Top Banner
STEM CELLS AND REGENERATION RESEARCH ARTICLE Niche-mediated BMP/SMAD signaling regulates lung alveolar stem cell proliferation and differentiation Mei-I Chung 1 , Melissa Bujnis 1 , Christina E. Barkauskas 2 , Yoshihiko Kobayashi 1 and Brigid L. M. Hogan 1, * ABSTRACT The bone morphogenetic protein (BMP) signaling pathway, including antagonists, functions in lung development and regeneration of tracheal epithelium from basal stem cells. Here, we explore its role in the alveolar region, where type 2 epithelial cells (AT2s) and Pdgfrα + type 2-associated stromal cells (TASCs) are components of the stem cell niche. We use organoids and in vivo alveolar regrowth after pneumonectomy (PNX) a process that requires proliferation of AT2s and differentiation into type 1 cells (AT1s). BMP signaling is active in AT2s and TASCs, transiently declines post-PNX in association with upregulation of antagonists, and is restored during differentiation of AT2s to AT1s. In organoids, BMP4 inhibits AT2 proliferation, whereas antagonists (follistatin, noggin) promote AT2 self-renewal at the expense of differentiation. Gain- and loss-of-function genetic manipulation reveals that reduced BMP signaling in AT2s after PNX allows self-renewal but reduces differentiation; conversely, increased BMP signaling promotes AT1 formation. Constitutive BMP signaling in Pdgfrα + cells reduces their AT2 support function, both after PNX and in organoid culture. Our data reveal multiple cell-type-specific roles for BMP signaling during alveolar regeneration. KEY WORDS: Lung, Alveolar epithelium, AT1, AT2, BMP, Smad1/5/8, Follistatin, Noggin, Regeneration, Compensatory regrowth, Pneumonectomy INTRODUCTION The lung is a complex organ comprising a highly branched system of air-conducting tubes terminating in millions of air-exchanging units called alveoli. The alveolar epithelium is composed of two distinct cell types: type 1 and type 2 cells. Type 1 alveolar epithelial cells (AT1s) are very large, thin squamous cells that cover about 95% of the internal surface of the lung and are important for gas exchange between the air and blood in the capillaries. Type 2 alveolar epithelial cells (AT2s) are cuboidal and characterized by the production and secretion of pulmonary surfactant, preventing lung collapse during exhalation. At steady state, alveolar cell turnover is low. However, efficient repair and regeneration has been reported following cellular damage or increased functional demand, both in animal models (see below) and humans (Butler et al., 2012; Kumar et al., 2011; Toufen et al., 2011). Adult AT2s, as a population, have been recognized as alveolar stem/progenitor cells capable of both self-renewal and differentiation into AT1s (Barkauskas et al., 2013; Desai et al., 2014; Evans et al., 1973; Hogan et al., 2014; Kapanci et al., 1969; Rock et al., 2011). The microenvironment in which AT2s reside encompasses a number of different cell types, including AT1s, Pdgfrα + and Pdgfrβ + stromal cells, endothelial cells, and immune cells. A number of studies have explored roles for these different components in models of lung repair and regeneration (Barkauskas et al., 2013; Chen et al., 2012; Ding et al., 2011; Jain et al., 2015a; Lechner et al., 2017; Lee et al., 2014; Liu et al., 2015, 2016; Nabhan et al., 2018; Rafii et al., 2015; Zacharias et al., 2018; Zepp et al., 2017). One such model is alveolar regrowth after pneumonectomy (PNX): the surgical removal of one or more lung lobes. This procedure, in different species, leads to compensatory regrowth of the remaining lung tissue, with formation of new blood vessels, epithelial and mesenchymal cells, and alveolar septa in order to restore alveolar number and surface area. In murine lungs, regrowth is achieved by 21 days after surgery, with the peak of AT2 proliferation occurring at around 7 days (Buhain and Brody, 1973; Butler et al., 2012; Green et al., 2016; Hsia et al., 1994; Thane et al., 2014). So far, several factors and signaling pathways have been identified that promote the proliferation of AT2s after PNX. These include mechanical tension-induced YAP activation, EGF-related peptides released from the extracellular matrix by metalloproteinases secreted by endothelial cells in response to platelet-derived SDF1 signaling, and paracrine signals from activated macrophages (Ding et al., 2011; Lechner et al., 2017; Liu et al., 2016; Rafii et al., 2015). Compensatory regrowth involves not only increased proliferation of AT2s but also formation of new AT1s to restore alveolar surface area and pulmonary function. This conclusion is supported by the fact that a reduction in both AT2 proliferation and AT2 to AT1 differentiation is associated with impaired compensatory regrowth in the absence of YAP or activated macrophages (Lechner et al., 2017; Liu et al., 2016). One outstanding issue relevant to the biology of alveolar regrowth is the identity of all of the niche factors that promote AT2 cell proliferation and differentiation, and the cells producing and receiving them. Here, we use both 3D organoid culture and in vivo studies to examine the role of BMP signaling in the AT2 stem cell niche. We find that post-PNX, Smad-dependent BMP signaling is transiently reduced in both AT2s and the Pdgfrα + cells adjacent to them [referred to here as TASCs (type 2-associated stromal cells)]. This modulation involves changes in both BMP receptor levels and the upregulation of genes encoding BMP antagonists. Gain- and loss- of-function genetic manipulation in vivo reveals that loss of BMP signaling in AT2s after PNX allows their self-renewal but significantly reduces their ability to give rise to AT1s; conversely, increased BMP signaling promotes AT1 differentiation. Focusing on the contribution of the stroma to AT2 behavior, we provide Received 3 January 2018; Accepted 4 April 2018 1 Department of Cell Biology, Duke University Medical School, Durham, NC 27710, USA. 2 Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Duke University Medical School, Durham, NC 27710, USA. *Author for correspondence ([email protected]) B.L.M.H., 0000-0002-7916-1573 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed. 1 © 2018. Published by The Company of Biologists Ltd | Development (2018) 145, dev163014. doi:10.1242/dev.163014 DEVELOPMENT
10

Niche-mediated BMP/SMAD signaling regulates lung alveolar ...STEM CELLS AND REGENERATION RESEARCH ARTICLE Niche-mediated BMP/SMAD signaling regulates lung alveolar stem cell proliferation

