Skip to main content
American Journal of Physiology - Lung Cellular and Molecular Physiology logoLink to American Journal of Physiology - Lung Cellular and Molecular Physiology
. 2014 Aug 22;307(8):L618–L631. doi: 10.1152/ajplung.00144.2014

Regulation of fibroblast lipid storage and myofibroblast phenotypes during alveolar septation in mice

Stephen E McGowan 1,, Diann M McCoy 1
PMCID: PMC4200388  PMID: 25150063

Abstract

Signaling through platelet-derived growth factor receptor-α (PDGFRα) is required for alveolar septation and participates in alveolar regeneration after pneumonectomy. In both adipose tissue and skeletal muscle, bipotent pdgfrα-expressing progenitors expressing delta-like ligand-1 or sex-determining region Y box 9 (Sox9) may differentiate into either lipid storage cells or myofibroblasts. We analyzed markers of mesenchymal progenitors and differentiation in lung fibroblasts (LF) with different levels (absent, low, or high) of pdgfrα gene expression. A larger proportion of pdgfrα-expressing than nonexpressing LF contained Sox9. Neutral lipids, CD166, and Tcf21 were more abundant in LF with a lower compared with a higher level of pdgfrα gene expression. PDGF-A increased Sox9 in primary LF cultures, suggesting that active signaling through PDGFRα is required to maintain Sox9. As alveolar septation progresses from postnatal day (P) 8 to P12, fewer pdgfrα-expressing LF contain Sox9, whereas more of these LF contain myocardin-like transcription factor-A, showing that Sox9 diminishes as LF become myofibroblasts. At P8, neutral lipid droplets predominate in LF with the lower level of pdgfrα gene expression, whereas transgelin (tagln) was predominantly expressed in LF with higher pdgfrα gene expression. Targeted deletion of pdgfrα in LF, which expressed tagln, reduced Sox9 in α-actin (α-SMA, ACTA2)-containing LF, whereas it increased the abundance of cell surface delta-like protein-1 (as well as peroxisome proliferator-activated receptor-γ and tcf21 mRNA in LF, which also expressed stem cell antigen-1). Thus pdgfrα deletion differentially alters delta-like protein-1 and Sox9, suggesting that targeting different downstream pathways in PDGF-A-responsive LF could identify strategies that promote lung regeneration without initiating fibrosis.

Keywords: stem cell, lung alveolarization, sex-determining region Y box 9, preadipocyte factor-1, stereology, adipocyte, platelet-derived growth factor, delta-like ligand-1 (Dlk1)


platelet-derived growth factor receptor-α (pdgfrα)-expressing cells are required for development of the lung, central nervous system, and heart and contribute to malignancies in the gastrointestinal tract, brain, and bone marrow (5, 7, 13, 26, 43, 52). PDGFRα resides in cells which assume divergent characteristics in the brain [oligodendrocyte precursors (OPC)], the heart (epicardial fibroblasts), stromal vascular cells in skeletal muscle, adipocytes in white or brown fat, and stellate cells in the liver (3, 18, 42, 50). During pulmonary alveolar development, pdgfrα-expressing cells are required for secondary septation and variably display either lipid storage or myofibroblastic phenotypes. The signaling pathways that direct PDGFRα+ alveolar cells along these two pathways remain undefined, although it is clear that the PDGFRα ligand PDGF-A directs cellular proliferation, survival, and migration.

Using mice with a green fluorescent protein (GFP) insertion controlled by the endogenous pdgfrα promoter regulatory region, both GFP-bright and GFP-dim septal cells have been observed, indicating that the level of pdgfrα gene expression varies among interstitial lung fibroblasts (LF) (32). What remains unclear, however, is how these two phenotypes of LF are regulated during secondary septation. Previous studies have established that the myofibroblastic population of LF is required for secondary septal formation and was posited to arise from a progenitor population that requires signaling through PDGFRα (the sole receptor for PDGF-A) (5). Boström and associates hypothesized that the progenitors fail to differentiate into myofibroblasts, thereby halting secondary alveolar septation (4, 5). It remains unclear how PDGF-A regulates the proliferative expansion of LF and their transition to myofibroblasts, although myocardin-like transcription factors (MRTF) may be involved (51). A second LF population is lipid-laden alveolar interstitial cells (now widely held to be fibroblasts). These cells display CD90 (Thy1), which is absent from fibroblasts lacking neutral lipids (60, 61). The lipid content can be modified in culture by exposure to peroxisome proliferator-activated receptor-γ (PPARγ) agonists or transfection with Thy1 cDNA (34, 61, 62). In addition, the G0-G1 switch 2 (G0S2) gene is abundant in adipocytes and is required for the development of cytoplasmic lipid droplets, a hallmark of mature adipocytes (20).

Myogenesis and adipogenesis are regulated by several gene products, including delta-like protein-1 (Dlk1, also known as preadipocyte factor-1 or fetal antigen-11), sex-determining region Y box 9 (Sox9), and Tcf21 (Pod1). Dlk1 is a transmembrane protein with an extracellular domain that may be cleaved by tumor necrosis factor-α-converting enzyme (TACE/ADAM17), releasing a biologically active fragment with paracrine effects that differ from those exerted by membrane-bound Dlk1 (56). ADAM17 complexes with CD166 and tetraspanin, which stabilizes CD166 and promotes cell adhesion through homotypic CD166-CD166 interactions (15). Sox9 is a high-mobility group-box domain transcription factor that regulates male sex determination and chondrogenesis (12). In the developing lung, Sox9 has been most extensively studied in proliferating distal epithelial cells, where it defines progenitors that differentiate into alveolar type 1 and type 2 cells (45). Sox9 is also expressed in the airway mesenchyme, and mice bearing a Sox9 gene deletion in airway cartilage die at birth (57). However, very little is known about how Sox9 may influence mesenchymal cells during alveolarization. Likewise Tcf21, a helix-loop-helix transcription factor, is essential for airway branching morphogenesis, but its contributions to alveolar formation have not been reported (44).

We hypothesized that PDGFRα signaling influences the progenitor state and bidirectional differentiation of the LF precursors during secondary septation. Using flow cytometry, we examined markers of the progenitor and differentiated states [i.e., CD34, stem cell antigen-1 (Sca1), CD166, Dlk1, Tcf21, and Sox9] in PDGFRα-nonexpressing (GFP), PDGFRα-GFP dim (GFPlow), and PDGFRα-GFP bright (GFPhigh) LF. We chose postnatal day (P) 8 to examine the bipotential state because both lipid-laden and non-lipid-laden fibroblasts are abundant, portions of both populations are proliferating, and both the PDGFRα-GFP dim and PDGFRα-GFP bright LF are observed (Fig. 1A). Studies were extended to mice with a conditional deletion of pdgfrα in alveolar cells that express transgelin (tagln, SM22α, Fig. 1B). PDGFRα signaling has divergent effects during differentiation into lipid storage or myofibroblastic LF.

Fig. 1.

Fig. 1.

Genetic manipulations used to label alveolar mesenchymal cells. Platelet-derived growth factor receptor (PDGFR) α-expressing lung fibroblasts (LF) were marked with enhanced green fluorescent protein (GFP), regulated by the endogenous PDGFRα promoter (PDGFRα-GFP, A). Cell surface CD140a (PDGFRα) increased with GFP fluorescence, and PDGFREα mRNA was more abundant in the GFPhigh cells. **P < 0.01, GFPlow or GFPhigh compared with GFP negative (GFP). *P < 0.05, GFPhigh compared with GFPlow. †P < 0.05, GFPhigh vs. GFPlow. B: 2 subpopulations were distinguished based on the intensity of GFP fluorescence: GFPhigh LF, which reside more distally in the elongating alveolar septa, and GFPlow LF, which contain lipid droplets that reside at the base of septa at postnatal day (P) 8 but regress by P15. C: a separate line of mice that expressed Cre recombinase controlled by the transgelin (TG) promoter was used to excise a stop codon and enable expression of tdTomato (dTom), which was expressed in pericytes and at P8 and in PDGFRα-GFP LF: TGCre;R26Tom; PDGFRα-GFP (contain both GFP and dTom, B). TG-Cre was also used to inactivate LoxP-flanked PDGFRα in cells where TG expression is abundant, which has functional consequences in PDGFRα-expressing LF but not in pericytes, which express PDGFRβ but not PDGFRα.

MATERIALS AND METHODS

Reagents

Primary antibodies.

Primary antibodies were mouse monoclonal anti-α-smooth muscle actin (α-SMA), clone IH8 (MA5-15805; Thermo Scientific, Rockford, IL); mouse monoclonal clone IA4, anti-α-SMA-PE (R&D Systems, Minneapolis, MN); mouse monoclonal α-SMA clone IA4-FITC (Sigma-Aldrich, St. Louis, MO); goat polyclonal anti-mouse CD166 (R&D Systems); mouse monoclonal IgG1 anti-Dlk1, clone 3A10 (MA5-15915; Thermo Scientific); goat anti-MRTF-A (sc-21558, Western 1:500 dilution; Santa Cruz Biotech, San Jose, CA); anti-Sox9 (rabbit polyclonal, AB5535, Western 1:2,000 dilution; Millipore, Temecula, CA); and mouse monoclonal anti-β-tubulin (clone TUB2.1; Sigma-Aldrich). We also used CD34 Brilliant Violet rat anti-mouse CD34, rat IgG2a clone RAM34, final 1.25 μg/ml; rat monoclonal IgG2a, 0.4 μg/ml anti-mouse Sca-1-allophycocyanin (APC)/Cy7 conjugate; rat monoclonal IgG2a antimouse CD45-APC conjugate 1 μg/ml (BD Biosciences, San Jose, CA). In addition clone 16A8 rat IgG2a AlexaFluor 647 anti-mouse Ki67 (0.4 μg/ml; Biolegend, San Diego, CA); polyclonal rabbit IgG anti-G0S2 (1 μg/ml, PA5–23697; Thermo Scientific); rabbit monoclonal IgG anti-p57kip2 (1 μg/ml) and rabbit polyclonal anti-Tcf21, 1 μg/ml (Abcam, Cambridge, MA); anti-pan cytokeratin-PE, mouse IgG1 monoclonal clone C-11 (Novus Biologicals, Littleton, CO); anti-adipocyte differentiation-related peptide (ADRP), rabbit IgG monoclonal 0.5 μg/ml (Abcam); and rat anti-mouse CD140a (PDGFRα)-APC, 1 μg/ml (BD-Biosciences) were employed.

