Abstract
Idiopathic pulmonary fibrosis is associated with a decreased expression of caveolin-1 (cav-1), yet its role remains unclear. To investigate the role of cav-1, we induced pulmonary fibrosis in wild-type (WT) and cav-1–deficient (cav-1−/−) mice using intratracheal instillation of bleomycin. Contrary to expectations, significantly less collagen deposition was measured in tissue from cav-1−/− mice than in their WT counterparts, consistent with reduced mRNA expression of procollagen1a2 and procollagen3a1. Moreover, cav-1−/− mice demonstrated 77% less α-smooth muscle actin staining, suggesting reduced mesenchymal cell activation. Levels of pulmonary injury, assessed by tenascin-C mRNA expression and CD44v10 detection, were significantly increased at Day 21 after injury in WT mice, an effect significantly attenuated in cav-1−/− mice. The apparent protective effect against bleomycin-induced fibrosis in cav-1−/− mice was attributed to reduce cellular senescence and apoptosis in cav-1−/− epithelial cells during the early phase of lung injury. Reduced matrix metalloproteinase (MMP)-2 and MMP-9 expressions indicated a low profile of senescence-associated secretory phenotype (SASP) in the bleomycin-injured cav-1−/− mice. However, IL-6 and macrophage inflammatory protein 2 were increased in WT and cav-1−/− mice after bleomycin challenge, suggesting that bleomycin-induced inflammatory response substantiated the SASP pool. Thus, loss of cav-1 attenuates early injury response to bleomycin by limiting stress-induced cellular senescence/apoptosis in epithelial cells. In contrast, decreased cav-1 expression promotes fibroblast activation and collagen deposition, effects that may be relevant in later stages of reparative response. Hence, therapeutic strategies to modulate the expression of cav-1 should take into account cell-specific effects in the regenerative responses of the lung epithelium to injury.
Keywords: caveolin-1, lung injury, fibrosis, cellular senescence, apoptosis
Idiopathic pulmonary fibrosis (IPF) is an interstitial lung pathological disease and is associated with severe respiratory consequences resulting in increased mortality (1). Besides familial inheritance and progressive aging, additional factors contribute to the development of IPF; however, they remain unknown. The pathobiology of IPF is associated with usual interstitial pneumonia and chronic inflammation (1). Studies conducted toward understanding the mechanism of onset and progression of the disease using experimental animals have suggested a positive correlation between epithelial injury along with apoptosis and development of pulmonary fibrosis (2). In addition, the occurrence of epithelial cell senescence has been reported in IPF, resulting in an aberrant interaction of epithelial cells with mesenchymal cells (3). Furthermore, telomere shortening and DNA damage through activation of p21/p53 is linked to the presence of senescent phenotypes in fibrotic lungs (3–5).
Recently, we have shown that genotoxic stress induced by hydrogen peroxide and bleomycin leads to alveolar epithelial cell senescence in a p16/pRB-dependent manner, thereby establishing the fact that age-associated lung pathologies such as chronic obstructive pulmonary disease and IPF may involve cellular senescence as a major contributor to increased pathological consequences and chronic inflammation (6). A plethora of proinflammatory cytokines, chemokines, proteases, growth factors, and matrix metalloproteinases (MMPs) constitute senescence-associated secretory phenotype (SASP), making senescence a paradoxical phenomenon leading to tumor development (7).
A membrane-associated scaffolding protein of the caveolae, caveolin-1 (cav-1), has been widely studied regarding senescence under in vitro and in vivo conditions (8–10). It is ubiquitously expressed in differentiated cells such as lung epithelial type I cells, endothelial cells, and fibroblasts (11–13). In the context of tumor development, increased expression of cav-1 blocks mitogenic signaling during differentiation, cav-1 expression is decreased during transformation, and its expression is increased in aggressive tumor development (14). Together, these observations suggest that cav-1 serves as an antagonist to this exaggerated inflammatory mediator production and tumor progression via activation of tumor suppressor protein p53.
In contrast, experimental studies have shown a loss of cav-1 in a radiation-induced model of pulmonary fibrosis in rats and in bleomycin-induced fibrosis in mice (15, 16). Expression of cav-1 is reduced in patients with IPF; more importantly, down-regulation is observed in fibroblasts and epithelial cells (13). The significance of down-regulation of cav-1 in these other cell types to fibrogenesis and to the repair responses to lung injury is unclear. Recent studies have shown that a peptide corresponding to the scaffolding domain of cav-1 (CSD) inhibits collagen production in normal lung fibroblasts. In addition, systemic administration of the CSD peptide into bleomycin-treated mice can reverse the fibrotic process by blocking epithelial cell apoptosis, inflammatory cell infiltration, and matrix deposition (17). Furthermore, cav-1 is an essential regulator of fibroblast proliferation and extracellular matrix deposition in IPF and systemic sclerosis (17, 18).
The lungs of cav-1−/− mice are known to have abnormalities such as thickened alveolar septa from uncontrolled endothelial cell proliferation and reduced airway function at an early age from increased collagen deposition (19, 20). cav-1 deficiency in fibroblasts has been shown to dramatically inhibit premature senescence induced by oxidants contained in cigarette smoke (21). It has also been shown that cav-1 is required for oxidative stress-induced premature senescence in fibroblasts (22).
Based on the current paradigms of a role for cav-1 in inhibiting profibrogenic signaling, the response of the cav-1−/− mice would be expected to augment bleomycin-induced fibrosis. However, due to the fact that cav-1 enhances senescent phenotypes in lung epithelium, we hypothesized that the absence of cav-1 may promote resistance to senescence in lung epithelial cells upon bleomycin challenge and the development of increased fibrotic lesions (10, 21, 22). Thus, we investigated the role of cav-1 in bleomycin-induced lung fibrosis by comparing susceptibility to lung fibrosis in cav-1−/− versus WT mice. Our results indicated that cav-1 deficiency protected the animals from fibrotic injury caused by bleomycin and was positively correlated with a decreased level of senescence. Cav-1 deficiency also resulted in decreased production of contributors of SASP, MMP-2 and MMP-9; conversely, release of proinflammatory cytokines IL-6 and macrophage inflammatory protein (MIP)-2 was not compromised in cav-1−/− mice and was comparable to the levels of their WT counterparts (7, 23).
