lung epithelial branching program antagonizes alveolar ... · lung epithelial branching program...

10
Lung epithelial branching program antagonizes alveolar differentiation Daniel R. Chang a , Denise Martinez Alanis a , Rachel K. Miller b,1 , Hong Ji b , Haruhiko Akiyama c , Pierre D. McCrea b , and Jichao Chen a,b,2 a Department of Pulmonary Medicine and b Department of Biochemistry and Molecular Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030; and c Department of Orthopedics, Kyoto University, Sakyo, Kyoto 606-8507, Japan This Feature Article is part of a series identied by the Editorial Board as reporting ndings of exceptional signicance. Edited by Brigid L. M. Hogan, Duke University Medical Center, Durham, NC, and approved August 28, 2013 (received for review June 21, 2013) Mammalian organs, including the lung and kidney, often adopt a branched structure to achieve high efciency and capacity of their physiological functions. Formation of a functional lung requires two developmental processes: branching morphogenesis, which builds a tree-like tubular network, and alveolar differenti- ation, which generates specialized epithelial cells for gas ex- change. Much progress has been made to understand each of the two processes individually; however, it is not clear whether the two processes are coordinated and how they are deployed at the correct time and location. Here we show that an epithelial branching morphogenesis program antagonizes alveolar differen- tiation in the mouse lung. We nd a negative correlation between branching morphogenesis and alveolar differentiation temporally, spatially, and evolutionarily. Gain-of-function experiments show that hyperactive small GTPase Kras expands the branching pro- gram and also suppresses molecular and cellular differentiation of alveolar cells. Loss-of-function experiments show that SRY- box containing gene 9 (Sox9) functions downstream of Fibroblast growth factor (Fgf)/Kras to promote branching and also sup- presses premature initiation of alveolar differentiation. We thus propose that lung epithelial progenitors continuously balance be- tween branching morphogenesis and alveolar differentiation, and such a balance is mediated by dual-function regulators, including Kras and Sox9. The resulting temporal delay of differentiation by the branching program may provide new insights to lung imma- turity in preterm neonates and the increase in organ complexity during evolution. A n optimal organ design maximizes not only the number of its basic functional units, for example alveoli in the lung and glomeruli in the kidney, but also the efciency of transporting gas and liquid to and from these functional units. This design is often achieved by building a branched, tree-like structure with functional units attached at the end, as seen in the lungs, kidneys, and mammary glands. Compared with a beads-on-a-string de- sign, where all functional units are connected sequentially on a linear duct, a branching design not only packs the maximal number of functional units within a given 3D organ space with- out entanglement of the ductal system, but also increases transport efciency by minimizing the average distance between each functional unit to the entry point of an organ. Two developmental processes underlie a branching design: branching morphogenesis, a process that builds the branched ducts, and cellular differentiation, a process that builds the functional units. These two processes have to be somehow coordinated to ensure a fundamental feature of a branching design: all functional units are only located at the termini of the branched ducts. Branching morphogenesis has been well studied both mor- phologically and genetically in the mouse lung. The entire airway lumen is bordered by epithelial cells that originate from the ventral foregut around embryonic day 10 (E10) (1). It has been shown that epithelial progenitors undergo a highly organized process of branching morphogenesis to give rise to the rst 5,000 or so airway branches within 5 d of development (E10 to E15) (2). Branching morphogenesis involves reciprocal interactions between the lung epithelium and the surrounding mesenchyme, and requires secretion of a broblast growth factor (FGF10) by the mesenchymal cells, activating its membrane receptor (FGFR2) and subsequently a small GTPase (KRAS) in the epithelial cells (35). Additional signaling pathways, including Wnt , Sonic hedgehog (Shh), and Tgf signalings, are also essential for normal branching (1, 6). Lung epithelial progenitors not only build a branched duct system via branching morphogenesis, but also differentiate into specialized alveolar cells required for gas exchange, a process named alveolar differentiation. There are two alveolar cell types: type I cells that are at and cover more than 90% of the alveolar surface, across which gases diffuse; and type II cells that are cuboidal and synthesize pulmonary surfactants, lipoprotein complexes that hydrate the alveolar surface and prevent alveolar collapsing by reducing surface tension (7). Genetic studies in mice have identied several transcription regulators specically required for alveolar differentiation (812). Although both branching morphogenesis and alveolar differ- entiation have been extensively studied, much less is known about whether and how they are coordinated. Each process is generally viewed as independently controlled and occurring during early versus late lung development, respectively. Al- though defects in alveolar differentiation are not expected to Signicance Mammalian organs, including the lung and the kidney, often use a branched design to maximize their functional capacity and efciency. Lung formation requires two developmental processes: branching morphogenesis to build a treelike tubular network, and alveolar differentiation to generate specialized epithelial cells for gas exchange. Although each process has been extensively studied, much less is known about whether and how the two processes are coordinated. We show that an epithelial branching morphogenesis program antagonizes al- veolar differentiation in the mouse lung. Our ndings may provide fresh insights to lung immaturity in preterm neonates and the increase in organ complexity during evolution. Author contributions: D.R.C. and J.C. designed research; D.R.C., D.M.A., R.K.M., H.J., and J.C. performed research; H.A. contributed new reagents/analytic tools; D.R.C. and J.C. analyzed data; and D.R.C., P.D.M., and J.C. wrote the paper. The authors declare no conict of interest. This article is a PNAS Direct Submission. See Commentary on page 18029. 1 Present Address: Department of Pediatrics, University of Texas Medical School at Hous- ton, Houston, TX 77030 2 To whom correspondence should be addressed. E-mail: [email protected]. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1311760110/-/DCSupplemental. 1804218051 | PNAS | November 5, 2013 | vol. 110 | no. 45 www.pnas.org/cgi/doi/10.1073/pnas.1311760110 Downloaded by guest on November 2, 2020

Upload: others

Post on 08-Aug-2020

10 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Lung epithelial branching program antagonizes alveolar ... · Lung epithelial branching program antagonizes alveolar differentiation Daniel R. Changa, Denise Martinez Alanisa, Rachel

Lung epithelial branching program antagonizesalveolar differentiationDaniel R. Changa, Denise Martinez Alanisa, Rachel K. Millerb,1, Hong Jib, Haruhiko Akiyamac, Pierre D. McCreab,and Jichao Chena,b,2

aDepartment of Pulmonary Medicine and bDepartment of Biochemistry and Molecular Biology, University of Texas MD Anderson Cancer Center, Houston,TX 77030; and cDepartment of Orthopedics, Kyoto University, Sakyo, Kyoto 606-8507, Japan

This Feature Article is part of a series identified by the Editorial Board as reporting findings of exceptional significance.

Edited by Brigid L. M. Hogan, Duke University Medical Center, Durham, NC, and approved August 28, 2013 (received for review June 21, 2013)

Mammalian organs, including the lung and kidney, often adopta branched structure to achieve high efficiency and capacity oftheir physiological functions. Formation of a functional lungrequires two developmental processes: branching morphogenesis,which builds a tree-like tubular network, and alveolar differenti-ation, which generates specialized epithelial cells for gas ex-change. Much progress has been made to understand each ofthe two processes individually; however, it is not clear whetherthe two processes are coordinated and how they are deployedat the correct time and location. Here we show that an epithelialbranching morphogenesis program antagonizes alveolar differen-tiation in the mouse lung. We find a negative correlation betweenbranching morphogenesis and alveolar differentiation temporally,spatially, and evolutionarily. Gain-of-function experiments showthat hyperactive small GTPase Kras expands the branching pro-gram and also suppresses molecular and cellular differentiationof alveolar cells. Loss-of-function experiments show that SRY-box containing gene 9 (Sox9) functions downstream of Fibroblastgrowth factor (Fgf)/Kras to promote branching and also sup-presses premature initiation of alveolar differentiation. We thuspropose that lung epithelial progenitors continuously balance be-tween branching morphogenesis and alveolar differentiation, andsuch a balance is mediated by dual-function regulators, includingKras and Sox9. The resulting temporal delay of differentiation bythe branching program may provide new insights to lung imma-turity in preterm neonates and the increase in organ complexityduring evolution.

