Inhibition of CLC-2 chloride channel expression interrupts expansion of fetal lung cysts

Carol J. Blaisdell,1,2 Marcelo M. Morales,3 Ana Carolina Oliveira Andrade,3 Penelope Bamford,1 Michael Wasicko,1 and Paul Welling2

Departments of 1Pediatrics and 2Physiology, University of Maryland, Baltimore, Maryland 21201; and 3Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21949-900 Rio de Janeiro, Brazil

Submitted 4 April 2003 ; accepted in final form 30 October 2003


    ABSTRACT
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
Normal lung morphogenesis is dependent on chloride-driven fluid transport. The molecular identity of essential fetal lung chloride channel(s) has not been elucidated. CLC-2 is a chloride channel, which is expressed on the apical surface of the developing respiratory epithelium. CLC-2-like pH-dependent chloride secretion exists in fetal airway cells. We used a 14-day fetal rat lung submersion culture model to examine the role of CLC-2 in lung development. In this model, the excised fetal lung continues to grow, secrete fluid, and become progressively cystic in morphology (26). We inhibited CLC-2 expression in these explants, using antisense oligonucleotides, and found that lung cyst morphology was disrupted. In addition, transepithelial voltage (Vt) of lung explants transfected with antisense CLC-2 was inhibited with Vt = -1.5 ± 0.2 mV (means + SE) compared with -3.7 ± 0.3 mV (means + SE) for mock-transfected controls and -3.3 ± 0.3 mV (means + SE) for nonsense oligodeoxynucleotide-transfected controls. This suggests that CLC-2 is important for fetal lung fluid production and that it may play a role in normal lung morphogenesis.

lung development; chloride secretion


DURING FETAL LIFE, the developing pulmonary epithelium secretes fluid that fills the potential air spaces (25, 26). Peak fluid secretion occurs in the canalicular and saccular phases of lung development (34). This secretion of fluid is crucial to the developing lung. Overexpansion or growth arrest of the lung can occur if fluid egress to the amniotic sac or lung fluid retention is interrupted (3, 7, 15, 39, 43). The fluid that fills the potential air spaces arises from continuous active secretion of chloride (Cl-) by the pulmonary epithelium starting during embryonic development (40) and continuing in the fetal lung (20, 25, 32) until just before birth (12, 19). However, the Cl- channel responsible for fetal lung Cl- secretion and resultant fluid production is unknown.

The cystic fibrosis transmembrane conductance regulator (CFTR) is an essential Cl- channel. Mutations of CFTR lead to recurrent lung infections and premature death in patients with cystic fibrosis. However, despite defects in CFTR expression and function, patients with cystic fibrosis are born with normal lung morphology (41), suggesting that another Cl- channel is responsible for Cl- and fluid secretion in the developing lung. Proposed alternative Cl- channels in the lung include the Ca2+-activated Cl- channel (11), the outwardly rectifying Cl- channel (16), or members of the voltage-gated CLC family of Cl- channels (CLC-2, -3, and -5) (8, 14, 21, 31).

CLC-2 is a member of the CLC family of voltage-gated Cl- channels that is widely expressed in epithelial cells, including those in the lung (42). Previous work in this laboratory revealed that protein expression of CLC-2 was most abundant in the canalicular and saccular phases of lung development and decreased significantly shortly after birth (31). In addition, immunocytochemical analysis revealed that CLC-2 protein is localized to the apical surface of the fetal airway epithelium (30, 31). The presence of CLC-2 protein in the epithelium of conducting airways during the canalicular and saccular phases of lung development coincides with the period of peak fluid secretion in the fetal lung (34).

The expression of CLC-2 during the fetal period supports the hypothesis that Cl- secretion by CLC-2 channels contributes to the secretion of fluid in the developing lung. In the present study, we tested this hypothesis in a fetal rat lung explant culture (24) by exposure to a CLC-2 antisense oligodeoxynucleotide (ODN). We found that fetal lungs in explant culture developed fluid-filled cysts over a 48-h period, as described by others (23, 26). In contrast, lungs treated with a CLC-2 antisense ODN for 48 h exhibited branching, but the size of the cysts was markedly reduced, suggesting fluid secretion was impaired. Transepithelial potential difference (PD) measurements across the membrane of the cysts were markedly reduced in the antisense-treated lungs, suggesting that inhibition of CLC-2 Cl- secretion was responsible for the reduction in fluid secretion. Analysis of CLC-2 protein and mRNA confirmed that CLC-2 channel formation was blocked by the antisense ODN after 48 h in culture. These results suggest that CLC-2 is an important factor in the regulation of fetal lung fluid secretion.


    MATERIALS AND METHODS
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
Fetal Rat Lung Explant Culture

Timed-pregnant Sprague-Dawley rats (Zivic Miller) were killed on day 14 of gestation (22 days = term) by a 3-min exposure to 100% CO2. The fetuses were aseptically removed from uterine decidua and placed in sterile Hanks' balanced salt solution (HBSS) at 4°C. Fetal lung rudiments were microdissected and washed in HBSS at 4°C for 5–10 min. Three to four lungs from a single litter were submerged in 500 µl of Matrigel (Becton Dickinson) and incubated for 1 h in a humidified atmosphere at 37°C in 5% CO2 and 21% O2 before transfection.

