and TRPC4 was greater in distal than proximal PASMC and PA. These results provide further support for the importance of SOCE in HPV and suggest that HPV is greater in distal than proximal PA because greater numbers and activation of SOCC in distal PASMC generate bigger increases in [Ca2+]i.

Keywords: hypoxic pulmonary vasoconstriction, calcium signaling, vascular smooth muscle, potassium chloride, stromal interacting molecule 1, canonical transient receptor potential, calcium ion


evidence from intact animals (45, 47), pulmonary arteries (PA) (24), and pulmonary arterial smooth muscle cells (PASMC) (25) indicates that hypoxic pulmonary vasoconstriction (HPV) is greater in distal (“resistance”) PA than in proximal (“conduit”) PA. This difference in HPV has been associated with other differences between proximal and distal PASMC, such as the change in intracellular Ca2+ concentration ([Ca2+]i) induced by hypoxia (5, 46, 57, 59), K+ channel function (3, 4, 26, 48, 51) and expression (4, 9), Cl channel function (48), Ca2+ channel density and O2 sensitivity (13), Ca2+ stores in sarcoplasmic reticulum (SR) (58), and expression and function of endothelin receptors (10, 54). One assumption of these studies is that differences between proximal and distal PASMC could yield clues to the mechanisms of HPV, which remain elusive.

Recent results suggest that [Ca2+]i responses to hypoxia in distal PASMC (31, 62) and HPV in isolated lungs (64) require Ca2+ entry through store-operated Ca2+ channels (SOCC). SOCC are activated by release of Ca2+ from stores in SR and are thought to be heterotetrameric assemblies of so-called canonical transient receptor potential (TRPC) proteins (33, 34). Of the seven TRPC isoforms (TRPC1–7) thus far identified (29), all have been detected in pulmonary arterial smooth muscle (14, 18, 21, 27, 30, 43, 53, 60, 61, 63, 65, 71, 72, 74); however, the pattern of expression has varied considerably among studies. TRPC1, TRPC4, and TRPC6 are the most consistently identified isoforms, whereas TRPC2, TRPC3, TRPC5, and TRPC7 have been detected less frequently. A single quantitative polymerase chain reaction analysis (qPCR) of mouse distal PASMC revealed that TRPC1 and TRPC6 were more abundant than other TRPC isoforms (65). Whether TRPC expression differs between proximal and distal PASMC is unknown.

Stromal interacting molecule 1 (STIM1), a 90-kDa transmembrane Ca2+-binding protein found in sarco(endo)plasmic reticulum and plasma membrane, is thought to play a pivotal role in activation of SOCC (15, 20, 28, 39). According to this hypothesis, decreased Ca2+ concentration ([Ca2+]) in the SR lumen caused by Ca2+ release leads to dissociation of Ca2+ from an EF-hand motif in the intralumenal NH2-terminal region of STIM1, aggregation of STIM1 proteins at “puncta” in portions of the SR membrane close to plasma membrane, and physical interaction of STIM1 with SOCC and/or associated regulatory proteins, leading to channel activation (15, 68, 76). Although originally demonstrated in HeLa (22) and Drosophila S2 cells (44), STIM1 is now known to be required for store-operated Ca2+ entry (SOCE) in many cell types, including smooth muscle (12, 36, 55); however, evidence of STIM1 expression in PASMC has not been reported.

In this study, we quantified expression of TRPC and STIM1 in primary cultures of proximal and distal PASMC and freshly isolated deendothelialized proximal and distal PA. To assess whether differences in myocyte expression of TRPC and STIM1 have functional consequences in these vessels, we also measured the effects of hypoxia or KCl on [Ca2+]i and the effects of hypoxia on SOCE in proximal and distal PASMC.

METHODS

Preparation of PA and PASMC.

Animal protocols were approved by the Animal Care and Use Committee of the Johns Hopkins Medical Institutions. As previously described (6163, 66), proximal (main trunk) and distal (>4th generation) intrapulmonary arteries were dissected from lungs removed from male Wistar rats (300–500 g body wt) after anesthetization with pentobarbital sodium (65 mg/kg ip). Endothelium was denuded from the isolated PA by opening the vessel longitudinally and rubbing the luminal surface with a cotton swab. PASMC were harvested from these vessels enzymatically, plated on 25-mm cover slips in six-well dishes, and cultured for 3–5 days to 40–60% confluence in smooth muscle growth media (Clonetics, Walkersville, MD) in a humidified atmosphere of 5% CO2-95% air at 37°C. Before an experiment (24 h), culture media were changed to smooth muscle basal media (Clonetics) containing 0.3% serum to stop cell growth. Cellular purity was >95%, as assessed by morphological appearance under phase-contrast microscopy and immunofluorescence staining for α-actin (61).

Measurement of intracellular [Ca2+].

As previously described (61, 66), cover slips with myocytes were incubated with 5 μM fura 2-AM (Invitrogen, Carlsbad, CA) for 60 min at 37°C under an atmosphere of 5% CO2-95% air, mounted in a closed polycarbonate chamber clamped in a heated aluminum platform (PH-2; Warner Instrument, Hamden, CT) on the stage of a Nikon TSE 100 Ellipse inverted microscope (Nikon, Melville, NY), and perfused at 0.5 ml/min with Krebs Ringer bicarbonate solution (KRBS) that contained (in mM) 118 NaCl, 4.7 KCl, 2.5 CaCl2, 0.57 MgSO4, 1.18 KH2PO4, 25 NaHCO3, and 10 glucose. Perfusate was equilibrated in heated reservoirs with 5% CO2 and either 16% O2 (normoxia) or 4% O2 (hypoxia) and led to the chamber through stainless steel tubing. For depolarization of cells, perfusate KCl was increased to 60 mM, and NaCl was decreased to 62.7 mM. Chamber temperature was maintained at 37°C with an in-line heat exchanger and dual-channel heater controller (models SF-28 and TC-344B; Warner Instrument).

After removal of extracellular dye by 10 min of normoxic perfusion, [Ca2+]i was determined at 30- to 60-s intervals from the ratio of fura 2 fluorescence emitted at 510 nm after excitation at 340 nm to that after excitation at 380 nm (F340/F380) measured in 20–30 cells using a xenon arc lamp, interference filters, electronic shutter, ×20 fluorescence objective, and a cooled charge-coupled device imaging camera. Data were collected online with InCyte software (Intracellular Imaging, Cincinnati, OH). [Ca2+]i was estimated from F340/F380 measured in vitro in calibration solutions with [Ca2+] of 0–1,350 nM (Invitrogen). As previously described, hypoxia did not alter behavior of fura 2 in PASMC (62).

Measurement of SOCE.

