Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Tidal Stretches Differently Regulate the Contractile and Cytoskeletal Elements in Intact Airways

Retraction

After publication of this article [1], concerns were raised about similarities between some GAPDH loading control bands (hereafter, LCs) in western blots in Figs 2, 3, and 7.

Specifically,

  • The LCs in Fig 2B, Fig 3B (when flipped horizontally), and the lefthand panel of Fig 7B appear similar.
  • The LC panels in Figs 2A, 2C, 3A, 7C, and the lefthand panel of 7D appear to show the same data; of note, Fig 7D represents different experimental conditions than the other panels listed here (mechanical loading with versus without constriction). Specific bands are labeled as representing different experimental groups across these figures, and for some panels the bands appear in different order and/or orientation. In the case of Fig 3A, the same band appears to represent both the T0 and D conditions.
  • The LCs for Fig 3C appear similar to the LCs in Fig 7A.
  • The LCs appear similar between the lefthand panel of Fig 7A and the righthand panel of Fig 7A.
  • The LCs appear similar between Fig 2D and Fig 3D.

During editorial follow up, the corresponding author provided the available underlying blot images for these published figures, as well as replicate blot data. Additional methodological information regarding the western blot experiments was also provided to clarify that some loading control panels (left- and right-hand panels of Fig 7A; Fig 2D and Fig 3D) were correctly duplicated because the same membrane was re-probed for different proteins of interest. However, some original blot data needed to address the concerns are now unavailable and so questions about the reliability of the published loading control blots could not be fully resolved. The corresponding author requested retraction of this article.

In light of the unresolved concerns about the reliability of loading control image data in the published figures, the PLOS ONE Editors retract this article.

EBS and BS agreed with retraction and stand by the article’s findings. ASLP, BCH, and KRL either did not respond directly or could not be reached.

22 Feb 2024: The PLOS ONE Editors (2024) Retraction: Tidal Stretches Differently Regulate the Contractile and Cytoskeletal Elements in Intact Airways. PLOS ONE 19(2): e0299553. https://doi.org/10.1371/journal.pone.0299553 View retraction

Abstract

Recent reports suggest that tidal stretches do not cause significant and sustainable dilation of constricted intact airways ex vivo. To better understand the underlying mechanisms, we aimed to map the physiological stretch-induced molecular changes related to cytoskeletal (CSK) structure and contractile force generation through integrin receptors. Using ultrasound, we measured airway constriction in isolated intact airways during 90 minutes of static transmural pressure (Ptm) of 7.5 cmH2O or dynamic variations between Ptm of 5 and 10 cmH20 mimicking breathing. Integrin and focal adhesion kinase activity increased during Ptm oscillations which was further amplified during constriction. While Ptm oscillations reduced β-actin and F-actin formation implying lower CSK stiffness, it did not affect tubulin. However, constriction was amplified when the microtubule structure was disassembled. Without constriction, α-smooth muscle actin (ASMA) level was higher and smooth muscle myosin heavy chain 2 was lower during Ptm oscillations. Alternatively, during constriction, overall molecular motor activity was enhanced by Ptm oscillations, but ASMA level became lower. Thus, ASMA and motor protein levels change in opposite directions due to stretch and contraction maintaining similar airway constriction levels during static and dynamic Ptm. We conclude that physiological Ptm variations affect cellular processes in intact airways with constriction determined by the balance among contractile and CSK molecules and structure.

Introduction

A hallmark characteristic of asthma is airway hyperresponsiveness (AHR) defined as an exaggerated constriction response of the airways to a variety of stimuli compared to non-asthmatics. While AHR is often considered a consequence of inflammation [1], the manifestation of AHR is through the contractile ability of the airway smooth muscle (ASM) itself [2]. It has been shown that cyclic stretching can ameliorate contraction of isolated trachealis smooth muscle [3][8] as well as ASM cells in culture [9]. By extrapolation, it has been proposed that physical forces due to tidal breathing and deep inspirations (DI) could work to attenuate the contractile force of ASM [10] and the converse, loss of the ability to impose such mechanical stimuli could amplify the ASM contractile force. Indeed, studies designed to limit airway stretch in normal subjects also result in amplified lung reactivity based on lung function tests [11]. Similarly, in animals when compared to static conditions, tidal breathing was shown to decrease airway responsiveness [12], [13] as well as to relax airways following constriction [14]. This has led to the notion that tidal stretches and DIs fluidize the cytoskeleton (CSK) of ASM cells in situ and the lack of the capacity to impose such stretching may be the basis of AHR in asthma [10].

Recently, LaPrad et al., however, challenged the notion that tidal stretches mitigate airway constriction in intact whole airways [15]. Specifically, they reported that tidal stretches delivered to bovine airways via physiologically realistic transmural pressures were not able to significantly reduce the extent of ASM contraction, measured as airway diameter reduction to acetylcholine (Ach) exposure. Interestingly, even large stretches mimicking DIs had only transient bronchodilatory effects. These findings have been independently confirmed by Noble et al. in airways from non-asthmatic human subjects [16]. Similarly, airways embedded in parenchymal slice obtained from human non-asthmatic lungs showed only a small reversal of contraction with stretches corresponding to tidal breathing [17]. Recently, Ansell et. al. found that ASM strain, rather than stress, is the critical determinant of bronchodilation but surprisingly, the rate of inflation during DIs also impacts on bronchodilation [18]. Furthermore, Harvey et. al. [19] showed that only supra-physiological stresses could impose strains sufficient to obliterate airway constriction in intact airways.

The aforementioned studies present compelling data showing a lack of influence of tidal stretch on intact airway function. However, it is unclear if and how tidal stretches affect the effector proteins and their interactions to regulate bronchial ASM contraction in the intact airway wall. The mechanical factors that determine airway constriction include the contractile force of the ASM, all passive loads against which contraction occurs, and the applied forces due to tidal breathing [20]. The contractile apparatus of ASM cells includes actin-myosin cross-bridges, which is regulated by various intracellular signaling pathways [21]. The extent of phosphorylation of the 20 kDa myosin light chain (MLC) by myosin light chain kinase (MLCK) and myosin light chain phosphatase (MLCP) is the central regulatory mechanism of smooth muscle contraction by initiating cross-bridge formation [22], [23]. Additionally, the ASM is embedded in the extracellular matrix (ECM) of the airway wall that is part of the passive mechanical load. When tidal stretches dilate the airways, both the ECM and the ASM experience mechanical forces and the force transmission between them occurs via integrins. Complexes of multiple integrins are expressed on ASM cells and they modulate connections between the contractile apparatus and the underlying ECM and hence ASM contraction [24]. Furthermore, in response to contractile stimuli, the cell membrane is strengthened by subcortical cytoskeletal actin polymerization [24]. Thus, whether or not tidal stretch-related forces reach the cross-bridge apparatus depends on the structure of the ASM-ECM complex and their mechanical interaction.