Transfection of ODNs

ODNs targeted for transfection were determined previously by Kajita et al. (18) to inhibit CLC-2 expression and function in porcine choroid plexus. The antisense CLC-2 ODN corresponded to nucleotide sequence 2,412–2,390 (accession number GI56705): 5'-TTC CAA TTC TGA CGT CGT CGA CT-3' and the random control ODN: 5'-AGA CGC TTG CGA CAG CAG ATC G-3'. The ODNs were synthesized by Oligos, Etc. (Wilsonville, OR) and modified by phosphorothiolation to improve stability against nuclease degradation in culture.

Transfection with either the antisense or the random control ODN into the lung explant was accomplished by a cation liposome transfection method. For each experiment the ODN was diluted in Opti-MEM (Life Technologies) and mixed with Lipofectamine 2000, at a final concentration of 45 µM ODN, and incubated at room temperature for 20 min. This oligo/Lipofectamine solution was carefully added to each well, and the cultures were allowed to incubate for 48 h at 37°C in 21% O2-5% CO2. For each litter, approximately six fetal lungs were used as mock-transfected controls, six were transfected with the antisense ODN, and six were transfected with the random control ODN. For mock-transfected control conditions, the transfection solutions were prepared as above, without any ODN added (only Lipofectamine in Opti-Mem was applied to the lung).

Lung Explant Morphology

Individual lung sections were photographed on a Nikon TS 100 inverted microscope with a x10 objective, using an Optronics camera and Twain 32 software. Images were imported into Photoshop 4.0.1. Morphological changes in lung cyst development were documented at 0, 24, and 48 h. These images were scanned into a densitometry program, MacBas, on a PowerMac G4. To measure the diameter of each air space, we identified the transect by cyst density and distance across the lumen measured in millimeters. The same cysts were measured for each condition over the 48-h period in culture.

CLC-2 mRNA Expression by Quantitative Polymerase Chain Reaction

Isolation of total RNA. To determine whether CLC-2 antisense ODNs inhibited CLC-2 mRNA expression, we performed semiquantitative reverse transcription polymerase chain reaction (RT-PCR) of explants at 48 h. Total RNA was extracted from pooled lung explants transfected on the same day with antisense CLC-2, random, or no ODN (mock-transfected controls, as described above) using TRIzol reagent (GIBCO-BRL, Grand Island, NY) according to the manufacturer's instructions. The isolated RNAs were treated with ribonuclease-free deoxyribonuclease DNase I (1 U/µl) for 1 h to eliminate contamination with genomic DNA.

RT-PCR. To prepare first-strand complementary deoxyribonucleic acid (cDNA), total RNA isolated from antisense, random, and mock-transfected lung explants above was first primed with oligodeoxythymidilic [oligo(dT)] primer and then reverse-transcribed with SuperScript (GIBCO-BRL) at 37°C for 60 min. Extraction with phenol-chloroform-isopropyl alcohol and precipitation with ethanol terminated the treatment.

PCR was used to amplify the synthesized cDNA with the following solution: 0.2 µmol primers, 0.2 µmol each deoxynucleotide triphosphate, 50 mmol KCl, 10 mmol Tris·Cl (pH 8.3), 1.5 mmol MgCl2, and 2.5 units AmpliTaq (Perkin Elmer). One pair of oligonucleotides was synthesized for CLC-2 amplification (A = 5'-TATGCCATCGCGTC TG-3' and B = 5'-GAAGTCGAGTCGGAACCG-3') corresponding, respectively, to nucleotides 345–362 and 943–960 of rat CLC-2 cDNA sequence (accession number GI8393137). PCR was performed with 35 cycles of denaturation (94°C, 1 min), annealing (55°C, 1 min), and extension (72°C, 1 min).

Semiquantitative RT-PCR was used to compare the expression of CLC-2 in 500 ng of total RNA isolated from mock-transfected control explants and cells treated with antisense or random ODNs. Oligo(dT) was used in the RT to obtain single-stranded DNA. PCR conditions were the same as described above. For the semiquantitative RT-PCR, GAPDH primers, predicted to amplify a 225-bp product, were added into the same RT-PCR reaction tubes, and their products were used as an internal control. The GAPDH primers were 5'-GTC TTC ACC ACC ATG GAG-3' and 5'-CGA TGC CAA AGT TGT CAT G-3', corresponding to nucleotides 301–318 and 508–526, respectively (accession number XM227654). PCR products were size fractionated with 10 g/l agarose gel electrophoresis. The expected CLC-2 and GAPDH bands from the same sample were densitometrically analyzed and normalized by dividing the CLC-2 values by the corresponding GAPDH values.