We perfused PASMC for at least 10 min with Ca2+-free physiological salt solution containing 0.5 mM EGTA to chelate residual Ca2+, 5 μM nifedipine (Sigma Chemical, St. Louis, MO) to prevent calcium entry through L-type voltage-operated Ca2+ channels (VOCC), and 10 μM cyclopiazonic acid (CPA; Sigma Chemical) to deplete SR Ca2+ stores. SOCE was assessed in two ways, as described previously (61). First, we measured [Ca2+]i before and after restoration of extracellular [Ca2+] to 2.5 mM. SOCE was evaluated from the peak increase in [Ca2+]i caused by restoration of extracellular Ca2+ in the continued presence of nifedipine and CPA. Second, we monitored fura 2 fluorescence excited at 360 nm at 30-s intervals before and after addition of MnCl2 (200 μM) to the cell perfusate; SOCE was evaluated from the rate at which fura 2 fluorescence was quenched by Mn2+, which enters the cell as a Ca2+ surrogate and reduces fura 2 fluorescence upon binding to the dye. Fluorescence excited at 360 nm is the same for Ca2+-bound and Ca2+-free fura 2; therefore, changes in fluorescence are caused by Mn2+ alone.

RNA isolation and measurement by real-time qPCR.

Total RNA in deendothelialized distal or proximal PA was isolated using the TRIzol method, as previously described (61, 63). Briefly, samples were placed in ice-cold TRIzol reagent (Invitrogen) at 1 ml/20–50 mg tissue and homogenized at 20,000 rpm (model PT3100; Polytron, Kinematica, Switzerland). After incubation at 30°C for 5 min and addition of chloroform (0.2 ml/ml TRIzol), samples were centrifuged at 4°C and 8,000 g for 15 min. The upper aqueous phase of centrifuged samples was mixed with 100% isopropanol (0.5 ml/ml TRIzol), incubated at 30°C for 10 min, and recentrifuged at 4°C and 12,000 g for 10 min. The clear gel-like RNA precipitate was washed one time with 1 ml of 75% ethanol, dissolved in diethylpyrocarbonate-treated water, and stored at −80°C. Total RNA in PASMC was extracted using the RNeasy kit (Qiagen, Valencia, CA). DNA contamination in RNA preparations was removed by on-column DNase digestion using an RNeasy column and RNase-free DNase (Qiagen).

Reverse transcription was performed using the iScript cDNA synthesis kit (Bio-Rad, Hercules, CA). The reaction mixture contained 1 μg total RNA in a 20-μl volume. cDNA was quantified by real-time qPCR using QuantiTect SYBR Green PCR Master Mix (Qiagen) in a iCyclerIQ real-time PCR detection system (Bio-Rad) using the following conditions: 95°C for 15 min and 45 cycles, each at 94°C for 15 s, 57.5°C for 20 s, and 72°C for 20 s. The volume of each real-time qPCR reaction mixture was 25 μl containing 300 nM forward and reverse primers and cDNA template from 50 ng RNA. Primer sequences specific for rat TRPC, STIM1, or β-actin were designed using Primer3 software (http://frodo.wi.mit.edu/cgi-bin/primer3/primer3_www.cgi) and are listed in Table 1. Identity of the qPCR products was confirmed by 1) a single sharp peak in the melting curve performed after cDNA amplification; 2) a single band of the expected size resolved by agarose gel electrophoresis; and 3) the correct DNA sequence. Melting curves were performed at 95°C for 1 min and 55°C for 1 min, followed by 80 increments of 0.5°C at 10-s intervals. Real-time qPCR detection threshold cycle values were generated by iCyclerIQ software. Relative concentrations of each transcript were calculated using the Pfaffl (37) method. Efficiency for each gene was determined from five-point serial dilutions of positive control cDNA samples (PA or PASMC for TRPC1, TRPC4, TRPC6, and STIM1; rat brain for TRPC2, TRPC3, TRPC5, and TRPC7). Mean efficiencies of each gene were used to quantify expression of TRPC and STIM1 relative to β-actin in the same sample.

Table 1.

Primers for rat TRPC, STIM1, and β-actin used in real-time quantitative PCR

Gene Accession No. Primer Sequence (Left/Right) Product Size, bp Location in Sequence
TRPC1 NM_053558 5′-AGCCTCTTGACAAACGAGGA-3′ 146 797–942
5′-ACCTGACATCTGTCCGAACC-3′
TRPC2 NM_022638 5′-TTGCTGGGAGAGTCTCTGGT-3′ 139 1235–1393
5′-CCTCGATCCACACCTCCTTA-3′
TRPC3 NM_021771 5′-GAGATCTGGAATCGGTGGAA-3′ 127 608–754
5′-AAAAGCTGCTGTTGGCAGTT-3′
TRPC4 NM_053434 5′-GACACGGAGTTCCAGAGAGC-3′ 142 771–912
5′-GTTGGGCTGAGCAACAAACT-3′
TRPC5 NM_080898 5′-CCGCAAGGAGGTGGTAGG-3′ 121 643–784
5′-TGTGATGTCTGGTGTGAACTC-3′
TRPC6 NM_053559 5′-TACTGGTGTGCTCCTTGCAG-3′ 141 1267–1407
5′-GAGCTTGGTGCCTTCAAATC-3′
TRPC7 XM_225159 5′-AACGAGACCTTCACAGACTAC-3′ 114 1270–1383
5′-GCATTCAGACCAGATCATTCC-3′
STIM1 XM_341896 5′-ATGCCAATGGTGATGTGGAT-3′ 97 848–944
5′-CCATGGAAGGTGCTGTGTTT-3′
β-actin NM_031144 5′-ACGGTCAGGTCATCACTATC-3′ 90 812–901
5′-TGCCACAGGATTCCATACC-3′

TRPC, canonical transient receptor potential; STIM1, stromal interacting molecule 1.

Protein isolation and measurement by Western blotting.

Deendothelialized proximal or distal PA or PASMC samples were homogenized by sonication in Laemmlie sample buffer containing 62.5 mM Tris·HCl (pH 6.8), 2% SDS, 10% glycerol, 5% protease inhibitor cocktail, 1 mM EDTA, and 200 μM 4-(2-aminoethyl)benzenesulfonyl fluoride hydrochloride. Total protein concentration in the homogenates was determined by the bicinchoninic acid protein assay (Pierce, Rockford, IL) using BSA as a standard. Homogenates were denatured by adding dithiothreitol to 150 mM and heating at 95°C for 3 min. Homogenate proteins were resolved by 10% SDS-PAGE calibrated with prestained protein molecular weight markers (Precision Plus; Bio-Rad, Carlsbad, CA). Separated proteins were transferred to polyvinylidene difluoride membranes (pore size 0.45 μM; Bio-Rad). Membranes were blocked with 5% nonfat dry milk in Tris-buffered saline containing 0.2% Tween 20, blotted with affinity-purified polyclonal antibodies specific for TRPC proteins, or monoclonal antibody specific for STIM1 or α-actin. The membranes were then washed for 15 min three times and incubated with horseradish peroxidase-conjugated goat anti-rabbit or anti-mouse IgG for 1 h. Bound antibodies were detected using an enhanced chemiluminescence system (ECL; GE Healthcare, Piscataway, NJ).

Materials and drugs.

Unless otherwise specified, all reagents were obtained from Sigma Chemical. TRPC antibodies other than TRPC1 were obtained from Alomone Laboratories (Jerusalem, Israel). STIM1 antibody was obtained from BD Biosciences (Franklin Lakes, NJ). Fura 2-AM (Invitrogen) was prepared on the day of the experiment as a 2.5 mM stock solution in pluronic dimethyl sulfoxide (DMSO). Stock solutions (30 mM) of CPA and nifedipine were made in DMSO.