Since the above studies suggested that tidal stretches minimally influence the responsiveness of whole bronchial airways [15], [16], we aimed to map the molecular responses of ASM within intact airways to physiologically realistic transmural pressure (Ptm) variations mimicking breathing. To this end, we measured the changes in airway diameter of intact bovine bronchi exposed to static Ptm and dynamic Ptm as well as following Ach stimulation. We determined the expression of the major molecules involved in contractility: ASMA, MLC, MLCK, myosin phosphatase target subunit 1 (MYPT1), smooth muscle myosin heavy chain 2 (SMMHC2); the major structural cytoskeletal molecules: α-actin and α-tubulin, and their connecting partner to ECM, integrin-β1, and its signaling molecule, the focal adhesion kinase (FAK). We found that both stretch patterns significantly alter molecular signaling and subcellular structure but these alterations counterbalance each other and result in a cancellation of their effect on tissue-level airway function, reported as airway luminal diameters.

Methods

Reagents

All chemicals were from Sigma-Aldrich (St Louis, MO) or otherwise stated.

Intact airway segment preparation and experimental setup

Bovine lungs were obtained from a local slaughterhouse (Research 87, Bolyston, MA) immediately after euthanasia and kept chilled. A bronchus of the right lung (generations 10–15, ∼35 mm long and internal radii at Ptm = 10 cmH2O ranging from 2.1 to 4 mm) was freed from parenchyma, and the side branches were closed off. The airways were cannulated at each end and mounted horizontally in a tissue bath containing gassed (95% O2–5% CO2) and heated (37°C) Krebs solution (121 mM NaCl; 5.4 mM KCl; 1.2 mM MgSO4; 25 mM NaHCO3; 5.0 mM sodium morpholinopropane sulphonic acid; 11.5 mM glucose; and 2.5 mM CaCl2). The airways were stretched longitudinally (∼110% of its resting length) and held fixed at its extended length for the entire experiment to ensure that only radial dilation occurs when the airway is pressurized. This amount of axial stretch mimics airway lengthening during tidal breathing [25]. Tissue viability was then confirmed with both electric field stimulation and Ach (10−5 M) challenge, as previously described [15], [25].

Measurement of airway reactivity in vitro - Ptm oscillation protocols

The details of the experimental system are described elsewhere [15]. Briefly, intact airways were mounted as described above in a custom designed system that combined mechanical loading via transmural pressure oscillations with ultrasound imaging. A pressure-controlled syringe pump delivered desired Ptm stimuli to the intact airways accordingly to the hydrostatic pressure applied. A portable ultrasound system (Terason 2000), consisting of a high-frequency linear array transducer (10L5) and an external beamformer module, was used to visualize the intact airway. The ultrasound transducer was mounted above the intact airway and partially submerged in the tissue bath. The airway was imaged with fixed ultrasound imaging settings (focal depth: 30 mm, focal length: 13 mm, gain: 0.2). Airways received either Ptm oscillations (5–10 cmH2O, 0.2 Hz, dynamic loading) or a static Ptm level (7.5 cmH2O, static loading) for 90 minutes with (n = 10) or without (n = 9) a single dose ACh (10−5 M) induced constriction. Ultrasound images were taken in real time. Images at the start (T0) and end (T90) of the protocol were used for normalization and for comparison. The airway lumen and the two airway walls were digitally segmented from the images and airway diameter was determined in the middle of the airway segment.

Inhibitor studies

To block polymerization of microtubules or actin filaments nocodazole (Noc, n = 8) or cytochalasin D (CD, n = 6) was used respectively. The length and diameter of each airway was measured and the inner volumes were calculated. The airways were mounted in the system horizontally as described above and both cannulated ends were closed off to insure constant concentration of the inhibitors. Noc (1µM final concentration) or CD (2µM final concentration) was injected into the lumen of the airways. After a 30 minute incubation period the closed ends were opened and the dynamic loading condition (5–10 cmH2O, 0.2 Hz) was applied for 1.5 hours with a single dose of ACh (10−5 M).

Tissue processing

Immediately after the physiological measurements airway samples were collected: the middle of ∼15 mm tissue was cut from ∼10–10 mm of each end and further divided into two pieces; ∼5 mm was dropped into 10%, neutral buffered formalin fixative (Fisher Scientific, Houston, TX) for histological processing and the other ∼10 mm of the tissue for biochemistry was dropped into T-PER Tissue Protein Extraction Reagent to ensure membrane bound protein solubilization (Thermo Scientific, Pierce Protein Biology Products, Rockford, IL) containing Halt Protease Inhibitor Cocktail and Halt Phosphatase Inhibitor Cocktail (PI, Thermo Scientific, Pierce Protein Biology Products, Rockford, IL) , both at 1.5X final concentration to avoid any protein degradation. The latter was immediately homogenized using PowerGen 125 (Fisher Scientific, Houston, TX) and frozen overnight at −80°C and re-homogenized the next day (with extra PIs, 1X final concentration). To ensure the complete solubilization of membrane bound proteins by the T-PER Tissue Protein Extraction Reagent sodium dodecyl sulfate (SDS) was added. The amount of protein in the supernatant samples was measured using BCA-Reducing Agent Compatible protein assay reagent kit (Pierce, Rockford, IL).