The semiquantitative method of RT-PCR was validated in preliminary experiments. First, the optimal PCR conditions that yielded a single band on agarose gel electrophoresis were determined for each gene in the same reaction tube. Second, to determine whether the method was semiquantitative, serial amounts (65.5, 125, 250, 500, 1,000, 2,000, and 4,000 ng) were used for RT-PCR amplification for both genes in the same reaction tube. Third, experiments were performed to determine the optimal number of PCR cycles that yielded PCR products in the linear phase of amplification. Finally, to ensure that the reactions were consistent, we performed PCR reactions at least twice. Only one of these reactions was included for final densitometric analysis, and the selection was arbitrary.

All reactions included a negative control RT(-) (cDNA made in the absence of reverse transcriptase). The identity of the amplification was confirmed by determination of the molecular size on agarose gel electrophoresis (1.6% agarose in buffer containing 40 mmol/l of Tris-acetate plus 1 mmol/l EDTA) and visualized by ethidium bromide staining (0.5 µg/ml) under ultraviolet light. The computer software Sigma Gel version 1.1 was used (Jandel Scientific) for densitometric analysis of the bands. Band density was calculated as densitometric mean values of CLC-2 compared with the mean of GAPDH.

CLC-2 Protein Expression by Western Analysis

To confirm that antisense ODN transfection inhibited not only CLC-2 mRNA but also protein expression, we performed Western analysis. Lung homogenates from explants transfected for 48 h were prepared in 250 mM sucrose-5 mM Tris-1 mM EDTA (STE) buffer with Complete Mini EDTA-free Protease Inhibitor Cocktail (Boehringer Mannheim). For each condition (antisense- vs. mock-transfected controls), approximately six fetal lungs were homogenized in 300 µl of STE buffer with a Polytron homogenizer (Brinkman Instruments). The homogenates were gently mixed for 1 h while maintained at 4°C and centrifuged at 10,000 g for 10 min, and the supernatant was removed for analysis.

Equivalent amounts of total protein (20 µg) from antisense and control homogenates were separated on an 8% SDS-PAGE gel. Proteins were transferred to nitrocellulose and immunoblotted with whole chicken serum CLC-2 COOH terminus antibody at a 1:3,000 dilution (31). Analysis of {beta}-actin, using monoclonal anti-{beta}-actin mouse ascites fluid (Sigma Immunochemicals), on the same immunoblots, served as a measure of equivalent loading of samples. Band density was quantified on MacBas.

Immunolocalization of CLC-2 in Explants

Explants prepared, transfected, and incubated for 48 h as described above were washed three times in phosphate-buffered saline (PBS) and fixed in 4% paraformaldehyde in PBS, blocked in paraffin, and sectioned at 10 µm.

Sections on slides were deparaffinized, hydrated, and mordanted in hot citrate buffer 30 min, cooled to room temperature, and blocked in 5% donkey serum, 0.3% Triton X in PBS for 1 h. Sections were exposed to chicken anti-CLC-2 (31) at 1:100 in block solution for 1 h at room temperature and washed five times in PBS. Second antibody was tetramethylrhodamine isothiocyanate-conjugated rabbit anti-chicken IgG (Sigma) at 1:1,000 in PBS was added for 1 h at RT. After five washes in PBS, Hoechst 33342 counterstain at 1:30,000 was added for 10 min at room temperature, and the sections were washed again five times in PBS and mounted in Fluoromount G (Southern Biotechnical Associates).

Sections were viewed under a Zeiss LSM410 scanning confocal microscope (Facility for Confocal Microscopy, Department of Physiology, University of Maryland Baltimore) for emission at 568 nm (rhodamine) and 364 nm (Hoechst 33342) and saved on a Zeiss LSM-PC Image processing system.

Measurement of Lung Cyst Transepithelial PD

Explants to be used for microelectrode studies were prepared as above, with the added step of mounting the fetal lung in Matrigel on a glass coverslip before transfection. The lungs were examined after incubation for 48 h in the transfection media. The lung on the coverslip was mounted in an open chamber and perfused with a solution containing (in mM): 145 NaCl, 1 CaCl2, 1 MgCl2, 5 glucose, 60 sucrose, and 5 HEPES. The microelectrode studies were performed at 21°C. Glass microelectrodes (resistance {approx}10–50 M{Omega}) were filled with 3 M KCl, placed in a microelectrode holder containing a Ag-AgCl pellet, and advanced under direct observation using a dissection microscope (Olympus, Melville, NY) into a fetal lung cyst with a three-dimensional hydraulic micromanipulator (Newport). Transepithelial voltage (Vt) was measured through a high-input impedance electrometer (725B; Warner, Hamden, CT) with respect to a bath electrode ground (a stable flowing 3 M KCl junction connected to a Ag-AgCl half cell) and recorded on a strip chart recorder (model TA 240; Gould, Valleyview, OH). Impalements were considered acceptable if: 1) there was an abrupt negative deflection of the electrical PD when entering the cyst, 2) the PD reached a stable plateau that was maintained for at least 1 min, and 3) there was an abrupt return to baseline on withdrawal from the cyst. On each experimental day, at least three recordings were made for each condition, and all three conditions were tested.