Materials And Methods
Animals and Induction of Pulmonary Fibrosis
Animal experiments were conducted using a protocol approved by the Institutional Animal Care and Use Committee of the University of Hawaii. WT C57BL/6J (WT) and Cavtm1Mls/J (cav-1−/−) mice were purchased from The Jackson Laboratory (Bar Harbor, ME). The genetic background for the cav-1−/− mice is a mixture of C57BL/6J and 129S6/SvEv. The resulting chimeric animals were then back-crossed into C57BL/6J background no more than six times due to a lower fertility. To induce pulmonary fibrosis, WT and cav-1−/− mice were administered with 1.3 U/kg body weight of bleomycin intratracheally. Control animals from both cohorts received PBS intratracheally. Animals were killed at the indicated time points. Lungs and blood samples were collected. Dynamic compliance of the lungs was analyzed as previously described by Jourdan-Le Saux and colleagues (19) as a measure of lung mechanics using a Flexivent instrument (Scireq Inc., Montreal, Quebec, Canada) (21, 22).
Assessment of Pulmonary Fibrosis Development and Lung Injury
The extent of fibrosis indices in the lungs of bleomycin-challenged mice were assessed with established markers such as collagen and α-smooth muscle actin (α-SMA) expression using immunohistochemical staining. The level of expression of the α-2 chain of type I procollagen (procol1A2) and the α1 chain of type III procollagen (procol3A1) was detected by quantitative RT-PCR using commercially available TaqMan probes (Applied Biosystems, Foster City, CA). Total collagen was assessed by the Sircol Collagen Assay kit (Biodye Science, Westbury, NY) according to the manufacturer's instructions.
For the assessment of lung injury and repair, levels of CD44v10 were analyzed by immunohistochemistry (IHC), and expression of tenascin-C (tnc) was analyzed at mRNA level. Using the specific probes available commercially, expression levels of cav-1 and actin were also analyzed at mRNA levels to understand their relationship with the development of fibrosis.
Analyses of Senescence and Apoptosis
Lung cell senescence was measured by senescence-associated β-gal activity as described by Debacq-Chainiaux and colleagues (24). The levels of senescence markers p16 and pRB and the heterochromatin-associated protein, macrohistone 2A (MH2A) were analyzed by immunohistochemical and immunofluorescent staining (6, 25). Quantification of apoptotic index in whole-lung paraffin-embedded sections was performed using terminal deoxynucleotide transferase-mediated dUTP nick end labeling (TUNEL) in the in situ cell death detection kit (Roche Diagnostics, Indianapolis, IN) according to the manufacturer's protocol. Quantification of apoptosis in epithelial cells was performed by flow cytometric analysis using an Annexin V/PI labeling kit (BD Pharmingen, San Diego, CA) and E-cadherin labeling (Cell Signaling, Danvers, MA) (26). Analysis was done on the isolated total lung cell population from WT and cav1−/− mice at 2, 4, and 6 days after bleomycin challenge. Results are represented as percent total and E-cadherin–gated cells population showing annexin-V positivity and PI negativity to derive the actual number of cells undergoing apoptosis as opposed to the total apoptotic cells quantified by TUNEL assay.
Statistical Analysis
Student's t test and two-way ANOVA were used to determine statistical significance between groups. Data entry, management, and statistical analysis were performed using Prism software (GraphPad Software, San Diego, CA).
RESULTS
Effects of Bleomycin Injury on Body Weight and Lung Compliance in cav-1−/− Mice
Bleomycin-treated WT mice were more symptomatic of the fibrotic disease process than the cav-1−/− mice. There was 50% attrition rate in the WT population over the 21-day observation period, with the majority of deaths occurring within Days 10 to 15, compared with a 100% survival rate in the cav-1−/− mice (see Figure E1A in the online supplement). WT mice showed a steady approximately 20% decrease in body weight by Day 12 (Figure E1B) before slowly recovering toward their original weight by Day 20. The bleomycin-treated cav-1−/− mice showed an initial rapid 10% decrease in body weight by Day 2, preceding a steady increase in weight to 96% of original at Day 11 and 100% of original at Day 21. There was no significant difference in lung compliance between cav-1−/− and WT mice treated with PBS (Figure E1C). However, in bleomycin-injured groups, WT mice at Day 11 demonstrated a 46% loss in compliance compared with only a 26% loss for cav-1−/− animals. Together, these data indicate that cav-1 deficiency protects the mice against bleomycin-induced loss of body weight and lung compliance.
Development of Bleomycin-Induced Pulmonary Fibrosis Is Attenuated in cav-1−/− Mice
Blind histological scoring of paraffin sections stained for collagen type I indicated that WT mice had a progressive and statistically significant increase in collagen deposition from Day 6 through Day 21 (Table 1). In contrast, in cav-1−/− mice, we measured no significant increase in collagen deposition at any of the time points evaluated. For α-SMA levels, WT mice showed a progressive, statistically significant increase in the presence of α-SMA from Day 6 to that measured at Days 11 21. Conversely, cav-1−/− mice showed a lack of response at Day 6 followed by a significant increase in α-SMA at Day 11 that returned to baseline levels by Day 21 (Table 1). Analysis of picrosirius red–stained tissue sections (Figure 1A) confirmed the data obtained through blind scoring of collagen type 1 deposition. In WT mice, the level of picrosirius red staining in lung parenchyma increased through Day 21. This increase was significant at all three measured time points compared with PBS-treated mice. However, in cav-1−/− mice, there was no significant increase in collagen deposition. Reminiscent of the scored samples, we observed an initial significant increase from PBS mice at Day 11, which was lost by Day 21. As previously described (19), we measured significantly more collagen in the lungs of PBS-treated cav-1−/− mice than in the lungs of their WT counterparts, indicating a higher background level of collagen in the cav-1−/− mice. Thereafter, WT mice showed significantly more collagen than cav-1−/− mice, whose collagen deposition had returned to baseline levels by Day 21 (Figure 1A). In addition, quantification of collagen was performed by SIRCOL assay and confirmed the reduced levels of collagen in bleomycin-challenged cav-1−/− mice (0.18 ± 0.05 in the WT versus 0.04 ± 0.002 in cav-1−/− mice on Day 21 after bleomycin treatment) (Figure 1B). Furthermore, to calculate whether the increase in collagen deposition correlated with a change in gene expression, we analyzed levels of procol1a2 and procol3a1 mRNA extracted from whole lung tissue (Figures 1C and 1D). Although not statistically significant, at Days 6 and 11 we detected a general increasing trend in procol1a2 mRNA expression in WT mice. This progressed into a statistically significant doubling (1.9 ± 1.4) of procol1a2 at Day 21 compared with PBS-treated mice. There was also an insignificant increase in procol3a1 expression at Day 21 in cav-1−/− mice, where we measured a 1.5 ± 0.5-fold increase above averaged controls, compared with a 2.0 ± 0.8-fold increase in WT mice. These data suggested a correlation between the increased collagen depositions measured in bleomycin-treated WT mice and increased gene expression for procol1a2 and procol3a1.