Secondary antibodies.

Secondary antibodies included goat-anti-rabbit IgG-AlexaFluor 568 or -647; goat anti-rabbit-peroxidase; rabbit anti-goat or goat-anti mouse peroxidase; Zenon mouse IgG1 primary antibody-labeling kits AlexaFluor 405 or AlexaFluor 488 or Zenon rabbit IgG labeling kits at AlexaFluor 405, 568, or 647 (Life Technologies, San Diego, CA); (E,E)-3,5-bis-(4-phenyl-1,3-butadienyl)-4,4-difluoro-4-bora-3a,4a-diaza-s-indacene, BODIPY 665/670 (1:2,000) (Life Technologies); and LipidTOX red (LTR) neutral lipid stain (Life Technologies) 1:400 dilution. We also used recombinant human PDGF-AA and TGF-β1 (R&D Systems), PoPo3-idodide (benzoxazolium, 2,2′-{1,3-propanediylbis[(dimethyliminio)-3,1-propanediyl-1(4H)-pyridinyl-4-ylidene-1-propen-1-yl-3-ylidene]}bis[3-methyl], tetra-iodide) (Life Technologies).

Mice

Mice bearing the PDGF-Rα-GFP construct have been described (27, 28). Production and nuclear localization of GFP is under the control of the endogenous pdgf-rα promoter (Fig. 1A). GFP expression in the PDGF-Rα-GFP mice spatially and temporally recapitulates endogenous pdgf-rα expression (16). The mice used in this study carried one pdgfrα-GFP allele (that does not encode for active PDGF-Rα) and one functional pdgf-rα allele. These heterozygous mice are phenotypically identical to wild-type (GFP) mice, except for nuclear GFP, which enables their identification (16). Mice with a targeted deletion of LoxP-pdgfrα (Fig. 1C) have also been described (37). The DNA coding Cre-recombinase was inserted into exon 1 of tagln (TG) and mediates Cre-recombination postnatally but not in the embryo (64). Transgelin is expressed in pulmonary myofibroblasts, pericytes, and smooth muscle cells. The PDGFRα-GFP mice were bred with mice possessing the transgelin-driven Cre-recombinase B6.129S6-Taglntm2(cre)Yec/J (Jackson Labs 00678) and the LoxP-flanked dTomato reporter, which bears a LoxP-flanked stop codon B6.Cg-Gt(ROSA)26Sortm14(CAG−tdTomato)Hze/J (Jackson Labs 007914). After Cre-mediated excision of the stop codon, these mice demonstrate the nuclear GFP marker in cells expressing pdgfrα and the dTomato cytoplasmic marker in cells expressing Tagln. Triple heterozygote PDGFRFP+/−;TGCre+/−;R26dTom+/− mice were used to distinguish myofibroblastic Tagln-bearing PDGFRFP+ LF from PDGFRFP+ LF, which do not express Tagln.

Isolation of primary mouse LF.

Lung fibroblasts were isolated from heterozygous mice on P8 or for primary cell culture on P12 using a previously reported method involving digestion with collagenase (35). The dispersed cells were resuspended in Ham's F-12 medium containing 10% FBS, fibroblasts were selected by their adherence to tissue culture dishes for 1 at 370C, and 34.0 ± 3.5% of the adherent cells were PDGFRα-GFP+ (mean ± SE, n = 10). The purity of the fibroblasts was assessed by staining for cellular markers specific for epithelial (cytokeratin 18), macrophage (CD206), and endothelial (CD31) cells, as previously published. Epithelial and endothelial cells comprised ∼2.5 and 1.6%, respectively, whereas macrophages comprised 8.4 ± 2.3% (n = 5) of the adherent cells and were only detected in the PDGFRα-GFP population (36). Using an anti-pan cytokeratin antibody, we observed that 3.4 ± 0.9, 3.3 ± 1.3, and 2.0 ± 0.2% of GFP, GFPlow, and GFPhigh LF, respectively, contained cytokeratin at P8 (mean ± SE, n = 4). Protocols for animal use were approved by the Iowa City Veterans Affairs Medical Center animal use committee (35).

Analysis of CD45, Sca1, Sox9, Dlk1, Tcf21, CD34, CD166, p57kip2, G0S2, and α-SMA in mouse LF using flow cytometry.

LF that had been freshly isolated at P8 were fixed; and for Sox9, Tcf21, p57kip2, G0S2, or α-SMA, the LF were permeabilized before immunostaining (27). Flow cytometry (FACS) was conducted using a LSR II flow cytometer (BD Biosciences), and at least 20,000 gated events were analyzed using CellQuest Software (BD Biosciences) (27, 35). More detailed information about the antibodies used for staining appears in the materials and methods section. The background fluorescence from the corresponding IgG isotype controls was subtracted to calculate the proportions of the different fibroblast populations. The isolated lung cells that stained with anti-CD45 (a marker of hematopoietic cells) were excluded from the quantitative analyses.

Analysis of gene expression in cultured LF.

The methods for culturing LF cells have been published (27, 35). After being washed with PBS, the cells were cultured in Opti-MEM containing 2% FBS, 2 mg/ml BSA, 3 mM CaCl2, 100 μg streptomycin, and 100 units penicillin G/ml for 16 h before adding the PDGF-A or TGF-β1. Total RNA was isolated using TRI-Reagent, subjected to reverse transcription. mrtf-A (Mm00461840_m1), mrtf-b (Mm00463877_m1), and β2-microglobulin (Mm00437762.m1) mRNA were quantified using TaqMan Gene Expression Assays (27). At least five independent experiments were performed. Values for Sox9 and MRTF gene expression were normalized to β2-microglobulin using the 2−ΔΔCT relative quantification method (30).

Selection of CD45 LF and gene expression analysis.

LF were isolated at P8 from PDGFRα-GFP+ mice, stained for 45 min at 4οC with APC-anti-CD45a, washed, and sorted on a FACS Aria (BD Biosciences). Parenchymal lung cells from five litters were separated into four populations (CD45+; CD45,GFP; CD45,GFP+low; and CD45,GFP+high). After being washed, the cell pellets were lysed, and the RNA was purified using the Quick RNA MicroPrep kit (Zymo Research, Irvine, CA). Following reverse transcription, the cDNA was purified, quantified, and subjected to real-time qRT-PCR using TaqMan probes for dlk1 (Mm00494477_m1), sox9 (Mm00448840_m1), tcf21 (Mm00448961_m1), and G0S2 (Mm00484537_g1). LF were isolated from TGCre+/−;PDGFRαF/F and littermate control mice, and CD45+ cells were removed using rat anti-CD45 attached to magnetic beads coated with sheep anti-rat IgG (DynaBeads Life Technologies, San Diego, CA). Periostin gene expression was analyzed using TaqMan probes, which detected all mRNA (Mm00450111_m1), or the unique splice product Mm01284919_m1, which spans the exon 20–22 junction (exon 21 spliced out). PPARγ mRNA was quantified using Mn01184322_m1. TaqMan qRT-PCR primers and probes targeting splice sites (underlined) in exon 5 of Dlk1 were designed to differentially amplify the alternatively spliced dlk1-A and dlk1-C isoforms and compared with a primer and probe set that was not subject to alternative spicing dlk1-A (Mn00494477_m1). Forward and reverse primers were 5′-CTGGATTCGTCGACAAGA-3′ and 5′-cttgcacagacactcgaa-3′, respectively; probe 5′-Fam-CCTCCAGCACACCCAGGT-3′ Iowa black (quencher). For Dlk1-C, forward was the same as for dlk1-A, and reverse primers 5′-caggatggtgaagcagat-3, probe 5′-Fam-CCAGCACACCCAGGGACA-3′ Iowa black were obtained from Integrated DNA Technologies (IDT, Coralville, IA). Reactions were performed using the standard TaqMan master mix, and amplification conditions recommended for standard TaqMan probes, except that the concentration of the forward primer was 900 nM, and the reverse primers were 300 nM for the dlk1-A and -C amplification reactions.

Sox9 and MRTF-A immunoblotting.

After being washed and lysed in the presence of phosphatase and proteinase inhibitors, equal quantities of protein were subjected to SDS-PAGE and transferred to nitrocellulose (35). Anti-Sox9 or MRTF-A were diluted 1:2,000 or 1:500, respectively. After being washed, the primary antibodies were detected using goat anti-rabbit peroxidase and enhanced chemiluminescence (GE Healthcare, Piscataway, NJ) or rabbit anti-goat peroxidase and SuperSignal West Pico Chemiluminescent Substrate (Thermo Scientific). Fluorography was performed, the film was imaged, and the densities of Sox9 or MRTF-A were normalized to the density of β-tubulin for each lane to account for differences in the amounts of protein that were loaded.

Laser-scanning confocal microscopy: Tissue preparation and imaging.

Lungs were uniformly inflated using 50 μl of fixative containing agarose per gram body weight and fixed for 6 h for laser-scanning confocal microscopy (32). For localization of Sox9 or MRTF-A 7-μm sections were used, whereas for enumeration of cells containing lipid droplets 100-μm sections were prepared (36). We determined the proportions of Sox9- or MRTF-A-containing cells within pdgfrα-expressing (GFP+) and pdgfrα-nonexpressing (GFP) cells. Nuclei were stained with PoPo3 diluted 1:15,000 from a stock in DMSO (InVitrogen; Molecular Probes). Incubations with the primary antibodies were overnight at 4°C. All secondary antibodies were AlexaFluor-labeled and used at a 1:2,000 dilution. The laser intensities and detector gains were optimized at the beginning and remained constant for each channel throughout the imaging session. Images of the stained sections were acquired from randomly selected fields using a Zeiss LSM710 laser-scanning confocal microscope at a 1,024 × 1,024 pixel density.