Statistical Analysis

These studies were approved by the Institutional Animal Care and Use Committee at the University of Maryland, Baltimore. The data are given as means ± SE of n observations. Comparisons between mean values of treated and control explants were determined by one-way analysis of variance and the significance of the difference between means by t-tests. Newman-Keuls adjustment was used in multiple comparison of group means of outcomes for RNA data, and Tukey-Kramer adjustment was used in multiple comparison of group means for PD data. Statistical significance was determined for P < 0.05.


    RESULTS
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
Lung Cyst Arrest

Transfection with CLC-2 antisense ODN resulted in marked changes in lung morphology over 48 h (Fig. 1). Under control conditions the fetal lungs developed fluid-filled cysts by 24 h that further enlarged by 48 h similar to previous investigators (36, 40). In the presence of antisense CLC-2 ODNs, the fetal lungs continued to differentiate, but cyst lumen size was markedly reduced. In contrast, in the presence of the randomized control ODN, cyst size was comparable to the mock-transfected control.



View larger version (87K):
[in this window]
[in a new window]
 
Fig. 1. Phase-contrast photomicrographs of 14-day fetal rat lungs grown in submersion explant culture over 48 h in the absence (control) or presence of antisense or nonsense oligodeoxynucleotides (ODNs). Note the enlarging intraluminal space of cysts of control and nonsense ODN-treated cysts at 24 and 48 h (large arrow within lumen). By 48 h, antisense-treated cysts have collapse of the cyst lumen (small arrow at collapsed lumen). Bar = 0.5 mm.

 

Quantitation of lung cyst size was accomplished by measurement of cyst lumen diameter in treated and untreated conditions at 0, 24, and 48 h. At 24 h, lung cysts increased 2.9, 1.8, and 2.3 times baseline in mock control, antisense-, and nonsense-transfected cysts (P < 0.01 antisense vs. nonsense and control). By 48 h, cyst diameter increased from baseline 4.9-fold in mock-transfected control cysts and 3.2-fold in random ODN-transfected controls compared with only 2.3 times in antisense CLC-2-treated cysts (P < 0.01 antisense vs. nonsense and control, Fig. 2, n = 4–6 cysts each condition from six separate experiments). Thus antisense treatment of CLC-2 interrupts lung cyst expansion, which presumably reflects a decrease in fluid production.



View larger version (20K):
[in this window]
[in a new window]
 
Fig. 2. Effect of antisense CLC-2 transfection on lung explant cyst diameter at 0, 24, and 48 h, expressed in arbitrary units. Control (n = 25) and nonsense ODN (n = 20) explant cysts increased 2.9- to 2.3-fold over 24 h and 4.9- to 3.2-fold at 48 h, whereas antisense-treated explants (n = 23) increased 1.8- to 2.0-fold over the same period (P = 0.01 antisense vs. control and P = 0.01 antisense vs. nonsense at 48 h by t-test).

 

Inhibition of CLC-2 mRNA Expression

To confirm that antisense ODNs inhibited CLC-2 mRNA expression, we used semiquantitative PCR. Amplification of CLC-2 cDNA from antisense and control explants yielded the expected 615-base pair product. Amplification of the internal control, GAPDH, yielded an expected 211-base pair product. Mean densitometric measurements for CLC-2 compared with GAPDH demonstrated that transfection with antisense CLC-2 ODN had the expected result and interrupted CLC-2 mRNA transcript expression (Fig. 3) compared with mock- and nonsense-transfected controls (P < 0.01 antisense vs. mock; P < 0.01 antisense vs. nonsense; P > 0.05 mock vs. nonsense, n = 4 separate experiments).



View larger version (41K):
[in this window]
[in a new window]
 
Fig. 3. Expression of CLC-2 mRNA in fetal lung explants in the absence and presence of antisense or random ODN at 48 h in submersion culture. Densitometric values of PCR band CLC-2 mRNA are compared with GAPDH within the same sample. CLC-2 mRNA is inhibited by antisense treatment compared with mock- and random ODN-treated controls (P < 0.01 antisense vs. mock and nonsense transfected controls, n = 4 separate experiments). Top: representative ethidium bromide stained PCR products for CLC-2 and GAPDH.

 

Antisense CLC-2 ODNs Reduce Protein Expression

To investigate whether arrest of explant lung cyst development was also associated with interruption of CLC-2 protein expression, we performed Western analysis of lung homogenates collected 48 h after transfection. Total protein homogenates from explants cultured for 48 h in the presence or absence of CLC-2 antisense ODN were analyzed for both immunoreaction to a polyclonal CLC-2 antibody and {beta}-actin to normalize for variation in loading. We detected a reduction in a 94-kDa band using the CLC-2 antibody in antisense-treated explants compared with the 25-kDa band detected with the {beta}-actin monoclonal antibody (Fig. 4). Quantification of CLC-2/{beta}-actin band intensity for antisense- and control-treated explants demonstrated that this reduction is significant (P = 0.03 by t-test, n = 5 experiments).