TABLE 1.
IMMUNOHISTOCHEMISTRY WAS USED TO ASSESS EXPRESSION OF COLLAGEN TYPE I AND α SMOOTH MUSCLE ACTIN
| Traits | Day 6 | Day 11 | Day 21 |
| WT-COL1 | 1.6 ± 0.3 | 7.7 ± 0.4** | 11.4 ± 1*** |
| cav-1−/−–COL1 | 1 ± 0.4 | 3.3 ± 0.4+ | 1.6 ± 0.3+++ |
| WT–α-SMA | 0 | 2.5 ± 0.3*** | 2.7 ± 0.3*** |
| cav-1−/−–α-SMA | 0 | 1.6 ± 0.3 | 0.9 ± 0.1+++ |
Definition of abbreviations: α-SMA = α smooth muscle actin (dilution 1800; Dunn, Labortechnik GmbH, Asbach, Germany); cav-1 = caveolin-1; COL1 = collagen type 1 (dilution 120; Quartett, Berlin, Germany); WT = wild type.
The COL1 score was expressed as: immunopositive area in % (10% = 1; 100% = 10) × staining intensity (0 = negative; 1 = weakly positive; 2 = strongly positive). The α-SMA score was expressed as: 0–5% myofibroblast positive = 1, 6–20% myofibroblast positive area = 2, and >20% myofibroblast positive area = 3. Histological scoring of deposition are given as mean ± SD. There was a time-dependent significant increase in COL1 and α-SMA in WT mice. However, in cav-1−/− mice there was no significant increase in COL1 deposition and only a significant increase for α-SMA at Day 11. A minimum of five sections per group were analyzed; significance was assessed by two-tailed t test.
Asterisks denote significance within groups (**P < 0.01; ***P < 0.001); plus symbols denote significance between strains at matched pairs (+P < 0.05; +++P < 0.001).
Figure 1.
Effect of caveolin-1 (cav-1) deficiency on the amount of collagen deposition and gene expression with time. (A) Representative histological sections from the whole left pulmonary lobe were taken from each animal and stained with picrosirius red. To minimize variation, samples were processed under identical conditions. Levels of picrosirius red staining were quantified with ImagePro software. Values given are mean ± SEM, and significance was measured by one-way ANOVA (n = 5 controls and minimum of 10 per treatment). (B) Soluble collagen content measured in the salt extracts of lung tissues using Sircol assay (n = 3 for each group) show a gradual increase and a significant increase on Day 21 in collagen levels in the wild-type (WT) mice as compared with the cav-1–deficient (cav-1−/−) mice after bleomycin challenge. Values given are mean ± SEM, and significance was measured by one-way ANOVA. (C) RT-PCR expression levels for mRNA from procol1a2 and procol3a1 (D). Values are arbitrary units ± SEM (two-tailed Student's t test; n = 5 individual specimens in triplicate). Asterisks denote significance within group (*P < 0.05; **P < 0.01; ***P < 0.001); + denotes significance between the traits at matched pairs (P < 0.05).
Reduced Injury and Repair Response in the Lungs of cav-1−/− Mice after Bleomycin Challenge
We measured expression levels of the matrix protein tnc mRNA as a marker of tissue repair (27–29). Tnc showed a marked increase in WT mice from Day 6 through Day 21 after bleomycin challenge. In addition, at Day 21, levels of tnc found in WT mice were significantly higher than those found in cav-1−/− mice. A significant but smaller increase in tnc was measured in cav-1−/− mice through Day 21 (Figure 2A). These data suggested the possibility that repair mechanisms are not triggered in cav-1−/− mice after bleomycin challenge. To analyze the level of epithelial injury, we assessed levels of CD44v10 by IHC. CD44v10 is a marker of alveolar epithelial type II cells that also stains the bronchial epithelium in mouse lungs. During fibrogenesis, epithelial alterations can be monitored with epithelial isoforms of CD44 (30). In most cases, alveolar epithelial type II cell morphology changes toward an (intermediate) alveolar epithelial type I cell–like cell type. In bleomycin-injured WT mouse lungs, CD44v10 staining was prominent, compared with that of PBS-treated WT controls, which showed no change over time. However, cav-1−/− mice showed mild positivity of CD44v10 upon bleomycin challenge, whereas no apparent difference was observed in the PBS-cav-1−/− controls. More importantly, the positive correlation observed between epithelial injury and repair process in bleomycin-injured WT mouse lungs versus cav-1−/− indicated that cav-1−/− mice might have experienced less injury and therefore less repair process (Figure 2B). Injury and repair profiles are reduced in bleomycin-challenged cav-1−/− mouse lungs.
Figure 2.
cav-1−/− mice exhibit reduced tissue damage and epithelial injury after bleomycin treatment. (A) RT-PCR for analysis of expression levels of tenascin-C tnc mRNA. Values are arbitrary units ± SEM (two-tailed Student's t test; n = 5 individual specimens in triplicate). Asterisks denote significance within strain (*P < 0.05; **P < 0.01; ***P < 0.001); ++ denotes significance between strains at matched pairs (P < 0.01). (B) Paraffin sections of mouse lung tissues from WT and cav-1−/− animals. Representative immunostaining for CD44v10 (undiluted supernatant; kind gift from Dr. U. Günthert, Basel, Switzerland) in untreated (A: WT; B: cav-1−/− mice) and bleomycin-treated (C: 21 d WT; D: 21 d cav-1−/−) mice. Scale bars in A and B = 50 μm; scale bars in C and D = 100 μm. (C) cav-1 expression in WT mouse lungs over time after bleomycin challenge. cav-1 and β-actin expression at mRNA level were determined by qRT-PCR. In WT mice treated with bleomycin, cav-1 expression decreases with time as fibrosis develops. Values are mean ± SEM (n ≥ 4). ***P < 0.001.
