Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Am J Physiol Cell Physiol. 2008 Sep; 295(3): C768–C778.
Published online 2008 Jul 2. doi: 10.1152/ajpcell.00174.2008
PMCID: PMC2544444
PMID: 18596213

Cytoskeletal remodeling in differentiated vascular smooth muscle is actin isoform dependent and stimulus dependent

Abstract

Dynamic remodeling of the actin cytoskeleton plays an essential role in the migration and proliferation of vascular smooth muscle cells. It has been suggested that actin remodeling may also play an important functional role in nonmigrating, nonproliferating differentiated vascular smooth muscle (dVSM). In the present study, we show that contractile agonists increase the net polymerization of actin in dVSM, as measured by the differential ultracentrifugation of vascular smooth muscle tissue and the costaining of single freshly dissociated cells with fluorescent probes specific for globular and filamentous actin. Furthermore, induced alterations of the actin polymerization state, as well as actin decoy peptides, inhibit contractility in a stimulus-dependent manner. Latrunculin pretreatment or actin decoy peptides significantly inhibit contractility induced by a phorbol ester or an α-agonist, but these procedures have no effect on contractions induced by KCl. Aorta dVSM expresses α-smooth muscle actin, β-actin, nonmuscle γ-actin, and smooth muscle γ-actin. The incorporation of isoform-specific cell-permeant synthetic actin decoy peptides, as well as isoform-specific probing of cell fractions and two-dimensional gels, demonstrates that actin remodeling during α-agonist contractions involves the remodeling of primarily γ-actin and, to a lesser extent, β-actin. Taken together, these results show that net isoform- and agonist-dependent increases in actin polymerization regulate vascular contractility.

Keywords: decoy peptide, β-actin, γ-actin, contractility

actin makes up ∼20% of total protein content in smooth muscle (4). Its importance is highlighted by the fact that it is highly conserved throughout evolution and across species (4, 21). Six isoforms, separate gene products, are present in vertebrate muscles (60). Even among these actin isoforms, there is a large degree of sequence conservation, with over 95% homology at the protein level (61). Most sequence differences are clustered at the NH2-terminal region (21, 61), and the sequence differences in the NH2-terminal region of actin result in a small difference (0.02 units) in the isoelectric point of the isoforms (18), which allows the separation of the isoforms by two-dimensional gel electrophoresis. The NH2-terminal region of actin isoforms is also known to be involved in the binding of many actin-binding proteins (52) and has been suggested to result in isoform-specific functions, largely based on experiments performed in cultured cells (21, 27).

Since the realization that multiple actin isoforms can be expressed within a single cell type, considerable interest has focused on the question of whether such actin isoforms are differentially localized and perform different functions. Despite the importance of actin in diverse cellular functions, the exact functions and locations of actin isoforms have yet to be fully understood. North et al. (46) have reported that actin isoforms in chicken gizzard cells differ in their spatial localization and implied isoform-specific functions based their localization. In contrast, the Murphy laboratory (12) has reported a homogeneous distribution of actin isoforms in mammalian vascular smooth muscle (VSM). Whether the apparent discrepancy is due to tissue, species, or methodological differences has not been clear.

The cytoskeleton in differentiated VSM (dVSM) has generally been assumed to be largely static, performing a solely structural role, compared with the dynamic cytoskeleton of migrating, proliferative VSM cells (VSMCs) (56). However, a considerable literature on airway smooth muscle cells has documented a significant actin cytoskeletal remodeling, particularly in the setting of airway narrowing (32, 35, 41). Recently, a few laboratories have also suggested that actin cytoskeletal remodeling may occur to a significant degree in dVSM, specifically during stretch and hydrodynamic stimuli, which have been linked to changes in actin polymerization patterns (7, 40). However, conflicting results on agonist-induced cytoskeletal remodeling have been reported. Flavahan et al. (16) reported that α-agonists had no effect on actin remodeling, whereas Ohanian et al. (48) and Ward et al. (62) reported that α-agonists induced the tyrosine phosphorylation of adhesion plaque proteins expected to lead to cytoskeletal remodeling. Latrunculin and cytochalasin, agents known to decrease actin polymerization, have also been reported to inhibit agonist-induced contractility and tone maintenance (2, 3, 53, 54, 64, 66). However, it is difficult to discriminate between the possible nonphysiological effects of latrunculin and cytochalasin on force via a direct alteration of contractile filament content versus effects on individual signal transduction pathways involving cytoskeletal remodeling.

We report here, by a combination of cellular and biochemical approaches, a stimulus- and isoform-dependent cytoskeletal remodeling of dVSMC. Furthermore, we show that alterations in the actin cytoskeleton caused by chemical agents or decoy peptides result in significant alterations in vascular tone in a stimulus-dependent manner.

MATERIALS AND METHODS

Tissue preparation and force measurements.

All procedures were performed according to protocols approved by the Institutional Care and Use Committee. As previously described (37), ferrets (Marshall Farms, North Rose, NY) were euthanized by an overdose of chloroform, and the aorta was quickly removed from animals and dissected in oxygenated (95% O2-5% CO2) physiological salt solution (PSS) consisting of (in mM) 120 NaCl, 5.9 KCl, 1.2 NaH2PO4, 25 NaHCO3, 11.5 dextrose, 1 CaCl2, and 1.4 MgCl2 (pH 7.4). The aorta was carefully dissected to remove all connective tissue and the endothelium under a dissecting microscope. The aorta was cut into circular rings (∼3 mm wide), which were then attached to a force transducer and stretched to ∼1.3× their resting length (a resting tension of ∼2.0 g; previously determined to estimate optimal length). The rings were allowed to equilibrate at 37°C for 1 h in oxygenated PSS and were then stimulated for up to 10 min with 51 mM KCl-PSS (51 mM of NaCl replaced with KCl in PSS). After the rings were allowed to reach maximal, steady-state force, the depolarizing solution was washed three times with PSS. The rings were then allowed to relax for 1 h before beginning the experiment. The aorta rings were quick-frozen by immersion into dry ice-acetone slurry containing 10 mM dithiothreitol. The concentrations of agonist used here were chosen to be exactly those used in our past studies using the same tissue and thus allow direct comparison. The concentrations used are also maximally effective concentrations and allow us to make accurate measurements with the techniques used in this article.

Differential ultracentrifugation of homogenates for ratios of filamentous- and globular-actin.

The protocol was according to manufacturer's instructions (Cytoskeleton, Denver, CO) with some modifications. Each muscle strip (∼20 mg wet weight) was placed into a glass homogenizer tube (Kimble Kontes, Vineland, NJ) and quickly homogenized by a motorized tissue grinder in 200 μl of lysis buffer (37°C); 50 mM PIPES (pH 6.9), 50 mM NaCl, 5 mM MgCl2, 5 mM EGTA, 5% (vol/vol) glycerol, 0.1% Nonidet P-40 (NP-40), 0.1% Triton X-100, 0.1% Tween 20, 0.1% 2-mercapto-ethanol, 0.001% antifoam C with 100 mM ATP and a cocktail of protease inhibitors; and 0.4 mM tosyl arginine methyl ester, 1.5 mM leupeptin, 1 mM pepstatin A, and 1 M benzamidine (Cytoskeleton). Tissue homogenates were placed on ice until all samples are completed and then incubated at 37°C for 10 min. Tissue homogenates were transferred to a prewarmed (37°C) ultracentrifuge and spun at 150,000 g for 1 h at 37°C to separate the globular (G)-actin (supernatant) and filamentous (F)-actin fractions (Beckman, Fullerton, CA). The pellets were resuspended with the same amount of volume of ice-cooled distilled water containing 10 μM cytochalasin D. The pellets were dissolved by trituration with a pipette and left on ice for 1 h, vortexing every 15 min for 1 min to dissociate F-actin. Resuspended solutions were centrifuged at 2,300 g for 5 min at 4°C. The second supernatants (F-actin) were collected, and all samples were diluted with appropriate loading buffer and boiled for 5 min. The samples were stored at −80°C until further separation by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and immunoblotting.