Western blot analysis

Equal amounts of total protein (7.8 µg/sample) were separated using 4–20% SDS-polyacrylamide gels and transferred onto polyvinylidene fluoride membranes (Millipore, Bedford, MA). Western blot analysis was carried out to assess the levels of β-actin, α-tubulin, MLC, the activated form phosho-MLC (ph-MLC), MLCK, MYPT1, SMMHC2, ASMA, integrin-β1 (Int), its phophorylated form (ph-Int), FAK and phospho-FAK (ph-FAK). GAPDH was used as loading control. All primary and secondary antibody incubation as well as the bovine serum albumin blocking step was done for one hour. All antibodies were from Abcam Inc. (Cambridge, MA) exept MYPT1 (Thermo Scientific, Pierce Protein Biology Products, Rockford, IL). Quantitative densitometry was performed after chemiluminescence detection using Pierce ECL substrates or SuperSignal West Pico Chemiluminescent Substrate (Thermo Scientific, Pierce Protein Biology Products, Rockford, IL) and corrections were made for background and loading control.

Single and double label immunohistochemistry (IHC)

Formalin (10%, neutral buffered) fixed, paraffin-embedded sections of airways were deparaffinized in xylene and rehydrated in decreasing alcohol series. Endogenous peroxidase activity was quenched by 1% H2O2 and sections were washed in 10 mM sodium phosphate buffer, 150 mM NaCl (PBS), pH 7.5. Protein blocking step was performed with horse serum and sections were incubated for 1 hr with anti-β-actin or anti-α-tubulin or anti-ASMA or anti-F-actin antibodies. Mouse or rabbit IgG (20 ng/ml) as well as omitting the primary or secondary antibodies were used as technical controls. All antibodies were from Abcam Inc. (Cambridge, MA). After PBS washes, the mouse or rabbit HRP conjugated secondary antibodies (Vector Lab, Burlingame CA) were applied for 1 hr. Sections were washed in PBS and incubated for 30 min in VECTASTAIN ABC reagent (Vector Lab). Enzyme substrates (Vector Lab) were applied until the right colors developed: DAB (brown), Vector SG (blue/gray). After this step, either counter staining (Nuclear Fast Red, Vector Lab) and dehydration-clearing-mounting or, in the case of ASMA, a second antigen labeling with F- actin antibody was applied following the protocol as above on a second set of sections. All conditions were processed simultaneously for each antibody (n = 30/condition). Images were captured by a Nikon Eclipse 50i microscope and SPOT camera (Micro Video Instruments, Avon, MA) and histological evaluation was performed.

Statistical analysis

All data were expressed as means ±SE. One-way repeated measures ANOVA was used to determine the effect of the Ptm loading conditions on luminal radii. One-way ANOVA and unpaired t-tests were used on the cytoskeletal, adhesion and contractile molecules of intact airways for no Ach and Ach conditions, respectively. When the normality test failed, the corresponding non-parametric statistical procedures were used. Post hoc comparisons included Holm-Sidak and Tukey tests for parametric and non-parametric one-way ANOVA, respectively. Statistical significance was accepted at p<0.05.

Results

In intact airways, Ach and cytoskeleton, but not dynamic Ptm determine airway diameters

We measured the changes in airway diameter of intact bovine bronchi during static Ptm and dynamic Ptm applying physiologically realistic pressures to mimic breathing. Figure 1A shows representative ultrasound images at time zero (T0) and after 90 minutes (T90) in the presence or absence of Ach stimulation during static (7.5 cmH2O) or dynamic (±2.5 cmH2O sinusoidal pressure variations around a mean Ptm of 7.5 cmH2O) loading conditions. Applying dynamic or static mechanical stresses resulted in similar diameters of intact airways without constriction. The application of Ach reduced airway diameter to about 55% of its value before constriction (p<0.05) independent of the mechanical loading condition consistent with our earlier findings [15]. To assess how the main cytoskeletal load bearing proteins contribute to airway responsiveness, the experiments were repeated in the presence of cytochalasin D (CD), a drug that disrupts actin polymerization, as well as nocodazole (Noc), which inhibits microtubule polymerization (Figure 1B). As expected, disassembling the actin fibers substantially mitigated ASM contraction. However, inhibition of microtubules enhanced responsiveness of intact airways further reducing the airway diameter by 30%. Airway wall circumferential strain, defined as the difference between internal radii at Ptm of 10 and 5 cmH2O divided by the radius at Ptm = 5 cmH2O at baseline, showed a decreasing trend with time without Ach and without inhibitors (2.6% at T0 vs 2.1% at T90; p = 0.07) but it did not change with Ach, CD or Noc. On average, the strain was 2.7±0.8% in agreement with previous findings [15].

thumbnail
Figure 1. In intact airways, Ach and cytoskeleton, but not dynamic Ptm determine airway diameters.

Effects of static or dynamic mechanical loading conditions are shown during acetylcholine-induced (ACh) constriction in intact bovine airways in the absence (A) or presence (B) of cytoskeletal de-depolymerizing agents. Top panels show representative ultrasound images. Bottom graphs show the mean and SD of airway radii relative to pre-treatment condition (A; n: 9 and 10 and B; n: 6 and 8) CD: cytochalasin D, an actin depolymerizing agen; Noc: nocodazole, a microtubule depolymerizing agent.

https://doi.org/10.1371/journal.pone.0094828.g001

Tidal stretch differently regulates the tensile and compressive cytoskeletal elements

To evaluate the involvement of the cytoskeleton in airway responsiveness, we characterized the levels of the tensile element β-actin and the compressive element α-tubulin, the two major load bearing components of the cytoskeleton [26]. Figure 2 shows example Western blots and the corresponding statistics of β-actin (A and B) and α-tubulin (C and D) with (B and D) and without (A and C) Ach-induced constriction during both static and dynamic loading conditions. In the absence of constriction, the amount of β-actin was not significantly different between T0 and either loading condition. However, compared to static loading, the amount of β-actin was statistically significantly lower by 24% during dynamic loading (Figure 2A). At the end of the 90 min Ach-induced constriction, dynamic loading resulted in a very similar 27% lower β-actin compared to static loading (Figure 2B; p<0.001). Tubulin slightly but statistically significantly increased (∼23%) in the unconstricted airway compared to T0 (p<0.001) for both loading conditions (Figure 2C). During constriction, there was no difference between the tubulin levels (Figure 2D). Thus, assessment of the CSK load-bearing elements suggests that Ptm did not affect tubulin regardless of conditions but the levels of β-actin were lower during dynamic Ptm oscillations.

thumbnail
Figure 2. Tidal stretch differently regulates the tensile and compressive cytoskeletal elements.