An optimal organ design maximizes not only the number of itsbasic functional units, for example alveoli in the lung and

glomeruli in the kidney, but also the efficiency of transportinggas and liquid to and from these functional units. This design isoften achieved by building a branched, tree-like structure withfunctional units attached at the end, as seen in the lungs, kidneys,and mammary glands. Compared with a beads-on-a-string de-sign, where all functional units are connected sequentially ona linear duct, a branching design not only packs the maximalnumber of functional units within a given 3D organ space with-out entanglement of the ductal system, but also increasestransport efficiency by minimizing the average distance betweeneach functional unit to the entry point of an organ.Two developmental processes underlie a branching design:

branching morphogenesis, a process that builds the branchedducts, and cellular differentiation, a process that builds thefunctional units. These two processes have to be somehowcoordinated to ensure a fundamental feature of a branchingdesign: all functional units are only located at the termini of thebranched ducts.Branching morphogenesis has been well studied both mor-

phologically and genetically in the mouse lung. The entire airwaylumen is bordered by epithelial cells that originate from theventral foregut around embryonic day 10 (E10) (1). It has beenshown that epithelial progenitors undergo a highly organized

process of branching morphogenesis to give rise to the first 5,000or so airway branches within 5 d of development (E10 to E15)(2). Branching morphogenesis involves reciprocal interactionsbetween the lung epithelium and the surrounding mesenchyme,and requires secretion of a fibroblast growth factor (FGF10) bythe mesenchymal cells, activating its membrane receptor (FGFR2)and subsequently a small GTPase (KRAS) in the epithelial cells(3–5). Additional signaling pathways, including Wnt, Sonichedgehog (Shh), and Tgf signalings, are also essential for normalbranching (1, 6).Lung epithelial progenitors not only build a branched duct

system via branching morphogenesis, but also differentiate intospecialized alveolar cells required for gas exchange, a processnamed alveolar differentiation. There are two alveolar cell types:type I cells that are flat and cover more than 90% of the alveolarsurface, across which gases diffuse; and type II cells that arecuboidal and synthesize pulmonary surfactants, lipoproteincomplexes that hydrate the alveolar surface and prevent alveolarcollapsing by reducing surface tension (7). Genetic studies inmice have identified several transcription regulators specificallyrequired for alveolar differentiation (8–12).Although both branching morphogenesis and alveolar differ-

entiation have been extensively studied, much less is knownabout whether and how they are coordinated. Each process isgenerally viewed as independently controlled and occurringduring early versus late lung development, respectively. Al-though defects in alveolar differentiation are not expected to

Significance

Mammalian organs, including the lung and the kidney, oftenuse a branched design to maximize their functional capacityand efficiency. Lung formation requires two developmentalprocesses: branching morphogenesis to build a treelike tubularnetwork, and alveolar differentiation to generate specializedepithelial cells for gas exchange. Although each process hasbeen extensively studied, much less is known about whetherand how the two processes are coordinated. We show that anepithelial branching morphogenesis program antagonizes al-veolar differentiation in the mouse lung. Our findings mayprovide fresh insights to lung immaturity in preterm neonatesand the increase in organ complexity during evolution.

Author contributions: D.R.C. and J.C. designed research; D.R.C., D.M.A., R.K.M., H.J., andJ.C. performed research; H.A. contributed new reagents/analytic tools; D.R.C. and J.C.analyzed data; and D.R.C., P.D.M., and J.C. wrote the paper.

The authors declare no conflict of interest.

This article is a PNAS Direct Submission.

See Commentary on page 18029.1Present Address: Department of Pediatrics, University of Texas Medical School at Hous-ton, Houston, TX 77030

2To whom correspondence should be addressed. E-mail: [email protected].

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1311760110/-/DCSupplemental.

18042–18051 | PNAS | November 5, 2013 | vol. 110 | no. 45 www.pnas.org/cgi/doi/10.1073/pnas.1311760110

Dow

nloa

ded

by g

uest

on

Nov

embe

r 2,

202

0

Page 2: Lung epithelial branching program antagonizes alveolar ... · Lung epithelial branching program antagonizes alveolar differentiation Daniel R. Changa, Denise Martinez Alanisa, Rachel

affect early branching morphogenesis because of their temporalseparation, defects in branching morphogenesis in early-stagelungs have been associated with either an increase or decrease inalveolar differentiation in late-stage lungs, exemplified in severalrecent studies (13–15). This association has been partly attrib-uted to altered proximal/distal patterning of the epithelium asa result of defective branching, which in turn affects the differ-entiation of distal epithelial progenitors into alveolar cells.However, it is not obvious why and how alterations in the spatialpatterning of progenitors will lead to defects in their cellulardifferentiation. It is unknown whether genes required for branch-ing morphogenesis can regulate alveolar differentiation directly,rather than indirectly via regulation of proximal/distal patterning.In this integrated analysis of branching and differentiation

over the entire course of mouse embryonic lung development, weprovide evidence that branching morphogenesis and alveolardifferentiation are two alternative processes that lung epithelialprogenitors need to balance throughout development, and thatsuch a balance is mediated by dual-function regulators, includingKras and SRY-box containing gene 9 (Sox9).

ResultsNegative Correlations Between Branching Morphogenesis and AlveolarDifferentiation. Because both branching morphogenesis and al-veolar differentiation occur in the distal region of the developingairway tree, we carried out a time-course transcriptome analysisof purified distal lung epithelial cells and found a temporalnegative correlation between the two developmental processes.We used a combination of lineage marking and genetic reportersto label the distal lung epithelium: the entire lung epithelium waslineage-marked by a ShhCre allele (16, 17) and a RosatdT redfluorescence Cre reporter (18), and the proximal conductingairway epithelium was labeled by a Sox2EGFP allele expressinga GFP only in conducting airway cells (19, 20). We enzymaticallydissociated Sox2EGFP/+;RosatdT/+;ShhCre/+ embryonic lungs tosingle cells and used FACS to purify distal epithelial cells thatexpressed red, but not green, fluorescent proteins (Fig. S1).Microarray expression comparison of these distal epithelial cellsfrom E14 through E19 showed up-regulation of a number ofmarkers for alveolar cells, including Napsa, Aquaporin 1 (Aqp1),Surfactant protein B (Sftpb), Lamp3, Ager, Ctsh, Abca3, andSurfactant protein C (Sftpc), consistent with alveolar differentia-tion in late-stage lungs (Fig. 1A and Dataset S1). We also founddown-regulation of genes expressed at the branch tips and as-sociated with branching morphogenesis, including Sox9, Wnt7b,Clusterin (Clu), Melanoma inhibitory activity 1 (Mia1), and Etv4(21–25). Additional down-regulated genes were cell cycle-related,presumably because of a decrease in the ratio of proliferativeepithelial cells undergoing branching morphogenesis and differ-entiated alveolar cells in the purified distal epithelium. Therefore,this transcriptome analysis showed an increase in the alveolardifferentiation program and a decrease in the branching mor-phogenesis program in the distal lung epithelium over time.The gradual decline in the expression of branching-related

genes led us to investigate whether and where in the airway treeepithelial progenitors maintain their branching activity after E17,when alveolar differentiation also occurs. We used a 3D imagingtechnology, optical projection tomography (OPT) (26), to examinebranching morphogenesis in E17 to E19 lungs morphologicallyand molecularly by immunostaining for ECAD (E-Cadherin), anepithelial marker, and SOX9, a marker expressed in branch tipsof early lungs (21). We found that, similar to early-stage lungs(see, for example, Fig. 3), E17 lungs had SOX9+ branch tips atthe termini of the airway tree and their number increased fromE17 to E18 (Fig. S2). In E19 lungs, branch tips are very smallwith only a few cells expressing a high level of SOX9. Thus, weconcluded that branching continues at the distal end of the air-way tree in the late stages of lung development.

We examined how this branching process in late-stage lungswas spatially related to alveolar differentiation and found thatSOX9+ cells at the branch tips were the least differentiated. Wecostained sections from E17 to E19 lungs for SOX9 and a panelof alveolar markers for both alveolar type I [receptor for ad-vanced glycosylation of end products (RAGE), AQP1] and typeII (ABCA3, LAMP3, SFTPB) cells. Because branching occurs ina 3D space and new branches can grow internally away from thesurface of the lung, tissues in the center of a physical section canbe at the topologically distal edge of the airway tree; therefore,we assessed the spatial distributions of these alveolar markersonly in regions where the airway lumen extended continuouslyfrom the proximal conducting airways to the distal branch tips.Consistent with our time-course transcriptome analysis (Fig. 1A),expression of alveolar markers increased over time. Notably, thisincrease mostly occurred in the nonbranch tip regions of thedistal epithelium, whereas SOX9+ cells at the branch tipsexpressed a very low level of alveolar markers (Fig. 1B and Fig.S3). Alveolar differentiation expanded distally over time and atE19 most epithelial cells, except for a few SOX9+ cells at thebranch tips, were differentiated.This difference in alveolar differentiation in branch tip versus

nonbranch tip regions was also supported by cell morphologyanalysis: ECAD staining showed that undifferentiated cells at thebranch tips were clustered and cuboidal, whereas differentiatedalveolar type I and type II cells at the nonbranch tip regions wereflat and isolated cuboidal cells, respectively (Fig. 1B and Fig. S2).Our analyses revealed an unexpected negative correlation be-tween branching and alveolar differentiation along the proximal/distal axis of the airway tree, suggesting that in late-stage lungs,branching continues at the distal edge of the airway tree andleaves behind cells undergoing alveolar differentiation.We extended the temporal and spatial negative correlations

between branching and alveolar differentiation to the Xenopuslung, one of most primitive lungs in evolution. Similar to themouse lung, the Xenopus lung arises from the foregut, expressesthe lung lineage factor Nkx2.1 (27), and initially forms one tubeon each side of the embryo (Fig. 1C and Fig. S4). Unlike themouse lung, which branches extensively after its specification, theinitial two tubes of the Xenopus lung simply grew posteriorly andthe entire lung epithelium differentiated to express alveolarmarkers, including Sftpb and Sftpc (Fig. 1C and Fig. S4) (28).Importantly, although Sox9 was present in the Xenopus genomeand was detected in the pharyngeal arches, it was not expressedin the Xenopus lungs at all stages examined (Fig. 1C and Fig. S4).Therefore, the Xenopus lung lacks the branching program andSox9 expression, and initiates alveolar differentiation immedi-ately after lung specification.