Immunoblotting.

Quick-frozen tissue samples were homogenized and analyzed via immunoblotting as previously published (37). Briefly, the homogenized samples were boiled in Laemmli sample buffer. The samples were separated using SDS-PAGE and transferred to a polyvinylidene difluoride membrane (Millipore). Protein transfer was confirmed by staining the membrane with Ponceau S red. The membrane was stained with an appropriate primary antibody overnight at 4°C. After extensive wash with Tris-buffered saline-Tween 20, the membrane was stained with an appropriate secondary antibody. Densitometry of the actin bands was performed with the software for an Odyssey infrared imaging system (LI-COR Biosciences).

Single cell isolation from aorta.

Cells were freshly enzymatically dissociated from aorta tissue by a previously published method (37). Briefly, the aorta was carefully dissected to remove all connective tissue and the endothelium under a dissecting microscope. About 100 mg (wet weight) of aorta tissue was then cut into 2 × 2-mm pieces and placed into a silicon-coated flask containing 7.5 ml Ca2+, Mg2+-free Hanks' balanced salt solution (HBSS), ∼100 U/ml type II collagenase (Worthington), ∼1.0 U/ml elastase (Roche Diagnostics), and 0.2% BSA (Sigma). The flask was secured in a shaking water bath (rate, ∼100 cycles/min; and temperature, ∼34°C) under an atmosphere of 100% oxygen for 80 min. The tissue pieces were filtered on a nylon mesh (pore size, ∼0.5 mm) and washed with ∼12 ml Ca2+, Mg2+-free HBSS, and 0.2% BSA. The wash buffer containing the dissociated cells was collected and poured over glass coverslips on ice under an atmosphere of 100% oxygen. Cells were allowed to adhere to the coverslips for ∼1 h. After the first digestion, three additional digestions were performed with 50% less elastase for ∼20–30 min. After each of these additional digestions, the tissue pieces were filtered and washed, and the dissociated cells were allowed to adhere to coverslips for ∼1 h. The coverslips were washed with Ca2+, Mg2+-free HBSS, and 0.2% BSA, and Ca2+ and Mg2+ were then reintroduced to the dVSMCs by gradually adding CaCl2 and MgCl2 into the medium surrounding the coverslips until they reached concentrations of 1 mM and 1.2 mM, respectively. In one coverslip of each batch, it was confirmed that the cells were capable of shortening in sucrose-free PSS. Other coverslips were put in a PSS containing 300 mM sucrose, which increases the tonicity of the solution and prevents shortening directly at the cross bridge level (20), allowing use of the coverslips for imaging. Sucrose HBSS has been shown to cause no change in resting or activated Ca2+ levels, PKC signaling, ERK activation time courses, cytoskeletal structure, or resting cell length in differentiated smooth muscle cells (dSMCs) (10, 28, 31, 50, 51).

Cell staining and imaging.

All stimulated and unstimulated dSMCs were fixed with 4% paraformaldehyde for 10 min in 100 mM sodium phosphate buffer. The fixative was quenched by washing the cells with 0.1 mM glycine in 1% BSA HBSS. The cell membranes were then permeabilized using 0.1% Triton X-100 in 1% BSA HBSS. Nonspecific binding sites were blocked by incubating the cells with 10% goat serum in 0.05% Triton X-100 and 1% BSA HBSS. After blocking, the cells were incubated with the desired primary antibody diluted in 2% goat serum, 0.05% Triton X-100, and 1% BSA HBSS. Cells were incubated with the appropriate secondary antibody diluted in HBSS. Cells were washed six times with HBSS and mounted with FluorSave (Calbiochem). Cells were costained with DNase I (Alexa Fluor 488; 300 nM) and phalloidin (Alexa Fluor 568; 1:50 dilution) for 20 min. Latrunculin B (30 nM) was used for pretreatement for 45 min. Images of immunostained dSMCs were taken with a Kr/Ar laser (BioRad Radiance 2000) scanning confocal microscope equipped with a Nikon X-60 [numerical aperature (NA), 1.4] oil immersion objective. Images were recorded with Laser Sharp 2000 (Bio-Rad) software. In all cases, it was confirmed that there was no detectable background fluorescence by staining cells in the absence of a primary antibody. In addition, for all colabeling experiments, it was confirmed that there was no detectable cross talk between fluorescent labels by exchanging excitation/emission filters on single-labeled cells.

Digital confocal microscopy for phalloidin-to-DNase I ratio.

Ratio measurements for phalloidin-to-DNase I ratios followed a modification of previously published methods (17). Phalloidin has been shown to be highly specific for F-actin, and DNase I was used to identify G-actin and discriminate it from F-actin (34). Images were acquired using a Kr/Ar laser (Bio-Rad Radiance 2000) scanning confocal microscope equipped with a Nikon X-60 (NA, 1.4) oil immersion objective. All images were digitally scanned at 166 lines/s, Kalman averaged three times with minimal pinhole size, and quantified with Bio-Rad Laser Sharp 2000 for Windows NT. For each cell, the single central nucleus was first located at the widest and thickest part of the cell. In previously published studies, nuclear staining confirmed that visual inspection gives accurate identification of the nuclear location (29). The position of the nucleus was especially obvious in the current study, since the phalloidin staining was characteristically decreased in the nuclear region. Others have also note that nuclear stains give an identical image to that seen in the bright field image of fixed/permeabilized cells (26). The stage was moved so that the nucleus was positioned to one side of the field of view. We used center optical sections and drew each box to extend from cell edge to cell edge. We used three regions of interest (ROIs) per cell in relatively straight tail regions and excluded the nucleus area. In most cases, we covered the majority of the straight, nonnuclear space (Fig. 2A, inset). The average integrated intensity per pixel within each ROI was determined for each wavelength. The ratio of the value for phalloidin was divided by that for DNase I. The ratio values for the three ROIs for each cell were averaged, and the final number was taken as an index of the ratio of F-actin-to-G-actin (F/G ratio) for that cell as previously described (34). One advantage of the phalloidin/DNase I method used here is that the relative abundance of structures too small to resolve by confocal microscopy (such as G-actin monomers) can be quantitated by integrating the fluorescent signal.

An external file that holds a picture, illustration, etc.
Object name is zh00090857010002.jpg

Single cell F/G actin turnover demonstrated by phalloidin and DNase I costaining. A: illustrative cell images stained with Alexa 488-DNase I (G-actin; green; left), Alexa 568-phalloidin (F-actin; red; middle), and merged images (right) in unstimulated, Lat (0.03 μM), KCl (51 mM), DPBA (10 μM), PE (10 μM), or jasplakinolide (Jas; 0.5 μM)-treated dVSM cell (dVSMC). Inset: diagram of cell with white boxes to illustrate how regions of interest (ROIs) were defined. B: quantitative analysis of ROIs stained with fluorescent phalloidin and DNase I in cells. **P < 0.01, compared with unstimulated cells. Average is ± SE for 8–41 cells.

Two-dimensional gel electrophoresis.