Analysis of Images Obtained Using Confocal Microscopy

The Sox9- or MRTF-A-containing cells in the alveolar walls were classified as follows: nuclear GFP+ (pdgf-rα expressing, green), PoPo3+ (all nuclei, red), Sox9 or MRTF-A (AlexaFluor 647, pseudocolor blue) in cells without GFP (PoPo3, but without GFP), and Sox9 or MRTF-A in cells containing GFP and PoPo3. With the use of uniform segmentation criteria, the pixel regions contained 1) both red and blue, but not green, (nuclei that were Sox9+ and GFP), 2) red, blue, and green (nuclei that were Sox9+ and GFP+), and 3) all PoPo3+ nuclei (red). The nuclei of cells that were Sox9+, GFP, or Sox9+, GFP+ were enumerated and expressed relative to the total of pdgfrα-expressing alveolar cells (GFP+) or nonexpressing alveolar cells (GFP). Because MRTF-A was located in the cytoplasm, a circle with a diameter of 10 μm was placed over each nucleus. The cell was classified as MRTF-A+ if the blue pixels met the segmentation criteria and were within the nuclear-centered circle. The percentages of nuclei in Sox9, GFP+ or Sox9, GFP groups were calculated for each field, and a mean was determined for each mouse. The mean data from four mice were combined to obtain a mean and SE for each age.

Distinguishing myofibroblast-like LF using reporter mice bearing both dtomato and eGFP.

PDGFRα-GFP+;TGCre+/−;R26dTom+/− mice were killed at P8, P15, and P21 and as adults (4 mo or older). The lungs were fixed at uniform inflation (50 μl/g body wt), sectioned at 100 μm, and stained with BODIPY 665/676 (InVitrogen Molecular Probes) to identify neutral lipid droplets after excitation at 647 nm. Image stacks were acquired at a 2.5-μm z-interval and analyzed using the optical fractionator and StereoInvestigator (MBF Bioscience, Williston, VT). Randomly selected alveolar regions (lacking airways and blood vessels) were imaged at 1,024 × 1,024 pixel resolution using the Zeiss LSM710 averaging three scans per field and capturing three image stacks at each age. One mouse at each age was used within one staining and imaging session, and four different mice were used at each age over the four sessions. PDGFRα-GFP+ cells were subclassified based on GFP intensity (GFP high or low) as well as the presence of dTomato (dTom+ or dTom) and whether or not they contained neutral lipid droplets. Cells were classified as containing lipids if the droplets were in close proximity to the nucleus.

Assessment of respiratory mechanics in control and TGCre+/−;pdgfrαF/F mice.

Static and dynamic pulmonary mechanics were assessed using a FlexiVent (Scireq, Montreal, Quebec, Canada) ventilator (33). Five adult mice (ages ranging from 4 to 6 mo) were deeply anesthetized, tracheotomized, paralyzed, and ventilated with an end-expiratory pressure of 2 cmH2O. After volume history was standardized, an 8-s perturbation (Prime-8) consisting of randomly generated mutually prime oscillations was delivered while pressure and volume were monitored continuously. The impedance to the oscillatory wave was calculated and partitioned into the airway resistance, tissue damping (G), and tissue elastance (H) components using the constant-phase model (17). Static volume-pressure measurements were performed using the FlexiVent algorithm with the prescribed weight-adjusted volumes. The data for each subject were expressed as the mean of four determinations. The mice were killed, and the lungs were uniformly inflated and maintained at 25 cmH2O pressure for 16 h in 4% paraformaldehyde. Volume was measured by displacement; the lungs were embedded in paraffin, sectioned, and stained with hematoxylin and eosin or Verhoeff-van Gieson. The surface area was quantified using the cycloids for Sv stereological probe (StereoInvestigator, Williston, VT) and normalized to the displacement volume (37).

Statistical Methods

Data were expressed as means ± SE of the number of different mice that were used or the number of different experiments that were performed using cell cultures (46). ANOVA (either 1, 2, 3 way as indicated in the legends for Figs. 110) was performed using Systat (Chicago, IL) and Student's t-test using Microsoft Excel. Additional details describing how the data from different images or image stacks were combined appear in the method for a particular stereological procedure. Post hoc tests are described in the legends for Figs. 110. Values of P < 0.05 were considered significant.

Fig. 10.

Fig. 10.

pdgfrα gene deletion alters respiratory mechanics. Representative microscopic fields for control (A) and TGCre+/−;PDGFRαF/F (B) mice; magnification ×50. Insets, elastic fibers in the alveolar walls (arrows), which were thinner and disconnected in mice with PDGFRα disruption. C: static pressure-volume curves for control (n = 8, closed circles) and TGCre+/−;PDGFRαF/F (n = 5, open squares) mice showing that targeted pdgfrα gene deletion increased the compliance of the respiratory system (combined lung and chest wall).

RESULTS

LF Differing in pdgfrα Expression (GFP Fluorescence Intensity) Exhibit Divergent Phenotypes

Our objective was to determine whether variations in PDGFRα levels identify distinct populations of lung progenitor cells using GFP fluorescence intensity as a surrogate for PDGFRα expression. LF that had been isolated from PDGFRα-GFP mice were stained for CD140a (PDGFRα) without permeabilization and examined using FACS to correlate the intensity of GFP fluorescence with the abundance of cell surface PDGFRα. The results in Fig. 1A show that the abundance of CD140a positively correlated with GFP intensity, indicating that GFP accurately represents the PDGFRα gene product. When LF were permeabilized, 100% of the GFP+ LF contained CD140a (data not shown). Alternatively using FACS, we sorted LF from PDGFRα-GFP mice and observed that PDGFRα mRNA was more abundant in GFPhigh than in GFPlow LF (Fig. 1A).

We first performed flow cytometry on LF that had been fixed immediately after isolation and stained for various antigens (Fig. 2A). Based on the intensity of GFP fluorescence, three subpopulations of LF were observed (GFP, GFPlow, and GFPhigh). Whereas the dot plots shown in Fig. 2A depict all adherent cells, for subsequent quantitative comparisons, CD45+ cells that had adhered to tissue culture plastic during selection (including macrophages and potentially CD45+ bone marrow fibrocytes) were excluded from the analyses. After gating on CD45-negative cells, which do not stain with APC-anti-CD45, we identified three populations based on GFP fluorescence intensity (R3: GFP, R4: GFPlow, and R5: GFPhigh; Fig. 2Bc). We observed differential distribution of Sca1+, CD34+, and Sca1+,CD34+ cells in the R3, R4, and R5 gates (Fig. 2, Bd-Bf). Specifically, fewer Sca1+ cells were observed in the GFPlow subpopulation (Fig. 2Be). The proportion of CD45+ cells in the isolated GFP and GFP+ cells was 49.0 ± 3.5 and 1.6 ± 0.6%, respectively (mean ± SE, n = 5). These data demonstrate that CD45 LF contain three distinct populations of PDGFRα-expressing cells as determined by GFP fluorescence intensity.

Fig. 2.

Fig. 2.

Representative plots from flow cytometric (FACS) analysis of LF. Pdgfrα-expressing (GFP+) and -nonexpressing (GFP) LF were isolated from mice bearing the pdgfrα-driven GFP marker. A: representative dot plots showing the distribution of events for LF that are GFPlow, GFPhigh, or are not expressing PDGFRα GFP. Panels a, e, g, i, and k show staining with isotype control immunoglobulins (IgG), whereas the remaining panels show staining with the designated immune IgGs or LipidTOX red (LTR). All adherent cells meeting the gating criteria for side scatter and forward scatter width are shown. CD45+ cells were not excluded from these plots but were excluded for the combined data shown in the bar graphs in Figs. 36. The numbers show percentage of total gated cells (R2 in Bb) in the three populations distinguished by GFP intensity. B: gating strategy used to evaluate Sca1 and CD34 in CD45 LF. Forward scatter area (FSC-A) vs. side scatter area (SSC-A) (a) or forward scatter width (FSC-W) (b) were used to eliminate potential aggregates or small dead cells. c, Negative selection using allophycocyanin (APC)-anti-CD45 identified LF with different levels of GFP fluorescence (R3, R4, R5). CD45+ LF were only observed in the GFP population and were excluded from subsequent analyses. df, LF stained with Brilliant violet (Brill-Viol 421) anti-CD34 and APC-Cy7 anti-Sca1 within the 3 CD45 populations (R3-R5). Percentages of total CD45 cells in each quadrant are shown.

We next compared differentiation and progenitor markers in the three LF populations differing in GFP fluorescence intensity. Markers for neutral lipid droplets (stain with LTR, Fig. 3A) or ADRP (perilipin 2; Fig. 3B) were more abundant in the GFPlow population. By contrast, a larger proportion of GFPhigh than GFPlow cells contained α-SMA (Fig. 3C). Next, cell surface markers of mesenchymal progenitors (Sca1, CD34, and CD166) were assessed. Sca1+ LF were observed in both the GFP+ and GFP populations, although CD34+ LF were more prevalent among GFP+ LF (Fig. 3, D and E). A significantly larger fraction of the GFPlow population displayed CD166 (Fig. 3, E and F).

Fig. 3.

Fig. 3.