Statistical analysis.

Data are expressed as means ± SE; n is the number of experiments, which equals the number of animals providing arteries or cells. When fura 2 fluorescence was measured, the number of cells in each experiment was 20–30, as indicated in results and the legends for Figs. 17. Statistical analyses were performed using analysis of variance and Student's t-test. If significant F-ratios were obtained with the former, pairwise comparison of means was conducted with t-tests. When necessary, a Bonferroni correction was used to adjust the level of significance for multiple comparisons. Otherwise, differences were considered significant when P < 0.05.

Fig. 1.

Fig. 1.

Changes in intracellular Ca2+ concentration ([Ca2+]i) caused by 4% O2 in rat distal pulmonary arterial smooth muscle cells (PASMC; Distal, n = 7 experiments in 205 cells) and proximal PASMC (Proximal, n = 7 experiments in 201 cells; A); or 60 mM KCl in rat distal (n = 7 experiments in 198 cells) and proximal (n = 7 experiments in 190 cells) PASMC (B). Responses to hypoxia, but not KCl, were different in distal and proximal PASMC (P < 0.001 and 0.95, respectively). Brackets indicate ± SE.

Fig. 7.

Fig. 7.

Expression of STIM1 protein relative to α-actin in rat distal and proximal PASMC (top) and PA (bottom). A and C: representative blots for STIM1 and α-actin in proximal (Prox) and distal (Dist) PASMC and PA. B and D: mean protein expression for STIM1 relative to α-actin (n = 3 experiments on samples from 3 animals). Brackets indicate ± SE.

RESULTS

[Ca2+]i responses to hypoxia and KCl in PASMC.

Acute hypoxia (4% O2) caused a rapid sustained increase in [Ca2+]i in both proximal and distal PASMC (Fig. 1A); however, the increase in distal PASMC was ∼2.5-fold greater than that in proximal PASMC (peak Δ[Ca2+]i = 123 ± 18 vs. 32 ± 5 nM; P < 0.001). In contrast, responses to KCl (Fig. 1B) were the same in proximal and distal cells (peak Δ[Ca2+]i = 307 ± 84 vs. 313 ± 63 nM).

SOCE in PASMC.

SOCE in proximal and distal PASMC was assessed in two ways. First, we measured the peak increase in [Ca2+]i caused by restoration of extracellular [Ca2+] to 2.5 mM in PASMC perfused with Ca2+-free KRBS containing CPA and nifedipine. CPA caused an initial transient increase in [Ca2+]i during Ca2+-free perfusion (Fig. 2, A and B), which tended to be greater in distal than proximal PASMC (220 ± 45 vs. 140 ± 19 nM, P = 0.06; Fig. 2C). This response was not altered by hypoxia, which began at the same time as CPA. The increase in [Ca2+]i caused by restoration of extracellular Ca2+ was greater in distal than proximal PASMC (562 ± 65 vs. 410 ± 83 nM during normoxia and 798 ± 65 vs. 471 ± 33 nM during hypoxia, P < 0.03; Fig. 2D). Hypoxia increased the response to restoration in distal PASMC (P < 0.02; Fig. 2D), but not proximal PASMC.

Fig. 2.

Fig. 2.

Effects of restoration of extracellular Ca2+ concentration ([Ca2+]) to 2.5 mM in proximal and distal PASMC perfused with Ca2+-free Krebs Ringer bicarbonate solution (KRBS; 0 Ca2+) containing cyclopiazonic acid (CPA, 10 μM) and nifedipine (5 μM) during normoxia (16% O2; n = 9 experiments in 265 cells for proximal PASMC, 9 experiments in 261 cells for distal PASMC; A) and hypoxia (4% O2; n = 5 experiments in 138 cells for proximal PASMC, 5 experiments in 141 cells for distal PASMC; B). C: maximum initial increase in [Ca2+]i in response to CPA during Ca2+-free perfusion tended to be greater in distal PASMC (P = 0.06). D: maximum increase in [Ca2+]i after restoration of extracellular [Ca2+] was greater in distal PASMC (P < 0.03) and further increased by hypoxia in distal cells (P < 0.02). Brackets indicate ± SE.

Because the increase in [Ca2+]i caused by restoration of extracellular Ca2+ can be affected by factors other that SOCE, such as changes in efflux and membrane potential, we also measured the rate at which Mn2+ quenched fura 2 fluorescence, which is thought to be a more specific index of Ca2+ influx (Fig. 3). Mn2+ quenching, expressed as the percentage decrease in fluorescence at 5 min, was greater in distal than proximal PASMC during both normoxia and hypoxia (20 ± 2 vs. 11 ± 1% during normoxia and 44 ± 7 vs. 19 ± 2% during hypoxia, P < 0.01; Fig. 3B). Acute hypoxia, quenching increased in both distal and proximal PASMC (P < 0.002; Fig. 3B), and the increase was significantly greater in distal cells (P < 0.04; Fig. 3B).

Fig. 3.

Fig. 3.

Quenching of fura 2 fluorescence at 360 nm by 200 μM Mn2+ in PASMC perfused with Ca2+-free KRBS (0 Ca2+) containing nifedipine (5 μM) and CPA (10 μM). A: time course of fluorescence normalized to fluorescence at time 0 (F/F0) during normoxia (n = 8 experiments in 211 cells for proximal PASMC and 8 experiments in 218 cells for distal PASMC) and hypoxia (n = 4 experiments in 98 cells for proximal PASMC and 4 experiments in 92 cells for distal PASMC). B: Mn2+ quenching, expressed as the percentage decrease in fluorescence from time 0, was greater in distal PASMC (P < 0.01) and increased by hypoxia (P < 0.002). The increase caused by hypoxia was also greater in distal cells (P < 0.04). Brackets indicate ± SE.

TRPC and STIM1 expression in PASMC and PA.

Using qPCR, we detected transcripts of TRPC1, TRPC3, TRPC4, TRPC5, and TRPC6 in proximal and distal PASMC and PA (Fig. 4), but not TRPC2 or TRPC7. In all preparations, the rank order of abundance was TRPC1 > TRPC6 > TRPC4 ≫ TRPC3 ≈ TRPC5. In particular, TRPC1 was ∼2- to 4-fold more abundant than TRPC6, 10- to 30-fold more abundant than TRPC4, and 1,000- to 10,000-fold more abundant than TRPC3 or TRPC5. Expression of TRPC1, TRPC6, and TRPC4 mRNA was about twofold greater in distal than proximal PASMC and PA (P < 0.01; Fig. 4). Expression of other TRPC mRNA was not different with respect to vessel locus except TRPC5 in PA, which was less abundant in distal arteries. We did not compare TRPC mRNA expression between PASMC and PA because reference gene (β-actin) expression was different in these preparations. In proximal and distal PASMC and PA, Western blotting confirmed protein expression of TRPC1, TRPC4, and TRPC6, the most abundant TRPC isoforms identified by qPCR (Fig. 5). In all cases, distal expression exceeded proximal expression. We were unable to detect TRPC3 and TRPC5 proteins, presumably because of low protein abundance and/or inadequate antibody specificity. Expression of TRPC2 and TRPC7 proteins was not tested because mRNA was not detected. Differences in antibody-antigen interactions in Western blotting precluded determination of the rank order of abundance among TRPC proteins.