Two-dimensional gel electrophoresis was performed following previously published methods (15, 46, 47). Briefly, samples were homogenized in a buffer containing 9.5 M Urea, 2.0% NP-40, 5% β-mercaptoethanol, 1.6% ampholines (pH 4–6.5), and 0.4% ampholines (pH 3–10). The isoelectric focusing gels were cast according to O′Farrell's method (47) except that the ampholyte mixture was restricted to the 4–6.5 intervals [9.2 M Urea, 4% acrylamide, 2% NP-40, 1.6% ampholines (pH 4–6.5), 0.4% ampholines (pH 3–10), 0.01% ammonium persulfate, and 0.1% N,N,N′,N′-tetramethylethylenediamine]. The gels were allowed to polymerize overnight at room temperature and were prefocused at room temperature using 20 mM NaOH (anode solution) and 20 mM phosphoric acid (cathode solution) for 15 min at 200 V, 15 min at 300 V, and 15 min at 400 V. Isoelectric focusing was performed for 3 h at 400 V and 15 to 16 h at 500 V using Hoefer Scientific tube gel equipment. The second-dimension gels were run as a regular SDS-PAGE. The first-dimension tube gel was transferred to the slab gel by running buffer containing 250 mM Tris, 1.92 M glycine, and 1% SDS.

Synthesis of NH2-terminal peptide of actin isoforms.

Five tetra- or penta-peptides representing the NH2-terminal actin isoform sequence were synthesized by solid-state peptide synthesis using fluorenylmethoxycarbonyl chemistry. The isoforms, acetylated at their NH2 termini, were α-skeletal muscle actin, Ac-DEDE; γ-smooth muscle actin, Ac-EEETT; γ-cytoplasmic actin, Ac-EEEIA; β-cytoplasmic actin, Ac-DDDIA; and α-smooth muscle actin, Ac-EEED. All isoforms were synthetically coupled to a TAT protein transduction domain comprising residues YGRKKRRQRRR.

To add a fluorescent label to the peptide, a bridge consisting of the sequence GGG-β-alanine-FITC was manually added as a side chain at the first lysine residue of the TAT sequence. The ɛ-amino group of this residue was protected during the initial synthesis by the acid labile 4-methyltrityl (MTT) group. The MTT was selectively removed from the peptide by five consecutive 1-min treatments with 2% trifluoracetic acid (TFA) plus 3% tri-isopropylsilane, leaving the peptide still attached to the resin with all other reactive side chains still protected. The three glycine residues and the β-alanine were added to this site with capping by acetic anhydride carried out after coupling each bridge residue. The FITC was coupled to the amino-group of the β-alanine. The peptides were purified to >95% using reversed-phase HPLC.

Loading of NH2-terminal peptide of actin isoforms.

An NH2-terminal FITC tag was added to monitor loading efficiency, and a TAT sequence (GYGRKKRRQRRR) was added to facilitate loading (1). α-Skeletal muscle actin peptides were used as a negative control peptide. Aorta tissue was loaded with the peptides as following protocol. At the beginning of all experiments, the tissue was challenged with 51 mM KCl-PSS to test for viability and to normalize for differences in the size or strength of each strip. As Kitazawa and his colleagues mentioned in their publications (11, 33, 63), most blood vessels show an increase in the amount of contraction observed to KCl challenges with time during the experiment. The data in this study were normalized to the first KCl challenge at the start of the day, and thus the response to a later KCl challenge was generally greater than 100% of the initial challenge. The KCl-PSS was washed out several times with PSS. The aorta tissues were allowed to relax for 1 h. The aorta tissues were then incubated for 3 h with 50 μM of each peptide in PSS aerated in oxygen. The peptide solution was changed with fresh peptide solution every hour. A parallel experiment without peptide in the solution was used as a sham. After the incubation period, the tissues were stimulated with 12-deoxyphorbol-13-isobutyrate-20-acetate (DPBA; 10 μM, for 20 min), phenylephrine (PE; 3 μM, for 10 min), or KCl-PSS (51 mM, for 10 min). The tissues, after peptide loading and extensive washing, are visually inspected, before proceeding with the experiment, for the yellowish-green color, confirming the successful loading of the FITC-labeled peptide. Details of this method are published in past studies from our laboratory using the same tissue and successfully loading TAT-peptides and peptides with similar TAT-like sequences (17, 24, 37).

Reagents.

Jasplakinolide, phalloidin, and DNase I were purchased from Molecular Probes (Eugene, OR). Latrunculin B was purchased from A. G. Scientific (San Diego, CA). A G-actin/F-actin assay kit (BK 037) was purchased from Cytoskeleton. DPBA was purchased from LC Laboratories (Woburn, MA). Pharmalytes for isoelectric focusing were purchased from Amersham Pharmacia Biotech AB (Uppsala, Sweden). Paraformaldehyde was purchased from Electron Microscopy Sciences (Hatfield, PA). General laboratory reagents used from analytical grade or better and were purchased from Sigma (St. Louis, MO) and Bio-Rad (Hercules, CA).

Antibodies.

The following antibodies were used: rabbit polyclonal G-actin antibody (1:500, 0.5 mg/ml; Cytoskeleton); monoclonal anti-α-smooth muscle actin (clone 1A4; 1:10,000, 4.4 mg/ml) and monoclonal anti-β-actin (clone AC-15; 1:150, 1.6 mg/ml) (Sigma); sheep anti-actin, γ-polyclonal antibody (1:1,000; Chemicon); Alexa Fluor 488 goat-anti mouse IgG (2 mg/ml), Alexa Fluor 568 goat-anti-mouse IgG (2 mg/ml), Alexa Fluor 488 donkey anti-sheep IgG (1:2,500, 2 mg/ml), Alexa Fluor 680 goat-anti mouse IgG (2 mg/ml), and Alexa Fluor 680 donkey anti-sheep IgG (1:2,500, 2 mg/ml; Molecular Probes); and IR-Dye 800 goat anti-rabbit IgG (1.0 mg/ml), IR-Dye 800 goat anti-mouse IgG (1.0 mg/ml), and IR-Dye 700 goat anti-mouse IgG (1.0 mg/ml; Rockland). The concentration of antibodies is in stock.

Data analysis.

All values given in the article are means ± SE. Differences between means were evaluated using a Student's t-test. Statistical significant differences were taken at the P < 0.05 level. The n values given represent numbers of cells or animals used in each experiment.

RESULTS

Contractile agonists increase the F/G ratio in dVSM.

F/G ratios were determined by two methods. First, F/G ratios were measured by quantifying results from the differential centrifugation of homogenates of dVSM (Fig. 1). Latrunculin, an agent known to associate with G-actin, thereby preventing actin polymerization (44), and phalloidin, an agent known to stabilize F-actin (9), were used as positive controls by directly adding them to homogenates of unstimulated aortic strips. As shown in Fig. 1, A and B, the addition of phalloidin significantly increased the F/G ratio, indicating that the technique measures changes in the F/G ratio when they do occur. Interestingly, the addition of latrunculin causes no significant change in the F/G ratio.

An external file that holds a picture, illustration, etc.
Object name is zh00090857010001.jpg

Differential ultracentrifugation of differentiated vascular smooth muscle (dVSM) indicates agonist-dependent changes in ratio of filamentous (F)-actin to globular (G)-actin (F/G ratios). A: immunoblot of actin in homogenates of ferret aorta. F- and G-actin were probed with a pan-actin antibody in unstimulated, phorbol ester 12-deoxyphorbol-13-isobutyrate-20-acetate (DPBA; 10 μM) or phenylephrine (PE; 10 μM)-stimulated aorta strip homogenates. Latrunculin (Lat; 10 μM) or phalloidin (Phal; 4 μM) was directly added to homogenates. B: F/G ratio from densitometry of actin bands. *P < 0.05; **P < 0.01, compared with unstimulated. Average is ± SE (n = 4–6).