Oct 11, 2020

Download

Documents

dariahiddleston
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Niche-mediated BMP/SMAD signaling regulates lung alveolar ...STEM CELLS AND REGENERATION RESEARCH ARTICLE Niche-mediated BMP/SMAD signaling regulates lung alveolar stem cell proliferation

STEM CELLS AND REGENERATION RESEARCH ARTICLE

Niche-mediated BMP/SMAD signaling regulates lung alveolarstem cell proliferation and differentiationMei-I Chung1, Melissa Bujnis1, Christina E. Barkauskas2, Yoshihiko Kobayashi1 and Brigid L. M. Hogan1,*

ABSTRACTThe bone morphogenetic protein (BMP) signaling pathway, includingantagonists, functions in lung development and regeneration oftracheal epithelium from basal stem cells. Here, we explore its role inthe alveolar region, where type 2 epithelial cells (AT2s) and Pdgfrα+

type 2-associated stromal cells (TASCs) are components of the stemcell niche. We use organoids and in vivo alveolar regrowth afterpneumonectomy (PNX) – a process that requires proliferation of AT2sand differentiation into type 1 cells (AT1s). BMP signaling is active inAT2s and TASCs, transiently declines post-PNX in association withupregulation of antagonists, and is restored during differentiation ofAT2s to AT1s. In organoids, BMP4 inhibits AT2 proliferation, whereasantagonists (follistatin, noggin) promote AT2 self-renewal at theexpense of differentiation. Gain- and loss-of-function geneticmanipulation reveals that reduced BMP signaling in AT2s after PNXallows self-renewal but reduces differentiation; conversely, increasedBMP signaling promotes AT1 formation. Constitutive BMP signalingin Pdgfrα+ cells reduces their AT2 support function, both after PNXand in organoid culture. Our data reveal multiple cell-type-specificroles for BMP signaling during alveolar regeneration.

KEYWORDS: Lung, Alveolar epithelium, AT1, AT2, BMP, Smad1/5/8,Follistatin, Noggin, Regeneration, Compensatory regrowth,Pneumonectomy

INTRODUCTIONThe lung is a complex organ comprising a highly branched systemof air-conducting tubes terminating in millions of air-exchangingunits called alveoli. The alveolar epithelium is composed of twodistinct cell types: type 1 and type 2 cells. Type 1 alveolar epithelialcells (AT1s) are very large, thin squamous cells that cover about95% of the internal surface of the lung and are important for gasexchange between the air and blood in the capillaries. Type 2alveolar epithelial cells (AT2s) are cuboidal and characterized by theproduction and secretion of pulmonary surfactant, preventing lungcollapse during exhalation.At steady state, alveolar cell turnover is low. However, efficient

repair and regeneration has been reported following cellular damageor increased functional demand, both in animal models (see below)and humans (Butler et al., 2012; Kumar et al., 2011; Toufen et al.,

2011). Adult AT2s, as a population, have been recognized asalveolar stem/progenitor cells capable of both self-renewal anddifferentiation into AT1s (Barkauskas et al., 2013; Desai et al.,2014; Evans et al., 1973; Hogan et al., 2014; Kapanci et al., 1969;Rock et al., 2011). The microenvironment in which AT2s resideencompasses a number of different cell types, including AT1s,Pdgfrα+ and Pdgfrβ+ stromal cells, endothelial cells, and immunecells. A number of studies have explored roles for these differentcomponents in models of lung repair and regeneration (Barkauskaset al., 2013; Chen et al., 2012; Ding et al., 2011; Jain et al., 2015a;Lechner et al., 2017; Lee et al., 2014; Liu et al., 2015, 2016; Nabhanet al., 2018; Rafii et al., 2015; Zacharias et al., 2018; Zepp et al.,2017). One such model is alveolar regrowth after pneumonectomy(PNX): the surgical removal of one or more lung lobes. Thisprocedure, in different species, leads to compensatory regrowth ofthe remaining lung tissue, with formation of new blood vessels,epithelial and mesenchymal cells, and alveolar septa in order torestore alveolar number and surface area. In murine lungs, regrowthis achieved by 21 days after surgery, with the peak of AT2proliferation occurring at around 7 days (Buhain and Brody, 1973;Butler et al., 2012; Green et al., 2016; Hsia et al., 1994; Thane et al.,2014).

So far, several factors and signaling pathways have been identifiedthat promote the proliferation of AT2s after PNX. These includemechanical tension-induced YAP activation, EGF-related peptidesreleased from the extracellular matrix by metalloproteinases secretedby endothelial cells in response to platelet-derived SDF1 signaling,and paracrine signals from activated macrophages (Ding et al., 2011;Lechner et al., 2017; Liu et al., 2016; Rafii et al., 2015).Compensatory regrowth involves not only increased proliferation ofAT2s but also formation of new AT1s to restore alveolar surface areaand pulmonary function. This conclusion is supported by the fact thata reduction in both AT2 proliferation and AT2 to AT1 differentiationis associated with impaired compensatory regrowth in the absence ofYAPor activatedmacrophages (Lechner et al., 2017; Liu et al., 2016).One outstanding issue relevant to the biology of alveolar regrowthis the identity of all of the niche factors that promote AT2 cellproliferation and differentiation, and the cells producing andreceiving them.

Here, we use both 3D organoid culture and in vivo studies toexamine the role of BMP signaling in the AT2 stem cell niche. Wefind that post-PNX, Smad-dependent BMP signaling is transientlyreduced in both AT2s and the Pdgfrα+ cells adjacent to them[referred to here as TASCs (type 2-associated stromal cells)]. Thismodulation involves changes in both BMP receptor levels and theupregulation of genes encoding BMP antagonists. Gain- and loss-of-function genetic manipulation in vivo reveals that loss of BMPsignaling in AT2s after PNX allows their self-renewal butsignificantly reduces their ability to give rise to AT1s; conversely,increased BMP signaling promotes AT1 differentiation. Focusingon the contribution of the stroma to AT2 behavior, we provideReceived 3 January 2018; Accepted 4 April 2018

1Department of Cell Biology, Duke University Medical School, Durham, NC 27710,USA. 2Division of Pulmonary, Allergy and Critical Care Medicine, Department ofMedicine, Duke University Medical School, Durham, NC 27710, USA.