Fig. 4.

Fig. 4.

Expression of canonical transient receptor potential (TRPC) mRNA relative to β-actin in proximal and distal PASMC (top; n = 6 in each group) and pulmonary arteries (PA; bottom; n = 11 for TRPC1, TRPC4, and TRPC6 in each group, and n = 6 for TRPC5 and TRPC3 in each group) as determined by real-time quantitative PCR. TRPC2 and TRPC7 were not detected. Note expanded scales for TRPC5 and TRPC3. Brackets indicate ± SE.

Fig. 5.

Fig. 5.

Expression of TRPC protein relative to α-actin in distal (Dist) and proximal (Prox) PASMC (top) and PA (bottom) as determined by Western blotting. A and C: representative blots for TRPC1, TRPC4, TRPC6, and α-actin in proximal (Prox) and distal (Dist) PASMC and PA. B and D: mean protein expression for TRPC1, TRPC4, and TRPC6 relative to α-actin (n = 3 experiments on samples from 3 animals). Brackets indicate ± SE.

STIM1 mRNA was detected by qPCR in proximal and distal PASMC and PA. In both PASMC and PA, distal expression was two- to threefold greater than proximal expression (P < 0.001; Fig. 6). Western blotting confirmed that STIM1 protein expression was greater in distal PASMC and PA (Fig. 7).

Fig. 6.

Fig. 6.

Expression of stromal interacting molecule 1 (STIM1) mRNA relative to β-actin in proximal and distal PASMC (top; n = 6 in each group) and PA (bottom; n = 6 in each group) as determined by real-time quantitative PCR. Brackets indicate ± SE.

DISCUSSION

Acute hypoxia increased [Ca2+]i in both proximal and distal PASMC, but the increase was greater in distal cells (Fig. 1A). This result is consistent with previous observations in PASMC (25) and suggests that differences in the magnitude of HPV between proximal and distal PA (24, 45, 47) could be caused by differences in Ca2+ signaling in PASMC. In contrast, [Ca2+]i responses to KCl were the same in proximal and distal PASMC (Fig. 1B). This result suggests that activation of VOCC by depolarization was similar in proximal and distal cells and that another signaling pathway must have been responsible for the different [Ca2+]i responses to hypoxia.

Recent evidence indicates that the [Ca2+]i response to hypoxia requires SOCE in distal PASMC (31, 62). To determine if SOCE was different in proximal and distal PASMC, we measured the effects of restoration of extracellular Ca2+ (Fig. 2) or addition of extracellular Mn2+ (Fig. 3) on fura 2 fluorescence in PASMC perfused with Ca2+-free KRBS. The perfusate contained CPA to deplete SR Ca2+ stores and activate SOCC and nifedipine to block Ca2+ entry through L-type VOCC. Responses to both interventions were greater in distal than proximal PASMC, indicating greater SOCE in distal cells. Furthermore, hypoxia increased Mn2+ quenching in proximal and distal PASMC, and this effect was greater in distal cells (Fig. 3). These results indicate that SOCE and the effects of hypoxia on SOCE were greater in distal than proximal PASMC. This difference could explain why [Ca2+]i responses to hypoxia were greater in distal cells (Fig. 1A).

SOCE could be increased because of increases in 1) Ca2+ release from SR or 2) the number and activation of SOCC. With respect to the first possibility, the transient increase in [Ca2+]i caused by CPA during perfusion with Ca2+-free KRBS was not different during normoxia and hypoxia (Fig. 2), suggesting similar SR Ca2+ release and therefore similar store depletion. One possible explanation for this finding is that CPA caused complete depletion of SR Ca2+ stores. In this case, the increase in SOCE caused by hypoxia in the presence of CPA (Fig. 3) could be the result of enhanced activation of SOCC through effects of hypoxia on the channels themselves or transduction mechanisms linking store depletion to channel activation (62). In contrast to hypoxia, the effect of vessel locus on CPA-induced Ca2+ release approached significance, i.e., Δ[Ca2+]i caused by CPA during perfusion with Ca2+-free KRBS tended to be greater in distal than proximal PASMC (P = 0.06; Fig. 2C), suggesting greater release and greater store depletion in distal cells. Potential reasons for such a difference include larger stores and/or more leakage of SR Ca2+ in distal PASMC. Of course, an augmented CPA response could also be the result of lower Ca2+ efflux via sarcolemmal Ca2+-ATPase or Na+/Ca2+ exchange, lower Ca2+ uptake in mitochondria, or less Ca2+ buffering in distal PASMC. Further investigation is needed to confirm the different effects of CPA in proximal and distal PASMC, and to clarify underlying mechanisms.

To evaluate the second possibility, we quantified expression of TRPC, which are thought to compose SOCC (33, 34), and STIM1, which is thought to sense SR Ca2+ and transduce activation of SOCC (12, 15, 20, 28, 36, 39, 55). Consistent with our previous results (61, 63) and reports of other investigators (18, 21, 65, 71, 72), we detected mRNA and protein expression for TRPC1, TRPC4, and TRPC6 in distal PASMC and PA (Figs. 4 and 5). We also detected TRPC3 and TRPC5, which were not identified in our previous studies (61, 63). This inconsistency is probably because of the use of real-time qPCR in the present study, which facilitated detection of less abundant isoforms. TRPC1 was by far the most abundant, followed by TRPC6 and TRPC4, and then much more distantly by TRPC3 and TRPC5 (Fig. 4). This rank order was the same in PASMC and PA, indicating that relative abundance was not altered by cell isolation and culture. It was also similar to that measured by qPCR in mouse distal PASMC (65) and standard PCR in human PASMC (71), but slightly different from standard PCR measurements in rat distal PASMC, where TRPC3 but not TRPC4 was detected (21). Consistent with our previous results (61, 63), but not those of others (21, 65, 71), we did not detect TRPC2 or TRPC7 by qPCR in PASMC or PA, even though both isoforms were detected in positive control tissue (rat brain). The reasons for these discrepancies are not clear.