It has been reported that cav-1 expression is decreased with the development of pulmonary fibrosis in mice and human IPF (15). To reconcile our findings with these previous published data, we determined that in our animal model, cav-1 expression levels were significantly decreased by over 2-fold in the lungs of WT mice after bleomycin challenge only starting at Day 11 through Day 21 (Figure 2C). Indeed, at Day 6 after injury, cav-1 expression was not affected when compared with PBS control samples. This decrease in cav-1 was a time-dependent process concomitant with the progression of fibrosis.
Absence of cav-1 Protects against Bleomycin-Induced Epithelial Cell Senescence and Apoptosis
Cellular senescence was evaluated using four well accepted senescence markers: the expression of SA-β gal activity, p16, pRB, and MH2A. The injury and repair processes showed distinct responses in the two bleomycin-challenged animal groups. Therefore, we focused our analyses of epithelial cell senescence at a midpoint on Day 11 after bleomycin challenge. Bleomycin-injured WT lung tissues presented an increase of SA-β gal activity after Day 11 (Figure 3A). In contrast, no evidence of the activation of this enzyme was detected in samples from bleomycin-injured cav-1−/− lung tissue. Examination of nuclear localization of pRB by immunofluorescence staining also depicted a similar phenotype (Figure 3B). We then analyzed the profile of epithelial cell senescence on Days 6, 11, and 21 in bleomycin-injured lungs by IHC analysis of the senescence markers p16, pRB, and MH2A. The senescence nuclear localization of p16, pRB, and MH2A was not yet observed on Day 6 after injury on WT and cav1−/− mice but showed a clear cytosolic staining in the alveolar lining and bronchial epithelial cells (Figure E2). On Days 11 and 21, challenged WT lung sections showed prominent nuclear localization of p16, pRB, and MH2A along with alveolar wall thickening and fibroblasts recruitment (Figure 3C and Figure E2). In contrast, in cav1−/− mice, nuclear localization of senescence markers was reduced (Figure 3C and Figure E2).
Figure 3.
Reduced cellular senescence in bleomycin-injured cav-1−/− lung tissues. (A) Representative photomicrographs on frozen sections of WT mice challenged with bleomycin for 11 days show SA-β galactosidase activity (blue staining shows senescence foci versus the clear areas, which are normal). No SA-β gal activity was detected in the cav-1−/− mice challenged with bleomycin. (B) Immunofluorescence staining of pRB-positive nuclei (red) for the presence of senescent cells versus the normal nuclei stained only with DAPI (blue). Original magnification: ×200. A histogram demonstrates the percentage of pRB-positive nuclei per field of view in the lung sections collected from WT and cav-1−/− mice treated with PBS or bleomycin for 11 days. (C) Representative paraffin-embedded lung sections immunohistochemically stained for p16, pRb, and MH2A. Immunohistochemistry analyses showed nuclear localization of senescence markers p16, pRB, and MH2A. Hematoxylin was used as a negative counterstain. Original magnification: ×400. Arrow points to positive stain.
Because senescence is decreased in the lungs of bleomycin-challenged cav-1−/− mice, we assessed the impact of cav-1 during bleomycin-induced apoptosis. To understand if the apoptotic process was prominent during the early response to the bleomycin-induced injury to the lungs, we performed a TUNEL assay on lung tissue sections at 2, 4, and 6 days after bleomycin challenge. Two days after injury, we counted a near 3-fold increase in TUNEL-positive cells in bleomycin-challenged WT mice (15.6 ± 1.9%) compared with PBS-treated WT mice (5.3 ± 1.6%). At Day 4 and Day 6, apoptosis was still increased in bleomycin-challenged WT mice at 12 ± 2.3% and 7.8 ± 2.2%, respectively (Figure 4A). There was also a moderate increase in apoptosis at 2 days (7.0 ± 1.2%) and 4 days (5.7 ± 1.5%) after bleomycin treatment as compared with that of PBS-treated cav-1−/− mice (2.6 ± 1.5%). By Day 6, the level of apoptosis in bleomycin-challenged cav-1−/− mice had returned to baseline levels. At Day 4, approximately 50 to 60% of TUNEL-positive cells also stained positive for the epithelial marker K19 (data not shown) in bleomycin-challenged mice from both WT and cav-1−/− groups.
Figure 4.
Rate of apoptosis is altered in cav-1−/− mice during bleomycin-induced lung injury. (A) Representative photomicrographs of the terminal deoxynucleotide transferase-mediated dUTP nick end labeling (TUNEL) assay performed on lung frozen lung tissue sections from Days 2, 4, and 6 after bleomycin challenge of WT and cav-1−/− mice. Note a decreased rate of TUNEL positivity in cav-1−/− mice compared with bright nuclear TUNEL positivity in the WT mice, which is reduced over time. To calculate the apoptosis index, the data are expressed as the percentage of cells positively expressing fluorescein verses the total cells (DAPI) present for each field of view. Significant increases in apoptosis were observed at Days 2 and 4 in bleomycin-treated WT mice and in cav-1−/− mice. There was significantly less apoptosis in cav-1−/− mice than in their WT counterparts. Values are mean ± SEM (Student's t test; n = 3 different specimens with three fields of view analyzed per section). Asterisks denote significance within group (P < 0.05; **P < 0.01; ***P < 0.001); + denotes significance between the traits at matched pairs (P < 0.05). (B) Flow cytometric sorting of E-cadherin–positive apoptotic cells showed a significant shift in the Annexin-V+/PI− cell population on Day 4 after bleomycin challenge. The percentage of E-cadherin/AnnexinV+/PI− cells is reduced cav1−/− mice.