*Author for correspondence ([email protected])

B.L.M.H., 0000-0002-7916-1573

This is an Open Access article distributed under the terms of the Creative Commons AttributionLicense (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use,distribution and reproduction in any medium provided that the original work is properly attributed.

1

© 2018. Published by The Company of Biologists Ltd | Development (2018) 145, dev163014. doi:10.1242/dev.163014

DEVELO

PM

ENT

Page 2: Niche-mediated BMP/SMAD signaling regulates lung alveolar ...STEM CELLS AND REGENERATION RESEARCH ARTICLE Niche-mediated BMP/SMAD signaling regulates lung alveolar stem cell proliferation

evidence that they are a source of BMP antagonists and thatconstitutive BMP signaling in Pdgfrα+ fibroblasts reduces theability of these cells to support AT2 proliferation, both in vivo andin vitro. Taken together, our studies thus establish multiple,dynamic, cell-type-specific roles for BMP signaling in regulatingadult alveolar regeneration.

RESULTSDynamic BMP signaling in the AT2 niche during alveolarregrowth in vivoTo explore whether BMP signaling plays a role in regulating lungalveolar regrowth after PNX, we examined canonical BMPsignaling before and after removing the left lobe. At steady state,immunofluorescence analysis for phospho-Smad1/5/8 (pSmad1/5/8)shows that BMP signaling is active in many alveolar cells, includingAT2s (77.9±0.8% of SFTPC+ cells) and Pdgfrα-H2B:GFP+ TASCsadjacent to them (66.9±2.1%) (Fig. 1A). Confocal microscopyshows that these TASCs have a characteristic morphology, withlong cellular extensions (Fig. S1 and Movie 1). pSmad1/5/8expression was also seen in AT1s (85.0±1.5% of HOPX+ cells),endomucin+ endothelial cells (64.5±0.9%) and Pdgfrb+ alveolarstromal cells (46.2±0.9%) (Fig. S2).At 7 days post-PNX, around the peak of AT2 proliferation (Brody

et al., 1978; Ding et al., 2011), the number of pSmad1/5/8+ AT2s

was significantly reduced (Fig. 1B). However, by 2 weeks post-PNX, when proliferation is minimal and AT2s are robustlydifferentiating into AT1s (see Fig. 5 and Fig. S3), the proportionof pSmad1/5/8+ AT2s has returned closer to steady state levels(Fig. 1B). A similar result was seen in Pdgfrα+ TASCs in which thelevel of pSmad1/5/8+ expression was reduced on day 7 and restoredon day 14 (Fig. 1B). A small reduction in pSmad1/5/8 was also seenin endomucin+ endothelial cells but no significant change wasobserved in either Pdgfrb+ stromal cells or AT1 cells (Fig. S2). Thedynamic change in overall BMP signaling was confirmed usingwestern blot analysis of pSmad1/5/8 levels in whole-lung lysates(Fig. 1C).

To better understand the mechanisms underlying the transientchange in BMP signaling, qPCR analysis was used to follow thedifferential expression of pathway components, including ligands,receptors and antagonists. As shown in Fig. 1D, the expression ofBmp6 and Bmpr2was significantly reduced in AT2s on days 4, 7 and14 post-PNX, whileBmp2 andBmpr1a levels were reduced on days 4and 7. A similar trend was also seen in the expression of Bmp6 andBmpr2 in Pdgfrα+ cells. Significantly, transcripts encoding BMPantagonists, including follistatin (Fst) and follistatin-like 1 (Fstl1),were strongly upregulated in Pdgfrα+ cells. Some increase in the lowlevels of Grem1 transcripts was detected (Fig. S2) but there was noapparent change in the expression of Grem2 (which encodes an

Fig. 1. Dynamic BMPsignaling in theAT2 niche during alveolarregrowth in vivo. (A) Expression of phospho-Smad1/5/8(pSmad1/5/8), SFTPC, Pdgfrα-H2b:GFPandHOPX in the alveolarregion of the adult mouse lung, as revealed byimmunofluorescence and confocal microscopy. Left: SFTPC, H2b:GFP and nuclei (DAPI). Middle: same field of view, exposed toshow pSmad1/5/8 and DAPI. Yellow arrows indicate Pdgfrα+ cellsthat are pSmad1/5/8+, while the yellow arrowheads indicate aPdgfrα+ cell that is pSmad1/5/8−. Right: HOPX is both cytoplasmicand nuclear. White arrows indicate pSmad1/5/8+ AT1s. Scale bars:20 μm. (B) Expression of pSmad1/5/8 (magenta) in SFTPC+ cells(red) and Pdgfrα-H2b:GFP+ cells in the alveolar region at steadystate and 7 and 14 days after PNX. Arrows indicate AT2s adjacentto Pdgfrα+ TASCs. Scale bars: 50 μm. (C) pSmad1/5/8 levels inextracts of lungs after PNX. β-Actin is loading control.(D) qRT-PCR for transcripts of BMP signaling genes in lineage-labeled AT2s (left) and Pdgfrα-H2b:GFP stromal cells (right) atsteady state and at times after PNX. The mRNA expression levelsof PNX samples were normalized to steady state. Data are mean±s.e.m. *P<0.05, **P<0.01, ***P<0.001.

2

STEM CELLS AND REGENERATION Development (2018) 145, dev163014. doi:10.1242/dev.163014

DEVELO

PM

ENT

Page 3: Niche-mediated BMP/SMAD signaling regulates lung alveolar ...STEM CELLS AND REGENERATION RESEARCH ARTICLE Niche-mediated BMP/SMAD signaling regulates lung alveolar stem cell proliferation

antagonist implicated by others in promoting AT2 growth (Zeppet al., 2017) (Fig. 1D).