Hyperactive Kras Suppresses Alveolar Differentiation. To examinewhether there is a causal relationship in the observed negativecorrelations between branching and alveolar differentiation, weaugmented the branching program in epithelial progenitors byexpressing a hyperactive Kras allele (29) at the time of alveolardifferentiation and found that hyperactive Kras suppressed mo-lecular and cellular differentiation of the progenitors. We useda Sox9CreER allele (30) to only target lung progenitors late indevelopment to avoid difficulty in interpreting potentially sec-ondary phenotypes by targeting the early lung epithelium, forexample, with a ShhCre allele. Expressing the hyperactive Kras inlung progenitors after E15 resulted in an increase in thebranching program with SOX9 expression, normally restricted toa few cells at the branch tips of E19 lungs, expanded to half ofthe distal lung epithelium (Fig. 2A). This phenotype was similarto that in lungs expressing the hyperactive Kras throughout theepithelium at an earlier stage (see, for example, Fig. 4B). Thisexpansion in the branching program was accompanied by a re-striction of alveolar differentiation to regions near the conducting

Chang et al. PNAS | November 5, 2013 | vol. 110 | no. 45 | 18043

DEV

ELOPM

ENTA

LBIOLO

GY

FEATU

REART

ICLE

SEECO

MMEN

TARY

Dow

nloa

ded

by g

uest

on

Nov

embe

r 2,

202

0

Page 3: Lung epithelial branching program antagonizes alveolar ... · Lung epithelial branching program antagonizes alveolar differentiation Daniel R. Changa, Denise Martinez Alanisa, Rachel

airways, as assessed by expression of alveolar type I (AQP1 andRAGE) and type II (ABCA3 and LAMP3) markers (Fig. 2Band Fig. S5). We note that expression of SFTPB was more uni-form throughout the distal epithelium in the Kras mutant lung,although cells in the expanded branching regions were clustered,a similar arrangement to that of cells undergoing branchingmorphogenesis (Fig. 2C and Fig. S5).On the cellular level, hyperactive Kras suppressed cell flat-

tening, a morphological hallmark of alveolar type I cells. ECADstaining showed that although the control lung epithelium at E19mostly consisted of isolated cuboidal type II cells intermingledwith presumable flat type I cells, the expanded branching regionsof the Kras mutant lung consisted of closely packed epithelialcells, an organization reminiscent of that in undifferentiatedbranch tips of the early lungs (Fig. 2C). ECAD is a cell-junctionprotein and does not outline the entire cell; therefore, to visu-alize the morphology of individual epithelial cells, we used a Cre

reporter (RosamTmG) that expresses GFP throughout the cellmembrane upon recombination (31). Under our experimentalconditions, the RosamTmG allele was less sensitive to the Crerecombinase than the hyperactive Kras allele, and therefore la-beled a subset of Kras mutant cells. This process allowed visu-alization of cell morphology at a single-cell resolution andrevealed that the expanded branching regions of the Kras mutantlung lacked flat alveolar type I cells (Fig. 2C).Although Kras is well known as an oncogene for many cancers

(32), it is also an essential component of receptor tyrosine kinasesignalings during development, including the Fgf signaling inlung morphogenesis (5, 33). Therefore, the Kras gain-of-functionexperiments suggested that an augmented branching program inlate developing lungs suppresses alveolar differentiation andalters the observed spatial negative correlation between branch-ing and alveolar differentiation (Fig. 1B).

Fig. 1. Temporal (A), spatial (B), and evolutionary (C) negative correlations between branching morphogenesis and alveolar differentiation. (A) Time-coursemicroarray expression profiling of FACS-purified distal lung epithelial cells. Gene expression values (log2) in E14 samples are used as a baseline for comparison.Horizontal histograms for E15–E19 samples are the frequency distributions of average fold-changes for all genes; the units are shown as the number of genes(# genes). Alveolar markers are up-regulated (red), and branching (green) and cell cycle (blue) related genes are down-regulated over time. Down-regulatedgenes appear to have a smaller fold-change than up-regulated genes, possibly because of perdurance of transcripts. (B) Confocal images of immunostainedE18 lung sections in areas where the airway lumen can be continuously traced from the proximal conducting airways (black/white long dashed lines) to thedistal nonbranch tip (magenta) and branch tip (green) regions, as illustrated in the schematics. Branch tips are outlined with dashed lines. The boxed regionsare enlarged as insets outlined in corresponding colors, showing a spatial negative correlation between branching and alveolar differentiation. AQP1 labelsalveolar type I cells, the vasculature and the mesothelium, which are separated by dashed lines (Insets). There is a very low level of SOX9 expression innonbranch tip epithelium, possibly because of protein perdurance. (Scale bars, 20 μm.) (C) Whole-mount in situ hybridization and immunostaining of Xenopusembryos of indicated stages (St) showing the absence of Sox9 expression and branching in the Xenopus lung. The lungs are indicated with dashed lines ifstained or arrowheads if unstained. Open arrowheads indicate Sox9 expression in the pharyngeal arches. The two Xenopus Sox9 homologs (Sox9a and Sox9b)are 95% identical on the nucleotide level. OPT images of immunostained embryos (Left, Bottom) are shown as maximal intensity projection (Left) andsectional view (Right). (Scale bar, 200 μm.)

18044 | www.pnas.org/cgi/doi/10.1073/pnas.1311760110 Chang et al.

Dow

nloa

ded

by g

uest

on

Nov

embe

r 2,

202

0

Page 4: Lung epithelial branching program antagonizes alveolar ... · Lung epithelial branching program antagonizes alveolar differentiation Daniel R. Changa, Denise Martinez Alanisa, Rachel

Sox9 Is Required for Branching Morphogenesis. Suppression of al-veolar differentiation by the gain-of-function Kras-inducedbranching program led us to hypothesize that there are positiveregulators of branching morphogenesis, the loss-of-function mu-tations of which will cause premature alveolar differentiation.One intriguing candidate is Sox9, which is highly expressed indistal epithelial cells undergoing active branching (21) (Fig. 3)and required for the development of two other branched organs,the kidney and the pancreas (34, 35). To test the role of Sox9 inmouse lung development, we deleted it specifically in the epi-thelium with a ShhCre allele (16, 17). Sox9 expression was abol-ished in the mutant lung epithelium before E12, but was notaffected in the cartilages surrounding the tracheal epithelium(Fig. 3A). In contrast to a previous study (21), in which deletionof Sox9 did not affect lung development and function, epithelialdeletion of Sox9 using ShhCre resulted in a smaller lung withfewer and dilated airway branches. This branching phenotypewas apparent as early as E13 and persisted throughout embry-onic development (Fig. 3A and Fig. S6). No viable Sox9 mutantmice were obtained postnatally. Despite significant disruption of

branching, the coalignment between the epithelial branches andthe arterial vasculature was not affected (Fig. S6).Besides branch number and morphology, Sox9 regulates

a subset of branching-related genes in a cell-autonomous man-ner. In a microarray expression profiling experiment comparingcontrol and Sox9 mutant lungs (Dataset S2; described in detailbelow), we identified several genes down-regulated in the Sox9mutant, including Clu and Mia, which have been implicated inlung development and cancer (25, 36). In situ hybridization andimmunostaining experiments showed that these genes wereexpressed at the branch tips and down-regulated in the Sox9mutant as early as E11 (Fig. 3B and Fig. S7 A and B). Using anNkx2.1CreER allele (37), we deleted SOX9 in a subset of epi-thelial cells and observed a mosaic expression pattern ofClusterin that matched perfectly with that of SOX9 (Fig. 3Cand Fig. S7E), demonstrating a cell-autonomous regulation ofClu by Sox9.The expression of many other genes that are normally re-

stricted to the branch tips, including Bmp4, Spry2, Id2, Thbs1,and Dlk1, were not affected in the Sox9 mutant as assayed viaboth microarray and in situ hybridization experiments (Fig. S7C