FACS analysis of progenitor markers in fibroblasts (LF) with different levels of pdgfrα gene expression. LF isolated from PDGFRα-GFP mice at P8 were stained for the antigens shown on the y-axes: LTR, neutral lipid binding LTR (A); ADRP, adipocyte differentiation related protein, perilipin-2 (B); α-SMA, α-smooth muscle actin, acta2; (C); Sca1, stem cell antigen-1 (D); CD34, podocalyxin (E); and CD166, activated leukocyte adhesion molecule (ALCAM), activated-leukocyte cell adhesion molecule (F); the CD45 cell populations were selected as shown in Fig. 2 and expressed as a percentage of all CD45 LF within a particular population based on the intensity of GFP fluorescence (GFP, GFPlow, GFPhigh). **P < 0.01 and *P < 0.05, mean ± SE, n = 6 mice, 2-way ANOVA, Student-Newman-Keuls post hoc test comparing GFPlow or GFPhigh LF with GFP. †P < 0.01 comparing GFPlow with GFPhigh.

Next, we investigated whether PDGFRα expression can distinguish between proliferative and quiescent LF. We found that Ki-67, an estimator of cellular proliferation, was more abundant in GFPhigh compared with GFPlow LF at both P4 (Fig. 4A) and P8 (Fig. 4B), with a higher proportion of Ki67+,GFPhigh LF at P4 than at P8 (P < 0.01, 3-way ANOVA with Student-Newman-Keuls post hoc test). A converse expression pattern was observed for markers of quiescence. Specifically, p57kip2 (Fig. 4C) and G0S2 (Fig. 4D) were more abundant in the GFPlow than in the GFPhigh LF populations at P8. There were no differences in G0S2 comparing P4 with P8 (at P4 15.5 ± 5.4%, mean ± SE of the GFPlow LF). We also observed that the progenitor cell transcription factor Tcf21 was more abundant in the GFPlow population at both P4 (data not shown) and P8 (Fig. 4E). Likewise, G0S2 and tcf21 mRNA levels were higher in GFPlow than in either the GFP or GFPhigh populations (Fig. 4F). Thus, the balance tilts toward proliferation in PDGFRα-high LF, whereas it tilts toward quiescence in PDGFRα-low LF.

Fig. 4.

Fig. 4.

Ki-67 is diminished, whereas p57kip2, G0S2, and Tcf21 are increased in GFPlow LF. FACS analysis of intracellular proteins [A, Ki67 (MKI67) P4; B, Ki67 P8; C, p57kip2 cyclin-dependent kinase inhibitor (CDKN1c); D, G0-G1 switch protein-2 (G0S2); E, Tcf21 (Pod1)] were analyzed by FACS at P8, except for A which shows LF at P4. The proportions of CD45 LF containing the various antigens are expressed as a percentage of all CD45 within a particular population. F: quantitative real-time PCR (qRT-PCR, normalizing to constitutive β2-microglobulin expression) analysis of G0S2 and Tcf21 mRNA in GFPlow and GFPhigh expressed relative to PDGFRα-GFP LF obtained after sorting by FACS (n = 4). **P < 0.01 and *P < 0.05, mean ± SE, n = 4 mice, 2-way ANOVA, Student-Newman-Keuls post hoc test comparing GFPlow or GFPhigh LF with GFP. †P < 0.05 and ††P < 0.01, comparing GFPlow with GFPhigh.

Active expression of particular genes is required to sustain progenitor cells and allay differentiation. Sox9 delays the differentiation of mesenchymal progenitors into smooth muscle cells, whereas Dlk1 modulates differentiation into either adipocytes or myofibroblasts (31, 56, 65). There was a trend toward greater abundance of Dlk+ LF in the GFPhigh population (P = 0.06, 2-way ANOVA, Student-Newman-Keuls post hoc test; Fig. 5A). Dlk1+, Sca1+ LF comprised a significantly larger proportion of the GFPhigh compared with either the GFP or GFPlow populations (Fig. 5A, inset). A larger proportion of GFP+ LF (either high or low) contained Sox9 compared with GFP LF (Fig. 5B). We next gated on the CD45, Sca1+ cells and found that a larger proportion of Sox9+ LF resided in the GFPhigh compared with either the GFP or GFPlow populations (Fig. 5B, inset). Therefore, although Sca1+, CD45 GFP-negative LF were observed, Sca1+ LF within the GFPhigh population exhibited a higher abundance of cell surface Dlk1 and Sox9. Analysis of mRNA levels demonstrated that Sox9 mRNA expression was more abundant in GFPhigh but not in GFPlow compared with GFP LF (P = 0.15, 2-way ANOVA; Fig. 5C). Splice-independent Dlk1 mRNA was higher in the GFPhigh subpopulation than in GFP LF, whereas GFPlow LF had significantly less dlk1 mRNA compared with GFPhigh LF and GFP LF (Fig. 5C). This shows that Sox9 and Dlk1 are predominantly found in the Sca1+, GFPhigh cells, identifying the progenitor characteristics of this LF population.

Fig. 5.

Fig. 5.

The abundance of sex-determining region Y box 9 (Sox9) correlates with pdgfrα gene expression. FACS analysis of the 3 LF subpopulations bearing cell surface delta-like protein-1 (Dlk1) (n = 7) (A) and intracellular Sox9 (n = 5) (B) were assessed at P8 using FACS. The proportions of CD45 LF containing Dlk1 or Sox9 are expressed as a percentage of all CD45 within a particular population. Insets show the cells that are both Sca1+ and CD45 relative to the total CD45 LF within each subpopulation defined by GFP intensity. C: qRT-PCR showing Sox9 and Dlk1 mRNA in GFPlow and GFPhigh expressed relative to PDGFRα-GFP LF (n = 4). **P < 0.01 and *P < 0.05, mean ± SE, 2-way ANOVA, Student-Newman-Keuls post hoc test comparing GFPlow or GFPhigh LF with GFP. †P < 0.01, comparing GFPlow with GFPhigh.

PDGFRα is Required to Maintain Sox9 in LF

PDGFRα and Sox9 may coincidently mark the same LF subpopulation, or PDGFRα signaling may directly influence Sox9 gene expression. To examine these possibilities, we studied LF from mice bearing a targeted deletion of pdgfrα in cells that express tagln, a marker of smooth muscle-like cells (Fig. 1C). We compared lipid-rich and myofibroblastic cells using the lipophilic fluorescent dye LTR after gating out CD45+ LF. Although pdgfrα deletion had no impact on the proportion LTR+ LF (Fig. 6A, bars on right), pdgfrα deletion increased the proportion of LF with Dlk1 on the cell surface within LTR+, Sca1+, or CD34+ cells (Fig. 6A, bars on left). Whereas the proportion CD34+ LF was reduced in pdgfrα-deficient mice, both CD166+ and the double-positive CD166+ and Dlk1+ LF were elevated after pdgfrα deletion (Fig. 6, B and C), showing that features of the PDGFRα GFPlow population were preserved.

Fig. 6.

Fig. 6.

Targeted pdgfrα gene deletion alters the abundance of Dlk1, Sox9, and Tcf21. AD: FACS analysis of LF isolated at P8 from 5 mice bearing the transgelin-mediated (TG-Cre) conditional deletion of loxP-flanked pdgfrα (PDGFRαF/F) or 5 littermate controls, gating on CD45 cells. A: %Dlk1+, LTR+, or Dlk1+,Sca1+ gating on CD45 (left); or %LTR+ within CD45 (axis on right). B: Dlk1+ LF within the CD45, CD34+ population (axis on left); %CD45, CD34+ relative to all CD45 LF (axis on right). C: %CD166+ (axis on left) or both CD166+, Dlk1+ (axis on right) in the CD45 population. *P < 0.05, n = 5, mean ± SE, t-test, paired variables. D: %CD45 LF containing α-SMA in control (open bars) or TGCre+/−;PDGFRαF/F (solid bars, axis on right). %Sox9+, CD45, α-SMA+ subpopulation, expressed relative to all α-SMA+ LF (axis on left). E: qRT-PCR analysis of periostin (Postn), periostin-lacking exon 21 (Postn 20–21), tcf21, and pparγ in PDGFRα-deleted and controls within the same litter, n = 5 litters. *P < 0.05 and **P < 0.01, mean ± SE, t-test, paired variables comparing PDGFRαF/F LF with those from their respective littermate controls. F: qRT-PCR using a probe that is not influenced by splicing dlk1-T (total), the A and C splice variants (dlk1-A and dlk1-C, respectively) comparing control and TGCre+/−;PDGFRαF/F mice (mean ± SE, n = 3 for each group); *P < 0.05, n = 3. dlk1-T was normalized to β2-microglobulin mRNA, whereas the dlk1-A and -C splice variants were normalized to dlk1-T for each respective sample.

We then examined the consequences of transgelin-mediated pdgfrα deletion on the myofibroblast marker α-SMA, which predominated in the GFPhigh myofibroblasts. In line with findings in Fig. 3, we observed a reduction in α-SMA + LF (Fig. 6D, bars on right). Similarly, deletion of pdgfrα reduced the proportion of α-SMA+, Sox9+ LF within both the CD45 and the fraction that was both CD45 and Sca1+ (Fig. 6D). To further examine effects of PDGFRα gene deletion on myofibroblasts, we examined periostin (postn), a matricellular protein produced by myofibroblasts and cardiac fibroblasts (53). Whereas total postn mRNA was unchanged following pdgfrα gene deletion, periostin transcripts lacking exon 21 were diminished (Fig. 6E), demonstrating that the splice isoforms are differentially regulated. Figure 6F compares expression of the splice-independent dlk1-T (total) and the dlk1-A and dlk1-C splice isoforms in MLF isolated from control and TGCre+/−;PDGFRαF/F mice. Although dlk1-T was unchanged, dlk1-C was increased in mice bearing the gene deletion. The Dlk1-C isoform cannot be cleaved and therefore would be preferentially identified using FACS for which only the cell surface pool was analyzed. This would explain why Dlk1 was increased in the retained lipid-laden LF after pdgfrα deletion (Fig. 6A). By contrast, tcf21 mRNA (which was more abundant in the GFPlow, lipid-laden LF) and pparγ mRNA (induces adipocyte differentiation) increased after pdgfrα gene deletion (Fig. 6E). Therefore, tagln-mediated deletion of pdgfrα preferentially depleted myofibroblasts while maintaining the population of LF that assume a lipid storage phenotype.