Pharmacological modulation of BMP signaling alters AT2proliferation and differentiation in 3D organoid culturesThe transient downregulation of BMP signaling in AT2s early in theregeneration process suggests that the pathway regulates either theproliferation or differentiation of AT2s, or both. To explorethese possibilities, we used an ‘alveolosphere’ organoid assay(Barkauskas et al., 2013) in which AT2s, lineage labeled usingSftpc-CreERT2; Rosa26-tdTomato alleles, are co-cultured in 3Dwith Pdgfrα-H2b:GFP+ stromal cells, with or without recombinantBMP ligands or antagonists in the medium. We then determined thecolony-forming efficiency (CFE) on day 14 post culture bycounting the number of spheres >45 μm in diameter (Barkauskaset al., 2013). We observed a significant decrease in CFE in thepresence of 20-50 ng/ml BMP4 (Fig. 2A) and a similar effect wasseen with BMP2 (Fig. S4). By contrast, there was no significanteffect with either BMP5 or BMP6 (Fig. S4A). At both day 7 and 14,the colonies incubated with 50 ng/ml BMP4 were much smallerthan controls (Fig. 2A,B). EdU incorporation during a short pulse(2 h before harvest) on day 7 showed that AT2 proliferation issignificantly reduced (50%) in the presence of BMP4 comparedwith controls (Fig. 2B).Our expression studies indicated that genes encoding the BMP

antagonists Fst and Fstl1 are dynamically expressed in regeneratingalveolar niche cells.We therefore tested the effect of BMPantagonistsin the alveolar organoid system. Analysis of cultures at day 14indicated no apparent difference in CFE and organoid size (diameter)after treatment with FST, FSTL1 and NOGGIN, compared with

controls (Fig. 2C). However, immunohistochemistry of histologicalsections clearly showed that all three antagonists gave a ∼50%reduction in the percentage of tdTomato+ lineage-labeled cells that areHOPX+ AT1s (Fig. 2D).

In the course of these studies, we observed that within 2 days oflineage-labeled AT2s being placed in the organoid culture system, thecells co-express both the AT2 marker SFTPC and the AT1 markerAGER (advanced glycosylation end product-specific receptor).AGER was assayed by both immunohistochemistry and byexpression of a new Ager-H2b:Venus knock-in allele (Fig. S8).Most of the cells remain dual positive at day 7. By day 14, however,AGER+ cells, that are also HOPX+, are predominantly found in theinterior of the spheres with the characteristic elongated morphology ofAT1s. By contrast, cuboidal SFTPC+ cells are found towards theoutside. At day 14, only a small proportion (about 2.6%±0.6%) ofthe total cells in control spheres are dual positive. By contrast, in theorganoids treated with the BMP antagonist FST, not only is theproportion of SFTPC+ AT2s increased relative to controls but so too isthe proportion of dual positive cells: to 5.5±0.9%of the total (Fig. 2D).

Taken together, our results suggest that in the organoid assayexcess BMP ligand negatively regulates AT2 proliferation. Bycontrast, inhibiting the BMP signaling pathway reduces thedifferentiation of AT2s to AT1s.

Enhanced BMPR1a-dependent signaling increasesdifferentiation of AT2s to AT1s in organoids, upregulates AT1genes and reduces trophic activity of Pdgfrα+ fibroblastsAs both AT2 and Pdgfrα+ stromal cells express Bmp receptors(Fig. 1D), each cell type has the potential to be affected by exogenousBMP ligands and antagonists in the organoid assay. We therefore

Fig. 2. Effect of BMP ligands and antagonists on AT2 cellproliferation and differentiation in 3D organoid culture. (A) Leftthree panels: typical day 14 alveolosphere cultures, with andwithout BMP4. Graphs quantitate the effect of BMP4 on CFE andorganoid size. (B) Effect of 50 ng/ml BMP4 on proliferation ofSFTPC+ cells in spheres at 7 days, as judged by incorporation ofEdU over a 2 h period. Scale bars: 20 μm. (C) Day 14 spherescultured with BMP antagonists FST and FSTL1 (500 ng/ml) andNoggin (1 μg/ml). No significant difference in CFE was seen. (D)Immunofluorescence analysis of SFTPC+ (AT2s) and HOPX+

(AT1s) revealed a reduction in AT2 to AT1 differentiation in spheresexposed to different BMP antagonists for 14 days. Left graphshows the percentage of total cells in multiple spheres that areHOPX+. Right graph shows the percentage of total cells that areSFTPC+, HOPX+, and SFTPC+ HOPX+, as judged byimmunofluorescence of sections. For all experiments, n=3animals. Data are mean±s.e.m. *P<0.05, ***P<0.001; n.s., notsignificant. Scale bars: 50 μm.

3

STEM CELLS AND REGENERATION Development (2018) 145, dev163014. doi:10.1242/dev.163014

DEVELO

PM

ENT

Page 4: Niche-mediated BMP/SMAD signaling regulates lung alveolar ...STEM CELLS AND REGENERATION RESEARCH ARTICLE Niche-mediated BMP/SMAD signaling regulates lung alveolar stem cell proliferation

used genetic strategies to determine the effect of upregulating orinhibitingBMP signaling in each cell population separately (Fig. 3A).To constitutively activate BMPR1a-dependent signaling in AT2s andto simultaneously lineage trace them, we generated mice with thegenotype Sftpc-CreERT2; Rosa26-tdTm/Rosa26-caBmpr1a andexposed them to tamoxifen (Tmx). Enhanced BMP signaling in theAT2 population (hereafter AT2CAB) was confirmed by qPCRanalysis, which indicated significantly increased transcripts of BMPdownstream genes, including Id1, Id2 and Smad6, compared withcontrol AT2CTRL isolated from mice lacking the Rosa26-caBmpr1aallele (Fig. S5A). When AT2CAB were co-cultured with wild-typePdgfrα+ cells, CFE was reduced at 14 days compared with controls(Fig. 3B). It was noted that AT2CAB gave rise to two populations ofalveolospheres based on sphere diameters (Fig. 3B). Analysis ofpSmad1/5/8 expression suggested that the larger colonies are derivedfromAT2s that had recombined theRosa26-tdTm but not theRosa26-caBmpr1a allele (Fig. S5D). Significantly, immunofluorescenceanalysis of the AT2CAB organoids found to have high pSmad1/5/8signals revealed a 1.5-fold increase in AT1 differentiation comparedwith AT2CTRL (Fig. 3B and Fig. S5D). To complement these results,we performed RNA sequencing analysis on AT2CAB cells isolated byFACS. As shown in Fig. S6, we found that 119 genes wereupregulated more than twofold compared with AT2CTRL. Of thesegenes, 10% are normally preferentially expressed in AT1 cells (Wanget al., 2018; www.lungmap.net). Of the 25 genes downregulatedmore than twofold in AT2CAB cells, 28% are preferentially expressedin AT2 cells (Fig. S6).The organoid experiments suggest that upregulation of BMP