Stimulation with the phorbol ester DPBA or the α-agonist PE to aortic tissues increases the F/G ratio in a consistent and statistically significant manner compared with that of unstimulated aortic tissues (Fig. 1, A and B). It is of note that we also attempted to measure F/G ratios in the presence of KCl-PSS but obtained highly variable results that showed no clear trend. Since KCl is a rapidly reversing stimulus, we were concerned that the method (involving hour-long centrifugation of nondenatured samples) is too slow to preserve any labile changes that might be caused by KCl-PSS.

To determine whether the above results could be reproduced with an independent second method with better time control, we also quick-fixed freshly enzymatically dissociated single muscle cells and costained them with fluorescent-labeled phalloidin to monitor the distribution of F-actin (red) and DNase I to monitor the distribution of G-actin (green) (34). DNase I, phalloidin, and merged images of illustrative individual stained cells are shown in Fig. 2A. In all cases, both red phalloidin staining, in a filamentous pattern, representing F-actin, and green DNase I staining, in a more patchy, diffuse pattern representing G-actin, were seen.

To quantify these data, we used a ratiometric approach within defined ROIs spanning the cell width but excluding the nuclear area. The position of the nucleus was identified by phase optics and is also afferent here from the characteristic decrease of the phalloidin staining in the nuclear region (Fig. 2A). The ratio of the value for phalloidin divided by that for DNase I was taken as an index of the F/G ratio as previously described in nonmuscle cells (34). Figure 2B shows the average quantitative results. In general, the data in freshly dissociated smooth muscle cells are qualitatively consistent with the data from the differential centrifugation of dVSM tissue. Stimulation with DPBA, PE, and also KCl increases phalloidin-to-DNase I ratios significantly compared with that of unstimulated isolated single smooth muscle cells (Fig. 2B). Note that latrunculin has no significant effect on the phalloidin-to-DNase I ratio, consistent with the data from differential ultracentrifugation. Jasplakinolide, a membrane-permeable stabilizer of F-actin (5), was also added to live cells before fixation, as a positive control. As expected, the addition of jasplakinolide to intact cells also significantly increases the phalloidin-to-DNase I ratio (Fig. 2B).

The absolute ratios obtained with the fluorescence labeling method cannot be compared directly with the absolute ratios from the differential centrifugation method, since additional factors (such as the strength of staining, the laser power, and photomultiplier voltage on the different channels necessary to prevent fluorophore cross talk, etc.) modify the absolute numbers for the fluorescence method. However, taken together, these data indicate that contractile agonist stimulation induces a significant net increase of polymerization of actin in dVSMCs.

Alterations of actin polymerization state modulate contractility in a stimulus-dependent manner.

To test the hypothesis that the net increases of actin polymerization described above cause changes in contractility, we pretreated live aortic smooth muscle tissue with either latrunculin or jasplakinolide. As shown in Fig. 3A, steady-state tonic contractile force in response to DPBA is significantly inhibited by pretreatment with latrunculin. In contrast, the average tonic contractile force response to DPBA is slightly increased by jasplakinolide pretreatment, but the difference is not statistically significant.

An external file that holds a picture, illustration, etc.
Object name is zh00090857010003.jpg

Modulation of polymerization regulates contractility. Contractile responses to DPBA (1 μM; A) or KCl (51 mM; B) in Lat (0.1 μM)- or Jas (0.5 μM)-pretreated aorta tissues. Vehicle (V) for Lat or Jas was ethanol or DMSO, respectively. **P < 0.01, compared with vehicle. Average is ± SE (n = 5 to 6).

The above results could simply be the nonspecific result of latrunculin-induced alterations of cellular cytoskeleton content. However, we performed the same experiment in preparations activated by 51 mM KCl-PSS, and neither latrunculin nor jasplakinolide pretreatment has any significant effect on force generation (Fig. 3B). These results also point to a difference in the mechanism by which KCl-PSS increases net actin polymerization, since this must involve a latrunculin-insensitive mechanism, i.e., one that is not affected by the sequestering of G-actin monomers.

It is worth noting that the treatment of aortic tissues with latrunculin or jasplakinolide alone has no detectable effect on intrinsic basal tone, even though the ferret aorta has previously been shown to display an active, intrinsic, basal tone (23). This further supports the selectivity of these effects.

Inhibition of net polymerization by latrunculin is agonist dependent.

To confirm that the stimulus-dependent modulations of contractility caused by latrunculin or jasplakinolide pretreatment are paralleled by changes in actin polymerization, we pretreated freshly dissociated dVSMCs with the same agents as above and quantitated the effect on F/G ratios. Figure 4A shows illustrative images of individual freshly dissociated dVSMCs under different conditions. Although considerable cell-to-cell variability was noticed, when the results were quantitated (Fig. 4B) by ratiometric analysis, it was found that latrunculin alone causes no significant change in the phalloidin-to-DNase I staining ratio. However, latrunculin pretreatment significantly inhibits both PE- and DPBA-induced increases in the phalloidin-to-DNase I ratios.

An external file that holds a picture, illustration, etc.
Object name is zh00090857010004.jpg

Actin remodeling by DPBA or PE, but not by KCl, is inhibited by Lat. A: illustrative images stained with Alexa 488-DNase I (G-actin; green; left), Alexa 568-phalloidin (F-actin; red; middle), and merged images (right) in cells without or with Lat (0.03 μM) pretreatment. B: quantitative analysis of ROI of freshly dissociated cells costained with fluorescent phalloidin and DNase I. **P < 0.01, compared with unstimulated (white bars); ##P < 0.01, compared with Lat-untreated cells (black bars). Average is ± SE for 18–40 cells.

In contrast, when 51 mM KCl is used as a stimulus, pretreatment with latrunculin causes no significant inhibition of the KCl-induced increase in the F/G ratio (Fig. 4B). Thus these results are consistent with the fact that latrunculin pretreatment has no significant effect on the depolarization-induced increases in contractility (Fig. 3) and, furthermore, point to a difference in the mechanism by which KCl depolarization, versus DPBA, causes a net increase in actin polymerization with the former being latrunculin insensitive and the latter being latrunculin sensitive.

Multiple actin isoforms coexist in dVSM.

Whether actin isoforms are differentially distributed within individual smooth muscle cells remains controversial. Thus we first determined the actin isoform subtype composition in intact ferret aorta tissue. Homogenates of intact aorta tissues display three predominant spots at 42 kDa, when run on two-dimensional gels (Fig. 5A). These three spots were confirmed to be α-smooth muscle actin, β-cytoplasmic actin, and γ-actin (from acidic to basic; Fig. 5A) by mass spectrometry (Tufts University Core Facility, Boston, MA). γ-Smooth muscle actin and γ-cytoplasmic actin cannot be separated by two-dimentional gels, but the relative amounts of proteins in the three spots were quantified by densitometry after staining the two-dimensional gels with Proteom IQ Blue. The major actin component in aorta is α-smooth muscle actin (∼66.2% of total actin), followed by β-cytoplasmic actin (∼21.1% of total actin), and γ-actin (∼12.7% of total actin). This result is consistent with a previous study on vascular actin isoform composition from different species (15).

An external file that holds a picture, illustration, etc.
Object name is zh00090857010005.jpg

Actin isoforms in aorta. A: 2-dimensional gel of aorta homogenate stained with Coomassie blue. B: images of illustrative dVSMCs immunostained with isoform-specific actin antibodies. Scale bars, 10 μm. IEF, isoelectric focusing.