PDGF-AA Increases Sox9 in Cultured LF

We next examined whether the ligand PDGF-A stimulates Sox9 gene expression by quantifying Sox9 mRNA and protein in primary cultures of LF, which assume a myofibroblastic phenotype in culture. In parallel, we analyzed MRTF-A because it is inversely related to Sox9 gene expression (31). PDGF-A increased Sox9 mRNA (Fig. 7A) and protein (Fig. 7B) expression compared with untreated control LF, whereas TGF-β1 did not alter Sox9 although it did increase MRTF-A protein (Fig. 7C). Therefore, PDGFRα signaling enhances Sox9 expression but does not enhance the transition to myofibroblasts.

Fig. 7.

Fig. 7.

PDGFR-A increases Sox9 mRNA and protein in cultured LF. Primary isolates of LF were cultured and exposed to 20 ng PDGF-AA or 2.5 ng TGF-β1/ml for 24 h. A: RNA was isolated, and quantitative real-time PCR was performed using TaqMan probes. mRNA was compared with LF cultured in the absence of PDGF or TGF-β1 (control) within each respective experiment using the 2−(ΔΔCT) method. Representative immunoblots are shown after probing for Sox9 (B) and myocardin-like transcription factor-A (MRTF-A, C) using β-tubulin (βTub) as a loading control. The densities for Sox9 and MRTF-A were compared relative to the density of the corresponding unexposed control culture after normalizing for the density of βtub for each respective condition; n = 6 independent experiments using different primary LF isolations for both RNA and protein. *P < 0.05, mean ± SE, 1-way ANOVA, Student-Newman-Keuls post hoc test.

Sox9+ pdgfrα-Expressing LF Decrease, Whereas MRTF-A+ LF Increase, During Alveolar Septation

Our next step was to examine whether there was an inverse relationship between Sox9 and MRTF-A in PDGFRα-expressing alveolar cells. Because more α-SMA+ myofibroblasts accumulate between P8 and P12, this interval should encompass the transition to myofibroblasts (27). Sox9 and MRTF-A were assessed in cells bearing the PDGFRα-GFP marker, and cells lacking GFP served as an internal control for cells that do not express PDGFRα. However, because both epithelial and mesenchymal cells contributed to GFP cells in lung tissue, we could not specifically assess Sox9 in the GFP LF population as we had done using FACS. Consistent with a PDGFRα-mediated effect, we observed a decrease in Sox9 and a concomitant increase in MRTF-A at P12 relative to P8 within GFP+ but not GFP alveolar cells (Fig. 8). Sox9 immunostaining was observed in alveolar cells that did not express PDGFRα, of which many are likely to be epithelial cells (8). These data demonstrate that the transition of PDGFRα-GFP+ LF to myofibroblasts is accompanied by a decrease in Sox9.

Fig. 8.

Fig. 8.

Sox9 decreases, whereas MRTF-A increases, during septal elongation. Lungs from PDGFRα-GFP+ mice were uniformly inflated and fixed at P8 or P12 and immunostained for Sox9 or MRTF-A; n = 4 or 3 mice, respectively. A and B: nuclei containing Sox9 (blue), which is more readily observed in B, where green was changed to a yellow pseudocolor for better contrast. Yellow and green arrows show GFPlow and GFPhigh nuclei, respectively, containing Sox9. C and D: MRTF-A (blue) surrounding nuclei (GFP+, arrow). The proportions of pdgfrα-expressing (GFP+) or -nonexpressing (GFP) alveolar cells within their respective parent populations (all GFP+ LF, hatched bars, axes on right; or GFP LF, open bars, axes on left) at P8 were compared with those at P12 for Sox9 or MRTF-A in E or F, respectively. *P < 0.05, mean ± SE, 3-way ANOVA, Student-Newman-Keuls post hoc test.

Lipid-Laden, PDGFRα-GFPlow LF Diminish After P8

We next examined whether PDGFRα gene expression regressed after secondary septation (which is maximal before P15) and whether the kinetics of regression vary with the level of PDGFRα gene expression. For these studies, we marked myofibroblasts as those that express tagln (tagln-Cre activated tdTomato), and cells containing lipid droplets were marked with a blue fluorescent lipophilic dye. GFP levels serve as a surrogate for PDGFRα expression. Lipid droplets were not observed in cells that contained tagln-dTomato (Fig. 9A), although they were observed in GFP alveolar cells that did not express tagln-dTomato (data not shown). The tagln-dTomato, GFP cells were excluded from analysis because this population likely contains alveolar type II cells in addition to LF. A higher proportion of alveolar cells that express both tagln-dTomato and PDGFRα-GFPhigh was observed at P8 (Fig. 9B), but this proportion decreased with age. Stereological analysis of tagln-dTomato, PDGFRα-GFPhigh (GFPhigh,dTom+) cells demonstrated that at P8 83 ± 13% (mean ± SE, n = 4) of the cells were positioned nearer the alveolar duct, i.e., within the proximal 40% of the distance extending from the duct to the alveolar base. Whereas a large majority (∼4.5-fold more) of GFPlow LF contained lipid droplets at P8, GFPhigh LF accounted for most of the lipid storage in GFPhigh LF at P15 and older (Fig. 9C). Similar age-related relationships were observed when the data were expressed relative to lung volume as determined by displacement (data not shown). Therefore, PDGFRα and tagln are only transiently coexpressed when septation is most active, and the GFPlow lipid-laden cells are nearly completely absent at P15 when septation starts to diminish.

Fig. 9.

Fig. 9.

Lipid droplets and transgelin (TG) are observed in different LF subpopulations. Lungs from four TG Cre+/−;Rosa26dTomato (Tom)+/−;PDGFRαGFP+ mice at various postnatal days or adults were stained with BODIPY 665/670 (to visualize neutral lipids) and imaged using confocal microscopy (A). Alveolar cells containing dTomato (dTom+) and/or GFP were enumerated and expressed/mm3 of lung tissue. B: only GFP-bright (high, Hi) cells are shown because no GFP-dim (low) cells also contained dTom (expressed transgelin). C: only cells not expressing transgelin (dTom) contained BODIPY 665/676-staining lipid droplets (LD). LD+ cells were distinguished and enumerated based on the intensity of GFP fluorescence (bright, Hi or dim, low). *P < 0.05, mean ± SE, n = 4 at each age (**P < 0.01), 1-way ANOVA for age, Student-Newman-Keuls post hoc test comparing P8 with older mice within each determination (shown by same symbol).

Alveolar Surface Area is Decreased and Compliance is Increased in TGCre+/−;PDGFRαF/F Mice

Some TGCre+/−;PDGFRαF/F mice with a less severe phenotype survived to adulthood. Their lungs showed (Fig. 10) a reduction in alveoli and alveolar ducts (A and B) and significantly greater static compliance (C). Elastic fibers (shown in the insets) were diminished. TGCre+/−;PDGFRαF/F mice had a higher pressure-volume curve shape constant, lower dynamic elastance, increased hysteresivity (η = G/H), and a smaller alveolar surface area compared with control lungs (Table 1). These findings are consistent with a loss of α-SMA and elastin-producing myofibroblasts.

Table 1.

Lung mechanics and gas exchange surface area in control and TGCre+/−;PDGFRαF/F mice

Impedance Oscillometry
Static PV Curve
Rn, cmH2O·s·ml−1 G, cmH2O/ml H, cmH2O/ml η K/cmH2O Cst, ml/cmH2O Alveolar Surface Area, cm2
Control 0.202 ± 0.016 4.504 ± 0.183 23.283 ± 1.688 0.198 ± 0.011 0.139 ± 0.003 0.097 ± 0.008 308.0 ± 12.8
TGCre+/−; PDGFRαF/F 0.327 ± 0.116 4.153 ± 0.739 11.732 ± 1.146** 0.344 ± 0.018** 0.196 ± 0.017* 0.128 ± 0.002** 101.1 ± 4.8**

Data are means ± SE; n =5 mice in each group except for surface area where n = 3.

Rn, Newtonian resistance; G, tissue damping; H, tissue elastance; η, hysteresivity; K, shape constant for pressure-volume curve; Cst, static compliance; PV, pressure/volume.

**

P < 0.01 and

*

P < 0.05, unpaired t-test, Bonferroni correction for multiple comparisons.

DISCUSSION

LF Bear Markers Characteristic Of Mesenchymal Progenitors

Current therapies for pulmonary emphysema or fibrosis reduce complications but do not allay the progression of these diseases, which involve alveolar destruction. Alveolar regeneration, activated to repair damaged alveoli and to restore the gas exchange surface area, requires restoration and repopulation of resident cells and/or an influx of circulating stem cells. During alveolar development, contractile interstitial fibroblasts control air entry and support the thin capillary-epithelial interface of alveoli. To explore how resident progenitors could be harnessed to restore alveoli, we studied how fibroblast subpopulations cooperate during alveolar development and have summarized their contrasting properties in Table 2. In other tissues (adipose, bone, heart, and skeletal muscle), CD45 bi- or oligopotent PDGFRα+ mesenchymal cells assume adipogenic, myogenic, or osteogenic phenotypes and display markers of mesenchymal progenitor cells (Sca1, CD34, CD166) (25, 42, 58). Using mice that express GFP under the control of the PDGFRα promoter, we examined mesenchymal progenitor cell markers as well as the divergent proliferative and differentiation capabilities in PDGFRα-expressing and -nonexpressing LF in neonatal mice. As others have observed in lungs from adult mice, Sca1+ LF were identified in GFP LF not expressing pdgfrα, as well as in GFP+ LF (Fig. 3C) (38). Within the Sca1+ population, Dlk1+ and Sox9+ were more abundant among PDGFRα-GFPhigh myofibroblasts (Fig. 5), consistent with their involvement in both skeletal and smooth muscle programming and differentiation (2, 58). A larger proportion of PDGFR-expressing than -nonexpressing LF exhibited the mesenchymal progenitor marker CD34 (Fig. 3E), and this fraction diminished when pdgfrα was deleted (Fig. 6B) (49). Although pdgfrα deletion did not reduce Sca1+ LF, the proportion of Sox9 + LF decreased, whereas cell surface-associated Dlk1+ LF increased within the Sca1+ population (Fig. 6, A and D). This differential effect suggests that downstream effectors such as Sox9 could be targeted to fine tune the regenerative function of fibroblasts.