signaling specifically in AT2s leads to a reduction in both CFE and

colony size. However, the values are still higher than in the organoidstreated with the highest dose of BMP4 ligand (Fig. 2A). Oneexplanation for this result is that BMP ligand also acts directly onPdgfrα+ stromal cells, reducing their ability to act as trophic support forAT2s. We tested this hypothesis by generating Pdgfrα-CreERT2;Rosa26-tdTomato/Rosa26-caBmpr1a mice (hereafter PdgfrαCAB),treating them with Tmx, isolating lineage-labeled Pdgfrα+ cells andusing them in co-culture assays. As shown in Fig. 3C, the PdgfrαCAB

stromal cells were much less efficient than PdgfrαCTRL cells insupportingCFE ofwild-typeAT2s. However, the relative proportion ofAT1s toAT2s in the organoids supported by PdgfrαCTRL or PdgfrαCAB

was the same, as analyzed by SFTPC and HOPX expression (Fig. 3C).Taken together, these results indicate that enhanced BMP signaling canfunction independently in both AT2s and stromal cells, with thecombined effect in organoid assays of reducing the self-renewal of AT2cells and promoting AT1 differentiation.

Cell type-specific deletion ofBmpr1a reduces differentiationof AT2s to AT1s in the organoid assayTo test the effect of loss of BMPR1a-mediated signaling in AT2s,we isolated lineage-labeled AT2s from Sftpc-CreERT2; Rosa26-tdTomato; Bmpr1afx/fx mice treated with Tmx (hereafter referred toas AT2Bmpr1afx/fx) and co-cultured them with wild-type Pdgfrα+

cells. Control studies confirmed that AT2Bmpr1afx/fx cells as apopulation have reduced expression of transcripts for Bmpr1a anddownstream target genes (Fig. S5B). As shown in Fig. 3D, reducedsignaling through BMPR1a receptor decreased the CFE ofAT2Bmpr1afx/fx cells, compared with control AT2Bmpr1afx/+ cells,and resulted in attenuated AT1 differentiation. By contrast, when

Fig. 3. Effect of BMP pathway activation orinhibition in AT2 or Pdgfrα+ cells on AT2 cellbehavior in organoid assays. (A) Schematic ofexperiments designed to co-culture different classesof AT2s and Pdgfrα+ cells in organoid assays. Mostcells were also lineage labeled using Rosa26-tdTomato. (B-E) Left: typical organoid cultures andsections immunostained for SFTPC (green) andHOPX (red). Right: quantification of CFEs andpercentage of total cells that are HOPX+.(B) Constitutively active BMP signaling in AT2sresults in lower CFE and enhanced AT1differentiation. (C) Constitutively active BMPsignaling in Pdgfrα+ cells reduces CFE but has noeffect on AT1 differentiation. (D) Conditional deletionof Bmpr1a in AT2s results in reduced CFE andattenuated AT1 differentiation. (E) Conditionaldeletion of Bmpr1a in Pdgfrα+ cells has no effects onCFE or differentiation. For all experiments, n=3animals. Data are mean±s.e.m. *P<0.05, **P<0.01;n.s., not significant. Scale bars: 50 μm.

4

STEM CELLS AND REGENERATION Development (2018) 145, dev163014. doi:10.1242/dev.163014

DEVELO

PM

ENT

Page 5: Niche-mediated BMP/SMAD signaling regulates lung alveolar ...STEM CELLS AND REGENERATION RESEARCH ARTICLE Niche-mediated BMP/SMAD signaling regulates lung alveolar stem cell proliferation

Bmpr1a was deleted in Pdgfrα+ cells, we observed no difference ineither CFE or AT1 differentiation (Fig. 3E).Together, our data show that either an increase or a decrease in

BMP signaling in AT2s results in reduced self-renewal, suggestingthat only a narrow range of BMP signaling is effective for AT2 self-renewal in the organoid assay. In addition, BMP signaling in AT2sis required for their efficient differentiation into AT1s.

In vivo, enhanced BMP signaling in Pdgfrα+ cells but not inAT2s reduces AT2 proliferation following PNXTo examine the effect of gain or loss of BMP signaling on AT2proliferation during alveolar regrowth in vivo, AT2CTRL, AT2CAB,AT2Bmpr1afx/+ and AT2Bmpr1afx/fx mice were pretreated with Tmx 2weeks before PNX. Given the strong effect of enhanced BMPsignaling on the CFE of isolated AT2s in vitro (Figs 2A and 3B), wehypothesized that constitutively active BMP signaling in AT2sin vivowould lead to a decrease in the number of EdU+ AT2s at day7 post-PNX. Surprisingly, at day 7 post-PNX, the number of EdU+

lineage-labeled AT2s was not significantly changed between

AT2CAB and AT2CTRL lungs, and in both cases there was asimilar increase compared with sham-operated controls (Fig. 4B,E).Likewise, we found no significant differences in the percentage ofEdU+ lineage-labeled AT2s in the AT2Bmpr1afx/fx lungs comparedwith AT2Bmpr1afx/+ (Fig. 4C,F).

We next asked whether activating BMP signaling in Pdgfrα+

cells, by pretreating Pdgfrα-CreERT2; Rosa26-tdTomato/Rosa26-caBmpr1amicewith Tmx, would have an effect on AT2 proliferationafter PNX. In PdgfrαCTRL mice, 8.6±0.7% of SFTPC+ cells wereEdU+ at 7 days post-PNX. This number was reduced to 5.5±0.6% inPdgfrαCAB lungs (Fig. 4D,G). Significantly, there was no change inthe EdU labeling of PdgfrαCAB cells compared with PdgfrαCTRL,suggesting that the reduced AT2 proliferation is independent ofstromal proliferation (Fig. 4H, Fig. S7).