View larger version (11K):
[in this window]
[in a new window]
 
Fig. 4. CLC-2 protein expression in antisense (A) vs. control (C) lung explants at 48 h by immunoblotting. Antisense-treated explants have >50% reduction in CLC-2/{beta}-actin expression compared with mock-transfected controls (*P < 0.03). Inset: reduction of a 94-kDa band detected using a polyclonal anti-CLC-2 antibody (top bands) and a 25-kDa band using a monoclonal {beta}-actin antibody (bottom bands) in a representative Western blot.

 

Antisense Inhibition of Apical CLC-2 Staining

CLC-2 expression was also examined in fetal lung explants by immunocytochemistry. Lung cysts that were treated with antisense CLC-2 oligos were smaller than controls. Immunostaining with CLC-2 polyclonal antibody demonstrated predominant localization in the apical compartment of the cyst epithelium (Fig. 5A). This immunolocalization was reduced in antisense-treated cysts (Fig. 5B). Localization of CLC-2 protein along the apical surface of the cysts supports the idea that this Cl- channel could play a role in the secretion of Cl- and fluid into the developing lung lumen.



View larger version (73K):
[in this window]
[in a new window]
 
Fig. 5. Immunolocalization of CLC-2 in explant cysts. Control cysts (A) demonstrate that CLC-2 is predominantly expressed in the apical compartment of the epithelium lining the cyst lumen (red immunofluorescence). Antisense ODN-treated lung cysts (B) have a smaller cyst lumen, and apical CLC-2 expression is reduced. Nuclei are counterstained with Hoechst. Bar = 10 microns.

 

Inhibition of Vt

We also measured the impact of CLC-2 knockout on the transepithelial PD across the fluid-filled cysts to determine whether the decrease in lung cyst development was the result of inhibition of CLC-2 Cl- transport. A total of 38 recordings were made in mock-transfected control cysts, 32 in antisense ODN-treated cysts, and 30 in random ODN-treated control cysts. Figure 6 shows the mean results of these measurements. In mock-transfected explants, the PD recorded was -3.74 ± 0.3 mV (mean + SE). In lungs treated with the CLC-2 antisense ODN, the transepithelial PD was reduced to -1.49 ± 0.2 mV (mean + SE), which was significantly less than the mock-transfected control (Vt = -1.49 vs. -3.74 P < 0.0001). Finally, in lungs treated with the random ODN, the PD was -3.36 ± 0.3 mV (mean + SE), which was significantly greater than lungs treated with the antisense ODN (Vt = -1.49 vs. -3.36, P < 0.0001) but not significantly different from the untreated control (P > 0.05). Thus blocking the expression of CLC-2 resulted in a reduction of ion movement across the cyst membrane. Because treating the lungs with the random control ODN had no effect on transepithelial PD, the negative charge is likely carried by Cl- through CLC-2 Cl- channels. We conclude that Cl- movement through CLC-2 channels is an important contributor to fluid secretion in the developing lung.



View larger version (8K):
[in this window]
[in a new window]
 
Fig. 6. Transepithelial potential difference (PD) measurements of antisense and control lung explants 48 h after transfection in submersion culture. PD is -3.5 mV in mock-transfected lung explants and -3.3 mV in nonsense ODN-treated explants compared with a depolarization of PD to -1.5 mV in antisense-transfected lung explant cysts (*P < 0.0001 antisense vs. control and nonsense). Vt, transepithelial voltage.

 


    DISCUSSION
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
The fetal lung secretes fluid, which is essential for normal lung development. Interruption of fetal lung fluid production or retention leads to lung dysgenesis (1). Fetal lung fluid has high concentrations of Cl- (2), and inhibition of lung fluid production with Cl- channel blockers interrupts lung morphogenesis (25, 26, 34, 35). Cl- secretion is the predominant ion transport mechanism in early and midgestation lung development both in lung explants (25, 26, 40) and monolayers of airway epithelial cells (37, 44). The Cl- channel responsible for lung fluid production has not previously been reported.

The airway epithelium is a complex system with multiple ion transport mechanisms necessary for lung development. Cl- transport across the basolateral surface of the airway epithelial cell is linked to Na+ and K+ via the Na+-K+-2Cl- cotransporter (NKCC1). The Na+-K+-ATPase on the basolateral surface of the airway epithelium exchanges Na+ for K+. The Cl- gradient generated by this energy-dependent process leads to passive secretion of Cl- across the apical surface of the cell. The best-studied Cl- channel, the CFTR, is expressed in the fetal lung (28) and is developmentally regulated (29). In the postnatal lung, CFTR is essential for host defense and, when defective, is responsible for chronic respiratory infections, lung destruction, and shortened lifespan. Although cAMP-mediated CFTR Cl- secretion is dysfunctional in utero (27, 28), its role in the developing lung is not crucial, because individuals with cystic fibrosis have normal lung morphogenesis (41). In addition, CFTR knockout mice have Cl- and fluid secretory pathways that can compensate for defective CFTR (10, 11). These non-CFTR Cl- secretory pathways (46, 22) may contribute to fetal lung development; however, the molecular identity of such fetal Cl- channels has not been elucidated.