Effects of static and dynamic Ptm on β-actin (act, ∼42 KDa) (A and B) and α-tubulin (tub, ∼50 KDa) (C and D) in intact bovine airways after 90 min of mechanical loading with (B and D) or without (A and C) constriction assessed by Western blots. T0: time 0 before stretch, St: static Ptm. D: dynamic Ptm oscillation. GAPDH (∼35 KDa) is loading control. Data are normalized such that the mean of the dynamic group is unity.

https://doi.org/10.1371/journal.pone.0094828.g002

Tidal stretch has opposite effects on ASMA and SMMHC2 levels in Ach activated airways than in non-activated airways

Next, we assessed how the two major contractile cross-bridge forming proteins, ASMA and SMMHC2, respond to stretch and stimulation. Both protein levels were significantly influenced by stretch pattern and Ach stimulation (Figure 3). Without contractile stimulation, ASMA (Figure 3A) was up-regulated by dynamic loading (i.e., in the opposite direction as β-actin (Figure 2A)) whereas during Ach-induced stimulation, ASMA was down regulated by dynamic loading (Figure 3B) (i.e., in the same direction as β-actin (Figure 2B)). Surprisingly, SMMHC2 levels changed exactly in the opposite direction as ASMA both with stretch and Ach activation: in the absence of Ach, static loading significantly upregulated SMMHC2 (Figure 3C) compared to both T0 and dynamic loading (p<0.001) whereas during Ach stimulation, SMMHC2 level was 44% higher (Figure 3D) during dynamic compared to static loading (p<0.001).

thumbnail
Figure 3. Tidal stretch has opposite effects on ASMA and SMMHC2 levels in constricted airways than in non-constricted airways.

Effects of Ptm on the cross-bridge forming contractile proteins α-smooth muscle actin (ASMA, ∼42 KDa) (A and B) and smooth muscle myosin heavy chain 2 (SMMHC2, ∼230 KDa) (C and D) in intact bovine airways after 90 min of mechanical loading with (B and D) or without (A and C) constriction assessed by Western blot. T0: time 0 before stretch, St: static Ptm. D: dynamic Ptm oscillation. GAPDH (∼35 KDa) is loading control. Data are normalized such that the mean of the dynamic group is unity.

https://doi.org/10.1371/journal.pone.0094828.g003

Tidal stretch increases MLC activation during contraction

We also determined the level and activation of MLCK, MYPT1 and MLC. While MLCK activates the regulatory 20 kDa MLC through phosphorylation allowing the formation of actomyosin and subsequently initiating ASM contraction via the sliding filament process, MLCP attenuates this process [27]. However, phosphorylation of MYPT1, the myosin binding subunit of MLCP, hinders MLCP activity which in turn leads to MLC phosphorylation [28]. In our experiments, MLCK followed the same pattern as SMMHC2 in the absence of Ach, static loading significantly upregulated MLCK (Figure 4A) compared to both T0 and dynamic loading (p<0.001) whereas during Ach stimulation, MLCK level was 51% higher (Figure 4B) during dynamic compared to static loading (p<0.03). MYPT1 was not sensitive to stretch in the absence of Ach (Figure 4C), but increased during dynamic loading in the presence of Ach (p<0.001, Figure 4D). MLC, on the other hand, did not change with stretch pattern or stimulation (Figure 4E) whereas its phosphorylated form was significantly upregulated by dynamic loading during Ach challenge only (Figure 4F; p<0.001) following the pattern of MYPT1. Thus, even though an increased molecular motor activity can be observed (up-regulation of SMMHC2, MLCK and hence phMLC) during Ptm oscillations, the limiting factor in force generation appears to be ASMA which is required for cross-bridge formation but is down-regulated (Figure 3B).

thumbnail
Figure 4. MLC activated only during dynamic loading with Ach challenge.

Effects of Ptm on the cross-bridge activating contractile proteins MLCK (∼210 KDa) (A and B), MYPT1 (∼140 KDa) (C and D) MLC (∼20 KDa), and its activated form phMLC (∼20 KDa) (E and F) of intact bovine airway after 90 min of mechanical loading with (B, D and F) or without (A, C and E) constriction assessed by Western blot. Notice that the same membrane was used in B as in figure 3D. T0: time 0 before stretch, St: static Ptm. D: dynamic Ptm oscillation. GAPDH (∼35 KDa) is loading control. Data are normalized such that the mean of the dynamic group is unity.

https://doi.org/10.1371/journal.pone.0094828.g004

The subcellular structures of actin and microtubule during static and dynamic loading

Figure 5 shows examples of ASMA, β-actin and α-tubulin following stretch in the absence of constriction. Figure 5A demonstrates that ASMA (presented as blue color) is more homogeneous at T0 than following both static and dynamic loading conditions under which it displays straight, fibrous structures. Cell nuclei especially within the muscle bundle are elongated with their long axis parallel to the fibers with both stretches. The spatial distribution of β-actin (presented as brown color) is also homogeneous at T0 and becomes patchy in appearance spanning through multiple neighboring cells throughout the smooth muscle layer in both loading conditions (Figure 5B). Similarly, the α-tubulin labeling (presented as blue color) appears stronger and more heterogeneous in both static and dynamic loading conditions compared to T0 (Figure 5C).

thumbnail
Figure 5.

The subcellular structures of actin and microtubule are similar during static and dynamic loading. Immunohistochemistry (IHC) shows the distribution of ASMA (A), β-actin (B) and α-tubulin (C) on representative sections of bovine intact airways at time 0 (before stretch, T0) and after 90 min of static or dynamic Ptm loading conditions without constriction. β-actin represented as brown (B), ASMA, and α-tubulin are represented as blue (A and C) and nuclei as pink (A, B and C). The inset in the top rows shows the technical control for IHC and counterstained for nuclei. All conditions were processed simultaneously for each antibody (n = 30/condition). The insets in the bottom row are enhanced images for better cellular visualization. Scale bars are 100 µm on the top row sections and 50 µm on the bottom rows.

https://doi.org/10.1371/journal.pone.0094828.g005

Following Ach challenge (Figure 6A), both loading conditions display straight, strong fiber structures for α- and β-actin (Figure 6, Aa, Ab, Ac and Ad), but the nuclei appear shorter and less elongated than without Ach in Figure 5. Sections from only static loading for both β-actin and ASMA show circular patchy appearance spanning through multiple neighboring cells throughout the smooth muscle layer (Figure 6, Aa and Ac). Under static loading, ASMA patches also show strong co-localization with patches of F-actin positive fibers (Figure 6, Ae). Under dynamic loading, there was little evidence of F-actin formation (Figure 6, Af). However, the staining for α-tubulin appears more homogeneously distributed under static condition and patchy under dynamic condition (Figure 6, Ag and Ah) during constriction.

thumbnail
Figure 6. Airway constriction changes the subcellular structures of actin and microtubule.