BMP signaling in AT2s in vivo regulates their differentiationinto AT1cellsDuring alveolar regeneration in response to PNX, AT2s bothproliferate and give rise to AT1s (Fig. S3) (Lechner et al., 2017; Liu

Fig. 4. Manipulating BMP signaling in Pdgfrα+ cells, butnot in AT2s, reduces AT2 proliferation in vivo post-PNX.(A) Tmx was used to lineage label and conditionally activate ordelete Bmpr1a in AT2s or Pdgfrα+ cells. After an interval, PNXor sham surgery was performed and 7 days later mice weregiven a 3 h pulse of EdU. (B-D) Immunofluorescence of lungsections was then used to calculate the percentage of EdU+

lineage-labeled SFTPC+ AT2s (lineage labeling is not shown)and Pdgfrα+ cells. Lower panels are higher magnifications ofboxed areas in the panels above. (E,F) Analysis revealed nodifference in proliferation between AT2CAB and AT2CTRL cellsand AT2Bmpr1afx/+ and AT2Bmpr1afx/fx cells. (D,G) By contrast,activation of Bmpr1a in Pdgfrα+ cells led to reducedproliferation of AT2s in PdgfrαCAB versus PdgfrαCTRL lungs.(H) There is no difference in proliferation of stromal cells inPdgfrαCTRL and PdgfrαCAB lungs. For all experiments, n≥3animals/group. Data are mean±s.e.m. n.s., not mentioned.*P<0.05. n.s., not significant. Scale bars: 50 μm.

5

STEM CELLS AND REGENERATION Development (2018) 145, dev163014. doi:10.1242/dev.163014

DEVELO

PM

ENT

Page 6: Niche-mediated BMP/SMAD signaling regulates lung alveolar ...STEM CELLS AND REGENERATION RESEARCH ARTICLE Niche-mediated BMP/SMAD signaling regulates lung alveolar stem cell proliferation

et al., 2016), the majority of which are pSmad1/5/8 positive atsteady state (Fig. 1A). We therefore examined the effects ofenhanced BMP signaling on AT2 to AT1 differentiation in AT2CAB

versus AT2CTRL lungs after PNX (Fig. 5A). SFTPC and LAMP3, acomponent of lamellar bodies, were used to mark AT2s, whileHOPX identified AT1s. By 7 days post-PNX, 5.5±1.2% of lineage-traced AT2s in control lungs lost expression of an AT2 marker andhad differentiated into AT1s, as judged by immunofluorescenceanalysis for HOPX (Fig. 5B). Significantly, AT2CAB gave rise tomore AT1s (11.9±0.9%) at this time (Fig. 5D), a result consistentwith our observations in organoid cultures. However, this differencewas no longer apparent at day 21 (Fig. 5D).To further investigate the role of BMP signaling in AT1

differentiation, we then pretreated AT2Bmpr1afx/+ and AT2Bmpr1afx/fx

mice with Tmx and analyzed lungs 21 days after PNX. While22.9±3.9% of lineage-labeled AT2Bmpr1afx/+ cells gave rise to AT1s(LAMP-3− HOPX+), loss of Bmpr1a in AT2Bmpr1afx/fx resulted in asignificant decrease in AT1 differentiation (9.6±2.7%) (Fig. 5C,E).The phenotype of reduced AT1 differentiation is consistent with theresults seen in the organoid assay after treatment with BMPantagonists and downregulating Bmpr1a (Figs 2D and 3D).

DISCUSSIONA number of studies in mammalian systems have shown thatcanonical BMP signaling plays complex and differential roles inprogenitor cell proliferation and differentiation in tissues such as theepidermis, hair follicles and intestine (Arenkiel et al., 2011;Genander et al., 2014; Haramis et al., 2004; He et al., 2004; Hsuet al., 2014; Lewis et al., 2014). In adult mouse trachea undergoingrepair after deletion of luminal cells, BMP signaling inhibits theproliferation of basal stem cells but has no effect on theirdifferentiation into ciliated versus secretory lineages. Moreover, itappears that upregulation of BMP antagonists plays a key role inregulating the stem cell niche (Tadokoro et al., 2016). Here, wepresent evidence for dynamic changes in BMP signaling during theregrowth of the distal gas-exchange region of the lung followingPNX. In this model, formation of new alveoli involves theproliferation and differentiation of AT2s, with about 20% oflineage-labeled AT2 cells generating AT1 cells over 21 days(Fig. S3). In the quiescent adult lung, where there is little cellturnover, immunohistochemistry for nuclear pSmad1/5/8 clearlyshows that BMP signaling is active in the majority of AT2s andAT1s, as well as in about half of the Pdgfrα+ TASCs located adjacent

Fig. 5. BMP signaling is required for AT1 differentiation.(A) Tmx was used to lineage label and conditionally activate ordelete Bmpr1a in AT2s. After 14 days, PNX or sham surgerywas performed and 7 or 21 days later lungs were harvestedand analyzed by immunofluorescence for lineage label (usingan antibody to RFP), SFTPC or LAMP-3 (AT2 markers), andHOPX (an AT1 marker). (B) Top panels show expression oflineage label and SFTPC at 7 days post sham surgery or post-PNX in control lungs and lungs in which Bmpr1a wasconstitutively activated. Bottom panels are the same viewsexposed to show lineage trace and HOPX. Arrowheadsindicate cells losing an AT2 marker and gaining AT1expression and elongating morphology. (D) Quantificationshows that, at 7 days post-PNX, twofold more lineage-labeledAT2CAB cells had given rise to AT1s compared with AT2CTRL.However, at 21 days there was no significant difference.(C) Immunohistochemistry 21 days post-PNX of AT2Bmpr1afx/+

and AT2Bmpr1afx/fx lungs for RFP (lineage label), LAMP3 andHOPX. (E) Fewer AT1s were derived from AT2Bmpr1afx/fx thanfrom AT2Bmpr1afx/+ and AT2CTRL. For all experiments, n≥3animals/group. Data are mean±s.e.m. *P<0.05. n.s., notsignificant. Scale bars: 50 μm.