In earlier studies, we have shown that acidic pH-activated Cl- channels in the fetal lung exist (8) that can be inhibited by the CLC-2 channel blocker cadmium chloride (38). In addition, CLC-2 is gestationally regulated and is predominantly expressed in fetal lung compared with the postnatal lung (30, 31). The expression of CLC-2 on the luminal surface of airway epithelia is consistent with a role in Cl- and fluid secretion in the developing lung. Keratinocyte growth factor induces CFTR-independent fluid accumulation in fetal lung explants (45) and is essential to CLC-2-like pH-sensitive Cl- secretion in fetal airway epithelia (8). We demonstrate here that CLC-2 Cl- secretion contributes to lung cyst expansion in a fetal lung explant model. The 14-day gestation fetal rat lung explant has a distal epithelial cell layer that has characteristics of type II cells (26). Using antisense ODN strategies similar to other investigators (18), we successfully inhibited CLC-2 mRNA and protein expression in the developing lung. Apical expression of CLC-2 in the epithelial cells of the explant cysts supports the potential role of CLC-2 in the developing lung. This CLC-2 knockout model led to a reduction in transepithelial PD and arrest of lung cyst development, suggesting that CLC-2 contributes to fetal lung fluid production.

On the basis of the findings of this study, we believe that CLC-2 may be an important fetal lung Cl- channel. Knockout of the CLC-2 channel in this explant model leads to inhibition of lung cyst development, which is linked to decreased Vt. CLC-2 knockout, however, may be insufficient to lead to complete lung dysgenesis, since the CLC-2 knockout mouse has no obvious respiratory distress at birth (9). Similarly, although the importance of NKCC1 for Cl- entry through the basolateral membrane of the fetal lung has been demonstrated, NKCC1 knockout mice are born with apparently normal lung function and have no detectable respiratory problems well into adulthood (17). There is likely redundancy to fetal ion and fluid transport as demonstrated in several knockout models, such that additional channels may compensate when CLC-2, CFTR, or NKCC1 is defective. Future studies using double knockout models may be able to address the contributions of these airway epithelial Cl- channels to lung development. In addition, knockout mice may inadequately model the consequences of ion transport abnormalities in the human (10, 13, 17, 33) and should not inhibit further pursuit of the pathogenesis of ion and fluid transport in human disease.


    ACKNOWLEDGMENTS
 
GRANTS

This work was funded by National Heart, Lung, and Blood Institute Grant K08-HL-03469 (C. J. Blaisdell).


    FOOTNOTES
 

Address for reprint requests and other correspondence: C. Blaisdell, Dept. of Pediatrics, Univ. of Maryland at Baltimore, 655 W. Baltimore St., BRB 10-021, Baltimore, MD 21201 (E-mail: cblaisdell{at}peds.umaryland.edu).