Single and double-labeled immunohistochemistry showing the distribution of β-actin, ASMA, the polymerized actin (F-actin) and α-tubulin on representative sections of bovine intact airways after static and dynamic Ptm conditions (90 min mechanical loading) and with constriction (A) and actin or microtubulin depolymerization (B). β-actin and F-actin are shown in brown; ASMA and α-tubulin are blue and nuclei are pink (A, and B). All conditions were processed simultaneously for each antibody (n = 30/condition). Scale bars are 100 µm on the top row sections and 50 µm on the bottom rows.

https://doi.org/10.1371/journal.pone.0094828.g006

The impacts of actin or microtubule depolymerization on subcellular structures

Since inhibition of actin and microtubules resulted in substantial physiological responses (Figure 1), we next examined how their molecular organization is affected by static and dynamic loading. In the presence of CD, ASMA (blue) becomes disorganized and fragmented with shortened and wavy fibers (Figure 6, Ba and Bb) suggesting that they do not carry force. The cytoskeletal β-actin (brown) appears more pronounced and heterogeneously distributed after CD treatment due to fragmentation and retraction of the polymers; the fibers are less elongated and found mostly around the nuclei while leaving unstained spaces between nuclei (Figure 6, Bc and Bd). The connections between cells and the patchy distribution disappear for both ASMA and β-actins. The microtubule fibers are also affected by CD treatment: they appear less pronounced and patchy (Figure 6, Be and Bf). Application of Noc resulted in disorganized α-tubulin (blue): the fibers appear fragmented, shortened and wavy (Figure 6, Be and Bf). The actin fibers are also affected by Noc treatment: ASMA appears more pronounced, less elongated around the nuclei (Figure 6, Ba and Bb), while β-actin staining is greatly diminished (Figure 6, Bc and Bd). The nuclei also appear less elongated (Figure 6B).

Tidal stretch increases the level and activity of integrin-β1 and activates its signaling partner FAK during contraction

All the above molecular responses and the subcellular structures are influenced by cell-ECM interactions through integrins. We thus examined these interactions by assessing the effects of stretch and Ach challenge on integrin-β1 and one of its signaling kinase FAK as well as their activation through phosphorylation, summarized in Figure 7. Integrin-β1 did not change with stretch in the absence of Ach (A), but increased during dynamic loading compared to static loading when the airway was constricted (p<0.001) (B). The phosphorylated form of integrin-β1 slightly increased with stretch compared to T0 (p<0.02) but this increase did not depend on stretch pattern (A) whereas during Ach challenge, dynamic loading resulted in a 28% higher phosphorylation of integrin-β1 than static loading (p<0.001) (B). The phosphorylated form of FAK was downregulated by stretch compared to T0 (p<0.001) independent of stretch pattern (C), whereas they were both upregulated by dynamic loading especially the phosphorylated form (p<0.001) after constriction (D). Thus, without constriction, integrin and its activation are similar during both mechanical loading conditions and hence integrins maintain stretch pattern-independent cell-ECM interactions. However, during constriction and dynamic stretch, enhanced coupling occurs perhaps to support the larger peak stresses than during the static condition.

thumbnail
Figure 7. Tidal stretch increases the level and activity of integrin-β1 and activates its signaling partner FAK during contraction.

Western blots are shown for integrin-β1 (Int, ∼140 KDa) and its phosphorylated form (ph-Int) (A and B) and for focal adhesion kinase (FAK, ∼125 KDa) and its phosphorylated form (ph-FAK) (C and D) in intact bovine airways after 90 min of mechanical loading with (B and D) or without (A and C) constriction. T0: time 0 before stretch, St: static Ptm. D: dynamic Ptm oscillation. GAPDH (∼35 KDa) is loading control. Data are normalized such that the mean of the dynamic group is unity.

https://doi.org/10.1371/journal.pone.0094828.g007

Discussion

In this study, we characterized the cellular and molecular determinants of airway responsiveness to stretch and constriction in intact bovine bronchi having radii between 2 and 4 mm corresponding to human airway generations of 4 to 9 [29]. The main results showed that following 90 min of stretch during Ach stimulation, the stretch pattern (static vs dynamic) had significant impacts on the expression of 1) the cytoskeletal stiffness-related protein β-actin, 2) the most important contractile proteins (ASMA, SMMHC2, MLC, MLCK and MYPT1) and their activated phosphorylated forms, 3) cell-ECM coupling proteins (integrin- β1 and FAK) and their activated forms as well as 4) the spatial organization of the contractile apparatus. An important implication of these results is that imposing tidal levels of Ptm stress on intact airways do initiate changes in subcellular expressions of proteins from the static state despite the small circumferential strains (2–4%). This further implies that these small dynamic strains imposed on the airways following constriction trigger various molecular machineries that could potentially mitigate constriction. However, as we explain below, our results show a unique up- and down-regulation of key proteins suggesting that these molecular processes are compensatory and self-canceling at the airway level. Therefore, the net result is zero change in airway diameter due to dynamic stretch implying that larger strains may be required to break the compensatory pathways which could then lead to substantial lasting dilation [19]. Furthermore, since the strains were on average the same before and after stimulation, any difference in molecular expressions between the unconstructed and constricted conditions during tidal stretching is primarily due to the direct signaling elicited by Ach.

Our results showed that the cytoskeletal β-actin was not different at the beginning and at the end of the protocol without constriction (Figure 2A). However, compared to static loading, dynamic loading slightly but statistically significantly down regulated β-actin both in the absence and presence of Ach (Figure 2A and 2B) by 24% and 27%, respectively. In fact, the difference between the 24% and 27% was not statistically significant suggesting that the down regulation of β-actin was due to the dynamic loading independent of whether the muscle was activated by Ach. Since β-actin is an important determinant of cell stiffness [30] and α-tubulin did not change with stretch pattern (Figure 2C and D), our results imply that sinusoidal stretching around a mean Ptm leads to a softening of ASM cells in the intact airway in agreement with cell culture studies [31] which would present a slightly reduced passive load against muscle contraction.