Like SOCE, expression of TRPC1, TRPC6, and TRPC4 was greater in distal than proximal PASMC and PA (Figs. 4 and 5). Accumulating evidence suggests that these TRPC isoforms may function as SOCC in pulmonary arterial smooth muscle (7, 11, 42). For example, treatment with an antisense oligonucleotide targeted to TRPC1 reduced TRPC1 gene expression and SOCE in human PASMC (53), whereas overexpression of human TRPC1 in rat pulmonary arterial rings enhanced contractile responses to CPA, but not KCl (19). Knockdown of TRPC1 with small-interfering RNA (siRNA) in rat PASMC attenuated cation influx induced by thapsigargin (21). Decreasing TRPC6 expression with an antisense oligonucleotide or increasing TRPC6 expression with mitogens decreased or increased, respectively, Ca2+ entry induced by CPA in rat distal PASMC (72). siRNA targeted to TRPC4 attenuated ATP-mediated increases in TRPC4 expression and SOCE and inhibited ATP-induced proliferation in human PASMC (75). If TRPC1, TRPC4, and TRPC6 were components of SOCC in our PASMC, the data shown in Figs. 4 and 5 suggest that the number of SOCC could be higher in distal cells and lead to greater SOCE and thus greater [Ca2+]i responses to hypoxia. Perhaps inconsistent with this possibility, expression of TRPC5 mRNA was lower in distal than proximal PA tissue (Fig. 4). The functional significance of this result is difficult to predict because TRPC5 mRNA expression was low in both PA and PASMC and equivalent in proximal and distal PASMC (Fig. 4). Nevertheless, TRPC5 can function as a SOCC in systemic vascular smooth muscle (69); therefore, its function in PASMC needs to be determined.

Other evidence suggests that TRPC6 plays an essential role in HPV as a receptor-operated channel. In rat PASMC, siRNA knockdown of TRPC6 decreased cation influx induced by the diacylglycerol (DAG) 1-oleoyl-2-acetyl-sn-glycerol but did not alter influx induced by thapsigargin (21). These results suggested that TRPC6 mediated receptor-operated but not SOCE. In distal PASMC isolated from TRPC6−/− knockout mice and “primed” with endothelin-1 or ANG II, hypoxia had no effect on [Ca2+]i, Mn2+ quenching of fura 2 fluorescence, or cation currents through pathways other than K+ channels or L-type VOCC, whereas all of these variables were increased by hypoxia in wild-type PASMC (65). In addition, the transient early (0–30 min) phase of HPV was abolished in lungs from TRPC6−/− mice. Because R-59949, a DAG kinase inhibitor, increased [Ca2+]i in PASMC from wild-type but not TRPC6−/− mice and caused sarcolemmal accumulation of DAG in both wild-type and TRPC6−/− cells, the authors concluded that TRPC6 knockout abolished early HPV by eliminating DAG-dependent influx of Ca2+ through receptor-operated channels composed of TRPC6. We do not know whether receptor-operated mechanisms contributed to augmentation of the [Ca2+]i response to hypoxia in distal PASMC (Fig. 1A), which exhibited enhanced TRPC6 expression (Figs. 45) but were not primed with vasoactive agents.

Several hypotheses have been proposed to explain how depletion of SR Ca2+ activates SOCC, including 1) SR release of “Ca2+ influx factor,” which liberates sarcolemmal lysophospholipids through activation of phospholipase A2 (41, 49, 50, 56); 2) decreased [Ca2+]i in a restricted space between SOCC and SR, which reverses Ca2+-dependent SOCC inactivation (6, 38); 3) fusion of SOCC-containing vesicles with sarcolemma, which increases the number of SOCC at the cell surface (1, 70); and 4) decreased binding of Ca2+ to regions of inositol trisphosphate receptors within the depleted SR lumen, which leads to “conformational coupling” of the receptor's cytoplasmic regions with sarcolemmal SOCC (8, 17, 35, 40). None of these hypotheses, however, has achieved general acceptance (34).

Recently, it was proposed that depletion of Ca2+ in endoplasmic reticulum (ER) is sensed by the Ca2+-binding protein STIM1, which spans the ER membrane and undergoes structural alteration when Ca2+ dissociates from its EF-hand region within the ER lumen (15, 20, 22, 28, 39, 44). This conformational change is thought to trigger aggregation of STIM1 proteins in regions of ER membrane close to the plasma membrane, where interaction with SOCC or its associated regulatory proteins causes channel activation (15, 52, 68, 76). Thus STIM1 may be both the sensor of ER Ca2+ and transducer of SOCC activation.

Much of the work on STIM1 has been carried out in nonexcitable cells; however, recent evidence indicates that STIM1 is also required for SOCE in excitable cells, including smooth muscle. In human airway, STIM1 was highly expressed in smooth muscle, and siRNA targeted to STIM1 decreased STIM1 mRNA and protein, reduced SOCE, and virtually abolished CPA-induced inward cation currents (36). In human coronary arterial smooth muscle, STIM1 protein was localized predominantly to SR but was also detected in sarcolemma (55). Furthermore, knockdown of STIM1 with STIM1-targeted siRNA drastically decreased SOCE. In mouse aortic smooth muscle cells, RNA interference decreased STIM1 expression and inhibited thapsigargin-induced store-operated cation influx (12). As far as we know, expression of STIM1 in PASMC has not been documented.

We found STIM1 to be highly expressed in both PA and PASMC (Figs. 56). Furthermore, like SOCE (Fig. 23) and expression of TRPC1, TRPC6, and TRPC4 (Fig. 45), STIM1 expression was greater in distal than proximal smooth muscle. If STIM1 sensed SR Ca2+ and transduced SOCC activation in PASMC, as it is thought in other smooth muscles, this difference could contribute to greater SOCE (Figs. 23), greater augmentation of SOCE by hypoxia (Fig. 3), and greater [Ca2+]i responses to hypoxia in distal PASMC.

The coordinate upregulation of STIM1 and TRPC in distal pulmonary arterial smooth muscle is interesting, since interaction of STIM1 with TRPC seems to be essential for the latter to function as SOCC (2, 16, 32, 55, 67, 73). In HEK cells, the structural features necessary for STIM1 to activate SOCC were found to be the same as those required for STIM1 to bind and activate TRPC1 channels (16). In human platelets, Ca2+ store depletion stimulated rapid STIM1 surface expression and association with endogenously expressed TRPC1 (23). In human coronary arterial smooth muscle, an EF-hand mutant of STIM1, STIM1(E87A), markedly increased SOCE, and this increase was abolished by cotransfection with siRNA targeted to TRPC1 (55). These findings indicate that SOCC composed of TRPC1 require STIM1 for activation, and the same may be true for other TRPC (67). Whether similar conclusions apply to PASMC remains to be determined.

In summary, we found that basal SOCE, the increases in SOCE and [Ca2+]i caused by hypoxia, and expression of STIM1 and the most abundant TRPC isoforms (TRPC1, TRPC6, TRPC4) were greater in distal than proximal pulmonary arterial smooth muscle. Because SOCC may be composed of TRPC and activated by STIM1, and SOCE is required for [Ca2+]i responses to hypoxia in PASMC and HPV in isolated lungs, these results suggest that HPV is greater in distal PA because greater numbers and activation of SOCC generate bigger [Ca2+]i responses to hypoxia in distal PASMC. Further investigation of the roles played by SOCE, TRPC, and STIM1 in HPV is warranted.

GRANTS

This work was supported by National Heart, Lung, and Blood Institute Grants HL-51912 (J. T. Sylvester), HL-75113 (J. T. Sylvester), and HL-67191 (L. A. Shimoda), an American Heart Association Scientist Development Grant (J. Wang), National Institutes of Health Independent Scientist Award HL-079981 (J. Wang), and an American Lung Association of Maryland Research Grant (J. Wang).