Table 2.

Comparison of interstitial mesenchymal cells bearing markers for progenitors or particular differentiated phenotypes

Cell Type PDGFR Lipid (LTR, ADRP) α-SMA, Tagln Progenitor Markers Proliferation Quiescence Regulate Differentiation
Lipofibroblast α-Low High α-SMA: 45 Sca1: 4 Ki67: 15 Dlk1: 5
CD34: 90 CDKN1c: 50 Sox9: 50
Tagln: neg CD166: 65 G0S2: 25 Tcf21: 25
Myofibroblast α-High Low or absent α-SMA: 90 Sca1: 10 Ki67: 30 Dlk1: 5
CD34: 90 CDKN1c: 10 Sox9: 50
Tagln: pos CD166: 5 G0S2: 5 Tcf21: 5
Pericyte α-Neg., β+ NE Tagln pos NE NE NE

Nos. are approximate mean percentages of cells bearing specified antigens within a particular cell type at postnatal day 8.

Lipofibroblast, lipid interstitial fibroblast; LTR, LipidTox red; ADRP, adipocyte differentiation related peptide; α-SMA, α-smooth muscle actin; Tagln, transgelin; CDKN1c, p57kip2; G0S2, G0-G1 switch protein 2; Dlk1, delta-like protein-1; Sox9, sex-determining region Y box 9; NE, not examined; neg, negative; pos, positive.

Whereas Sca1+ cells were less abundant in GFPlow compared with GFP-negative LF (Fig. 3D), significantly more GFPlow cells exhibited CD166 expression (Fig. 3F). A larger fraction of the GFPlow than either GFP or GFPhigh LF contained lipid droplets (Fig. 3A). This differs from the findings of McQualter and associates who observed that CD166-negative lung stromal cells were more likely to acquire lipid droplets in culture (39). Because both the level of CD166 expression and the phenotype change with culture of mesenchymal cells, our freshly isolated LF are not equivalent to cultured stromal cells from adult lungs, although cells from both sources were CD34+ and CD166+ (Fig. 3, E and F) (59). We found that LF bearing the progenitor markers Sca1 and CD34 reside within both the lipid-laden and myofibroblastic subpopulations, which is similar to others' observations in the adult lung and other organs (21, 49, 58).

Tcf21 (Pod-1) is required during branching morphogenesis, but little is known about tcf21 gene expression during alveolar septation (44). In response to cardiac stress, Tcf21+ mesenchymal cells migrate from the epicardium into the myocardium, where resident cardiac fibroblasts mount fibrotic reactions (6, 52). We found that tcf21 gene expression was greater in PDGFRα-low, lipid-laden LF than in PDGFRα-high myofibroblastic LF (Fig. 4). Similarly, other investigators observed that PDGFRα expression is required to sustain epicardial fibroblasts and adipocytes from white fat, but not coronary artery smooth muscle cells or brown adipocytes (1, 52, 55).

The proliferation marker Ki-67 was lower, whereas the growth arrest proteins G0S2 and p57kip2 were higher, in PDGFRα-low than in PDGFRα-high LF, suggesting that the lipid-laden population becomes quiescent (Fig. 4). G0S2 is expressed in the lung and is abundant in adipose cells, and its expression increases during differentiation from preadipocytes to mature adipocytes. In these mature cells, G0S2 inhibits adipose triglyceride lipase and controls the size of cytoplasmic lipid droplets (11, 19, 20). Others have reported that the expression of G0S2 is diminished at P8 in lungs from mice bearing a compound deletion of both FGFR3 and FGFR4, which exhibit defective alveolarization (54). Concordant with our observation, growth arrest in PDGFRα-expressing oligodendrocyte precursors, smooth muscle cells, and cardiomyocytes is also regulated by p57kip2 (CDKN1c) (22, 24).

CD166+, Dlk1+ LF are Maintained When PDGFRα is Reduced

CD166, also termed activated leukocyte adhesion molecule, has been identified on adipocyte-derived mesenchymal stem cells, fetal lung cells, bronchial fibroblasts, and adipocyte or chondrocyte progenitors, particularly during proliferation and migration (15, 47). When pdgfrα was deleted, Dlk1-positive, CD166-positive, and Dlk1,CD166 double-positive LF were more prevalent within the Sca1+ or CD34+ subpopulations (Fig. 6, A and B). Others have shown that CD166 is predominantly found in progenitor populations and that Dlk1 can suppress differentiation, enabling Dlk1+ progenitors to accumulate (10, 56). Taken with our observations in pdgfrα-deficient mice, it is possible that CD166 and Dlk1 work in concert to regulate differentiation.

Dlk1 gene expression and function are regulated in a complex fashion variously involving paternal imprinting, alternative mRNA splicing, and extracellular protein cleavage (56). Thereby, the protein assumes divergent characteristics during development that have been best characterized in adipocytes and myocytes (2). Most notably, the two major splice variants termed here dlk1-A and dlk1-C involve inclusion (dlk1-A) or exclusion (dlk1-C) of a juxtamembrane extracellular TACE cleavage site, releasing a soluble peptide (Dlk1S) that increases preadipocyte proliferation (41). In contrast, the uncleaved membrane-associated form (Dlk1M) suppresses proliferation. Comparably in myocytes, the Dlk1S form inhibits, whereas the Dlk1M form promotes, myocyte fusion and myotube formation, and Dlk1-C mRNA and Dlk1M predominate during postnatal muscle growth in the mouse (48). This complex regulation contributes to the paradoxical observation that Dlk1 promotes myogenesis during embryogenesis but inhibits myogenic repair in adult mice (2). To our knowledge, our findings using qRT-PCR are the first demonstration that PDGFRα influences alternative splicing of Dlk1 during development.

The PDGFRα-high population contained more α-SMA+ LF than the PDGFR-low population (Fig. 3C). Alveolar cells containing both PDGFRα- and tagln-driven dTomato localized near the alveolar entry rings, indicating that tagln-mediated PDGFRα deletion targets the distal septal tips where α-SMA is most abundant (32). In contrast, LF bearing markers of the PDGFRα-low subpopulation (LTR+, CD166+, and Tcf21+) were preserved. Within Sca1+ LF, the proportion of Sox9+ LF diminished, whereas Dlk+ LF increased, suggesting that PDGFRα deletion has a differential effect on LF populations bearing Sox9 or Dlk1. Observation of diminished postn and increased pparγ mRNA in the PDGFRα-deficient LF further supports a shift from the myofibroblastic toward a lipid storage phenotype.

Sox9 is Involved in the Differentiation of PDGFRα-Expressing LF to Myofibroblasts

Our novel finding that pdgfrα and sox9 are coexpressed in alveolar myofibroblast progenitors parallels observations made in OPC and epicardial fibroblast precursors. PDGFRα and the SoxE proteins, Sox9 and/or Sox10, mark a population of NG2 chondroitin sulfate protein-bearing OPC in the periventricular zone of the brain and the spinal cord and regulate their progenitor state and migration (23). It remains unclear whether the Sox9 and PDGFRα coincidently mark the same cellular population or whether they interact to enhance proliferation and migration (14). PDGFRα is required to maintain Sox9 gene expression in embryonic epicardial cells, for migration into the myocardium, and for differentiation (52). Forced expression of Sox9 enables epicardial cells to overcome defects in migration and differentiation that are acquired after pdgfrα gene deletion (52). These data suggest that Sox9 acts downstream from PDGFRα.

Consistent with findings in cardiac fibroblasts and OPC, we observed that Sox9 was more abundant in Sca1+ GFP+ LF when PDGFRα gene expression was higher (Fig. 5B) (14, 52). PDGF-A, which exclusively signals through PDGFRα, increased Sox9 mRNA and protein levels, suggesting a pretranslational effect (Fig. 7, A and B). Important PDGFRα downstream signaling mediators include PI3K-Akt, p65Rel A, and MEK1/2-Erk, which have all been shown to regulate Sox9 gene expression (29). In addition, we observed a decrease in the numbers of PDGFRα-GFP+ alveolar cells containing Sox9 and a reciprocal increase in cells containing MRTF-A from P8 to P12 (Fig. 8), which is accompanied by an increase in α-SMA (32). There is a similar reciprocal relationship between Sox9 and myocardin in smooth muscle cells. Sox9 suspends smooth muscle cell differentiation by complexing with serum response factor (SRF) and interfering with the binding of SRF to CarG box elements or by complexing with the transcription factor TSHZ3 (31, 63).

PDGFRα and Modulation of LF Phenotypes During Alveolar Restoration

We observed that the lipid-storing PDGFRα-low population was abundant in neonates but virtually absent in adults where PDGFRα-high lipid-laden LF predominate (Fig. 9) (27, 32). However, following pneumonectomy in adult PDGFRα-GFP mice, Chen and associates observed that α-SMA was more abundant in the PDGFRα (GFP)-low subpopulation (9). One possible explanation is that pneumonectomy restores the PDGFRα-low LF subpopulation, which in that context contains α-SMA and participates in alveolar regeneration.