The contractile machinery was significantly influenced by the nature of stretch including the cross-bridge forming ASMA and SMMHC2 (Figure 3) as well as the regulators MLC, MLCK, MYPT1 and their phosphorylated forms (Figure 4). During dynamic loading compared to static loading, ASMA decreased in the presence of and increased in the absence of Ach which, if acting alone, would imply a larger and a smaller airway diameter, respectively. In contrast, SMMHC2, a key determinant of the contractile force, followed an inverse pattern implying alone a smaller and a larger airway diameter with and without Ach, respectively. The simultaneous up and down regulations of these contractile proteins are reversed during static stretch. This double inverse regulation of the contractile proteins would effectively cancel each other's effect on ASM force.

Interestingly, the changes in integrin- β1 and FAK also appeared to follow an inverse pattern of the β-actin suggesting that while muscle stiffness may have decreased during dynamic loading, the cells tended to interact more strongly with the ECM potentially also canceling each other's effect.

These inverse effects during static and dynamic loading can thus maintain the airway at the same luminal diameter following Ach-induced stimulation. A proposed mechanism is summarized in Figure 8. Furthermore, this mechanism is robust since it likely holds for both human and bovine isolated whole airways under a range of dynamic stresses (from 0 to 10 cmH2O) with and without intermittent DIs, at least at two mean Ptm levels (5 and 7.5 cmH2O) and a wide range of Ach doses (from 10−7 to 10−3 M) since little difference was found in airway responsiveness during static and dynamic loading for these conditions in two independent studies [15], [16]. Given the relatively short 90 min protocol, a likely explanation is that the molecular changes seen in this study are a result of posttranslational alterations such as regulated protein degradation.

thumbnail
Figure 8. Proposed mechanism for the lack of stretch-induced airway dilation.

The cytoskeleton and connection to ECM by integrins determine and regulate airway diameter through double inverse regulation of ASMA and SMMHC2 by stretch and agonist stimulation. Notice that without constriction (left), changes in protein levels are relative to their values at T0, whereas during constriction (right), changes in protein levels in the static condition are compared to their levels during dynamic conditions.

https://doi.org/10.1371/journal.pone.0094828.g008

The internal structure of ASM visualized in Figures 5 and 6 suggests that mechanical stimuli, static or dynamic, reinforce actin and tubulin fibers in the absence of Ach. Since the cell nuclei appear elongated, the intracellular actin fibers must be under tension suggesting the presence of inherent muscle tone. Following Ach-induced contraction, the major difference in structure is seen on the ASMA (F-actin double labeled tissue in Figure 6A e and f). Specifically, the cell nuclei appear less elongated outside the muscle bundle in the case of the static loading and the cell nuclei are less elongated even inside muscle bundle following dynamic loading. These observations suggest a heterogeneous distribution of mechanical forces within the airway wall as well as within the ASM cell. Furthermore, whereas F-actin co-localizes with ASMA following static loading, little F-actin is seen after dynamic loading. The latter implies that tidal stretches do strain ASM cells and do break up the actin inside the ASM within the intact airway consistent with the notion of fluidization [32]. Yet, the reduced levels of phosphorylated MLC (Figure 4F) and myosin motors (Figure 3D) during static loading inhibits the muscle from constricting more than during dynamic loading.

What determines the final level of constriction for a given Ach dose as seen in Figure 1? We attempted to answer this question by inhibiting the polymerization of actin or tubulin during dynamic loading. As expected, depolymerization of actin resulted in dilation because fibrous actin, which is required for force generation, was disintegrated (Figure 6 A and B). However, depolymerization of microtubules enhanced airway responsiveness (Figure 1). The nocodazole treatment resulted in disorganized microtubules and washed out β-actin but it generated relatively strong ASMA fibers (Figure 6B). Thus, the amount and activation level of myosin motors together with the amount and organization ASMA fibers determine the total contractile force within a single ASM cell. This force has to be balanced by other structures in the cell and the elastic links between cells as well as the coupling of the cell to the ECM. Our results suggest that the network organization of microtubules is an important intracellular structure limiting cell contraction. The mechanism maybe related to the idea that microtubules can carry compressive loads [33] and hence the resistance of the cell against shortening might partially be due to the compressive modulus of the microtubule network. However, another possibility is that nocodazole directly influences phMLC. Indeed, microtubule disruption has been shown to increase MLC phosphorylation in isolated fibroblasts [34] as well as in porcine coronary arterial rings [35]. To test this possibility, we carried out a complementary experiment to determine the levels of phMLC following CD and Noc treatment during Ach-induced constriction. The data in Figure 9 demonstrate that increased MLC phosphorylation does occur in intact airways following both microtubule disruption by Noc and actin disruption by CD. These results confirm that microtubule structure plays a role in MLC activation, although we cannot exclude the possibility that microtubules also participate in the force balance without Noc, Therefore, airway diameter is ultimately governed by how the total contractile force (determined by SMMHC2 and ASMA) is balanced by the compressibility of structures within the ASM cells and internal to the entire ASM layer as well as the elasticity of the ECM external to the ASM cells at a given Ptm.

thumbnail
Figure 9. MLC activation during Ach challenge and dynamic loading in the presence of CD or Noc.

phMLC is significantly increased by the end of 90: time 0 before stretch, CD+Ach and Noc+Ach correspond to phMLC at the end of 90 min stretch in the presence of CD or Noc, respectively. GAPDH (∼35 KDa) is loading control. Data are presented in arbitrary units (AU). Statistical significance is given for the comparisons between T0 CD and CD+Ach and between T0 Noc and Noc+Ach.