Fig. 2. Hyperactive Kras suppresses alveolar differentiation. Confocal images of immunostained E19 lung sections from embryos of indicated genotypes andtheir respective littermate controls, showing that hyperactive Kras causes expansion of the SOX9+ branching region (A) and corresponding restriction ofalveolar differentiation based on molecular markers (B) and cell morphology (C). Cre Recombination is induced at E15 to specifically target epithelial pro-genitors in late stage lungs. Shown are areas where the airway lumen can be continuously traced from the proximal conducting airways (black/white longdashed lines) to the distal nonbranch tip (magenta) and branch tip (green) regions, as illustrated in the schematics. The boxed regions are enlarged (Insets)outlined in corresponding colors. Dashed green boxes indicate low branching activity with few SOX9+ cells. GFP staining (C) allows visualization of themorphology of isolated alveolar type I (square brackets) and type II (arrowheads) cells by outlining the cell membrane; ECAD staining only labels cell junctions.Under our experimental conditions, the RosamTmG allele is less sensitive to the Cre recombinase than the hyperactive Kras allele, and therefore labels a subsetof Kras mutant cells. (Scale bars, 20 μm.)

Chang et al. PNAS | November 5, 2013 | vol. 110 | no. 45 | 18045

DEV

ELOPM

ENTA

LBIOLO

GY

FEATU

REART

ICLE

SEECO

MMEN

TARY

Dow

nloa

ded

by g

uest

on

Nov

embe

r 2,

202

0

Page 5: Lung epithelial branching program antagonizes alveolar ... · Lung epithelial branching program antagonizes alveolar differentiation Daniel R. Changa, Denise Martinez Alanisa, Rachel

and Dataset S2). In addition, there are no significant changes incell polarity in the Sox9 mutant lung as assessed by apical[Zonula occluden 1 (ZO1) and Podocalyxin-like (POD)] (17),

basement membrane [Entactin (Ent) and Perlecan (Perl)] andcytoskeleton [acetylated-Tubulin (AcT)] marker expression (Fig.S8). These results suggested that these genes are regulated by

Fig. 3. Epithelial deletion of Sox9 impairs branching morphogenesis. (A) OPT images of whole-mount lungs from littermate control (Left) and Sox9CKO/CKO;ShhCre/+ mutant (Right) embryos at indicated embryonic stages, immunostained for SOX2, SOX9, and ECAD. The RCd.L3 branch lineages (2) in the boxed areasare enlarged and traced with dashed lines. SOX9 expression is lost before E12. Fewer branches form in the Sox9 mutant lungs as early as E13. Branches of theRCd.L3 branch lineages start to dilate at E13 in the Sox9 mutant. E, ECAD; S, SOX9. (Scale bar, 250 μm.) (B) OPT images of whole-mount immunostained lungsfrom E15 Sox9CKO/CKO;ShhCre/+ mutant and littermate control embryos. Similar to SOX9, CLU is normally expressed in branch tips and this expression is lost inthe Sox9 mutant. CLU is also expressed at a lower level in airway smooth muscles and this expression is not affected and becomes apparent in the Sox9mutant. (Scale bar, 250 μm.) (C) Mosaic deletion of Sox9. Confocal images of branch tips of whole-mount immunostained lungs from E15 Sox9CKO/CKO;Nkx2.1CreER/+ mutant and littermate control embryos. The boxed regions are enlarged in subsequent panels. SOX9 is deleted in a subset of epithelial cellsoutlined with dashed lines and these Sox9 mutant cells do not express CLU, demonstrating a cell autonomous requirement of Sox9 for CLU expression (Fig.S7E). (Scale bars, 10 μm.)

18046 | www.pnas.org/cgi/doi/10.1073/pnas.1311760110 Chang et al.

Dow

nloa

ded

by g

uest

on

Nov

embe

r 2,

202

0

Page 6: Lung epithelial branching program antagonizes alveolar ... · Lung epithelial branching program antagonizes alveolar differentiation Daniel R. Changa, Denise Martinez Alanisa, Rachel

Sox9-independent signals and may be responsible for theremaining branching program in the Sox9 mutant lung (Fig. 3A).Because the Sox9 mutant lung was smaller, we examined

molecules and cellular processes related to cell proliferation andapoptosis. One candidate was Cyclin D1 (CCND1), which isknown to be transcriptionally activated by growth factor-inducedRas signaling (38). We found a high level of CCND1 expressionin the branching regions of the epithelium, and significantlyfewer cells expressing a normal level of CCND1 in the Sox9

mutant lung (P < 0.001, χ2 test) (Fig. S9A). However, the per-centage of proliferating cells measured by the incorporation ofa nucleotide analog (EdU, 5-ethynyl-2′-deoxyuridine) was notsignificantly lower in the Sox9mutant branch tips, compared withtheir matched branch tips in littermate control lungs (P > 0.05,Student t test) (Fig. S9B), suggesting either compensation byother Cyclin D proteins or a small proliferation defect that wasundetectable using the current method but accumulated overtime to affect the size of the lung.

Fig. 4. Sox9 functions downstream of the Fgf/Kras branching signal. (A) OPT images of E13 whole-mount immunostained lungs from littermate control andFgfr2CKO/CKO;ShhCre/+ mutant embryos. The boxed areas are shown in a cross-section view. Arrowheads indicate that SOX9 expression in the branching ep-ithelium is lost in the Fgfr2 mutant lung. Arrows indicate that SOX9 expression in the cartilage precursors is not affected. (Scale bar, 200 μm.) (B) OPT imagesof E13 whole-mount immunostained lungs from control, Sox9CKO/CKO;ShhCre/+ mutant, KrasLSL-G12D/+;ShhCre/+ mutant, and KrasLSL-G12D/+;Sox9CKO/CKO;ShhCre/+

mutant embryos. Regions of the tracheas (brackets) are shown in a cross-section view. Epithelial Sox9 deletion suppresses the overgrowth and ectopic trachealbranch phenotypes in the Krasmutant lung. We note that Sox9 and Kras double-mutant branches remain cystic, possibly because of the presence of the Sox9-independent branching program (Fig. S7 C and D). (Scale bars, 200 μm.)

Chang et al. PNAS | November 5, 2013 | vol. 110 | no. 45 | 18047

DEV

ELOPM

ENTA

LBIOLO

GY

FEATU

REART

ICLE

SEECO

MMEN

TARY

Dow

nloa

ded

by g

uest

on

Nov

embe

r 2,

202

0

Page 7: Lung epithelial branching program antagonizes alveolar ... · Lung epithelial branching program antagonizes alveolar differentiation Daniel R. Changa, Denise Martinez Alanisa, Rachel

In addition to its requirement in the epithelium, Sox9 in-directly regulates mesenchymal cell survival and Fgf10 expres-sion, key components of the epithelial-mesenchymal interactionsrequired for branching morphogenesis (1). When examiningexpression of cleaved Caspase-3 (Casp3), an apoptotic marker,we found that the branching regions of both control and Sox9mutant epithelia had no apoptotic cells. However, apoptotic cellswere scattered throughout the distal mesenchyme of the Sox9mutant lung, but not the control lung, as early as E13 (Fig.S10A). Furthermore, the Sox9 mutant mesenchyme spanneda wider region between the epithelium and the mesothelium atthe surface of the lung, and had a higher level of Fgf10 expres-sion (Fig. S10B). A possible explanation for the observed mes-enchymal phenotype is that the Sox9-dependent branchingprogram also controls the production of a trophic or signalingfactor necessary for cell survival and epithelial feedback-regu-lation of Fgf10 expression in the mesenchyme. Dysregulation ofFgf10 expression might contribute to the cystic airway branchphenotype in the Sox9 mutant (Fig. 3A). Indeed, phosphorylatedERK (pERK), a mediator of the Kras branching signal, appearedmore diffuse in the Sox9mutant lung (Fig. S10C). Based on all ofthe branching-related phenotypes of Sox9 mutant lungs, weconcluded that Sox9 is not only a marker, but also a positiveregulator of the branching morphogenesis program.