To quantify the rate of apoptosis in epithelial cells from WT and cav1−/− mouse lungs, we performed a flow cytometric Annexin-V/PI labeling assay by sorting the cells with the epithelial cell marker E-cadherin on Day 2, 4, and 6 after bleomycin-induced injury. A significant decrease in E-cadherin–sorted apoptotic cells was measured in the cav1−/− samples (8.80 ± 0.92% in WT versus 4.42 ± 0.75% in cav-1−/− mice; P = 0.002), with no significant difference between the two strains on Day 2 or 6.
Cav-1−/− Mice Exhibit a Low Profile of SASP
We further examined the secretory phenotype of senescent cells (SASP) in bleomycin-challenged WT and cav-1−/− mice by analyzing the levels of two of the most characteristic proteins of SASP, MMP-2 and MMP-9 (6, 7). The marked increase in MMP-2 levels in the perivascular and alveolar spaces observed in the bleomycin WT lungs sections was attenuated in cav-1−/− counterparts at Day 11 after injury (Figure 5A). MMP-9 levels were more concentrated in the peribronchial areas and alveolar spaces in the WT bleomycin lung sections, and there was a reduced staining in the lung sections of cav-1−/− mice (Figure 5A). MMP-2 and MMP-9 levels were only increased over time in WT lungs after bleomycin injury (Figure 5B). Tissue IL-6 levels were similarly increased in bleomycin-challenged WT mice (P ≤ 0.001) and cav-1−/− mice (P = 0.002). In contrast, no significant difference was observed in MIP-2 levels in the lung homogenates of both strains after challenge with bleomycin (Figure 5C).
Figure 5.
A subset of the senescence-associated secretory phenotype profile shows a decreased level in bleomycin-challenged cav-1−/− mice. (A) Representative photomicrographs of immunohistochemical analyses of matrix metalloproteinase (MMP)-2 and MMP-9 using deparaffinized lung tissue sections of Day 11 after bleomycin injury in WT and cav-1−/− mice. Note the increased perivascular and alveolar staining of the MMPs in the lung sections of belomycin-challenged mice versus the reduced staining in those of cav-1−/− mice. (B) Western blot analysis of MMP-2 and MMP-9 using the lung tissue homogenates from Days 6, 11, and 21 after bleomycin injury show decreased levels in the cav-1−/− mice as compared with the WT mice. (C) Levels of IL-6 and MIP-2 as measured by ELISA in the lung homogenates and serum samples obtained from WT and cav-1−/− mice challenged with bleomycin and their corresponding PBS-treated controls. Values are represented as an average pg/mg tissue or pg/ml serum ± SD (n = 5).
Discussion
The lung responds to injury in a stereotypic manner with activation of inflammatory and reparative host processes. The intratracheal instillation of the chemotherapeutic agent bleomycin has been used as a model of studying the responses of the lung to noninfectious lung injury and promoting lung fibrosis (31). In this study, we demonstrate that cav-1−/− mice are protected from bleomycin-induced lung injury. Our studies suggest that this protection is related to reduced epithelial cell senescence and apoptosis with an associated reduction in the fibrotic response. Pulmonary fibrosis pathobiology is perhaps best described as an aberrant wound-healing response to recurrent or stress-induced alveolar injury. Normal healing requires proliferation and repopulation of denuded alveolar epithelial cells (AECs). The restitution of epithelium integrity is also associated with apoptosis of AECs and fibroblasts (32, 33). Repeated insults, however, lead to aberrant responses where AECs can no longer proliferate and instead excessive apoptosis and replicative senescence is observed. Subsequently, fibroblasts proliferate and become resistant to apoptosis, leading to the development of progressive fibrosis (34, 35). Modeling pulmonary fibrosis, such as IPF, in animals is complicated because the etiology and natural history of the pathology is unclear and because chronic diseases have proven to be difficult to model in animals. None of the animal models currently available mimics the progressive and irreversible nature of this chronic disease (36, 37). Histological and biochemical markers of fibrosis, such as intraalveolar buds and collagen deposition, are present in bleomycin-treated animals, similar to patients with IPF; however, unlike in human IPF, they are reduced over time in animals. The bleomycin model is the standard of experimental pulmonary fibrosis in animals (31, 36). Previous studies have shown that cav-1 participates in the regulation of inflammation and fibrosis of pulmonary fibrosis. Therefore, it would be expected that, in cav-1−/− mice, bleomycin would have an increased fibrogenic effect (13, 17). Our study indicates that bleomycin induced much less fibrosis in cav-1−/− mice relative to that in WT animals, as assessed by lung collagen content, histopathology, and α−SMA expression in the lungs. Mice deficient in cav-1 did not develop fibrosis after bleomycin injury, most likely because the cascade of events initiated after injury was not promoted. Along with the absence of epithelial injury as revealed by the CD44v10 marker, the levels of tnc and procol3a1 gene expression were not increased in cav-1−/− lung samples. Both of these extracellular matrix proteins are distributed in alveolar septal walls and secondary septal tips in the areas of tissue damage as part of the early response to pulmonary injury induced by bleomycin (19, 27). As the development of fibrosis proceeded, tnc mRNA expression decreased, whereas expression of procol3a1 persisted into the reparative phase. The deficiency in epithelial repair associated with fibrotic lung disorders has been shown to be a strong contributor to a persistent deposition of ECM components including tnc (38).Therefore, the expression of tnc has been proposed as a unique early-response ECM component and can provide information on sites of active foci of developing fibrotic lesions (27). Newer mechanisms involved during early stages of pathobiology in bleomycin-induced lung injury as well as in pulmonary fibrosis include apoptosis and senescence.
We measured AEC apoptosis in bleomycin-injured WT mice. In the absence of cav-1, the number of TUNEL-positive and E-cad/Annexin V–positive cells was reduced on Day 4. This suggested that cav-1 might be a regulator of apoptosis. cav-1 has been weakly linked to apoptosis but has been linked strongly with cellular senescence (14, 22). Cellular senescence is not well understood in lung injury and fibrosis; it is a phenomenon observed in cells that can no longer proliferate. It is the ultimate and irreversible loss of replicative capacity occurring in primary somatic cells (39). The two common factors that link senescence with lung injury and fibrosis are telomere shortening and reactive oxygen species–mediated stress (40, 41). Our data strongly suggested that cellular senescence not only contributes to the chronic inflammation that affects the lungs of patients with chronic obstructive pulmonary disease, as previously reported (42), but may also lead to impaired reepithelialization affecting epithelial repair responses associated with lung injury and fibrosis. The known mechanisms of senescence involve the tumor suppressor proteins p53/p21 and p16/pRB/MH2A, which arrest the cell cycle (43, 44). Similar mechanisms are suggested for progressive aging in the lungs, which include telomere shortening associated with the DNA damage response, resulting in the impaired regeneration of epithelial cells; this is also observed in pulmonary fibrosis diseases (5, 45). Indeed, senescence-associated β-gal activity and the expression of p16, pRB, and MH2A were observed in WT but not in cav-1−/− lung tissue after bleomycin treatment. The importance of senescence has also been pointed out by Xu and colleagues, who found that in a senescence-accelerated mouse model, development of fibrosis was more pronounced (46).