Our observations about the divergent characteristics of PDGFRα-expressing LF have potential therapeutic relevance to human diseases. In bronchopulmonary dysplasia, the transition from LF to myofibroblasts results in thickened alveolar septa and reduced oxygen diffusion in the alveolar walls via increased collagen deposition. Current salutary therapies, including positive pressure ventilation and supplemental oxygen, may further exacerbate these alveolar structural abnormalities. However, the PPARγ agonist rosiglitazone improves alveolar formation and reduces collagen (62). These findings suggest that directing fibroblasts from a myofibroblast to a lipid storage phenotype may normalize alveolar architecture during bronchopulmonary dysplasia. A similar strategy may benefit adults with pulmonary fibrosis based on data in cultured human lung fibroblasts in which pioglitazone, another PPARγ agonist, reduces collagen content and bleomycin-induced fibrosis (40). Focusing on specific targets of PDGF-A signaling, such as Sox9, may help select a restorative phenotype that avoids excessive fibrosis.

GRANTS

This research was supported by Merit Review funding from the Department of Veterans Affairs Research Service. Flow cytometry was performed at the Flow Cytometry Facility, which is a Carver College of Medicine/Holden Comprehensive Cancer Center core research facility at the University of Iowa. The Facility is funded through user fees and the generous financial support of the Carver College of Medicine, Holden Comprehensive Cancer Center, and Iowa City Veteran's Administration Medical Center. The Aria flow cytometer was funded by a grant from the National Center for Research Resources of the National Institutes of Health under Award No. 1S10-RR-27219.

DISCLOSURES

No conflicts of interest, financial or otherwise are declared by the authors.

AUTHOR CONTRIBUTIONS

S.E.M. conception and design of research; S.E.M. and D.M.M. performed experiments; S.E.M. and D.M.M. analyzed data; S.E.M. interpreted results of experiments; S.E.M. prepared figures; S.E.M. drafted manuscript; S.E.M. and D.M.M. edited and revised manuscript; S.E.M. and D.M.M. approved final version of manuscript.

ACKNOWLEDGMENTS

We are grateful to Kristina W. Thiel for valuable assistance in revising and preparing the manuscript for publication.