Sox9 Functions Downstream of the Fgf/Kras Branching Signal. TheSox9 expression pattern and mutant phenotypes (Fig. 3) led us toexamine the role of Sox9 in the known Fgf/Kras branchingmorphogenesis program. We examined expression of Sox9 in theabsence of the Fgf/Kras branching signal by genetically deletingthe Fgf receptor (Fgfr2) from the epithelium. The Fgfr2 mutantlung failed to branch and notably, epithelial expression of SOX9

was lost and the entire epithelium expressed SOX2 (Fig. 4A),a marker and regulator of the proximal conducting airways (19,39). Conversely, expression of the hyperactive Kras allelethroughout the epithelium led to an overgrown lung with ex-panded Sox9 expression domain (Fig. 4B). These data suggestedthat Sox9 is a downstream component of the Fgf/Kras branch-ing program.To determine whether Sox9 functionally mediates the Fgf/Kras

branching program, we examined the genetic interaction be-tween Sox9 and Kras. When we activated the hyperactive Krasallele throughout the lung epithelium, the Kras mutant lung waslarger and most of the epithelium was actively branching (Fig.4B). We noted that overactivation of Kras increased the size butnot the number of branch tips, suggesting that normal branchformation requires precise regulation of Kras activity. Never-theless, ectopic branches formed from the normally nonbranchingtracheal epithelium, comparable to the ectopic tracheal branchesinduced by the Fgf-expressing lung mesenchyme in a tissue graftexperiment (40). Genetic deletion of Sox9 in the hyperactive Krasbackground reduced the size of the lung and suppressed the for-mation of ectopic tracheal branches (Fig. 4B and Fig. S11). Thegenetic interaction between Sox9 and Kras, together with the Sox9mutant phenotypes, showed that Sox9 is a positive mediator of theFgf/Kras branching program.

Sox9 Suppresses Premature Initiation of Alveolar Differentiation. Amicroarray expression profiling experiment provided the initialclue that Sox9 is a dual-function regulator of both branchingmorphogenesis and alveolar differentiation (Fig. 5A and DatasetS2). In addition to the down-regulated genes (Fig. 3 and Fig. S7),the Sox9 mutant lung also up-regulated a number of genes, in-cluding Sftpb, Sftpc, Lamp3, Napsa, and Ctsh, that are known or

Fig. 5. Sox9 suppresses alveolar differentiation. (A) Average fold-change in gene expression on a log2 scale comparing Sox9CKO/CKO;ShhCre/+ mutant and lit-termate control lungs at indicated embryonic stages by microarray expression profiling. Genes up- and down-regulated in the Sox9mutant lungs are shown inred and green, respectively. The diameters of the circles represent the differences in fold-change between two biological replicates. The numbers in parenthesisrepresent the rank orders of the genes based on fold-change at E15. Horizontal histograms represent the frequency distributions of fold-changes for all geneson the microarray at indicated embryonic stages; the units are shown as the number of genes (# genes). (B) Sftpb whole-mount in situ hybridization of lit-termate control and Sox9CKO/CKO;ShhCre/+mutant lungs at indicated embryonic stages. Sftpb is up-regulated in the branch tips (dashed lines) of the Sox9mutantlung as early as E11 before appearance of any defects in branch morphology. (Scale bars, 200 μm.) (C) Confocal images of immunostained sections from E15Sox9CKO/CKO;ShhCre/+ mutant and littermate control lungs, showing up-regulation in SFTPB and LAMP3 protein expression in the branch tips (dashed lines) ofSox9 mutant lungs. LAMP3 is localized to intracellular vesicle-like structures (arrowheads). (Scale bar, 20 μm.) (D) The branching program antagonizes thealveolar differentiation program and this antagonization is mediated by dual-function regulators including Kras and Sox9 that promote branching andsuppress alveolar differentiation. The branching program contains additional Sox9 and Fgf/Kras independent genes, including Bmp4 and Id2 (dashed arrows).

18048 | www.pnas.org/cgi/doi/10.1073/pnas.1311760110 Chang et al.

Dow

nloa

ded

by g

uest

on

Nov

embe

r 2,

202

0

Page 8: Lung epithelial branching program antagonizes alveolar ... · Lung epithelial branching program antagonizes alveolar differentiation Daniel R. Changa, Denise Martinez Alanisa, Rachel

were shown (Fig. S12A) to be expressed by differentiatedalveolar cells and involved in the production and processing ofsurfactant proteins (41, 42).We confirmed the microarray results by in situ hybridization

and immunostaining experiments, showing that several alveolarmarkers increased dramatically in Sox9 mutant lungs comparedwith control lungs (Fig. 5 B and C, and Fig. S12 B and C).Strikingly, this premature alveolar differentiation occurred at theearliest stage of branching morphogenesis (E11), when the lungconsisted of only two tubes and before any morphological dif-ference between Sox9 control and mutant lungs (Fig. 5B). Thisaspect showed that the alveolar differentiation phenotype is notsecondary to the branching defects in the Sox9 mutant lung. Wenote that precocious activation of alveolar genes was restrictedto the distal branching epithelium, suggesting the presence ofnegative regulators (e.g., Sox2) of alveolar differentiation in theproximal conducting airways.Although a number of alveolar genes were expressed pre-

maturely in Sox9 mutant lungs, the full alveolar differentiationprogram were not activated, suggesting the presence of addi-tional negative regulators or lack of the complete set of positiveregulators of alveolar differentiation in early developing lungs.Surfactant proteins involved in immune defense (Sftpa andSftpd) (43) and markers for alveolar type I cells (RAGE andAQP1) were not precociously expressed in the Sox9 mutant lung(Dataset S2). An alternative possibility for this incomplete de-repression of alveolar differentiation is that the most ancientalveolar differentiation program during lung evolution is tosynthesize surfactants to reduce alveolar surface tension and thisbasic program is suppressed by Sox9, whereas immune defense-related surfactant proteins and alveolar type I cells are functionalimprovements that arise later in evolution and are controlled bySox9-independent mechanisms. Taking these data together, weconcluded that Sox9 promotes branching morphogenesis andalso suppresses premature initiation of alveolar differentiation.

DiscussionThe experiments presented herein reveal a previously unap-preciated link between branching morphogenesis and alveolardifferentiation in lung epithelial progenitors (Fig. 5D): thebranching program antagonizes the alveolar differentiationprogram, and dual-function regulators including Kras and Sox9promote branching and suppress alveolar differentiation. Thismodel does not exclude the possibility that Kras and Sox9 func-tion to maintain cell shape and proliferation, cellular processesthat are necessary for branching morphogenesis and affected inthe Kras and Sox9 mutant lungs. The cellular mechanisms un-derlying the balance between branching and alveolar differenti-ation require further investigation.Such a model suggests a new area of research in lung de-

velopment in which the two developmental processes are studiednot only individually, but also as a balanced choice of the pro-genitors. Our data imply that genes other than Kras and Sox9that are typically regarded as branching-related based on ex-pression patterns and mutant phenotypes (e.g., Id2, Bmp4, andSpry2) need to be investigated for their potential roles in alveolardifferentiation in gain and loss-of-function experiments similarto those in this study. Conversely, we speculate that genes con-trolling alveolar differentiation (e.g., Cebpa, Creb1, and Carm1)may suppress branching morphogenesis if overexpressed duringearly lung development. Indeed, the Creb1 mutant lung has ex-cessive SOX9+ epithelial cells (9), although it is unknown whetherthese cells form extra branches.The spatiotemporal transition from branching morphogenesis

to alveolar differentiation (Fig. 1) may provide new insights tomechanisms and therapies of lung defects in premature birth.This transition is interrupted by premature birth, especially invery/extremely low birth-weight infants born at the canalicular

stage of lung development (gestation week 24 in humans andE17 in mice) (44, 45). Interruption to this transition may lead tonot only cellular immaturity as a result of insufficient alveolardifferentiation, but also structural immaturity as a result of in-complete branching morphogenesis. Both cellular and structuralimmaturities need to be considered when optimizing currenttherapies, such as antenatal glucocorticoids and mechanicalventilation, and designing new therapies.The balance between branching morphogenesis and alveolar

differentiation (Fig. 5D) may be modulated during evolution toincrease the complexity and capacity of the lung. Although allvertebrate lungs mediate gas exchange with specialized alveolarcells, their complexity ranges from the two-tube amphibian lungto the tree-like mammalian lung. The Xenopus lung does notbranch and expresses alveolar genes at the beginning of lungdevelopment, whereas the mouse lung starts by branching ex-tensively and shifts to alveolar differentiation later in de-velopment (Fig. 1). We speculate that branching morphogenesisis an evolutionary addition to the ancestral alveolar differentia-tion program, and that the complex tree-like structure arises viaa developmental delay of alveolar differentiation to allow in-tegration of the branching morphogenesis program. Such a tem-poral coordination will also ensure positioning the alveolarregion at the termini of the airway tree. We further speculatethat the integration between the two developmental processes isachieved by dual-function regulators, including Kras and Sox9,and that modulation of the amount/activity of these dual-func-tion regulators in conjunction of developmental time will gen-erate airway trees of varying complexity in different species.Similar mechanisms may be applicable to the development

and evolution of other branched organs. The metanephrosdevelops in a centrifugal manner analogous to the lung: theproximal/medullary epithelia are older and more differentiatedthan the distal/cortical epithelia, and the cortical epithelia con-tinue to branch, whereas the medullary epithelia differentiateand become physiologically active. Interestingly, loss of β-cateninin the ureteric bud is shown to cause both branching defects andpremature differentiation, reminiscent of the lung phenotype inthe Sox9 mutant (46, 47).