MMP-2, MMP-9, and the proinflammatory cytokines IL-6 and MIP-2 levels were also increased in bleomycin-injured WT mice, suggesting a positive correlation between senescence and fibrosis, contributed by the SASP (23, 47). Despite a clear phenotypic difference between the WT and cav-1−/− for MMP-2 and MMP-9 expression, IL-6 and MIP-2 levels did not show a significant difference in the lungs of WT and cav1−/− mice after bleomycin injury. As evident from the photomicrographs, cav-1−/− mice display abundant lymphocyte infiltration 11 days after bleomycin challenge. In addition, generalized inflammatory status and an abnormal lung parenchymal architecture, which is inherent of cav-1−/− mice, without any apparent changes in lung cell function might also explain increased levels of MIP-2 (48). Moreover, the origin of IL-6 and MIP-2 is not exclusively from senescent cells. It is therefore a more delicate set of data to interpret because these data exclusively reflect the senescence status in the injured tissue.
Expression of cav-1 is reduced in IPF and bleomycin-treated mice and most specifically in epithelial cells, and it is associated with abnormal reepithelialization in lung fibrosis (49). In contrast, bleomycin-induced cellular senescence in epithelial lung cancer A549 cells is associated with the up-regulation of cav-1 expression (8, 50). In our system using cav-1−/− mice, we did not observe the increased fibrosis and collagen expression typically observed in a murine bleomycin model, and, as supported in studies by Linge and colleagues (8), nor did we observe excessive apoptosis and cellular senescence. The contradictory data might be due to the fact that the reduced expression of cav-1 occurred only 11 days after bleomycin injury in WT animals and was further reduced through Day 21. Moreover, the difference in senescence markers of cav-1−/− mice when compared with those of the bleomycin-challenged WT mice were in agreement with the low profiles of tnc expression on Day 11 after bleomycin challenge. In the early phase of the response to bleomycin, cav-1 appeared to be expressed, and eventually increased expression may be reported, as suggested by Linge and colleagues; therefore, we suspect cav-1 to contribute to apoptosis and cellular senescence in epithelial cells as proposed in Figure 6. In contrast, when fibroblasts are deficient for cav-1 expression, the fibrotic response is enhanced. In WT mice, the cav-1 status in epithelial cells is slightly increased in the early phase and promotes senescence in later phases, and cav-1 status in fibroblasts is reduced and promotes fibrosis (10).
Figure 6.
Schematic illustration of the role of cav-1 in the development of pulmonary fibrosis. Immediately after injury, there might be an increased expression of cav-1, leading to apoptosis and cellular senescence. Later in the progression of the disease, cav-1 expression decreased, and it is then associated with collagen deposition.
Previous work has suggested that treatment with the CSD peptide or adenoviral construct resulted in reduced inflammatory and profibrotic responses in the bleomycin model (13, 17). Our data tend to urge caution in the use of CSD. Depending on the cause of injury, an absence of cav-1 at an early stage might indeed be protective. cav-1 expression is increased in endothelial cells of fibrotic lung and liver, indicating that the role of cav-1 in the inflammatory responses is still unclear (16). This demonstrates that, in the response to initial injury and cellular response to injury, cav-1 may balance a positive or negative outcome. Timing, cause, and stage of the disease need to be considered to maximize the beneficial effects of targeting cav-1 as a therapeutic strategy.
Supplementary Material
Acknowledgments
The authors thank Drs. Tam and Orihuela for thoughtful and scientific comments, Ms. Calhoun for technical assistance for the pRB staining, and Karla D. Gorena, Research Core Facility Technologist, Flow Cytometry Core Facility Unit, UTHSCSA, for assisting in the Annexin V/PI labeling assay.
Footnotes
This work was supported by the American Lung Association of Hawaii (Breathing Room grant) (C.J.L.S.), the Leahi Foundation (Hawaii Community Foundation) (C.J.L.S.), the Janey Briscoe Center of Excellence in Cardiovascular Research (C.J.L.S.) and National Institutes of Health grant R01 HL067967 (V.J.T.).
This article has an online supplement, which is accessible from this issue's table of contents at www.atsjournals.org
Originally Published in Press as DOI: 10.1165/rcmb.2011-0349OC on February 23, 2012
Author disclosures are available with the text of this article at www.atsjournals.org.