REFERENCES

  • 1.Acharya A, Baek ST, Huang G, Eskiocak B, Goetsch S, Sung CY, Banfi S, Sauer MF, Olsen GS, Duffield JS, Olson EN, Tallquist MD. The bHLH transcription factor Tcf21 is required for lineage-specific EMT of cardiac fibroblast progenitors. Development 139: 2139–2149, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Andersen DC, Laborda J, Baladron V, Kassem M, Sheikh SP, Jensen CH. Dual role of delta-like 1 homolog (DLK1) in skeletal muscle development and adult muscle regeneration. Development 140: 3743–3753, 2013 [DOI] [PubMed] [Google Scholar]
  • 3.Berry R, Rodeheffer MS. Characterization of the adipocyte cellular lineage in vivo. Nat Cell Biol 15: 302–308, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Boström H, Gritli-Linde A, Betsholtz C. PDGF-A/PDGF alpha-receptor signaling is required for lung growth and the formation of alveoli but not for early lung branching morphogenesis. Dev Dynam 223: 155–162, 2002 [DOI] [PubMed] [Google Scholar]
  • 5.Boström H, Willetts K, Pekny M, Leveen P, Lindahl P, Hedstrand H, Pekna M, Hellstrom M, Gebre-Medin S, Schalling M, Nilsson M, Kurland S, Tornell J, Heath JK, Betsholtz C. PGDF-A signaling is a critical event in lung alveolar myofibroblast development and alveogenesis. Cell 85: 863–873, 1996 [DOI] [PubMed] [Google Scholar]
  • 6.Braitsch CM, Kanisicak O, van Berlo JH, Molkentin JD, Yutzey KE. Differential expression of embryonic epicardial progenitor markers and localization of cardiac fibrosis in adult ischemic injury and hypertensive heart disease. J Mol Cell Cardiol 65: 108–119, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Carter CS, Vogel TW, Zhang Q, Seo S, Swiderski RE, Moninger TO, Cassell MD, Thedens DR, Keppler-Noreuil KM, Nopoulos P, Nishimura DY, Searby CC, Bugge K, Sheffield VC. Abnormal development of NG2+PDGFR-alpha+ neural progenitor cells leads to neonatal hydrocephalus in a ciliopathy mouse model. Nat Med 18: 1797–1804, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Chang DR, Martinez AD, Miller RK, Ji H, Akiyama H, McCrea PD, Chen J. Lung epithelial branching program antagonizes alveolar differentiation. Proc Natl Acad Sci USA 110: 18042–18051, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Chen L, Acciani T, Le Cras T, Lutzko C, Perl AKT. Dynamic regulation of platelet-derived growth factor receptor α expression in alveolar fibroblasts during realveolarization. Am J Respir Cell Mol Biol 47: 517–527, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Chitteti BR, Cheng YH, Kacena MA, Srour EF. Hierarchical organization of osteoblasts reveals the significant role of CD166 in hematopoietic stem cell maintenance and function. Bone 54: 58–67, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Choi H, Lee H, Kim TH, Kim HJ, Lee YJ, Lee SJ, Yu JH, Kim D, Kim KS, Park SW, Kim JW. G0/G1 switch gene 2 has a critical role in adipocyte differentiation. Cell Death Differ 21: 1071–1080, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Dy P, Bhattaram P, Wang Q, Wang L, Ballock RT, Lefebvre V. Sox9 directs hypertrophic maturation and blocks osteoblast differentiation of growth plate. Dev Cell 22: 597–609, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Feng H, Hu B, Liu KW, Li Y, Lu X, Cheng T, Yiin JJ, Lu S, Keezer S, Fenton T, Furnari FB, Hamilton RL, Vuori K, Sarkaria JN, Nagane M, Nishikawa R, Cavenee WK, Cheng SY. Activation of Rac1 by Src-dependent phosphorylation of Dock180(Y1811) mediates PDGFRalpha-stimulated glioma tumorigenesis in mice and humans. J Clin Invest 121: 4670–4684, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Finzsch M, Stolt CC, Lommes P, Wegner M. Sox9 and Sox10 influence survival and migration of oligodendrocyte precursors in the spinal cord by regulating PDGF receptor alpha expression. Development 135: 637–646, 2008 [DOI] [PubMed] [Google Scholar]
  • 15.Gilsanz A, Sanchez-Martin L, Gutierrez-Lopez MD, Ovalle S, Machado-Pineda Y, Reyes R, Swart GW, Figdor CG, Lafuente EM, Cabanas C. ALCAM/CD166 adhesive function is regulated by the tetraspanin CD9. Cell Mol Life Sci 70: 475–493, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Hamilton TG, Klinghoffer RA, Corrin PD, Soriano P. Evolutionary divergence of platelet derived growth factor alpha receptor signaling mechanisms. Mol Cell Biol 23: 4013–4025, 2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Hantos Z, Daroczy B, Suki B, Nagy S. Low-frequency respiratory mechanical impedance in the rat. J Appl Physiol 63: 36–43, 1987 [DOI] [PubMed] [Google Scholar]
  • 18.Hayes BJ, Riehle KJ, Shimizu-Albergine M, Bauer RL, Hudkins KL, Johansson F, Yeh MM, Mahoney WM, Jr, Yeung RS, Campbell JS. Activation of platelet-derived growth factor receptor alpha contributes to liver fibrosis. PloS One 9: e92925, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Heckmann BL, Zhang X, Xie X, Saarinen A, Lu X, Yang X, Liu J. Defective adipose lipolysis and altered global energy metabolism in mice with adipose overexpression of the lipolytic inhibitor G0/G1 switch gene 2 (G0S2). J Biol Chem 289: 1905–1916, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Heckmann BL, Zhang Z, Xie X, Liu J. TheG0/G1 switch gene 2 (G0S2): regulating metabolism and beyond. Biochem Biophys Acta 1831: 276–281, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hittinger M, Czyz ZT, Huesemann Y, Maneck M, Botteron C, Kaeufl S, Klein CA, Polzer B. Molecular profiling of single Sca1+/CD34+,− Cells-the putative murine lung stem cells. PloS One 8: e83917, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Huang J, Elicker J, Bowens N, Liu X, Cheng L, Cappola TP, Zhu X, Parmacek MS. Myocardin regulates BMP10 expression and is required for heart development. J Clin Invest 122: 3678–3691, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Hughes EG, Kang SH, Fukaya M, Bergles DE. Oligodendrocyte progenitors balance growth with self-repulsion to achieve homeostasis in the adult brain. Nat Neurosci 16: 668–676, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Jadasz JJ, Rivera FJ, Taubert A, Kandasamy M, Sandner B, Weidner N, Aktas O, Hartung HP, Aigner L, Kury P. p57kip2 regulates glial fate decision in adult neural stem cells. Development 139: 3306–3315, 2012 [DOI] [PubMed] [Google Scholar]
  • 25.Joe AWB, Yi L, Natarajan A, Le Grand F, So L, Wang J, Rudnicki MA, Rossi FMV. Muscle injury activates resident fibro/adipogenic progenitors that facilitate myogenesis. Nat Cell Biol 12: 153–165, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Joses AV, Cross NCP. Oncogenic derivatives of platelet-derived growth factor receptors. Cell Mol Life Sci 61: 2912–2923, 2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Kimani PW, Holmes AJ, Grossmann RE, McGowan SE. PDGF-Rα gene expression predicts proliferation, but PDGF-A suppresses transdifferentiation of neonatal mouse lung myofibroblasts (Abstract). Respir Res 10: 19, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Klinghoffer RA, Hamilton TG, Hoch R, Soriano P. An allelic series at the PDGFalphaR locus indicates unequal contributions of distinct signaling pathways during development. Dev Cell 2: 103–113, 2002 [DOI] [PubMed] [Google Scholar]
  • 29.Ling S, Chang X, Schultz L, Lee TK, Chaux A, Marchionni L, Netto GJ, Sidransky D, Berman DM. An EGFR-ERK-SOX9 signaling cascade links urothelial development and regeneration to cancer. Cancer Res 71: 3812–3821, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time PCR and the 2(-DeltaDeltaC(T)) method. Methods 25: 402–408, 2001 [DOI] [PubMed] [Google Scholar]
  • 31.Martin E, Caubit X, Airik R, Vola C, Fatmi A, Kispert A, Fasano L. TSHZ3 and SOX9 regulate the timing of smooth muscle cell differentiation in the ureter by reducing myocardin activity. PloS One 8: e63721, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.McGowan SE, Grossmann RE, Kimani PW, Holmes AJ. Platelet-derived growth factor receptor-alpha-expressing cells localize to the alveolar entry ring and have characteristics of myofibroblasts during pulmonary alveolar septal formation. Anat Rec 291: 1649–1661, 2008 [DOI] [PubMed] [Google Scholar]
  • 33.McGowan SE, Holmes AJ. Vitamin A deficiency alters pulmonary parenchymal collagen and tissue mechanics. Respir Physiol Neurobiol 156: 312–319, 2007 [DOI] [PubMed] [Google Scholar]
  • 34.McGowan SE, Jackson SK, Doro MM, Olson PJ. Peroxisome proliferators alter lipid acquisition and elastin gene expression in neonatal rat lung fibroblasts. Am J Physiol Lung Cell Mol Physiol 273: L1249–L1257, 1997 [DOI] [PubMed] [Google Scholar]
  • 35.McGowan SE, McCoy DM. Fibroblasts expressing PDGF-receptor-alpha diminish during alveolar septal thinning in mice. Pediatr Res 70: 44–49, 2011 [DOI] [PubMed] [Google Scholar]
  • 36.McGowan SE, McCoy DM. Platelet-derived growth factor-A and sonic hedgehog signaling direct lung fibroblast precursors during alveolar septal formation. Am J Physiol Lung Cell Mol Physiol 305: L229–L239, 2013 [DOI] [PubMed] [Google Scholar]
  • 37.McGowan SE, McCoy DM. Platelet-derived growth factor-A regulates lung fibroblast S-phase entry through p27(kip1) and FoxO3a. Respir Res 14: 68, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.McQualter JL, Brouard N, Williams B, Baird BN, Sims-Lucas S, Yuen K, Nilsson SK, Simmons PJ, Bertoncello I. Endogenous fibroblastic progenitor cells in the adult mouse lung are highly enriched in the Sca-1 positive cell fraction. Stem Cells 27: 623–633, 2009 [DOI] [PubMed] [Google Scholar]
  • 39.McQualter JL, McCarty RC, Van der Velden J, O'Donoghue RJ, Asselin-Labat ML, Bozinovski S, Bertoncello I. TGF-β signaling in stromal cells acts upstream of FGF-10 to regulate epithelial stem cell growth in adult lung. Stem Cell Res 11: 1222–1233, 2013 [DOI] [PubMed] [Google Scholar]
  • 40.Milam JE, Keshamouni VG, Phan SH, Hu B, Gangireddy SR, Hogaboam CM, Standiford TJ, Thannickal vJ, Reddy RC. PPAR-γ agonists inhibit profibrotic phenotypes in human lung fibroblasts and bleomycin-induced pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 294: L891–L901, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Mortensen SB, Jensen CH, Schneider M, Thomassen M, Kruse TA, Laborda J, Sheikh SP, Andersen DC. Membrane-tethered delta-like 1 homolog (DLK1) restricts adipose tissue size by inhibiting preadipocyte proliferation. Diabetes 61: 2814–2822, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Pannerec A, Formicola L, Besson V, Marazzi G, Sassoon DA. Defining skeletal muscle resident progenitors and their cell fate potentials. Development 140: 2879–2891, 2013 [DOI] [PubMed] [Google Scholar]
  • 43.Pelczar P, Zibat A, van Dop WA, Heijmans J, Bleckmann A, Gruber W, Nitzki F, Uhmann A, Guijarro MV, Hernando E, Dittmann K, Wienands J, Dressel R, Wojnowski L, Binder C, Taguchi T, Beissbarth T, Hogendoorn PC, Antonescu CR, Rubin BP, Schulz-Schaeffer W, Aberger F, van den Brink GR, Hahn H. Inactivation of Patched1 in mice leads to development of gastrointestinal stromal-like tumors that express Pdgfralpha but not kit. Gastroenterology 144: 134–144, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Quaggin SE, Schwartz L, Cui S, Igarashi P, Deimling J, Post M, Rossant J. The basic-helix-loop-helix protein pod1 is critically important for kidney and lung organogenesis. Development 126: 5771–5783, 1999 [DOI] [PubMed] [Google Scholar]
  • 45.Rockich BE, Hrycaj SM, Shih HP, Nagy MS, Ferguson MA, Kopp JL, Sander M, Wellik DM, Spence JR. Sox9 plays multiple roles in the lung epithelium during branching morphogenesis. Proc Natl Acad Sci USA 110: E4456–E4464, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Rosner B. Fundamentals of Biostatistics. Boston, MA: PWS-Kent Publishing, 1990 [Google Scholar]
  • 47.Sabatini F, Petecchia L, Tavian M, Jodon de Villeroché V, Rossi GA, Brouty-Boye D. Human bronchial fibroblasts exhibit a mesenchymal stem cell phenotype and multilineage differentiating potentialities. Lab Invest 85: 962–971, 2005 [DOI] [PubMed] [Google Scholar]
  • 48.Shin S, Suh Y, Zerby HN, Lee K. Membrane-bound delta-like 1 homolog (Dlk1) promotes while soluble Dlk1 inhibits myogenesis in C2C12 cells. FEBS Lett 588: 1100–1108, 2014 [DOI] [PubMed] [Google Scholar]
  • 49.Sidney LE, Branch MJ, Dunphy SE, Dua HS, Hopkinson A. Evidence for CD34 as a common marker for diverse progenitors. Stem Cells 32: 1380–1389, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Sim FJ, McClain CR, Schanz SJ, Protack TL, Windrem MS, Goldman SA. CD140a identifies a population of highly myelinogenic, migration-competent and efficiently engrafting human oligodendrocyte progenitor cells. Nat Biotechnol 29: 934–941, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Small EN. The actin-MRTF-SRF gene regulatory axis in myofibroblast differentiation. J Cardiovasc Trans Res 5: 794–804, 2012 [DOI] [PubMed] [Google Scholar]
  • 52.Smith CL, Baek ST, Sung CY, Tallquist MD. Epicardial-derived cell epithelial-to-mesenchymal transition and fate specification require PDGF receptor signaling. Circ Res 108: e15–e26, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Snider P, Standley KN, Wang J, Azhar M, Doetschman T, Conway SJ. Origin of cardiac fibroblasts and the role of periostin. Circ Res 105: 934–947, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Srisuma S, Bhattacharya S, Simon DM, Solleti SK, Tyagi S, Starcher B, Mariani TJ. Fibroblast growth factor receptors control epithelial-mesenchymal interactions necessary for alveolar elastogenesis. Am J Respir Crit Care Med 181: 838–850, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Timmons JA, Wennmalm K, Larsson O, Walden TB, Lassmann T, Petrovic N, Hamilton DL, Gimeno RE, Wahlestedt C, Baar K, Nedergaard J, Cannon B. Myogenic gene expression signature establishes that brown and white adipocytes originate from distinct cell lineages. Proc Natl Acad Sci USA 104: 4401–4406, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Traustadottir GA, Kosmina R, Sheikh SP, Jensen CH, Andersen DC. Preadipocytes proliferate and differentiate under the guidance of Delta-like 1 homolog (DLK1). Adipocyte 2: 272–275, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Turcatel G, Rubin N, Menke DB, Martin G, Shi W, Warburton D. Lung mesenchymal expression of Sox9 plays a critical role in tracheal development. BMC Biol 11: 117, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Uezumi A, Ito T, Morikawa D, Shimizu N, Yoneda T, Segawa M, Yamaguchi M, Ogawa R, Matev MM, Miyagoe-Suzuki Y, Takeda S, Tsujikawa K, Tsuchida K, Yamamoto H, Fukada S. Fibrosis and adipogenesis originate from a common mesenchymal progenitor in skeletal muscle. J Cell Sci 124: 3654–3664, 2011 [DOI] [PubMed] [Google Scholar]
  • 59.Ullah M, Eucker J, Sittinger M, Ringe J. Mesenchymal stem cells and their chondrogenic differentiated and dedifferentiated progeny express chemokine receptor CCR9 and chemotactically migrate toward CCL25 or serum. Stem Cell Res Ther 4: 99, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Vaccaro C, Brody JS. Ultrastructure of developing alveoli. I. The role of the interstitial fibroblast. Anat Rec 192: 467–480, 1978 [DOI] [PubMed] [Google Scholar]
  • 61.Varisco BM, Ambalavanan N, Whitsett JA, Hagood JS. Thy-1 signals through PPARγ to promote lipofibroblast differentiation in the developing lung. Am J Respir Cell Mol Biol 46: 765–772, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Wang Y, Santos R, Sakurai R, Shin E, Cerny L, Torday JS, Rehan VK. Peroxisome proliferator-activated receptor γ agonists enhance lung maturation in a neonatal rat model. Pediatr Res 65: 150–155, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Xu Z, Ji G, Shen J, Wang X, Zhou J, Li L. SOX9 and myocardin counteract each other in regulating vascular smooth muscle cell differentiation. Biochem Biophys Res Commun 422: 285–290, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Zhang J, Zhong W, Cui T, Yang M, Hu X, Xu K, Xie C, Xue C, Gibbons GH, Liu C, Li L, Chen YE. Generation of an adult smooth muscle cell-targeted Cre recombinase mouse model. Arterioscler Thromb Vasc Biol 26: e23–e24, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Zhu NL, Asahina K, Wang J, Ueno A, Lazaro R, Miyaoka Y, Miyajima A, Tsukamoto H. Hepatic stellate cell-derived delta-like homolog 1 (DLK1) protein in liver regeneration. J Biol Chem 287: 10355–10367, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]

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

RESOURCES