Materials and MethodsMice. Mice carrying RosatdT, Sox2EGFP, ShhCre, Sox9CKO, Fgfr2CKO, RosamTmG,Sox9CreER, Nkx2.1CreER, and KrasLSL-G12D alleles have been previously de-scribed (16–18, 20, 29–31, 37, 48–50). The Sox9CKO allele was genotypedusing the following primers: 5′-GACAGCACTCCTGGCCAGAT-3′, 5′-CGCTTC-CTCGTGCTTTACG-3′, and 5′-TGGTAATGAGTCATACACAGTAC-3′. The day inwhich a vaginal plug was observed was denoted as E1. For Sox9CreER/+;KrasLSL-G12D/+ experiments, 2 mg tamoxifen (T5648; Sigma) dissolved in cornoil (C8267; Sigma) was given intraperitoneally to timed pregnant mice atE15. To activate the RosamTmG allele, 0.5 mg tamoxifen was injected to labelepithelial cells at a low density. For Sox9CKO/CKO;Nkx2.1CreER/+ experiments,3.5 mg tamoxifen was injected at E10. All animal experiments were ap-proved by the Institutional Animal Care and Use Committee at the TexasA&M Health Science Center Institute of Biosciences and Technology (pro-tocol no. 11004).

Xenopus Embryos. Xenopus laevis embryos were obtained and grown usingstandard methods (51). At Nieuwkoop and Faber stages 36, 38, 40, and42, embryos were fixed as previously described (52) and kept in methanolat −20 °C.

Antibodies. The following antibodies were used for immunostaining: rabbitanti–SOX9 (1:1,000; AB5535, Millipore; also recognizes the Xenopus SOX9),goat anti- SOX9 (1:1,000, AF3075; R&D Systems), goat anti-SOX2 (1:250;sc-17320; Santa Cruz Biotechnology), rat anti- ECAD (1:1,000; 131900,Invitrogen), rabbit anti-AQP1 (1:1,000; AB2219, Millipore), rabbit anti-CCND1 (1:500; RM9104S0, ThermoFisher), rabbit anti-CASP3 (1:500; 9661,Cell Signaling), rabbit anti- pERK (1:500; 4370, Cell Signaling), rabbit anti-SFTPC (1:1,000; AB3768, Millipore), rabbit anti-SFTPB (1:1,000; WRAB55522,Seven Hills Bioreagents), rabbit anti-Abca3 (1:1,000; WRAB70565, Seven Hills

Chang et al. PNAS | November 5, 2013 | vol. 110 | no. 45 | 18049

DEV

ELOPM

ENTA

LBIOLO

GY

FEATU

REART

ICLE

SEECO

MMEN

TARY

Dow

nloa

ded

by g

uest

on

Nov

embe

r 2,

202

0

Page 9: Lung epithelial branching program antagonizes alveolar ... · Lung epithelial branching program antagonizes alveolar differentiation Daniel R. Changa, Denise Martinez Alanisa, Rachel

Bioreagents), rat anti-Lamp3 (1:1,000; DDX0191, Imgenex), rat anti-RAGE(1:1,000; MAB1179, R&D Systems), chicken anti- GFP (1:2,500; AB13970,Abcam), Cy3 conjugated mouse anti-smooth muscle actin (SMA, 1:1,000;C6198, Sigma), goat anti-CLU (1:500; sc-6420, Santa Cruz Biotechnology),rabbit anti-ZO1 (1:50; 187430, Invitrogen), goat anti–POD (1:250; AF1556,R&D Systems), mouse anti-AcT (1:1,000; T6793, Sigma), rat anti-Perl (1:1,000;RT-794-P0, Thermo Scientific) and rat anti-Ent (1:1,000; RT-797-P0, ThermoScientific). Secondary antibody staining was performed using fluorescentdonkey secondary antibodies from Invitrogen and Jackson ImmunoResearchat 1:1,000 dilution.

Tissue Harvest and Immunostaining. Harvested embryos were fixed in PBS (pH7.4) with 0.5% paraformaldehyde (PFA; P6148, Sigma) for 3 h at roomtemperature. Adult lungs were inflated with 0.5% PFA in PBS at 25 cm H2Opressure, and then dissected and fixed with 0.5% PFA in PBS at room tem-perature for 3–6 h. Samples prepared for section immunostaining werecryoprotected in PBS with 20% (wt/vol) sucrose and 33% (vol/vol) OptimalCutting Temperature Compound (OCT; 4583; Tissue-Tek) at 4 °C overnightand then embedded in OCT. All section and whole-mount stainings werecarried out essentially as previously described (17, 53). Sections 10 μm inthickness were blocked in PBS + 5% (vol/vol) normal donkey serum (017-000-121; Jackson ImmunoResearch) for 1 h followed by incubation with primaryantibodies diluted in PBS + 0.3% Triton X-100 overnight at 4 °C. The nextday, primary antibodies were removed by washing slides with PBS for 90 minfollowed by incubation with secondary antibodies and DAPI diluted in PBS +0.1% Triton X-100 + 0.1% Tween-20 for 90 min at room temperature. Sec-ondary antibodies were removed by washing with PBS for 1 h followed byfinal mounting with Aquamount mounting medium (18606, Polysciences).

For whole-mount immunostaining, fixed samples were placed in PBSovernight at 4 °C, then dehydrated through a PBS/methanol gradient andbleached with 6% (vol/vol) hydrogen peroxide (H1009, Sigma) in methanolovernight at 4 °C. The next day, samples were rehydrated in a methanol/PBSgradient, then blocked in PBS + 0.3% Triton X-100 + 5% normal donkeyserum for 2 h, followed by addition of primary antibodies into blockingsolution and incubated overnight at 4 °C. Samples were then washed for 6 hwith PBS + 0.1% Triton X-100 + 0.1% Tween-20, followed by addition ofsecondary bodies diluted in PBS + 0.3% Triton X-100 at 4 °C overnight. Fi-nally, samples were washed again before fixation with 4% PFA in PBS for2–4 h. To minimize experimental variation, littermate control and mutantlungs were processed in the same tube throughout the experiment. Finalimages were captured on a fluorescence stereoscope with a 2× Plan Apo-chromat objective (M205C, Leica), an OPT microscope (Bioptonics), or aconfocal microscope (FV1000, Olympus).

OPT Microscopy. Following whole-mount immunostaining, samples wereembedded in 1% (wt/vol) low-melting agarose (16520-100, Invitrogen).Embedded samples were then dehydrated in three passages of methanol andcleared in two passages of BABB: one part benzyl alcohol (24122, Sigma) andtwo parts benzyl benzoate (B6630, Sigma). Samples were imaged using theBioptonics 3001 OPT scanner with GFP, TXR, and Cy5 filters at high resolution(1,024 pixels), according to the manufacturer’s instructions. Image stackswere reconstructed using the NRecon Reconstruction software (Bruker-microCT, BE) and visualized using maximal intensity projection or sectionview with the Bioptonics Viewer software (Bioptonics).

Section and Whole-Mount in Situ Hybridization. In situ hybridization was per-formed essentially as described previously (17, 53). To generate digoxigenin-labeled riboprobes (11277073910, Roche), cDNAs for target genes wereisolated as PCR products derived from mouse embryonic lung RNAs andXenopus stage 42 embryonic RNAs. Riboprobes were then transcribed witha T7 RNA polymerase (18033-019, Invitrogen) from the cDNAs. For sectionin situ hybridization, fixed adult lungs were incubated in PBS with 20%sucrose, 33% OCT, and 0.4% PFA overnight at 4 °C, followed by embed-ding in OCT. Embryonic lungs were quickly dissected and frozen in OCT.Ten-micrometer sections were fixed in 4% PFA in PBS for 10 min at roomtemperature, followed by treatment with fresh acetylation solution(diethylpyrocarbonate-treated H2O with 0.0133% triethynolamine and0.0025% acetic anhydride) for 10 min. Sections were then incubated inhybridization solution [50% (vol/vol) formamide, 5× SSC (sodium chlorideand sodium citrate), 5× Denhardt’s solution, 500 μg/uL salmon sperm DNA,250 μg/uL yeast tRNA] for 1 h at 62 °C, followed by overnight incubation in1 μg/mL riboprobes diluted in hybridization solution at 62 °C overnight ina slide oven (24100, Boekel). The next day, slides were incubated in 2× SSCfor 30 min at room temperature, followed by 0.2× SSC for 2 h at 65 °C.Slides were then blocked with TBST (100 mM Tris, 150 mM sodium

chloride, 0.1% Tween-20, pH 7.5) with 5% normal sheep serum (013-000-121; Jackson ImmunoResearch) followed by application of antidigoxigeninalkaline phosphatase Fab fragment (11093274910, Roche) diluted in TBST(1:1,000) at 4 °C overnight. The next day, slides were washed in TBST for2 h at room temperature followed by detection of alkaline phosphataseactivity with nitro-blue tetrazolium (N6639, Sigma) and 5-bromo-4-chloro-3′-indolyphosphate (B6149, Sigma).