The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

REFERENCES

  • 1.Alderton JM, Ahmed SA, Smith LA, Steinhardt RA. Evidence for a vesicle-mediated maintenance of store-operated calcium channels in a human embryonic kidney cell line. Cell Calcium 28: 161–169, 2000. [DOI] [PubMed] [Google Scholar]
  • 2.Ambudkar IS, Ong HL, Liu X, Bandyopadhyay B, Cheng KT. TRPC1: the link between functionally distinct store-operated calcium channels. Cell Calcium 42: 213–223, 2007. [DOI] [PubMed] [Google Scholar]
  • 3.Archer SL, Huang JM, Reeve HL, Hampl V, Tolarova S, Michelakis E, Weir EK. Differential distribution of electrophysiologically distinct myocytes in conduit and resistance arteries determines their response to nitric oxide and hypoxia. Circ Res 78: 431–442, 1996. [DOI] [PubMed] [Google Scholar]
  • 4.Archer SL, Wu XC, Thebaud B, Nsair A, Bonnet S, Tyrrell B, McMurtry MS, Hashimoto K, Harry G, Michelakis ED. Preferential expression and function of voltage-gated, O2-sensitive K+ channels in resistance pulmonary arteries explains regional heterogeneity in hypoxic pulmonary vasoconstriction: ionic diversity in smooth muscle cells. Circ Res 95: 308–318, 2004. [DOI] [PubMed] [Google Scholar]
  • 5.Bakhramov A, Evans AM, Kozlowski RZ. Differential effects of hypoxia on the intracellular Ca2+ concentration of myocytes isolated from different regions of the rat pulmonary arterial tree. Exp Physiol 83: 337–347, 1998. [DOI] [PubMed] [Google Scholar]
  • 6.Barritt GJ Does a decrease in subplasmalemmal Ca2+ explain how store-operated Ca2+ channels are opened? Cell Calcium 23: 65–75, 1998. [DOI] [PubMed] [Google Scholar]
  • 7.Beech DJ Emerging functions of 10 types of TRP cationic channel in vascular smooth muscle. Clin Exp Pharmacol Physiol 32: 597–603, 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Berridge MJ Capacitative calcium entry. Biochem J 312: 1–11, 1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Coppock EA, Tamkun MM. Differential expression of KV channel alpha- and beta-subunits in the bovine pulmonary arterial circulation. Am J Physiol Lung Cell Mol Physiol 281: L1350–L1360, 2001. [DOI] [PubMed] [Google Scholar]
  • 10.Davie N, Haleen SJ, Upton PD, Polak JM, Yacoub MH, Morrell NW, Wharton J. ETA and ETB receptors modulate the proliferation of human pulmonary artery smooth muscle cells. Am J Respir Crit Care Med 165: 398–405, 2002. [DOI] [PubMed] [Google Scholar]
  • 11.Dietrich A, Kalwa H, Fuchs B, Grimminger F, Weissmann N, Gudermann T. In vivo TRPC functions in the cardiopulmonary vasculature. Cell Calcium 42: 233–244, 2007. [DOI] [PubMed] [Google Scholar]
  • 12.Dietrich A, Kalwa H, Storch U, Mederos YSM, Salanova B, Pinkenburg O, Dubrovska G, Essin K, Gollasch M, Birnbaumer L, Gudermann T. Pressure-induced and store-operated cation influx in vascular smooth muscle cells is independent of TRPC1. Pflugers Arch 455: 465–477, 2007. [DOI] [PubMed] [Google Scholar]
  • 13.Franco-Obregon A, Lopez-Barneo J. Differential oxygen sensitivity of calcium channels in rabbit smooth muscle cells of conduit and resistance pulmonary arteries. J Physiol (Lond) 491: 511–518, 1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Golovina VA, Platoshyn O, Bailey CL, Wang J, Limsuwan A, Sweeney M, Rubin LJ, Yuan JX. Upregulated TRP and enhanced capacitative Ca2+ entry in human pulmonary artery myocytes during proliferation. Am J Physiol Heart Circ Physiol 280: H746–H755, 2001. [DOI] [PubMed] [Google Scholar]
  • 15.Hewavitharana T, Deng X, Soboloff J, Gill DL. Role of STIM and Orai proteins in the store-operated calcium signaling pathway. Cell Calcium 42: 173–182, 2007. [DOI] [PubMed] [Google Scholar]
  • 16.Huang GN, Zeng W, Kim JY, Yuan JP, Han L, Muallem S, Worley PF. STIM1 carboxyl-terminus activates native SOC, Icrac and TRPC1 channels. Nat Cell Biol 8: 1003–1010, 2006. [DOI] [PubMed] [Google Scholar]
  • 17.Irvine RF “Quantal” Ca2+ release and the control of Ca2+ entry by inositol phosphates–a possible mechanism. FEBS Lett 263: 5–9, 1990. [DOI] [PubMed] [Google Scholar]
  • 18.Kunichika N, Landsberg JW, Yu Y, Kunichika H, Thistlethwaite PA, Rubin LJ, Yuan JX. Bosentan inhibits transient receptor potential channel expression in pulmonary vascular myocytes. Am J Respir Crit Care Med 170: 1101–1107, 2004. [DOI] [PubMed] [Google Scholar]
  • 19.Kunichika N, Yu Y, Remillard CV, Platoshyn O, Zhang S, Yuan JX. Overexpression of TRPC1 enhances pulmonary vasoconstriction induced by capacitative Ca2+ entry. Am J Physiol Lung Cell Mol Physiol 287: L962–L969, 2004. [DOI] [PubMed] [Google Scholar]
  • 20.Lewis RS The molecular choreography of a store-operated calcium channel. Nature 446: 284–287, 2007. [DOI] [PubMed] [Google Scholar]
  • 21.Lin MJ, Leung GP, Zhang WM, Yang XR, Yip KP, Tse CM, Sham JS. Chronic hypoxia-induced upregulation of store-operated and receptor-operated Ca2+ channels in pulmonary arterial smooth muscle cells: a novel mechanism of hypoxic pulmonary hypertension. Circ Res 95: 496–505, 2004. [DOI] [PubMed] [Google Scholar]
  • 22.Liou J, Kim ML, Heo WD, Jones JT, Myers JW, Ferrell JE Jr, Meyer T. STIM is a Ca2+ sensor essential for Ca2+-store-depletion-triggered Ca2+ influx. Curr Biol 15: 1235–1241, 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Lopez JJ, Salido GM, Pariente JA, Rosado JA. Interaction of STIM1 with endogenously expressed human canonical TRP1 upon depletion of intracellular Ca2+ stores. J Biol Chem 281: 28254–28264, 2006. [DOI] [PubMed] [Google Scholar]
  • 24.Madden JA, Dawson CA, Harder DR. Hypoxia-induced activation in small isolated pulmonary arteries from the cat. J Appl Physiol 59: 113–118, 1985. [DOI] [PubMed] [Google Scholar]
  • 25.Madden JA, Vadula MS, Kurup VP. Effects of hypoxia and other vasoactive agents on pulmonary and cerebral artery smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 263: L384–L393, 1992. [DOI] [PubMed] [Google Scholar]