6

STEM CELLS AND REGENERATION Development (2018) 145, dev163014. doi:10.1242/dev.163014

DEVELO

PM

ENT

Page 7: Niche-mediated BMP/SMAD signaling regulates lung alveolar ...STEM CELLS AND REGENERATION RESEARCH ARTICLE Niche-mediated BMP/SMAD signaling regulates lung alveolar stem cell proliferation

to AT2s (Fig. 1). Significantly, this active signaling is transientlyreduced in both AT2s and TASCs early post-PNX, subsequentlyreturning to previous levels (Fig. 1B,C). This decline appears to bemediated by changes in the transcription of genes encoding someBMP ligands, but significantly also BMP receptors and BMPantagonists (Fig. 1D). Changes in multiple BMP signalingcomponents is a common feature of other examples of tissueremodeling, including the mouse trachea (Hsu et al., 2014; Kosinskiet al., 2007; Oshimori and Fuchs, 2012; Tadokoro et al., 2016).Taken together, our findings support the model summarized inFig. 6. According to this model, BMP signaling in AT2s helps tomaintain their quiescence and identity at steady state. A transientdecline in pSmad1/5/8 in AT2s early in the post-PNX regrowthphase enables them to transition to a more labile or permissive statein which they can respond to proliferative and differentiation signals.In this state, BMP signaling increases their propensity to differentiateinto AT1s.Initial support for our model came from organoid experiments in

which isolated AT2s are grown in 3D in the presence of Pdgfrα+

fibroblasts that, together with components of the culture medium,provide trophic support for AT2 self-renewal and differentiation intoAT1s. In this assay, the formation of organoids more than 45 μm indiameter depends on a balance between AT2 survival, proliferationand differentiation; signals promoting early AT1 differentiation willlikely inhibit CFE as the pool of AT2 cells is quickly exhausted. Inorganoid cultures, either activating or inhibiting BMP signaling inAT2s using constitutively active Bmpr1a or floxed null alleles leadsto an attenuated CFE (Fig. 3B,D), indicating that either too much ortoo little BMP signaling can disrupt colony formation. On the otherhand, in those organoids that do reach a scorable size, reduced BMPsignaling clearly inhibits AT2 to AT1 differentiation, whereasincreased BMP signaling promotes AT1 differentiation.Given our initial results with organoid assays, it was surprising that

manipulation of Bmpr1a-dependent signaling in AT2s in vivo, usingthe same inducible constitutively active Bmpr1a allele or Bmpr1afloxed null allele, did not apparently affect AT2 proliferation afterPNX (Fig. 4). One possible explanation is that the multiple signalingpathways acting in vivo after PNX can override or modulate thegenetically induced increases or decreases in Bmpr1a signaling, atleast in relation to cell proliferation. Such parallel pathways acting in

AT2s might include mechanical tension, and factors released byactivated macrophages and endothelial cells (Ding et al., 2011;Lechner et al., 2017; Liu et al., 2016; Rafii et al., 2015). Nevertheless,in vivo geneticmodulation of BMP signaling in AT2s does affect theirdifferentiation; loss of Bmpr1a in cells homozygous for the floxedalleles results in fewer AT1s, whereas activation of signaling inAT2CAB cells accelerates their differentiation into AT1s at 7 dayspost-PNX (Fig. 5B,D). The fact that no excess conversion of AT2s toAT1s is seen at 21 days suggests that only a fixed level of conversionis needed to restore lung homeostasis after PNX.

In the future, we need to know more about the mechanisms bywhich BMP signaling enhances the propensity of AT2s todifferentiate into AT1s and how the BMP pathway interacts withother pathways affecting self-renewal versus differentiation. Recentexperiments have indicated that canonical WNT signaling in AT2sinhibits AT1 fate choice (Frank et al., 2016; Nabhan et al., 2018),pointing to opposing effects between the two pathways. Suchantagonistic BMP/WNT effects have been described in variousother progenitors and stem cell niches, including intestinal stemcells, hair follicles and cardiomyoblasts (Genander et al., 2014; Jainet al., 2015b; Takeda et al., 2011). Studies with AT2s in culture havealso suggested that there is antagonism between BMP and TGFβsignaling, with the latter promoting AT1 differentiation (Zhao et al.,2013). Although our results with BMP signaling do not supporttheir model, it is of interest that AT2CAB cells upregulate Tgfb1expression about twofold, along with several genes that areexpressed in AT1 cells. Finally, during cardiomyogenesis, activepSmad1/5/8 complexes physically interact with HOPX to repressthe WNT pathway (Jain et al., 2015b). The fact that HOPX is highlyenriched in AT1s raises the possibility that BMP drives AT1differentiation through HOPX-mediated mechanisms.

One new observation reported here is that soon after lineage-labeled AT2 cells are placed in 3D culture, they co-express genestypically associated with AT2s (Sftpc) and AT1s (Ager), and this co-expression continues for up to 10 days (Fig. S8). We speculate thatthis phenotype is associated with the ‘priming’ or increased plasticityof AT2s proposed in our model (Fig. 6). In vivo, at steady state, fewerthan 1%ofAT2 cells are dual positive, but this value increases to 20%at day 7 post-PNX (Fig. S8). Future studies will need to test thefunctional significance of the dual positive phenotype.

Finally, another novel finding from our studies both in vitro andin vivo is that constitutive activation of BMP signaling in Pdgfrα+

mesenchymal cells inhibits their ability to support AT2 self-renewalboth in vivo and in vitro (Figs 3Cand 4D). This likely contributes to thelower CFE of AT2s in the organoid assay in the presence of BMP4ligand (Figs2Aand3C).Recent studies havepresented evidence that invivo there are at least two populations of stromal cells in the alveolarniche, onlyone ofwhich,mesenchymal alveolar niche cells (MANCs),promotes in vitro alveolar organoid growth (Zepp et al., 2017). Usingsingle-cell RNA-seq analysis, Zepp et al. identified a BMP antagonist,Grem2, as one of the regulators secreted by MANCs but not by othermesenchymal populations. By contrast, our data show that thetranscription of Grem2 does not change in Pdgfrα+ stromal cellspost-PNX (Fig. 1D), but rather indicate that FSTL1 and FST are themajorantagonists regulatingBMPsignaling, at least in thePNXmodel.Further experiments are required to localize transcripts and protein forthe multiple BMP ligands and antagonists at the single-cell level inalveoli at different times during the regrowth process. In addition, itwill be important to clarifywhether theAxin2+ stromal cells identifiedby Zepp et al. are identical to the Pdgfrα+ cells adjacent to AT2s thatwe have here termed TASCs. These cells have a very characteristicmorphology, with extended processes (Fig. S1 and Movie 1).