For whole-mount in situ hybridization, fixed lungs were dehydrated andstored in methanol at −20 °C. For E11 and E13 lungs, the whole lung wasstained intact; for E14 and E15 lungs, lobes were separated and stained in-dividually. Samples were rehydrated in a methanol/DEPC-PBS (diethylpyr-ocarbonate-treated PBS) gradient at room temperature and permeabilizedin DEPC-PBS with 10 μg/mL protease K for 10 min. Samples were then fixedin PBS with 4% PFA and 0.25% (vol/vol) glutaraldehyde for 20 min andwashed with DEPC-PBS afterward. Prehybridization solution (50% formam-ide, 4× SSC, 1× Denhardt’s solution, 250 μg/uL salmon sperm DNA, 250 μg/uLyeast tRNA, 50 μg/mL heparin, 0.1% Tween-20) was then applied and sam-ples were incubated at 62 °C for 1 h in a slide oven. Subsequently, 1 μg/mLriboprobes diluted in 1 mL of hybridization solution (prehybridization so-lution with 10% (wt/vol) >500 kDa dextran sulfate) was added to samplesfollowed by incubation at 62 °C in a slide oven overnight. The next day,samples were washed in 50% formamide with 4× SSC and 0.1% Tween-20 at65 °C for 1 h, incubated in 100 μg/mL RNase A diluted in 100 mM Tris (pH 7.5)with 500 mM sodium chloride and 0.1% Tween-20 at 37 °C for 1 h, andwashed in 50% formamide with 2× SSC and 0.1% Tween-20 at 65 °C over-night in a water bath. The following day, samples were washed in TBST for20 min followed by blocking with TBST+ 5% normal sheep serum for 2 h. Anantidigoxigenin alkaline phosphatase Fab fragment was then added to theblocking solution for 3 h followed by TBST washes for 3 h and an overnightwash in TBST with 0.5 mg/mL levamisole (L9756, Sigma) at 4 °C. The final day,samples were washed with TBST for 2 h followed by detection of alkalinephosphatase activity with nitro-blue tetrazolium and 5-bromo-4-chloro-3′-indolyphosphate. Following the alkaline phosphatase reaction, sampleswere fixed in 4% PFA in PBS overnight, cleared in 75% (vol/vol) glycerol andmounted on a depression-well slide with coverslip for imaging. Whole-mount in situ hybridization of Xenopus embryos was performed using thesame protocol for mouse lung staining. After staining, Xenopus embryoswere fixed with 4% PFA in PBS and bleached in 0.5× SSC with 5% formamideand 1.2% hydrogen peroxide for 2 h on a light box. Samples were thenwashed in 100% methanol three times in 1 d, followed by two washes ofBABB in 1 d. To minimize experimental variation, littermate control andSox9CKO/CKO;ShhCre/+ mutant lungs, or Xenopus embryos of different stageswere embedded in the same OCT block and processed on the same section,or processed in the same tube for each riboprobe throughout the in situhybridization experiment. Images of sections and whole-mount stainingswere taken on an upright microscope (BX60, Olympus), or a stereoscope(M80, Leica).

EdU Staining and CCND1 Cell Count. EdU staining was performed using a Click-iT EdU imaging kit (C10337, Invitrogen). E14 pregnant mice were given0.1 mg EdU by intraperitoneal injection and embryos were harvested 1.5 hafter injection. EdU section staining was incorporated with the sectionimmunostaining protocol described above. After removing secondary anti-bodies, EdU reaction was developed on slides for 30 min according to themanufacturer’s instructions. Confocal images were taken of matchingbranch tips [RCd lobar and RCd.L1 branches (2)] of littermate control andSox9CKO/CKO;ShhCre/+ mutant lungs. At least 200 cells from five sections werecounted for each branch tip. Branches of the L.L1–L5 branch lineages (2)from E13 littermate control and Sox9CKO/CKO;ShhCre/+ mutant lungs wereused for CCND1 cell count. At least 250 cells from seven branch tips werecounted. Cell counting was performed on epithelial cells of the distal half ofeach branch tip, where SOX9 was normally expressed at a high level. Imageswere analyzed on the FLUOVIEW Viewer software (Olympus).

FACS. Embryonic lungs were dissected free of extrapulmonary tissues, mincedinto 1-mm pieces with forceps, and digested with 2 mg/mL Collagenase type I(Worthington, CLS-1) and 0.2 mg/mL DNase I (Worthington, D) for 20 (E14lungs) to 40 (E19 lungs) min at 37 °C with frequent agitation. An equalvolume of solution containing 0.25% Trypsin/EDTA (Invitrogen, 25200–056)and 0.2 mg/mL DNase I (Worthington, D) were added and incubated foranother 8 min at 37 °C with frequent agitation. After digestion, FBS (Invi-trogen, 10082–139) were added to a final concentration of 10% (vol/vol) andthe samples were triturated by pipetting for 20∼40 times. Dissociated cellswere washed with PBS with 1% FBS and sorted on a BD Biosciences Influxsorter equipped with 488 and 561 lasers.

18050 | www.pnas.org/cgi/doi/10.1073/pnas.1311760110 Chang et al.

Dow

nloa

ded

by g

uest

on

Nov

embe

r 2,

202

0

Page 10: Lung epithelial branching program antagonizes alveolar ... · Lung epithelial branching program antagonizes alveolar differentiation Daniel R. Changa, Denise Martinez Alanisa, Rachel

Microarray Expression Profiling. RNA was extracted from at least 2 × 105 FACSpurified epithelial cells or lungs dissected free of extrapulmonary tissuesfrom E13, E14, and E15 littermate control and Sox9CKO/CKO;ShhCre/+ mutantembryos using TRIzol reagents (15596018, Invitrogen) and an RNeasy Minikit (74104, Qiagen). Labeled cRNAs were generated using an IlluminarTotalPrep RNA amplification kit (AMIL1791, Invitrogen) and hybridized toIllumina mouse WG-6 v2 expression beadchips (BD-201–0202, Illumina). Theexperiments were repeated with lungs from independent litters. The ex-pression values from the microarrays were normalized using rank invariantnormalization and analyzed using the R statistical software.

ACKNOWLEDGMENTS. We thank Richard Behringer and William Klein forsuggestions and comments on the manuscript, and David Pollock for assistancewith the microarray experiments. The University of Texas MD Anderson CancerCenter DNA Analysis Facility and Flow Cytometry and Cellular Imaging CoreFacility are supported by the Cancer Center Support Grant (CA 16672). Thiswork was supported by The University of Texas System Rising STARS (Scienceand Technology Acquisition and Retention) award, The University of Texas MDAnderson Cancer Center Start-up Fund, and The University of Texas MDAnderson Cancer Center SPORE (Specialized Programs of Research Excel-lence) in Lung Cancer Career Development Award (to J.C.). R.K.M. wassupported by National Institute of Diabetes and Digestive and Kidney Dis-eases Career Development Award K01 DK092320.

1. Morrisey EE, Hogan BL (2010) Preparing for the first breath: Genetic and cellularmechanisms in lung development. Dev Cell 18(1):8–23.

2. Metzger RJ, Klein OD, Martin GR, Krasnow MA (2008) The branching programme ofmouse lung development. Nature 453(7196):745–750.

3. De Moerlooze L, et al. (2000) An important role for the IIIb isoform of fibroblastgrowth factor receptor 2 (FGFR2) in mesenchymal-epithelial signalling during mouseorganogenesis. Development 127(3):483–492.

4. Sekine K, et al. (1999) Fgf10 is essential for limb and lung formation. Nat Genet 21(1):138–141.

5. Tang N, Marshall WF, McMahon M, Metzger RJ, Martin GR (2011) Control of mitoticspindle angle by the RAS-regulated ERK1/2 pathway determines lung tube shape.Science 333(6040):342–345.

6. CardosoWV, Lü J (2006) Regulation of early lung morphogenesis: Questions, facts andcontroversies. Development 133(9):1611–1624.