  • 26.McCulloch KM, Kempsill FE, Buchanan KJ, Gurney AM. Regional distribution of potassium currents in the rabbit pulmonary arterial circulation. Exp Physiol 85: 487–496, 2000. [PubMed] [Google Scholar]
  • 27.McDaniel SS, Platoshyn O, Wang J, Yu Y, Sweeney M, Krick S, Rubin LJ, Yuan JX. Capacitative Ca2+ entry in agonist-induced pulmonary vasoconstriction. Am J Physiol Lung Cell Mol Physiol 280: L870–L880, 2001. [DOI] [PubMed] [Google Scholar]
  • 28.Minke B TRP channels and Ca2+ signaling. Cell Calcium 40: 261–275, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Minke B, Cook B. TRP channel proteins and signal transduction. Physiol Rev 82: 429–472, 2002. [DOI] [PubMed] [Google Scholar]
  • 30.Ng LC, Gurney AM. Store-operated channels mediate Ca2+ influx and contraction in rat pulmonary artery. Circ Res 89: 923–929, 2001. [DOI] [PubMed] [Google Scholar]
  • 31.Ng LC, Wilson SM, Hume JR. Mobilization of SR stores by hypoxia leads to consequent activation of capacitative Ca2+ entry in isolated canine pulmonary arterial smooth muscle cells. J Physiol 563: 409–419, 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ong HL, Cheng KT, Liu X, Bandyopadhyay BC, Paria BC, Soboloff J, Pani B, Gwack Y, Srikanth S, Singh BB, Gill DL, Ambudkar IS. Dynamic assembly of TRPC1-STIM1-Orai1 ternary complex is involved in store-operated calcium influx. Evidence for similarities in store-operated and calcium release-activated calcium channel components. J Biol Chem 282: 9105–9116, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Parekh AB, Penner R. Store depletion and calcium influx. Physiol Rev 77: 901–930, 1997. [DOI] [PubMed] [Google Scholar]
  • 34.Parekh AB, Putney JW Jr. Store-operated calcium channels. Physiol Rev 85: 757–810, 2005. [DOI] [PubMed] [Google Scholar]
  • 35.Patterson RL, van Rossum DB, Gill DL. Store-operated Ca2+ entry: evidence for a secretion-like coupling model. Cell 98: 487–499, 1999. [DOI] [PubMed] [Google Scholar]
  • 36.Peel SE, Liu B, Hall IP. A key role for STIM1 in store operated calcium channel activation in airway smooth muscle (Abstract). Respir Res 7: 119, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Pfaffl MW A new mathematical model for relative quantification in real-time RT-PCR (Abstract). Nucleic Acids Res 29: e45, 2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Putney JW A model for receptor-regulated calcium entry. Cell Calcium 7: 1–12, 1986. [DOI] [PubMed] [Google Scholar]
  • 39.Putney JW Recent breakthroughs in the molecular mechanism of capacitative calcium entry (with thoughts on how we got here). Cell Calcium 42: 103–110, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Putney JW, Broad LM, Braun FJ, Lievremont JP, Bird GS. Mechanisms of capacitative calcium entry. J Cell Sci 114: 2223–2229, 2001. [DOI] [PubMed] [Google Scholar]
  • 41.Randriamampita C, Tsien RY. Emptying of intracellular Ca2+ stores releases a novel small messenger that stimulates Ca2+ influx. Nature 364: 809–814, 1993. [DOI] [PubMed] [Google Scholar]
  • 42.Remillard CV, Yuan JX. TRP channels, CCE, and the pulmonary vascular smooth muscle. Microcirculation 13: 671–692, 2006. [DOI] [PubMed] [Google Scholar]
  • 43.Resnik ER, Keck M, Sukovich DJ, Herron JM, Cornfield DN. Chronic intrauterine pulmonary hypertension increases capacitative calcium entry in fetal pulmonary artery smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 292: L953–L959, 2007. [DOI] [PubMed] [Google Scholar]
  • 44.Roos J, DiGregorio PJ, Yeromin AV, Ohlsen K, Lioudyno M, Zhang S, Safrina O, Kozak JA, Wagner SL, Cahalan MD, Velicelebi G, Stauderman KA. STIM1, an essential and conserved component of store-operated Ca2+ channel function. J Cell Biol 169: 435–445, 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Schwenke DO, Pearson JT, Umetani K, Kangawa K, Shirai M. Imaging of the pulmonary circulation in the closed-chest rat using synchrotron radiation microangiography. J Appl Physiol 102: 787–793, 2007. [DOI] [PubMed] [Google Scholar]
  • 46.Sham JS, Crenshaw BR, Deng LH, Shimoda LA, Sylvester JT. Effects of hypoxia in porcine pulmonary arterial myocytes: roles of KV channel and endothelin-1. Am J Physiol Lung Cell Mol Physiol 279: L262–L272, 2000. [DOI] [PubMed] [Google Scholar]
  • 47.Shirai M, Sada K, Ninomiya I. Effects of regional alveolar hypoxia and hypercapnia on small pulmonary vessels in cats. J Appl Physiol 61: 440–448, 1986. [DOI] [PubMed] [Google Scholar]
  • 48.Smani T, Iwabuchi S, Lopez-Barneo J, Urena J. Differential segmental activation of Ca2+-dependent CI- and K+ channels in pulmonary arterial myocytes. Cell Calcium 29: 369–377, 2001. [DOI] [PubMed] [Google Scholar]
  • 49.Smani T, Zakharov SI, Csutora P, Leno E, Trepakova ES, Bolotina VM. A novel mechanism for the store-operated calcium influx pathway. Nat Cell Biol 6: 113–120, 2004. [DOI] [PubMed] [Google Scholar]
  • 50.Smani T, Zakharov SI, Leno E, Csutora P, Trepakova ES, Bolotina VM. Ca2+-independent phospholipase A2 is a novel determinant of store-operated Ca2+ entry. J Biol Chem 278: 11909–11915, 2003. [DOI] [PubMed] [Google Scholar]
  • 51.Smirnov SV, Beck R, Tammaro P, Ishii T, Aaronson PI. Electrophysiologically distinct smooth muscle cell subtypes in rat conduit and resistance pulmonary arteries. J Physiol 538: 867–878, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Spassova MA, Soboloff J, He LP, Xu W, Dziadek MA, Gill DL. STIM1 has a plasma membrane role in the activation of store-operated Ca2+ channels. Proc Natl Acad Sci USA 103: 4040–4045, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Sweeney M, Yu Y, Platoshyn O, Zhang S, McDaniel SS, Yuan JX. Inhibition of endogenous TRP1 decreases capacitative Ca2+ entry and attenuates pulmonary artery smooth muscle cell proliferation. Am J Physiol Lung Cell Mol Physiol 283: L144–L155, 2002. [DOI] [PubMed] [Google Scholar]
  • 54.Takahashi H, Soma S, Muramatsu M, Oka M, Ienaga H, Fukuchi Y. Discrepant distribution of big endothelin (ET)-1 and ET receptors in the pulmonary artery. Eur Respir J 18: 5–14, 2001. [DOI] [PubMed] [Google Scholar]