https://doi.org/10.1371/journal.pone.0094828.g009

What are the possible implications of our results to asthma? The airway wall structure in asthmatics is certainly different from that of normal subjects and varies with asthma severity [36]. However, even in moderate asthmatics without discernible difference in the ECM within the ASM layer from normal subjects, deterioration in airway function positively correlated with increasing amounts of collagen I, collagen III and laminin [37] implying an enhanced ability of ASM to contract in the presence of increased amounts of ECM proteins within the ASM layer. There is also increased ASM mass in the airway walls of asthmatics [38]. Furthermore, alteration in the composition of integrin-β1 complexes was shown to modify ASM function: the α2β1, α4β1 and α5β1 integrins can mediate airway smooth muscle proliferation by collagen I through PDGF BB [39] whereas the integrin α9β1 suppressed exaggerated airway narrowing [40]. Additionally, methacholine sensitivity was found to inversely correlate with the expressions of ASMA and desmin whereas deep inspiration-induced bronchodilation was inversely related to desmin, MLCK and calponin [41]. Desai et al. also showed that IL-13 and mechanical stimuli interact at the integrin adhesion complexes and regulate SMMHC expression in airway smooth muscle tissue [42]. Our study further adds to the complexities of ASM regulation by suggesting that the contractile apparatus itself is under a delicate balance during Ach-induced stimulation and tidal stretching. We speculate that disturbance of this balance by sustained or altered mechanical and biochemical conditions may contribute to AHR in asthmatics. Therefore, the interruption of this newly developed pathological balance can be beneficial in asthma treatment. Indeed, the application of continuous airway pressure for seven days reduced airway reactivity in clinically stable asthmatics [43].

There are several limitations of the study. The experimental setup has fixed boundaries at the two ends of the airway whereas in vivo the airways can also shorten axially. Such axial shortening is not likely to be considerable during constriction because the pitch angle of the muscle spiral around the airway is only about 13 degrees [44]. The fixed boundaries also imposed fixed diameters at the two ends. To minimize the influence of the boundaries, we analyzed diameters only at the middle of the airway. Additionally, in an earlier study, airways were found to easily reach full collapse along most of its length at a sufficiently high Ach dose [15] which also suggests that the lack of surface tension in the fluid filled system is not a major limitation. The ex vivo preparation does not have innervation. We used Ach as a stimulant which is the neurotransmitter released from the parasympathetic nerve endings that regulate airway tone in situ. Our results are likely not species specific because Noble et al. found similar lack of sustained influence of stretch on constriction in human airways [16]. Nevertheless, these authors also used Ach; hence, our results may be specific to cholinergic excitation of the ASM. The biochemical assays measure the average expression of proteins from tissue homogenate. Thus, some of the protein expressions do not strictly represent ASM. For example, every adherent cell contains β-actin and integrin and fibroblasts also express some contractile proteins. The ASM is a major cellular component of the airway wall and our images show (Figs. 5 and 6) that most of the β-actin and the contractile proteins come from this cell type. To confirm the source of proteins, in three additional airways we removed the adventitia and the mucosa from the smooth muscle layer. We did not find significant differences in the biochemical evaluation between the whole airway and the dissected smooth muscle layer.

In summary, we found that the lack of airway responsiveness to tidal stretching in intact airways stimulated in vitro is related to an inverse regulation of contractile proteins by stretch pattern and contractile stimulation while the level of constriction is determined by the availability of ASMA/phMLC and the structure of both actin and tubulin. In order to be able to exploit the mechanisms revealed in this study for the treatment of asthma, experiments should validate the findings in animal models of asthma and ultimately in asthmatic patients.

Author Contributions

Conceived and designed the experiments: EBS ASL. Performed the experiments: EBS ASL BCH. Analyzed the data: EBS ASL BS. Wrote the paper: EBS BS KRL.