7. Ward HE, Nicholas TE (1984) Alveolar type I and type II cells. Aust N Z J Med 14(5,Suppl 3):731–734.

8. Martis PC, et al. (2006) C/EBPalpha is required for lung maturation at birth. De-velopment 133(6):1155–1164.

9. Bird AD, et al. (2011) cAMP response element binding protein is required for differ-entiation of respiratory epithelium during murine development. PLoS ONE 6(3):e17843.

10. O’Brien KB, et al. (2010) CARM1 is required for proper control of proliferation anddifferentiation of pulmonary epithelial cells. Development 137(13):2147–2156.

11. Manwani N, et al. (2010) Reduced viability of mice with lung epithelial-specificknockout of glucocorticoid receptor. Am J Respir Cell Mol Biol 43(5):599–606.

12. Habermehl D, et al. (2011) Glucocorticoid activity during lung maturation is es-sential in mesenchymal and less in alveolar epithelial cells. Mol Endocrinol 25(8):1280–1288.

13. Nechiporuk T, Klezovitch O, Nguyen L, Vasioukhin V (2013) Dlg5 maintains apicalaPKC and regulates progenitor differentiation during lung morphogenesis. Dev Biol377(2):375–384.

14. El-Hashash AH, Al Alam D, Turcatel G, Bellusci S, Warburton D (2011) Eyes absent 1(Eya1) is a critical coordinator of epithelial, mesenchymal and vascular morphogenesisin the mammalian lung. Dev Biol 350(1):112–126.

15. Shu W, et al. (2005) Wnt/beta-catenin signaling acts upstream of N-myc, BMP4, andFGF signaling to regulate proximal-distal patterning in the lung. Dev Biol 283(1):226–239.

16. Harris KS, Zhang Z, McManus MT, Harfe BD, Sun X (2006) Dicer function is essentialfor lung epithelium morphogenesis. Proc Natl Acad Sci USA 103(7):2208–2213.

17. Chen J, Krasnow MA (2012) Integrin Beta 1 suppresses multilayering of a simple ep-ithelium. PLoS ONE 7(12):e52886.

18. Madisen L, et al. (2010) A robust and high-throughput Cre reporting and character-ization system for the whole mouse brain. Nat Neurosci 13(1):133–140.

19. Que J, Luo X, Schwartz RJ, Hogan BL (2009) Multiple roles for Sox2 in the developingand adult mouse trachea. Development 136(11):1899–1907.

20. Arnold K, et al. (2011) Sox2(+) adult stem and progenitor cells are important for tissueregeneration and survival of mice. Cell Stem Cell 9(4):317–329.

21. Perl AK, Kist R, Shan Z, Scherer G, Whitsett JA (2005) Normal lung development andfunction after Sox9 inactivation in the respiratory epithelium. Genesis 41(1):23–32.

22. French LE, et al. (1993) Murine clusterin: Molecular cloning and mRNA localization ofa gene associated with epithelial differentiation processes during embryogenesis.J Cell Biol 122(5):1119–1130.

23. Rajagopal J, et al. (2008) Wnt7b stimulates embryonic lung growth by coordinatelyincreasing the replication of epithelium and mesenchyme. Development 135(9):1625–1634.

24. Liu Y, Hogan BL (2002) Differential gene expression in the distal tip endoderm of theembryonic mouse lung. Gene Expr Patterns 2(3–4):229–233.

25. Lin S, et al. (2008) Misexpression of MIA disrupts lung morphogenesis and causesneonatal death. Dev Biol 316(2):441–455.

26. Sharpe J, et al. (2002) Optical projection tomography as a tool for 3D microscopy andgene expression studies. Science 296(5567):541–545.

27. Small EM, Vokes SA, Garriock RJ, Li D, Krieg PA (2000) Developmental expression ofthe Xenopus Nkx2-1 and Nkx2-4 genes. Mech Dev 96(2):259–262.

28. Hyatt BA, Resnik ER, Johnson NS, Lohr JL, Cornfield DN (2007) Lung specific de-velopmental expression of the Xenopus laevis surfactant protein C and B genes. GeneExpr Patterns 7(1–2):8–14.

29. Jackson EL, et al. (2001) Analysis of lung tumor initiation and progression usingconditional expression of oncogenic K-ras. Genes Dev 15(24):3243–3248.

30. Soeda T, et al. (2010) Sox9-expressing precursors are the cellular origin of the cruciateligament of the knee joint and the limb tendons. Genesis 48(11):635–644.

31. Muzumdar MD, Tasic B, Miyamichi K, Li L, Luo L (2007) A global double-fluorescentCre reporter mouse. Genesis 45(9):593–605.

32. Kranenburg O (2005) The KRAS oncogene: Past, present, and future. Biochim BiophysActa 1756(2):81–82.

33. Johnson L, et al. (1997) K-ras is an essential gene in the mouse with partial functionaloverlap with N-ras. Genes Dev 11(19):2468–2481.

34. Reginensi A, et al. (2011) SOX9 controls epithelial branching by activating RET ef-fector genes during kidney development. Hum Mol Genet 20(6):1143–1153.

35. Seymour PA, et al. (2012) A Sox9/Fgf feed-forward loop maintains pancreatic organidentity. Development 139(18):3363–3372.

36. Panico F, Rizzi F, Fabbri LM, Bettuzzi S, Luppi F (2009) Clusterin (CLU) and lung cancer.Adv Cancer Res 105:63–76.

37. Taniguchi H, et al. (2011) A resource of Cre driver lines for genetic targeting ofGABAergic neurons in cerebral cortex. Neuron 71(6):995–1013.

38. Sherr CJ, Roberts JM (1999) CDK inhibitors: Positive and negative regulators of G1-phase progression. Genes Dev 13(12):1501–1512.

39. Tompkins DH, et al. (2009) Sox2 is required for maintenance and differentiation ofbronchiolar Clara, ciliated, and goblet cells. PLoS ONE 4(12):e8248.

40. Shannon JM (1994) Induction of alveolar type II cell differentiation in fetal trachealepithelium by grafted distal lung mesenchyme. Dev Biol 166(2):600–614.

41. Salaun B, et al. (2004) CD208/dendritic cell-lysosomal associated membrane protein isa marker of normal and transformed type II pneumocytes. Am J Pathol 164(3):861–871.

42. Ueno T, et al. (2004) Processing of pulmonary surfactant protein B by napsin andcathepsin H. J Biol Chem 279(16):16178–16184.

43. Whitsett JA, Wert SE, Weaver TE (2010) Alveolar surfactant homeostasis and thepathogenesis of pulmonary disease. Annu Rev Med 61:105–119.

44. Agrons GA, Courtney SE, Stocker JT, Markowitz RI (2005) From the archives of theAFIP: Lung disease in premature neonates: radiologic-pathologic correlation. Radio-graphics 25(4):1047–1073.

45. Warburton D, et al. (2010) Lung organogenesis. Curr Top Dev Biol 90:73–158.46. Marose TD, Merkel CE, McMahon AP, Carroll TJ (2008) Beta-catenin is necessary to

keep cells of ureteric bud/Wolffian duct epithelium in a precursor state. Dev Biol314(1):112–126.

47. Bridgewater D, et al. (2008) Canonical WNT/beta-catenin signaling is required forureteric branching. Dev Biol 317(1):83–94.

48. Harfe BD, et al. (2004) Evidence for an expansion-based temporal Shh gradient inspecifying vertebrate digit identities. Cell 118(4):517–528.

49. Bi W, et al. (2001) Haploinsufficiency of Sox9 results in defective cartilage primordiaand premature skeletal mineralization. Proc Natl Acad Sci USA 98(12):6698–6703.

50. Yu K, et al. (2003) Conditional inactivation of FGF receptor 2 reveals an essential rolefor FGF signaling in the regulation of osteoblast function and bone growth. De-velopment 130(13):3063–3074.

51. Sive HL, Grainger RM, Harland RM (2000) Early Development of Xenopus laevis: ALaboratory Manual (Cold Spring Harbor Lab Press, Cold Spring Harbor, NY).

52. Lyons JP, et al. (2009) Requirement of Wnt/beta-catenin signaling in pronephrickidney development. Mech Dev 126(3–4):142–159.

53. Chen J, Nathans J (2007) Estrogen-related receptor beta/NR3B2 controls epithelial cellfate and endolymph production by the stria vascularis. Dev Cell 13(3):325–337.

Chang et al. PNAS | November 5, 2013 | vol. 110 | no. 45 | 18051

DEV

ELOPM

ENTA

LBIOLO

GY

FEATU

REART

ICLE

SEECO

MMEN

TARY

Dow

nloa

ded

by g

uest

on

Nov

embe

r 2,

202

0