  • 55.Takahashi Y, Watanabe H, Murakami M, Ono K, Munehisa Y, Koyama T, Nobori K, Iijima T, Ito H. Functional role of stromal interaction molecule 1 (STIM1) in vascular smooth muscle cells. Biochem Biophys Res Commun 361: 934–940, 2007. [DOI] [PubMed] [Google Scholar]
  • 56.Trepakova ES, Csutora P, Hunton DL, Marchase RB, Cohen RA, Bolotina VM. Calcium influx factor directly activates store-operated cation channels in vascular smooth muscle cells. J Biol Chem 275: 26158–26163, 2000. [DOI] [PubMed] [Google Scholar]
  • 57.Urena J, Franco-Obregon A, Lopez-Barneo J. Contrasting effects of hypoxia on cytosolic Ca2+ spikes in conduit and resistance myocytes of the rabbit pulmonary artery. J Physiol (Lond) 496: 103–109, 1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Urena J, Smani T, Lopez-Barneo J. Differential functional properties of Ca2+ stores in pulmonary arterial conduit and resistance myocytes. Cell Calcium 36: 525–534, 2004. [DOI] [PubMed] [Google Scholar]
  • 59.Vadula MS, Kleinman JG, Madden JA. Effect of hypoxia and norepinephrine on cytoplasmic free Ca2+ in pulmonary and cerebral arterial myocytes. Am J Physiol Lung Cell Mol Physiol 265: L591–L597, 1993. [DOI] [PubMed] [Google Scholar]
  • 60.Walker RL, Hume JR, Horowitz B. Differential expression and alternative splicing of TRP channel genes in smooth muscles. Am J Physiol Cell Physiol 280: C1184–C1192, 2001. [DOI] [PubMed] [Google Scholar]
  • 61.Wang J, Shimoda LA, Sylvester JT. Capacitative calcium entry and TRPC channel proteins are expressed in rat distal pulmonary arterial smooth muscle. Am J Physiol Lung Cell Mol Physiol 286: L848–L858, 2004. [DOI] [PubMed] [Google Scholar]
  • 62.Wang J, Shimoda LA, Weigand L, Wang W, Sun D, Sylvester JT. Acute hypoxia increases intracellular [Ca2+] in pulmonary arterial smooth muscle by enhancing capacitative Ca2+ entry. Am J Physiol Lung Cell Mol Physiol 288: L1059–L1069, 2005. [DOI] [PubMed] [Google Scholar]
  • 63.Wang J, Weigand L, Lu W, Sylvester JT, Semenza GL, Shimoda LA. Hypoxia inducible factor 1 mediates hypoxia-induced TRPC expression and elevated intracellular Ca2+ in pulmonary arterial smooth muscle cells. Circ Res 98: 1528–1537, 2006. [DOI] [PubMed] [Google Scholar]
  • 64.Weigand L, Foxson J, Wang J, Shimoda LA, Sylvester JT. Inhibition of hypoxic pulmonary vasoconstriction by antagonists of store-operated Ca2+ and nonselective cation channels. Am J Physiol Lung Cell Mol Physiol 289: L5–L13, 2005. [DOI] [PubMed] [Google Scholar]
  • 65.Weissmann N, Dietrich A, Fuchs B, Kalwa H, Ay M, Dumitrascu R, Olschewski A, Storch U, Mederos YSM, Ghofrani HA, Schermuly RT, Pinkenburg O, Seeger W, Grimminger F, Gudermann T. Classical transient receptor potential channel 6 (TRPC6) is essential for hypoxic pulmonary vasoconstriction and alveolar gas exchange. Proc Natl Acad Sci USA 103: 19093–19098, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Whitman EM, Pisarcik S, Luke T, Fallon M, Wang J, Sylvester JT, Semenza GL, Shimoda LA. Endothelin-1 mediates hypoxia-induced inhibition of voltage-gated K+ channel expression in pulmonary arterial myocytes. Am J Physiol Lung Cell Mol Physiol 294: L309–L318, 2008. [DOI] [PubMed] [Google Scholar]
  • 67.Worley PF, Zeng W, Huang GN, Yuan JP, Kim JY, Lee MG, Muallem S. TRPC channels as STIM1-regulated store-operated channels. Cell Calcium 42: 205–211, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Wu MM, Buchanan J, Luik RM, Lewis RS. Ca2+ store depletion causes STIM1 to accumulate in ER regions closely associated with the plasma membrane. J Cell Biol 174: 803–813, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Xu SZ, Boulay G, Flemming R, Beech DJ. E3-targeted anti-TRPC5 antibody inhibits store-operated calcium entry in freshly isolated pial arterioles. Am J Physiol Heart Circ Physiol 291: H2653–H2659, 2006. [DOI] [PubMed] [Google Scholar]
  • 70.Yao Y, Ferrer-Montiel AV, Montal M, Tsien RY. Activation of store-operated Ca2+ current in Xenopus oocytes requires SNAP-25 but not a diffusible messenger. Cell 98: 475–485, 1999. [DOI] [PubMed] [Google Scholar]
  • 71.Yu Y, Fantozzi I, Remillard CV, Landsberg JW, Kunichika N, Platoshyn O, Tigno DD, Thistlethwaite PA, Rubin LJ, Yuan JX. Enhanced expression of transient receptor potential channels in idiopathic pulmonary arterial hypertension. Proc Natl Acad Sci USA 101: 13861–13866, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Yu Y, Sweeney M, Zhang S, Platoshyn O, Landsberg J, Rothman A, Yuan JX. PDGF stimulates pulmonary vascular smooth muscle cell proliferation by upregulating TRPC6 expression. Am J Physiol Cell Physiol 284: C316–C330, 2003. [DOI] [PubMed] [Google Scholar]
  • 73.Yuan JP, Zeng W, Huang GN, Worley PF, Muallem S. STIM1 heteromultimerizes TRPC channels to determine their function as store-operated channels. Nat Cell Biol 9: 636–645, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Zhang S, Patel HH, Murray F, Remillard CV, Schach C, Thistlethwaite PA, Insel PA, Yuan JX. Pulmonary artery smooth muscle cells from normal subjects and IPAH patients show divergent cAMP-mediated effects on TRPC expression and capacitative Ca2+ entry. Am J Physiol Lung Cell Mol Physiol 292: L1202–L1210, 2007. [DOI] [PubMed] [Google Scholar]
  • 75.Zhang S, Remillard CV, Fantozzi I, Yuan JX. ATP-induced mitogenesis is mediated by cyclic AMP response element-binding protein-enhanced TRPC4 expression and activity in human pulmonary artery smooth muscle cells. Am J Physiol Cell Physiol 287: C1192–C1201, 2004. [DOI] [PubMed] [Google Scholar]
  • 76.Zhang SL, Yu Y, Roos J, Kozak JA, Deerinck TJ, Ellisman MH, Stauderman KA, Cahalan MD. STIM1 is a Ca2+ sensor that activates CRAC channels and migrates from the Ca2+ store to the plasma membrane. Nature 437: 902–905, 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Physiology. Lung Cellular and Molecular Physiology are provided here courtesy of American Physiological Society

RESOURCES