To determine whether the subcellular distribution of actin is isoform dependent in dVSMCs, we stained freshly dissociated aorta cells with isoform-specific antibodies for α-smooth muscle actin, β-cytoplasmic actin, and α-/γ-actin (Fig. 5B). The α-smooth muscle actin is present in longitudinally or slightly tangentially oriented filamentous bundles (arrows) in the freshly isolated dVSMCs (top) in a manner similar to that shown previously for myosin-containing contractile filament bundles (50). In contrast, β-cytoplasmic actin was prominent in the cell cortex and in shorter punctuate spots (arrows) throughout the cell (middle). Unfortunately, there is no available entirely specific anti-γ-antibody. We found that the commonly used commercially available polyclonal γ-antibody (Chemicon, Temecula, CA) recognizes, on two-dimensional gels, both α- and γ-actin isoforms (data not shown). When cells are stained with this antibody, the distribution of staining includes some longitudinal filaments (arrows) expected from α-smooth muscle actin cross-reactivity (Fig. 5B, bottom), but a clear discrimination of the γ-actin staining from that due to α-smooth muscle actin could not be determined.

Isoform-specific NH2-terminal actin peptides depress contractile force.

To further test whether actin isoforms modulate the contractile function of smooth muscle, we synthesized NH2-terminal decoy peptides of actin isoforms (Table 1) coupled to a cell permeation TAT tag sequence and introduced them to intact dVSM. It has previously been reported that the NH2-terminal α-smooth muscle actin tetrapeptide inhibits the formation of stress fibers and wrinkles on silicone substrate in an isoform-specific manner in cultured cells (6, 22). The NH2-terminal actin sequences are also known to include sequences involved in the association with many actin-binding proteins, and since considerable evidence in nonmuscle cells indicates that, despite the large degree of sequence conservation, actin displays isoform-specific functions, we synthesized all of the relevant NH2-terminal peptides as putative decoy peptides. We also made the α-skeletal muscle peptide as a negative control to test the specificity of the approach.

Table 1.

Actin isoform-specific peptides

Actin IsoformPeptide
α-Smooth muscle actinAc-EEED-FITC-TAT
β-Cytoplasmic actinAc-DDDIA-FITC-TAT
γ-Smooth muscle actinAc-EEETT-FITC-TAT
γ-Cytoplasmic actinAc-EEEIA-FITC-TAT
α-Skeletal muscle actinAc-DEDE-FITC-TAT

TAT, GYGRKKRRQRRR; Ac, acetylation.

As expected, the α-skeletal muscle actin peptide has no significant effect on the steady-state contractile force induced by either DPBA or PE (Fig. 6). The introduction of the β-cytoplasmic actin peptide also has no significant effect on either DPBA- or PE-induced smooth muscle contraction. However, the contractile force induced by DPBA is significantly inhibited by the introduction of α-smooth muscle, γ-smooth muscle, and γ-cytoplasmic actin peptides (Fig. 6A). Interestingly, the contractile force induced by PE is significantly inhibited by the introduction of γ-smooth muscle, and γ-cytoplasmic actin peptides, but not in α-smooth muscle actin (Fig. 6B).

An external file that holds a picture, illustration, etc.
Object name is zh00090857010006.jpg

Decoy actin peptides inhibit contractility in an isoform-dependent manner. Steady-state force in the presence of DPBA (10 μM; A), PE (3 μM; B), or KCl (51 mM; C) in the presence of peptides or sham treated (Table 1) is shown. Contractile responses were normalized to force to a challenge with KCl physiological salt solution (PSS; 51 mM KCl) at the beginning of each experiment. *P < 0.05, effects of peptides compared with sham (Sham). Average is ± SE (n = 3–6). A-SM, α-smooth muscle actin; beta, β-cytoplasmic actin; G-SM, γ-smooth muscle actin; G-CYT, γ-cytoplasmic actin; A-SK, α-skeletal muscle actin.

In contrast, in the same experimental protocol, but with preparations activated by KCl-PSS, none of these peptides has any significant effect on force generation (Fig. 6C), further pointing to selectivity in the action of these peptides. There was also no detectable decrease in intrinsic basal tone with any of peptides.

Contractile agonists induce cytoskeletal remodeling in an actin isoform-dependent manner in dVSM.

The above results indicate that smooth muscle function can be regulated in an actin isoform-dependent manner. To further test the hypothesis that actin remodeling is actin isoform dependent, we used differential centrifugation combined with immunoblots. The F/G ratio of α-smooth muscle and β-cytoplasmic actin was obtained by using either anti-α-smooth muscle actin or anti-β-cytoplasmic actin antibodies to probe the samples. As shown in Fig. 7, the α-smooth muscle actin population shows no significant DPBA- or PE-induced changes in the F/G ratio (Fig. 7A). In contrast, stimulation with DPBA increases the F/G ratio of β-cytoplasmic actin significantly (Fig. 7B). However, the β-specific remodeling is far less pronounced than the preceding results on F/G ratios for total actin (Fig. 1B), which suggests that the γ-isoforms may remodel to a greater degree than other isoforms. It is also of note that the α-smooth muscle isoform appears to be the least dynamic isoform during contractile responses.

An external file that holds a picture, illustration, etc.
Object name is zh00090857010007.jpg

Isoform-specific changes in F/G ratios in differential ultracentrifugation samples. F- and G- actin probed in immunoblots for α-smooth muscle actin (A) or β-cytoplasmic actin (B) in unstimulated DPBA (10 μM) or PE (10 μM)-stimulated aorta. C: γ-actin in the F fraction after immunoblotting with γ-actin antibody after 2-dimensional separation of parallel homogenates. *P < 0.05, compared with unstimulated. Average is ± SE (n = 3–5).

We could not directly apply the same method to the γ-actin isoform, because of the lack of availability of a truly γ-specific actin antibody. Instead, we chose to run the F-actin sample on two-dimensional gels. The less abundant, G-actin samples on two-dimensional gels were not measurable because of lower signal-to-noise ratios of the bands. However, we were able to get reliable results comparing the F-actin samples from the γ-spots of two-dimensional gels. Samples were cut from individual gels and transferred to the same membrane so that they could be directly compared. The data are expressed as the densitometry of the γ-spot as a fraction of the total of the unstimulated actin spots. When analyzed in this way, as shown in Fig. 7C, the γ-F-actin population shows significant increases with PE stimulation. Taken together, these data point to isoform-specific roles in signaling pathways activated by different stimuli in dVSM, with γ-actin being the most dynamic, especially in the presence of PE.

DISCUSSION

In the present study we show, for the first time, that the actin cytoskeleton of dVSM undergoes remodeling during contractile activation in an actin isoform-dependent and stimulus-dependent manner. It is generally accepted that VSM contractility is regulated both by mechanisms that regulate actin-activated myosin ATPase activity, via changes in the phosphorylation state of the 20-kDa myosin light chain (55), as well as by mechanisms that regulate the availability of actin to interact with myosin, via the action of inhibitory actin-binding proteins such as caldesmon (43). It has also been proposed, but is more controversial, that signaling pathways exist by which the actin cytoskeleton is remodeled during agonist activation and that this might modulate vascular contractility (7, 49).

Cytoskeletal remodeling has previously been reported to occur in a few VSM studies (48, 58). However, what has not been clear is exactly how, or whether, cytoskeletal remodeling modulates contractility of smooth muscle. Some groups have suggested that cytoskeletal remodeling may alter the transmission of cross-bridge generated force to the cell membrane in airway smooth muscle (19). Other groups have suggested that it modulates force maintenance or optimizes the energetic cost of tension in the VSMC (25).

Using a method of differential centrifugation to quantify the F- and G-actin, we found that the F/G ratio of dVSM is agonist sensitive. Our results are qualitatively and quantitatively consistent with the F/G ratios, reported by Gunst and colleagues (65) for airway smooth muscle. Previously, Tang and Tan (58) reported similar increases in F/G ratios in VSM stimulated by the α-agonist norepinephrine but lower absolute F/G ratios of dVSM compared with our values. To confirm these results and also to develop a method that allows better time resolution, we have also quantitated F/G ratios, for the first time in dVSMCs, by costaining with fluorescently labeled phalloidin and DNase I in freshly enzymatically dissociated single dVSMCs. Qualitatively similar agonist-dependent results are obtained with both methods; however, it is important to emphasize that the absolute phalloidin-to-DNase I ratios are also affected by staining intensity, photomultiplier nonlinearity, and confocal settings and thus are not quantitatively identical to the F/G ratios obtained by differential centrifugation. Even though the two methods cannot be directly compared in a quantitative manner, the qualitative results in both cases clearly indicate that certain contractile stimuli induce a significant net increase of actin polymerization in dVSMCs.