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


    REFERENCES
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 

  1. Adams FH, Fujiwara T, and Rowshan G. The nature and origin of the fluid in fetal lamb lung. J Pediatr 63: 881-888, 1963.[ISI]
  2. Adamson TM, Boyd RDH, Platt HS, and Strang LB. Composition of alveolar liquid in the foetal lamb. J Physiol 204: 159-168, 1969.[ISI][Medline]
  3. Alcorn D, Adamson TM, Lambert TF, Maloney JE, Ritchie BC, and Robinson PM. Morphological effects of chronic tracheal ligation and drainage in the fetal lamb lung. J Anat 123: 649-660, 1977.[ISI][Medline]
  4. Barker PM, Boucher RC, and Yankaskas JR. Bioelectric properties of cultured monolayers from epithelium of distal human fetal lung. Am J Physiol Lung Cell Mol Physiol 268: L270-L277, 1995.[Abstract/Free Full Text]
  5. Barker PM, Brigman KK, Paradiso AM, Boucher RC, and Gatzy JT. Cl- secretion by trachea of CFTR (+/-) and (-/-) fetal mouse. Am J Respir Cell Mol Biol 13: 307-313, 1995.[Abstract]
  6. Barker PM and Gatzy JT. Effects of adenosine, ATP, and UTP on chloride secretion by epithelia explanted from fetal rat lung. Pediatr Res 43: 652-659, 1998.[Abstract]
  7. Blachford KG and Thurlbeck WM. Lung growth and maturation in experimental oligohydramnios in the rat. Pediatr Pulmonol 3: 328-333, 1987.[ISI][Medline]
  8. Blaisdell CJ, Edmonds RD, Wang XT, Guggino S, and Zeitlin PL. pH-regulated chloride secretion in fetal lung epithelia. Am J Physiol Lung Cell Mol Physiol 278: L1248-L1255, 2000.[Abstract/Free Full Text]
  9. Bosl MR, Stein V, Hubner C, Zdebik AA, Jordt SE, Mukhopadhyay AK, Davidoff MS, Holstein AF, and Jentsch TJ. Male germ cells and photoreceptors, both dependent on close cell-cell interactions, degenerate upon ClC-2 Cl- channel disruption. EMBO J 20: 1289-1299, 2001.[Abstract/Free Full Text]
  10. Clarke LL, Burns KA, Bayle JY, Boucher RC, and Van Scott MR. Sodium- and chloride-conductive pathways in cultured mouse tracheal epithelium. Am J Physiol Lung Cell Mol Physiol 263: L519-L525, 1992.[Abstract/Free Full Text]
  11. Clarke LL, Grubb BR, Yankaskas JR, Cotton CU, McKenzie A, and Boucher RC. Relationship of a non-cystic fibrosis transmembrane conductance regulator-mediated chloride conductance to organ-level disease in Cftr(-/-) mice. Proc Natl Acad Sci USA 91: 479-483, 1994.[Abstract]
  12. Dickson KA, Maloney JE, and Berger PJ. Decline in lung liquid volume before labor in fetal lambs. J Appl Physiol 61: 2266-2272, 1986.[Abstract/Free Full Text]
  13. Dorin JR, Dickinson P, Alton EW, Smith SN, Geddes DM, Stevenson BJ, Kimber WL, Fleming S, Clarke AR, Hooper and ML. Cystic fibrosis in the mouse by targeted insertional mutagenesis. Nature 359: 211-215, 1992.[CrossRef][ISI][Medline]
  14. Edmonds RD, Silva IV, Guggino WB, Butler RB, Zeitlin PL, and Blaisdell CJ. ClC-5: ontogeny of an alternative chloride channel in respiratory epithelia. Am J Physiol Lung Cell Mol Physiol 282: L501-L507, 2002.[Abstract/Free Full Text]
  15. Fewell JE, Hislop AA, Kitterman JA, and Johnson P. Effect of tracheostomy on lung development in fetal lambs. J Appl Physiol 55: 1103-1108, 1983.[Abstract/Free Full Text]
  16. Gabriel SE, Clarke LL, Boucher RC, and Stutts MJ. CFTR and outward rectifying chloride channels are distinct proteins with a regulatory relationship. Nature 363: 263-268, 1993.[CrossRef][ISI][Medline]
  17. Gillie DJ, Pace AJ, Coakley RJ, Koller BH, and Barker PM. Liquid and ion transport by fetal airway and lung epithelia of mice deficient in sodium-potassium-2-chloride transporter. Am J Respir Cell Mol Biol 25: 14-20, 2001.[Abstract/Free Full Text]
  18. Kajita H, Omori K, and Matsuda H. The chloride channel ClC-2 contributes to the inwardly rectifying Cl- conductance in cultured porcine choroid plexus epithelial cells. J Physiol 523: 313-324, 2000.[Abstract/Free Full Text]
  19. Kitterman JA, Ballard PL, Clements JA, Mescher EJ, and Tooley WH. Tracheal fluid in fetal lambs: spontaneous decrease prior to birth. J Appl Physiol 47: 985-989, 1979.[Abstract/Free Full Text]
  20. Krochmal EM, Ballard ST, Yankaskas JR, Boucher RC, and Gatzy JT. Volume and ion transport by fetal rat alveolar and tracheal epithelia in submersion culture. Am J Physiol Renal Fluid Electrolyte Physiol 256: F397-F407, 1989.[Abstract/Free Full Text]
  21. Lamb FS, Graeff RW, Clayton GH, Smith RL, Schutte BC, and McCray PB Jr. Ontogeny of CLCN3 chloride channel gene expression in human pulmonary epithelium. Am J Respir Cell Mol Biol 24: 376-381, 2001.[Abstract/Free Full Text]
  22. MacLeod RJ, Hamilton JR, Kopelman H, and Sweezey NB. Developmental differences of cystic fibrosis transmembrane conductance regulator functional expression in isolated rat fetal distal airway epithelial cells. Pediatr Res 35: 45-49, 1994.[Abstract]
  23. Massoud EA, Sekhon HS, Rotschild A, Puterman ML, Matsui R, and Thurlbeck WM. In vitro branching morphogenesis of the fetal rat lung. Pediatr Pulmonol 15: 89-97, 1993.[ISI][Medline]