References
- 1.Carbone R, Balleari E, Grosso M, Montanaro F, Bottino G, Ghio R. Predictors of mortality of idiopathic pulmonary fibrosis. Eur Rev Med Pharmacol Sci 2008;12:97–104 [PubMed] [Google Scholar]
- 2.Thannickal VJ, Horowitz JC. Evolving concepts of apoptosis in idiopathic pulmonary fibrosis. Proc Am Thorac Soc 2006;3:350–356 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Schissel SL, Layne MD. Telomerase, myofibroblasts, and pulmonary fibrosis. Am J Respir Cell Mol Biol 2006;34:520–522 [DOI] [PubMed] [Google Scholar]
- 4.Armanios MY, Chen JJ, Cogan JD, Alder JK, Ingersoll RG, Markin C, Lawson WE, Xie M, Vulto I, Phillips JA, III, et al. Telomerase mutations in families with idiopathic pulmonary fibrosis. N Engl J Med 2007;356:1317–1326 [DOI] [PubMed] [Google Scholar]
- 5.Minagawa S, Araya J, Numata T, Nojiri S, Hara H, Yumino Y, Kawaishi M, Odaka M, Morikawa T, Nishimura SL, et al. Accelerated epithelial cell senescence in ipf and the inhibitory role of sirt6 in tgf-beta-induced senescence of human bronchial epithelial cells. Am J Physiol Lung Cell Mol Physiol 2011;300:L391–L401 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Shivshankar P, Boyd AR, Le Saux CJ, Yeh IT, Orihuela CJ. Cellular senescence increases expression of bacterial ligands in the lungs and is positively correlated with increased susceptibility to pneumococcal pneumonia. Aging Cell 2011;10:798–806 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Freund A, Orjalo AV, Desprez PY, Campisi J. Inflammatory networks during cellular senescence: causes and consequences. Trends Mol Med 2011;16:238–246 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Linge A, Weinhold K, Blasche R, Kasper M, Barth K. Downregulation of caveolin-1 affects bleomycin-induced growth arrest and cellular senescence in a549 cells. Int J Biochem Cell Biol 2007;39:1964–1974 [DOI] [PubMed] [Google Scholar]
- 9.Chretien A, Piront N, Delaive E, Demazy C, Ninane N, Toussaint O. Increased abundance of cytoplasmic and nuclear caveolin 1 in human diploid fibroblasts in h(2)o(2)-induced premature senescence and interplay with p38alpha(mapk). FEBS Lett 2008;582:1685–1692 [DOI] [PubMed] [Google Scholar]
- 10.Kasper M, Barth K. Bleomycin and its role in inducing apoptosis and senescence in lung cells: modulating effects of caveolin-1. Curr Cancer Drug Targets 2009;9:341–353 [DOI] [PubMed] [Google Scholar]
- 11.Lee EK, Lee YS, Han IO, Park SH. Expression of caveolin-1 reduces cellular responses to tgf-beta1 through down-regulating the expression of tgf-beta type ii receptor gene in nih3t3 fibroblast cells. Biochem Biophys Res Commun 2007;359:385–390 [DOI] [PubMed] [Google Scholar]
- 12.Razani B, Zhang XL, Bitzer M, von Gersdorff G, Bottinger EP, Lisanti MP. Caveolin-1 regulates transforming growth factor (tgf)-beta/smad signaling through an interaction with the tgf-beta type i receptor. J Biol Chem 2001;276:6727–6738 [DOI] [PubMed] [Google Scholar]
- 13.Wang XM, Zhang Y, Kim HP, Zhou Z, Feghali-Bostwick CA, Liu F, Ifedigbo E, Xu X, Oury TD, Kaminski N, et al. Caveolin-1: a critical regulator of lung fibrosis in idiopathic pulmonary fibrosis. J Exp Med 2006;203:2895–2906 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Bartholomew JN, Volonte D, Galbiati F. Caveolin-1 regulates the antagonistic pleiotropic properties of cellular senescence through a novel mdm2/p53-mediated pathway. Cancer Res 2009;69:2878–2886 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Gabazza EC, Kasper M, Ohta K, Keane M, D'Alessandro-Gabazza C, Fujimoto H, Nishii Y, Nakahara H, Takagi T, Menon AG, et al. Decreased expression of aquaporin-5 in bleomycin-induced lung fibrosis in the mouse. Pathol Int 2004;54:774–780 [DOI] [PubMed] [Google Scholar]
- 16.Kasper M, Reimann T, Hempel U, Wenzel KW, Bierhaus A, Schuh D, Dimmer V, Haroske G, Muller M. Loss of caveolin expression in type i pneumocytes as an indicator of subcellular alterations during lung fibrogenesis. Histochem Cell Biol 1998;109:41–48 [DOI] [PubMed] [Google Scholar]
- 17.Tourkina E, Richard M, Gooz P, Bonner M, Pannu J, Harley R, Bernatchez PN, Sessa WC, Silver RM, Hoffman S. Antifibrotic properties of caveolin-1 scaffolding domain in vitro and in vivo. Am J .Physiol Lung Cell Mol Physiol 2008;294:L843–L861 [DOI] [PubMed] [Google Scholar]
- 18.Del Galdo F, Lisanti MP, Jimenez SA. Caveolin-1, transforming growth factor-beta receptor internalization, and the pathogenesis of systemic sclerosis. Curr Opin Rheumatol 2008;20:713–719 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Le Saux O, Teeters K, Miyasato S, Choi J, Nakamatsu G, Richardson JA, Starcher B, Davis EC, Tam EK, Jourdan-Le Saux C. The role of caveolin-1 in pulmonary matrix remodeling and mechanical properties. Am J Physiol Lung Cell Mol Physiol 2008;295:L1007–L1017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Drab M, Verkade P, Elger M, Kasper M, Lohn M, Lauterbach B, Menne J, Lindschau C, Mende F, Luft FC, et al. Loss of caveolae, vascular dysfunction, and pulmonary defects in caveolin-1 gene-disrupted mice. Science 2001;293:2449–2452 [DOI] [PubMed] [Google Scholar]
- 21.Volonte D, Galbiati F. Caveolin-1, cellular senescence and pulmonary emphysema. Aging (Albany NY) 2009;1:831–835 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Dasari A, Bartholomew JN, Volonte D, Galbiati F. Oxidative stress induces premature senescence by stimulating caveolin-1 gene transcription through p38 mitogen-activated protein kinase/sp1-mediated activation of two gc-rich promoter elements. Cancer Res 2006;66:10805–10814 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Coppe JP, Desprez PY, Krtolica A, Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol 2010;5:99–118 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Debacq-Chainiaux F, Erusalimsky JD, Campisi J, Toussaint O. Protocols to detect senescence-associated beta-galactosidase (sa-betagal) activity, a biomarker of senescent cells in culture and in vivo. Nat Protoc 2009;4:1798–1806 [DOI] [PubMed] [Google Scholar]