We have shown that three stimuli, KCl, PE, and DPBA, as well as an actin filament stabilizer, jasplakinolide, increase actin polymerization, as measured by F/G ratios. However, latrunculin, a stabilizer of G-actin, caused no significant change in the unstimulated tissue F/G ratio. These results suggest that an endogenous protein (for example, capping proteins, G-actin binding proteins, etc.) may stabilize G-actin so that the addition of latrunculin has no effect. However, latrunculin pretreatment is able to prevent stimulus-induced increases in the F/G ratio, suggesting that a stimulus-induced signal transduction pathway results in the liberation of free G-actin separate from the putative endogenous G-actin binding protein (40).

Interestingly, latrunculin was able to prevent only PE- and DPBA-induced increases in F/G ratios and not those induced by the depolarization of VSM. Although there are some studies that show either PE or DPBA stimulation on smooth muscle also induces membrane depolarization (13, 45), these results strongly suggest that a different mechanism of the regulation of the F/G ratio is being recruited by KCl-induced depolarization. Others have reported that depolarization can activate Rho/ROCK pathways that differ from the PKC-dependent pathways activated by phorbol esters and PE (42, 59), and this may point to distinct mechanisms by which actin remodeling may be regulated in dVSM.

The fact that latrunculin pretreatment causes a significant inhibition of the DPBA-induced contraction in parallel with the inhibition of actin polymerization links the observed cytoskeletal remodeling with an effect on contractility. One might conclude that the effects of latrunculin are due a nonspecific disruption or enhancement of the cytoskeleton, causing nonspecific effects on all types of smooth muscle contraction. However, the fact that in preparations activated by KCl-induced depolarization, neither latrunculin nor jasplakinolide had any effects on force generation, argues against this possibility.

It is worth noting that jasplakinolide had a small effect on the F/G ratio in cells but no significant effect on active force development in the intact tissue preparation. We assume that this can be explained by the fact that multiple redundant pathways regulate contractile force, whereas the F/G ratios directly measure a single pathway, so that we could not detect a change in the former case but could see a significant, but small change in the latter case. However, the possibility also exists that there could be some differences in function between the intact tissue and the dissociated cells (14).

We have previously reported a differential subcellular distribution of α-smooth muscle and β-cytoplasmic actin in portal vein VSMCs (50). North et al. (46) have also reported a differential distribution for β-cytoplasmic actin versus the unidentified (presumably γ-smooth muscle) actin isoform present in actomyosin-containing contractile bundles in chicken gizzard smooth muscle. A distinct population of actin filaments has also been reported by using immunoprecipitation for an anticalponin antibody to demonstrate the existence of an actin filament population lacking caldesmon (36). According to the model proposed by North et al. (46), nonmuscle β-actin in gizzard cells is localized in dense bodies and longitudinal channels linking consecutive dense bodies and membrane-associated dense plaques and lying along intermediate filaments. In contrast, Stromer et al. (57) have argued against any actin isoform-specific domains in gizzard or VSM. In the present study, we found that α-smooth muscle actin is present in longitudinally or slightly tangentially oriented filamentous bundles and β-actin is prominent in the cell cortex and in punctuate spots. These results are consistent with previous reports from Parker et al. (50) and qualitatively consistent with those of the North et al. study (46). The lack of availability of a truly specific anti-γ-actin antibody prevented us from imaging its subcellular distribution. However, differential centrifugation results indicate that the γ-actin isoform is the most dynamic actin and, since in nonmuscle cell types, cortical actin has been shown to be the site of the most dynamic actin turnover (8), one might suggest that the γ-actin in the dVSM cell fills this role.

In this study, for the first time, we introduced actin isoform-specific decoy peptides coupled to a cell permeation TAT tag sequence into dVSM. We observed a significant inhibition of dVSM contractility in a peptide-specific manner and in a stimulus-dependent manner. Negative results obtained with the skeletal α-actin sequence are consistent with the absence of skeletal muscle actin in these dVSM cells and confirm the specificity of the approach. The lack of a significant effect of these peptides on depolarization-mediated contractions of dVSM further supports specificity. We did, however, see significant inhibition of contractility with smooth muscle and cytoplasmic γ-actin peptides in both PE- and phorbol-induced contractions, consistent with our results with differential centrifugation.

We were somewhat puzzled by the effect of the α-smooth muscle peptide to inhibit phorbol ester-induced contractions but not PE-induced contractions. Since phorbol esters are nonphysiological stimuli, this may point to a difference in the mechanism of contraction caused by this stimulus versus the more relevant α-agonist PE. However, these results are also in contrast with the fact that we saw no change in the smooth muscle α-actin F/G ratio in the presence of any stimulus with differential centrifugation. The reason for this difference is not clear at present.

To further test the idea that actin remodeling is isoform dependent, with a major role played by the γ-isoform, we also analyzed results from differential centrifugation. Isoform-specific probing of the F- and G-actin fractions shows no evidence of involvement of α-smooth muscle actin in cytoskeletal remodeling. However, γ-actin does show significant isoform-dependent remodeling during PE contractions. Similarly, Mauss et al. (39) have shown that ileal smooth muscle, but not VSM, contractility is inhibited by botulinum C2 toxin. This toxin ADP ribosylates G-actin of γ- but not α-smooth muscle actin isoforms (38).

In summary, the results presented here indicate that the actin cytoskeleton of dVSMCs remodels in a stimulus-dependent manner by increasing net actin polymerization during contraction. Furthermore, we identify γ-actin as the most dynamically remodeled actin within the dVSMC cytoskeleton, and thus we predict that gamma actin, despite its lesser abundance, may play a critical role in the regulation of smooth muscle contractility.

GRANTS

This research was supported by National Heart, Lung, and Blood Institute Grants HL-86655, HL-31704, and HL-80003 and by National Institute of Child Health and Human Development Grant HD-043054 (to K. G. Morgan).