  24. McAteer JA, Cavanagh TJ, and Evan AP. Submersion culture of the intact fetal lung. In Vitro 19: 210-218, 1983.[ISI][Medline]
  25. McCray PB Jr, Bettencourt JD, and Bastacky J. Developing bronchopulmonary epithelium of the human fetus secretes fluid. Am J Physiol Lung Cell Mol Physiol 262: L270-L279, 1992.[Abstract/Free Full Text]
  26. McCray PB Jr, Bettencourt JD, and Bastacky J. Secretion of lung fluid by the developing fetal rat alveolar epithelium in organ culture. Am J Respir Cell Mol Biol 6: 609-616, 1992.[ISI][Medline]
  27. McCray PB Jr, Bettencourt JD, Bastacky J, Denning GM, and Welsh MJ. Expression of CFTR and a cAMP-stimulated chloride secretory current in cultured human fetal alveolar epithelial cells. Am J Respir Cell Mol Biol 9: 578-585, 1993.[ISI][Medline]
  28. McCray PB Jr, Reenstra WW, Louie E, Johnson J, Bettencourt JD, and Bastacky J. Expression of CFTR and presence of cAMP-mediated fluid secretion in human fetal lung. Am J Physiol Lung Cell Mol Physiol 262: L472-L481, 1992.[Abstract/Free Full Text]
  29. McGrath SA, Basu A, and Zeitlin PL. Cystic fibrosis gene and protein expression during fetal lung development. Am J Respir Cell Mol Biol 8: 201-208, 1993.[ISI][Medline]
  30. Murray CB, Chu S, and Zeitlin PL. Gestational and tissue-specific regulation of C1C-2 chloride channel expression. Am J Physiol Lung Cell Mol Physiol 271: L829-L837, 1996.[Abstract/Free Full Text]
  31. Murray CB, Morales MM, Flotte TR, McGrath-Morrow SA, Guggino WB, and Zeitlin PL. CIC-2: a developmentally dependent chloride channel expressed in the fetal lung and downregulated after birth. Am J Respir Cell Mol Biol 12: 597-604, 1995.[Abstract]
  32. O'Brodovich H. Epithelial ion transport in the fetal and perinatal lung. Am J Physiol Cell Physiol 261: C555-C564, 1991.[Abstract/Free Full Text]
  33. O'Brodovich H. Fetal lung liquid secretion: insights using the tools of inhibitors and genetic knock-out experiments. Am J Respir Cell Mol Biol 25: 8-10, 2001.[Free Full Text]
  34. Olver RE, Schneeberger EE, and Walters DV. Epithelial solute permeability, ion transport and tight junction morphology in the developing lung of the fetal lamb. J Physiol 315: 395-412, 1981.[ISI][Medline]
  35. Olver RE and Strang LB. Ion fluxes across the pulmonary epithelium and the secretion of lung liquid in the foetal lamb. J Physiol 241: 327-357, 1974.[ISI][Medline]
  36. Post M, Souza P, Liu J, Tseu I, Wang J, Kuliszewski M, and Tanswell AK. Keratinocyte growth factor and its receptor are involved in regulating early lung branching. Development 122: 3107-3115, 1996.[Abstract/Free Full Text]
  37. Rao AK and Cott GR. Ontogeny of ion transport across fetal pulmonary epithelial cells in monolayer culture. Am J Physiol Lung Cell Mol Physiol 261: L178-L187, 1991.[Abstract/Free Full Text]
  38. Schwiebert EM, Cid-Soto LP, Stafford D, Carter M, Blaisdell CJ, Zeitlin PL, Guggino WB, and Cutting GR. Analysis of ClC-2 channels as an alternative pathway for chloride conduction in cystic fibrosis airway cells. Proc Natl Acad Sci USA 95: 3879-3884, 1998.[Abstract/Free Full Text]
  39. Scurry JP, Adamson TM, and Cussen LJ. Fetal lung growth in laryngeal atresia and tracheal agenesis. Aust Paediatr J 25: 47-51, 1989.[ISI][Medline]
  40. Souza P, O'Brodovich H, and Post M. Lung fluid restriction affects growth but not airway branching of embryonic rat lung. Int J Dev Biol 39: 629-637, 1995.[ISI][Medline]
  41. Sturgess J and Imrie J. Quantitative evaluation of the development of tracheal submucosal glands in infants with cystic fibrosis and control infants. Am J Pathol 106: 303-311, 1982.[Abstract]
  42. Thiemann A, Grunder S, Pusch M, and Jentsch TJ. A chloride channel widely expressed in epithelial and non-epithelial cells. Nature 356: 57-60, 1992.[CrossRef][ISI][Medline]
  43. Wigglesworth JS, Desai R, and Hislop AA. Fetal lung growth in congenital laryngeal atresia. Pediatr Pathol 7: 515-525, 1987.[Medline]
  44. Zeitlin PL, Loughlin GM, and Guggino WB. Ion transport in cultured fetal and adult rabbit tracheal epithelia. Am J Physiol Cell Physiol 254: C691-C698, 1988.[Abstract/Free Full Text]
  45. Zhou L, Graeff RW, McCray PB Jr, Simonet WS, and Whitsett JA. Keratinocyte growth factor stimulates CFTR-independent fluid secretion in the fetal lung in vitro. Am J Physiol Lung Cell Mol Physiol 271: L987-L994, 1996.[Abstract/Free Full Text]




This Article
Abstract
Full Text (PDF)
Alert me when this article is cited
Alert me if a correction is posted
Citation Map
Services
Email this article to a friend
Similar articles in this journal
Similar articles in ISI Web of Science
Similar articles in PubMed
Alert me to new issues of the journal
Download to citation manager
Search for citing articles in:
ISI Web of Science (4)
Google Scholar
Articles by Blaisdell, C. J.
Articles by Welling, P.
Articles citing this Article
PubMed
PubMed Citation
Articles by Blaisdell, C. J.
Articles by Welling, P.


HOME HELP FEEDBACK SUBSCRIPTIONS ARCHIVE SEARCH TABLE OF CONTENTS
Visit Other APS Journals Online
Copyright © 2004 by the American Physiological Society.