- 25.Kreiling JA, Tamamori-Adachi M, Sexton AN, Jeyapalan JC, Munoz-Najar U, Peterson AL, Manivannan J, Rogers ES, Pchelintsev NA, Adams PD, et al. Age-associated increase in heterochromatic marks in murine and primate tissues. Aging Cell 2011;10:292–304 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Wilkes DS, Thompson LK, Cummings OW, Bragg S, Heidler KM. Instillation of allogeneic lung macrophages and dendritic cells cause differential effects on local ifn-gamma production, lymphocytic bronchitis, and vasculitis in recipient murine lungs. J Leukoc Biol 1998;64:578–586 [DOI] [PubMed] [Google Scholar]
- 27.Zhao Y, Young SL, McIntosh JC. Induction of tenascin in rat lungs undergoing bleomycin-induced pulmonary fibrosis. Am J Physiol 1998;274:L1049–L1057 [DOI] [PubMed] [Google Scholar]
- 28.Bosman FT, Stamenkovic I. Functional structure and composition of the extracellular matrix. J Pathol 2003;200:423–428 [DOI] [PubMed] [Google Scholar]
- 29.Chiquet-Ehrismann R, Chiquet M. Tenascins: regulation and putative functions during pathological stress. J Pathol 2003;200:488–499 [DOI] [PubMed] [Google Scholar]
- 30.Kasper M, Gunthert U, Dall P, Kayser K, Schuh D, Haroske G, Muller M. Distinct expression patterns of cd44 isoforms during human lung development and in pulmonary fibrosis. Am J Respir Cell Mol Biol 1995;13:648–656 [DOI] [PubMed] [Google Scholar]
- 31.Borzone G, Moreno R, Urrea R, Meneses M, Oyarzun M, Lisboa C. Bleomycin-induced chronic lung damage does not resemble human idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2001;163:1648–1653 [DOI] [PubMed] [Google Scholar]
- 32.Hagimoto N, Kuwano K, Nomoto Y, Kunitake R, Hara N. Apoptosis and expression of fas/fas ligand mrna in bleomycin-induced pulmonary fibrosis in mice. Am J Respir Cell Mol Biol 1997;16:91–101 [DOI] [PubMed] [Google Scholar]
- 33.Maher TM, Evans IC, Bottoms SE, Mercer PF, Thorley AJ, Nicholson AG, Laurent GJ, Tetley TD, Chambers RC, McAnulty RJ. Diminished prostaglandin e2 contributes to the apoptosis paradox in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2010;182:73–82 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Selman M, King TE, Pardo A. Idiopathic pulmonary fibrosis: prevailing and evolving hypotheses about its pathogenesis and implications for therapy. Ann Intern Med 2001;134:136–151 [DOI] [PubMed] [Google Scholar]
- 35.Maher TM, Wells AU, Laurent GJ. Idiopathic pulmonary fibrosis: multiple causes and multiple mechanisms? Eur Respir J 2007;30:835–839 [DOI] [PubMed] [Google Scholar]
- 36.Moore BB, Hogaboam CM. Murine models of pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 2008;294:L152–L160 [DOI] [PubMed] [Google Scholar]
- 37.Moeller A, Ask K, Warburton D, Gauldie J, Kolb M. The bleomycin animal model: a useful tool to investigate treatment options for idiopathic pulmonary fibrosis? Int J Biochem Cell Biol 2008;40:362–382 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Snyder JC, Zemke AC, Stripp BR. Reparative capacity of airway epithelium impacts deposition and remodeling of extracellular matrix. Am J Respir Cell Mol Biol 2009;40:633–642 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Hwang ES, Yoon G, Kang HT. A comparative analysis of the cell biology of senescence and aging. Cell Mol Life Sci 2009;66:2503–2524 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Alder JK, Chen JJ, Lancaster L, Danoff S, Su SC, Cogan JD, Vulto I, Xie M, Qi X, Tuder RM, et al. Short telomeres are a risk factor for idiopathic pulmonary fibrosis. Proc Natl Acad Sci USA 2008;105:13051–13056 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Walters DM, Cho HY, Kleeberger SR. Oxidative stress and antioxidants in the pathogenesis of pulmonary fibrosis: A potential role for nrf2. Antioxid Redox Signal 2008;10:321–332 [DOI] [PubMed] [Google Scholar]
- 42.Tsuji T, Aoshiba K, Nagai A. Alveolar cell senescence exacerbates pulmonary inflammation in patients with chronic obstructive pulmonary disease. Respiration 2010;80:59–70 [DOI] [PubMed] [Google Scholar]
- 43.Rovillain E, Mansfield L, Caetano C, Alvarez-Fernandez M, Caballero OL, Medema RH, Hummerich H, Jat PS. Activation of nuclear factor-kappa b signalling promotes cellular senescence. Oncogene 2011;30:2356–2366 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Voghel G, Thorin-Trescases N, Mamarbachi AM, Villeneuve L, Mallette FA, Ferbeyre G, Farhat N, Perrault LP, Carrier M, Thorin E. Endogenous oxidative stress prevents telomerase-dependent immortalization of human endothelial cells. Mech Ageing Dev 2010;131:354–363 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Ito K, Barnes PJ. COPD as a disease of accelerated lung aging. Chest 2009;135:173–180 [DOI] [PubMed] [Google Scholar]
- 46.Xu J, Gonzalez ET, Iyer SS, Mac V, Mora AL, Sutliff RL, Reed A, Brigham KL, Kelly P, Rojas M. Use of senescence-accelerated mouse model in bleomycin-induced lung injury suggests that bone marrow-derived cells can alter the outcome of lung injury in aged mice. J Gerontol A Biol Sci Med Sci 2009;64:731–739 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Mulder GD, Vande Berg JS. Cellular senescence and matrix metalloproteinase activity in chronic wounds: relevance to debridement and new technologies. J Am Podiatr Med Assoc 2002;92:34–37 [DOI] [PubMed] [Google Scholar]
- 48.Razani B, Engelman JA, Wang XB, Schubert W, Zhang XL, Marks CB, Macaluso F, Russell RG, Li M, Pestell RG, et al. Caveolin-1 null mice are viable but show evidence of hyperproliferative and vascular abnormalities. J Biol Chem 2001;276:38121–38138 [DOI] [PubMed] [Google Scholar]
- 49.Odajima N, Betsuyaku T, Nasuhara Y, Nishimura M. Loss of caveolin-1 in bronchiolization in lung fibrosis. J Histochem Cytochem 2007;55:899–909 [DOI] [PubMed] [Google Scholar]
- 50.Linge A, Morishima N, Kasper M, Barth K. Bleomycin induces caveolin-1 and -2 expression in epithelial lung cancer a549 cells. Anticancer Res 2007;27:1343–1351 [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.