Notes

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. Abes R, Arzumanov AA, Moulton HM, Abes S, Ivanova GD, Iversen PL, Gait MJ, Lebleu B. Cell-penetrating-peptide-based delivery of oligonucleotides: an overview. Biochem Soc Trans 035: 775–779, 2007. [PubMed] [Google Scholar]
2. Adler K, Krill J, Alberghini T, Evans J. Effect of cytochalasin D on smooth muscle contraction. Cell Motil 3: 545–551, 1983. [PubMed] [Google Scholar]
3. Battistella-Patterson AS, Wang S, Wright GL. Effect of disruption of the cytoskeleton on smooth muscle contraction. Can J Physiol Pharmacol 75: 1287–1299, 1997. [PubMed] [Google Scholar]
4. Bray D Cell Movements: From Molecules to Motility. New York: Garland Publishing, 2001.
5. Bubb M, Senderowicz A, Sausville E, Duncan K, Korn E. Jasplakinolide, a cytotoxic natural product, induces actin polymerization and competitively inhibits the binding of phalloidin to F-actin. J Biol Chem 269: 14869–14871, 1994. [PubMed] [Google Scholar]
6. Chaponnier C, Goethals M, Janmey PA, Gabbiani F, Gabbiani G, Vandekerckhove J. The specific NH2-terminal sequence Ac-EEED of alpha-smooth muscle actin plays a role in polymerization in vitro and in vivo. J Cell Biol 130: 887–895, 1995. [PMC free article] [PubMed] [Google Scholar]
7. Cipolla M, Gokina N, Osol G. Pressure-induced actin polymerization in vascular smooth muscle as a mechanism underlying myogenic behavior. FASEB J 16: 72–76, 2002. [PubMed] [Google Scholar]
8. Clement S, Hinz B, Dugina V, Gabbiani G, Chaponnier C. The N-terminal Ac-EEED sequence plays a role in α-smooth-muscle actin incorporation into stress fibers. J Cell Sci 118: 1395–1404, 2005. [PubMed] [Google Scholar]
9. Cooper JA Effects of cytochalasin and phalloidin on actin. J Cell Biol 105: 1473–1478, 1987. [PMC free article] [PubMed] [Google Scholar]
10. Dessy C, Kim I, Sougnez CL, Laporte R, Morgan KG. A role for MAP kinase in differentiated smooth muscle contraction evoked by α-adrenoceptor stimulation. Am J Physiol Cell Physiol 275: C1081–C1086, 1998. [PubMed] [Google Scholar]
11. Dimopoulos GJ, Semba S, Kitazawa K, Eto M, Kitazawa T. Ca2+-dependent rapid Ca2+ sensitization of contraction in arterial smooth muscle. Circ Res 100: 121–129, 2007. [PMC free article] [PubMed] [Google Scholar]
12. Drew JS, Moos C, Murphy R. Localization of isoactins in isolated smooth muscle thin filaments by double gold immunolabeling. Am J Physiol Cell Physiol 260: C1332–C1340, 1991. [PubMed] [Google Scholar]
13. Earley S, Straub SV, Brayden JE. Protein kinase C regulates vascular myogenic tone through activation of TRPM4. Am J Physiol Heart Circ Physiol 292: H2613–H2622, 2007. [PubMed] [Google Scholar]
14. Eddinger TJ, Schiebout JD, Swartz DR. Adherens junction-associated protein distribution differs in smooth muscle tissue and acutely isolated cells. Am J Physiol Gastrointest Liver Physiol 292: G684–G697, 2007. [PubMed] [Google Scholar]
15. Fatigati V, Murphy R. Actin and tropomyosin variants in smooth muscles. Dependence on tissue type. J Biol Chem 259: 14383–14388, 1984. [PubMed] [Google Scholar]
16. Flavahan NA, Bailey SR, Flavahan WA, Mitra S, Flavahan S. Imaging remodeling of the actin cytoskeleton in vascular smooth muscle cells after mechanosensitive arteriolar constriction. Am J Physiol Heart Circ Physiol 288: H660–H669, 2005. [PubMed] [Google Scholar]
17. Gallant C, You JY, Sasaki Y, Grabarek Z, Morgan KG. MARCKS is a major PKC-dependent regulator of calmodulin targeting in smooth muscle. J Cell Sci 118: 3595–3605, 2005. [PubMed] [Google Scholar]
18. Garrels JI, Gibson W. Identification and characterization of multiple forms of actin. Cell 9: 793–805, 1976. [PubMed] [Google Scholar]
19. Gerthoffer WT, Gunst SJ. Invited review: focal adhesion and small heat shock proteins in the regulation of actin remodeling and contractility in smooth muscle. J Appl Physiol 91: 963–972, 2001. [PubMed] [Google Scholar]
20. Godt RE, Kirby AC, Gordon AM. Effects of hypertonic solutions on contraction of frog tonic muscle fibers. Am J Physiol Cell Physiol 246: C148–C153, 1984. [PubMed] [Google Scholar]
21. Herman IM Actin Isoforms. Curr Opin Cell Biol 5: 48–55, 1993. [PubMed] [Google Scholar]
22. Hinz B, Gabbiani G, Chaponnier C. The NH2-terminal peptide of α-smooth muscle actin inhibits force generation by the myofibroblast in vitro and in vivo. J Cell Biol 157: 657–663, 2002. [PMC free article] [PubMed] [Google Scholar]
23. Jain M, Dai HB, Carrozza JP, Sellke FW, Morgan KG. Intrinsic tone as potential vascular reserve in conductance and resistance vessels. Circulation 94: 1083–1088, 1996. [PubMed] [Google Scholar]
24. Je HD, Gallant C, Leavis PC, Morgan KG. Caveolin-1 regulates contractility in differentiated vascular smooth muscle. Am J Physiol Heart Circ Physiol 286: H91–H98, 2004. [PubMed] [Google Scholar]
25. Jones KA, Perkins WJ, Lorenz RR, Prakash YS, Sieck GC, Warner DO. F-actin stabilization increases tension cost during contraction of permeabilized airway smooth muscle in dogs. J Physiol 519: 527–538, 1999. [PMC free article] [PubMed] [Google Scholar]
26. Kargacin GJ, Cooke PH, Abramson SB, Fay FS. Periodic organization of the contractile apparatus in smooth muscle revealed by the motion of dense bodies in single cells. J Cell Biol 108: 1465–1475, 1989. [PMC free article] [PubMed] [Google Scholar]
27. Khaitlina SY Functional specificity of actin isoforms. Int Rev Cytol 202: 35–98, 2001. [PubMed] [Google Scholar]
28. Khalil RA, Lajoie C, Morgan KG. In situ determination of the [Ca2+]i threshold for translocation of the α-protein kinase C isoform. Am J Physiol Cell Physiol 266: C1544–C1551, 1994. [PubMed] [Google Scholar]
29. Khalil RA, Lajoie C, Resnick MS, Morgan KG. Ca2+-independent isoforms of protein kinase C differentially translocate in smooth muscle. Am J Physiol Cell Physiol 263: C714–C719, 1992. [PubMed] [Google Scholar]
30. Khalil RA, Menice CB, Wang CL, Morgan KG. Phosphotyrosine-dependent targeting of mitogen-activated protein kinase in differentiated contractile vascular cells. Circ Res 76: 1101–1108, 1995. [PubMed] [Google Scholar]
31. Khalil RA, Morgan KG. PKC-mediated redistribution of mitogen-activated protein kinase during smooth muscle activation. Am J Physiol Cell Physiol 265: C406–C411, 1993. [PubMed] [Google Scholar]
32. Kim HR, Hoque M, Hai CM. Cholinergic receptor-mediated differential cytoskeletal recruitment of actin- and integrin-binding proteins in intact airway smooth muscle. Am J Physiol Cell Physiol 287: C1375–C1383, 2004. [PubMed] [Google Scholar]
33. Kitazawa T, Eto M, Woodsome TP, Khalequzzaman M. Phosphorylation of the myosin phosphatase targeting subunit and CPI-17 during Ca2+ sensitization in rabbit smooth muscle. J Physiol 546: 879–889, 2003. [PMC free article] [PubMed] [Google Scholar]
34. Knowles G, McCulloch C. Simultaneous localization and quantification of relative G and F actin content: optimization of fluorescence labeling methods. J Histochem Cytochem 40: 1605–1612, 1992. [PubMed] [Google Scholar]