References

  1. 1. Meurs H, Gosens R, Zaagsma J (2008) Airway hyperresponsiveness in asthma: lessons from in vitro model systems and animal models. Eur Respir J 32: 487–502.
  2. 2. An SS, Bai TR, Bates JH, Black JL, Brown RH, et al. (2007) Airway smooth muscle dynamics: a common pathway of airway obstruction in asthma. Eur Respir J 29: 834–860.
  3. 3. Fredberg JJ, Inouye D, Miller B, Nathan M, Jafari S, et al. (1997) Airway smooth muscle, tidal stretches, and dynamically determined contractile states. Am J Respir Crit Care Med 156: 1752–1759.
  4. 4. Fredberg JJ, Jones KA, Nathan M, Raboudi S, Prakash YS, et al. (1996) Friction in airway smooth muscle: mechanism, latch, and implications in asthma. J Appl Physiol 81: 2703–2712.
  5. 5. Latourelle J, Fabry B, Fredberg JJ (2002) Dynamic equilibration of airway smooth muscle contraction during physiological loading. J Appl Physiol 92: 771–779.
  6. 6. Naghshin J, Wang L, Pare PD, Seow CY (2003) Adaptation to chronic length change in explanted airway smooth muscle. J Appl Physiol 95: 448–453 discussion 435.
  7. 7. Oliver MN, Fabry B, Marinkovic A, Mijailovich SM, Butler JP, et al. (2007) Airway hyperresponsiveness, remodeling, and smooth muscle mass: right answer, wrong reason? Am J Respir Cell Mol Biol 37: 264–272.
  8. 8. Wang L, Pare PD, Seow CY (2000) Effects of length oscillation on the subsequent force development in swine tracheal smooth muscle. J Appl Physiol 88: 2246–2250.
  9. 9. An SS, Fabry B, Trepat X, Wang N, Fredberg JJ (2006) Do biophysical properties of the airway smooth muscle in culture predict airway hyperresponsiveness? Am J Respir Cell Mol Biol 35: 55–64.
  10. 10. Krishnan R, Trepat X, Nguyen TT, Lenormand G, Oliver M, et al. (2008) Airway smooth muscle and bronchospasm: fluctuating, fluidizing, freezing. Respir Physiol Neurobiol 163: 17–24.
  11. 11. Skloot G, Permutt S, Togias A (1995) Airway hyperresponsiveness in asthma: a problem of limited smooth muscle relaxation with inspiration. J Clin Invest 96: 2393–2403.
  12. 12. Shen X, Gunst SJ, Tepper RS (1997) Effect of tidal volume and frequency on airway responsiveness in mechanically ventilated rabbits. J Appl Physiol 83: 1202–1208.
  13. 13. Tepper RS, Shen X, Bakan E, Gunst SJ (1995) Maximal airway response in mature and immature rabbits during tidal ventilation. J Appl Physiol 79: 1190–1198.
  14. 14. Brown R, Mitzner W (2001) Effects of tidal volume stretch on airway constriction in vivo. J Appl Physiol 91: 1995–1998.
  15. 15. Laprad AS, Szabo TL, Suki B, Lutchen KR (2010) Tidal Stretches Do Not Modulate Responsiveness of Intact Airways in-Vitro. J Appl Physiol.
  16. 16. Noble PB, Jones RL, Needi ET, Cairncross A, Mitchell HW, et al. (2011) Responsiveness of the human airway in vitro during deep inspiration and tidal oscillation. J Appl Physiol 110: 1510–1518.
  17. 17. Lavoie TL, Krishnan R, Siegel HR, Maston ED, Fredberg JJ, et al. (2012) Dilatation of the constricted human airway by tidal expansion of lung parenchyma. Am J Respir Crit Care Med 186: 225–232.
  18. 18. Ansell TK, McFawn PK, Mitchell HW, Noble PB (2013) Bronchodilatory response to deep inspiration in bronchial segments: the effects of stress vs. strain. J Appl Physiol (1985) 115: 505–513.
  19. 19. Harvey BC, Parameswaran H, Lutchen KR (2013) Can Tidal Breathing with Deep Inspirations of Intact Airways Create Sustained Bronchoprotection or Bronchodilation? J Appl Physiol.
  20. 20. Bates JH, Maksym GN (2011) Mechanical determinants of airways hyperresponsiveness. Crit Rev Biomed Eng 39: 281–296.
  21. 21. Clarke D, Damera G, Sukkar MB, Tliba O (2009) Transcriptional regulation of cytokine function in airway smooth muscle cells. Pulm Pharmacol Ther 22: 436–445.
  22. 22. Horowitz A, Menice CB, Laporte R, Morgan KG (1996) Mechanisms of smooth muscle contraction. Physiol Rev 76: 967–1003.
  23. 23. Sieck GC, Han YS, Pabelick CM, Prakash YS (2001) Temporal aspects of excitation-contraction coupling in airway smooth muscle. J Appl Physiol (1985) 91: 2266–2274.
  24. 24. Zhang W, Gunst SJ (2008) Interactions of airway smooth muscle cells with their tissue matrix: implications for contraction. Proc Am Thorac Soc 5: 32–39.
  25. 25. Khangure SR, Noble PB, Sharma A, Chia PY, McFawn PK, et al. (2004) Cyclical elongation regulates contractile responses of isolated airways. J Appl Physiol 97: 913–919.
  26. 26. Stamenovic D (2008) Cytoskeletal mechanics in airway smooth muscle cells. Respir Physiol Neurobiol 163: 25–32.
  27. 27. Kamm KE, Stull JT (1985) The function of myosin and myosin light chain kinase phosphorylation in smooth muscle. Annu Rev Pharmacol Toxicol 25: 593–620.
  28. 28. Pfitzer G (2001) Invited review: regulation of myosin phosphorylation in smooth muscle. J Appl Physiol (1985) 91: 497–503.
  29. 29. Horsfield K, Dart G, Olson DE, Filley GF, Cumming G (1971) Models of the human bronchial tree. J Appl Physiol 31: 207–217.
  30. 30. Van Citters KM, Hoffman BD, Massiera G, Crocker JC (2006) The role of F-actin and myosin in epithelial cell rheology. Biophys J 91: 3946–3956.
  31. 31. Eldib M, Dean DA (2011) Cyclic stretch of alveolar epithelial cells alters cytoskeletal micromechanics. Biotechnol Bioeng 108: 446–453.
  32. 32. Trepat X, Deng L, An SS, Navajas D, Tschumperlin DJ, et al. (2007) Universal physical responses to stretch in the living cell. Nature 447: 592–595.
  33. 33. Stamenovic D, Coughlin MF (1999) The role of prestress and architecture of the cytoskeleton and deformability of cytoskeletal filaments in mechanics of adherent cells: a quantitative analysis. J Theor Biol 201: 63–74.
  34. 34. Kolodney MS, Elson EL (1995) Contraction due to microtubule disruption is associated with increased phosphorylation of myosin regulatory light chain. Proc Natl Acad Sci U S A 92: 10252–10256.
  35. 35. Paul RJ, Bowman PS, Kolodney MS (2000) Effects of microtubule disruption on force, velocity, stiffness and [Ca(2+)](i) in porcine coronary arteries. Am J Physiol Heart Circ Physiol 279: H2493–2501.
  36. 36. Pepe C, Foley S, Shannon J, Lemiere C, Olivenstein R, et al. (2005) Differences in airway remodeling between subjects with severe and moderate asthma. J Allergy Clin Immunol 116: 544–549.
  37. 37. Yick CY, Ferreira DS, Annoni R, von der Thusen JH, Kunst PW, et al. (2012) Extracellular matrix in airway smooth muscle is associated with dynamics of airway function in asthma. Allergy 67: 552–559.
  38. 38. Ebina M, Takahashi T, Chiba T, Motomiya M (1993) Cellular hypertrophy and hyperplasia of airway smooth muscles underlying bronchial asthma. A 3-D morphometric study. Am Rev Respir Dis 148: 720–726.
  39. 39. Nguyen TT, Ward JP, Hirst SJ (2005) beta1-Integrins mediate enhancement of airway smooth muscle proliferation by collagen and fibronectin. Am J Respir Crit Care Med 171: 217–223.
  40. 40. Chen C, Kudo M, Rutaganira F, Takano H, Lee C, et al. (2012) Integrin alpha9beta1 in airway smooth muscle suppresses exaggerated airway narrowing. J Clin Invest 122: 2916–2927.
  41. 41. Slats AM, Janssen K, van Schadewijk A, van der Plas DT, Schot R, et al. (2008) Expression of smooth muscle and extracellular matrix proteins in relation to airway function in asthma. J Allergy Clin Immunol 121: 1196–1202.
  42. 42. Desai LP, Wu Y, Tepper RS, Gunst SJ (2011) Mechanical stimuli and IL-13 interact at integrin adhesion complexes to regulate expression of smooth muscle myosin heavy chain in airway smooth muscle tissue. Am J Physiol Lung Cell Mol Physiol 301: L275–284.
  43. 43. Busk M, Busk N, Puntenney P, Hutchins J, Yu Z, et al. (2013) Use of continuous positive airway pressure reduces airway reactivity in adults with asthma. Eur Respir J 41: 317–322.
  44. 44. Lei M, Ghezzo H, Chen MF, Eidelman DH (1997) Airway smooth muscle orientation in intraparenchymal airways. J Appl Physiol 82: 70–77.