35. Laudadio RE, Millet EJ, Fabry B, An SS, Butler JP, Fredberg JJ. Rat airway smooth muscle cell during actin modulation: rheology and glassy dynamics. Am J Physiol Cell Physiol 289: C1388–C1395, 2005. [PubMed] [Google Scholar]
36. Lehman W Calponin and the composition of smooth muscle thin filaments. J Muscle Res Cell Motil 12: 221–224, 1991. [PubMed] [Google Scholar]
37. Marganski WA, Gangopadhyay SS, Je HD, Gallant C, Morgan KG. Targeting of a novel Ca2+/calmodulin-dependent protein kinase II is essential for extracellular signal-regulated kinase-mediated signaling in differentiated smooth muscle cells. Circ Res 97: 541–549, 2005. [PubMed] [Google Scholar]
38. Mauss S, Chaponnier C, Just I, Aktories K, Gabbiani G. ADP-ribosylation of actin isoforms by Clostridium botulinum C2 toxin and Clostridium perfringens iota toxin. Eur J Biochem 194: 237–241, 1990. [PubMed] [Google Scholar]
39. Mauss S, Koch G, Kreye V, Aktories K. Inhibition of the contraction of the isolated longitudinal muscle of the guinea-pig ilem by botulinum C2 toxin: evidence for a role of G/F-actin transition in smooth muscle contraction. Naunyn Schmiedebergs Arch Pharmacol 340: 345–351, 1989. [PubMed] [Google Scholar]
40. McGregor E, Kempster L, Wait R, Gosling M, Dunn MJ, Powell JT. F-actin capping (CapZ) and Other contractile saphenous vein smooth muscle proteins are altered by hemodynamic stress: a proteomic approach. Mol Cell Proteomics 3: 115–124, 2004. [PubMed] [Google Scholar]
41. Mehta D, Gunst SJ. Actin polymerization stimulated by contratile activation regulates force development in canine tracheal smooth muscle. J Physiol 519: 829–840, 1999. [PMC free article] [PubMed] [Google Scholar]
42. Mita M, Yanagihara H, Hishinuma S, Saito M, Walsh MP. Membrane depolarization-induced contraction of rat caudal arterial smooth muscle involves Rho-associated kinase. Biochem J 364: 431–440, 2002. [PMC free article] [PubMed] [Google Scholar]
43. Morgan KG, Gangopadhyay SS. Invited review: cross-bridge regulation by thin filament-associated proteins. J Appl Physiol 91: 953–962, 2001. [PubMed] [Google Scholar]
44. Morton WM, Ayscough KR, McLaughlin PJ. Latrunculin alters the actin-monomer subunit interface to prevent polymerization. Nat Cell Biol 2: 376–378, 2000. [PubMed] [Google Scholar]
45. Nelson MT, Standen NB, Brayden JE, Worley JF. Noradrenaline contracts arteries by activating voltage-dependent calcium channels. Nature 336: 382–385, 1988. [PubMed] [Google Scholar]
46. North AJ, Gimona M, Lando Z, Small JV. Actin isoform compartments in chicken gizzard smooth muscle cells. J Cell Sci 107: 445–455, 1994. [PubMed] [Google Scholar]
47. O′Farrell PH High resolution two-dimensional electrophoresis of proteins. J Biol Chem 250: 4007–4021, 1975. [PMC free article] [PubMed] [Google Scholar]
48. Ohanian V, Gatfield K, Ohanian J. Role of the actin cytoskeleton in G-protein-coupled receptor activation of PYK2 and paxillin in vascular smooth muscle. Hypertension 46: 93–99, 2005. [PubMed] [Google Scholar]
49. Opazo Saez A, Zhang W, Wu Y, Turner CE, Tang DD, Gunst SJ. Tension development during contractile stimulation of smooth muscle requires recruitment of paxillin and vinculin to the membrane. Am J Physiol Cell Physiol 286: C433–C447, 2004. [PubMed] [Google Scholar]
50. Parker CA, Takahashi K, Tang JX, Tao T, Morgan KG. Cytoskeletal targeting of calponin in differentiated, contractile smooth muscle cells of the ferret. J Physiol 508: 187–198, 1998. [PMC free article] [PubMed] [Google Scholar]
51. Parker CA, Takahashi K, Tao T, Morgan KG. Agonst-induced redistribution of calponin in contractile vascular smooth msucle cells. Am J Physiol Cell Physiol 267: C1262–C1270, 1994. [PubMed] [Google Scholar]
52. Prochniewicz E, Thomas DD. Site-specific mutations in the myosin binding sites of actin affect structural transitions that control myosin binding. Biochemistry 40: 13933–13940, 2001. [PubMed] [Google Scholar]
53. Saito S, Hori M, Ozaki H, Karaki H. Cytochalasin D inhibits smooth muscle contraction by directly inhibiting contractile apparatus. J Smooth Muscle Res 32: 51–60, 1996. [PubMed] [Google Scholar]
54. Shaw L, Ahmed S, Austin C, Taggart M. Inhibitors of actin filament polymerisation attenuate force but not global intracellular calcium in isolated pressurised resistace arteries. J Vasc Res 40: 1–10, 2003. [PubMed] [Google Scholar]
55. Somlyo AP, Somlyo AV. Ca2+ sensitivity of smooth muscle and nonmuscle myosin II: modulated by G proteins, kinases, and myosin phosphatase. Physiol Rev 83: 1325–1358, 2003. [PubMed] [Google Scholar]
56. Somlyo AV Ultrastructure of vascular smooth muscle. In: Handbook of Physiology. The Cardiovascular System. Vascular Smooth Muscle. Bethesda, MD: Am Physiol Soc, 1980, sect. 2, vol. II, p. 33–67.
57. Stromer MH, Mayes MS, Bellin RM. Use of actin isoform-specific antibodies to probe the domain structure in three smooth muscles. Histochem Cell Biol 118: 291–299, 2002. [PubMed] [Google Scholar]
58. Tang DD, Tan J. Role of Crk-associated substrate in the regulation of vascular smooth muscle contraction. Hypertension 42: 858–863, 2003. [PubMed] [Google Scholar]
59. Urban NH, Berg KM, Ratz PH. K+ depolarization induces RhoA kinase translocation to caveolae and Ca2+ sensitization of arterial muscle. Am J Physiol Cell Physiol 285: C1377–C1385, 2003. [PubMed] [Google Scholar]
60. Vandekerckhove J, Weber K. At least six different actins are expressed in a higher mammal: an analysis based on the amino acid sequence of the amino-terminal tryptic peptide. J Mol Biol 126: 783–802, 1978. [PubMed] [Google Scholar]
61. Vandekerckhove J, Weber K. The complete amino acid sequence of actins from bovine aorta, bovine heart, bovine fast skeletal muscle, and rabbit slow skeletal muscle. Differentiation 14: 123–133, 1979. [PubMed] [Google Scholar]
62. Ward DT, Alder AC, Ohanian J, Ohanian V. Noradrenaline-induced paxillin phosphorylation, ERK activation and MEK-regulated contraction in intact rat mesenteric arteries. J Vasc Res 39: 1–11, 2002. [PubMed] [Google Scholar]
63. Woodsome TP, Polzin A, Kitazawa K, Eto M, Kitazawa T. Agonist- and depolarization-induced signals for myosin light chain phosphorylation and force generation of cultured vascular smooth muscle cells. J Cell Sci 119: 1769–1780, 2006. [PubMed] [Google Scholar]
64. Zeidan A, Nordstrom I, Albinsson S, Malmqvist U, Sward K, Hellstrand P. Stretch-induced contractile differentiation of vascular smooth muscle: sensitivity to actin polymerization inhibitors. Am J Physiol Cell Physiol 284: C1387–C1396, 2003. [PubMed] [Google Scholar]
65. Zhang W, Wu Y, Du L, Tang DD, Gunst SJ. Activation of the Arp2/3 complex by N-WASp is required for actin polymerization and contraction in smooth muscle. Am J Physiol Cell Physiol 288: C1145–C1160, 2005. [PubMed] [Google Scholar]
66. Zhao Y, Xiao H, Long W, Pearce WJ, Longo LD. Expression of several cytoskeletal proteins in ovine cerebral arteries: developmental and functional considerations. J Physiol 558: 623–632, 2004. [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Physiology - Cell Physiology are provided here courtesy of American Physiological Society

-