Fig. 6. Schematicmodel for the role of BMP signaling in the alveolar nichebefore and after PNX. At steady state, pSmad1/5/8-dependent signaling ishigh in the alveolar niche. This helps to maintain the quiescence and identity ofAT2s. Following PNX there is a transient decrease in BMP signaling in the lung,with reduced levels of pSmad1/5/8 protein in the nuclei of AT2s and adjacentPdgfrα+ TASCs. This decrease is mediated mostly by changes in theexpression of genes encoding BMP receptors and BMP antagonists. Wesuggest that the reduced BMP signaling primes or permits individual AT2s tobe able to respond to trophic and proliferative signals in the niche, includingthose produced by adjacent TASCs. According to the signals they receive, theymay either self-renew or differentiate (e.g. give rise to two AT2s, two AT1s, orone AT2 and one AT1). As remodeling continues, pSmad1/5/8 expressionincreases in AT2s and this promotes their propensity to differentiate intopSmad1/5/8+ AT1s.

7

STEM CELLS AND REGENERATION Development (2018) 145, dev163014. doi:10.1242/dev.163014

DEVELO

PM

ENT

Page 8: Niche-mediated BMP/SMAD signaling regulates lung alveolar ...STEM CELLS AND REGENERATION RESEARCH ARTICLE Niche-mediated BMP/SMAD signaling regulates lung alveolar stem cell proliferation

Fibroblasts that appear to have a similar morphology and to makecontact with AT2s, AT1 and endothelial cells have been described inthe human lung (Sirianni et al., 2003).

MATERIALS AND METHODSMiceTo generate Ager-H2b:Venus mice, a DNA fragment containing 8 kbupstream of the first coding exon and exons 2-7 were retrieved from a BACclone (bMQ174, Source BioScience) and recombined into the vector pL25Bupstream of a HSV-TK cassette for negative selection. A cassette encodingH2b:Venus fusion protein followed by polyA (kindly provided by Dr Anna-Katerina Hadjantonakis, Sloan Kettering Cancer Center) and a neo cassetteflanked with FRT sites were recombined into the start codon. The constructwas electroporated into G4 (C57BL/6Ncr×129S6/SvEvTac) hybrid EScells. Two clones were injected into C57BL/6 blastocysts. Micewere bred to129S4-Gt(ROSA)26Sortm2(FLP*)Sor/J to remove the neo cassette. Ager-H2b:Venus mice are maintained on a C57BL/6 background.

A similar strategy was used to generate the Pdgfrα-CreERT2 ‘knock-in’allele. A CreERT2 poly-A cassette and a FRT-flanked neo cassette wererecombined into the start codon of Pdgfrα (BAC clone: bMQ123p11,Source BioScience). The construct was electroporated into TL1 (129S6/SvEvTac) ES cells and these were injected into C57BL/6 blastocysts. Theneo cassette was removed. Pdgfrα-CreERT2 mice were maintained on aC57BL/6 background.

Sftpctm1(cre/ERT)Blh (Sftpc-CreERT2) (Rock et al., 2011), Rosa26-CAG-lsl-tdTomato (Arenkiel et al., 2011), Rosa26-CAG-lsl-caBmpr1a(Rodriguez et al., 2010), Bmpr1a flox (Mishina et al., 2002) andPdgfratm11(EGFP)Sor (Pdgfra-H2b:GFP) (Hamilton et al., 2003) weremaintained on a C57BL/6 background. All experiments were performedaccording to IACUC-approved protocols.

PneumonectomyProcedures were performed as previously described (Lechner et al., 2017).Briefly, mice were anesthetized with 2% isofluorane and intubated using aHarvard mini-vent ventilator with 200 µl stroke volume at 200 strokes perminute. The left pulmonary vasculature and bronchus were ligated with atitanium clip and the left lobe was removed. After closing the ribs, anangiocath was inserted to remove air to re-establish negative pressure. Micewere disconnected from the ventilator when autonomous breathingrecovered. Sham animals underwent the same procedures withoutremoving the left lobe.

Lung dissociation and FACSLungs were inflated intratracheally with 1-1.5 ml of protease solutioncontaining collagenase type I (450 U/ml; Gibco #17100-017), elastase (4 U/ml;Worthington Biochemical Corporation #LS002279), dispase (5 U/ml; BDBiosciences #354235) and DNaseI (0.33 U/ml) in DMEM/F12. Lung lobeswere separated, cut into small pieces and incubated with 3 ml proteasesolution for 30 min at 37°C with frequent agitation. Equal amounts of mediacontaining 10% FBS was added to the tissue suspension and then filteredthrough a 100 µm strainer. The cell pellet was then resuspended andincubated with 2 ml of red blood cell lysis buffer (eBioscience) for 2 min atroom temperature. The cell suspension was washed with 10% FBS, filteredthrough a 40 µm strainer, centrifuged and resuspended in DMEM/F12+2%BSA. Sorting was performed using a FACS Vantage SE.

Organoid cultureFACS sorted cells were resuspended in MTEC/Plus media and mixed 1:1with growth factor-reduced Matrigel (BD Biosciences #356230). MTEC/Plus:Matrigel (90 µl) containing 5×103 AT2s and 5×104 stromal cells wasseeded in individual 24-well 0.4 µm Transwell inserts (Falcon). MTEC/Plus(500 µl) was placed in the lower chamber and media was changed everyother day. Spheres were counted and fixed on day 14. Recombinant proteinswere purchased from R&D systems and used as follows: BMP2 (50 ng/ml),BMP4 (50 ng/ml), FST (500 ng/ml), FSTL1 (500 ng/ml) and Noggin(1000 ng/ml). Each condition was tested in at least three wells and eachexperiment was repeated at least three times.

Histology and immunofluorescence analysisLungs were inflated with 4% paraformaldehyde to 25 cm H2O pressure for10 min and then removed and submerged in 4% paraformaldehyde in PBSfor 4 h at 4°C. For tissues used for phospho-Smad1/5/8 detection, PhosStop(Roche 4906845001) was added to the fixative. Tissue was dehydrated,embedded in paraffin and sectioned at 7 µm. Tissue sections underwent10 mMsodiumcitrate antigen retrieval andwere blockedwith 3%BSA, 10%donkey serum and 0.1% Triton X-100 for 1 h at room temperature. Primaryantibodies diluted in block were applied and incubated overnight at 4°C.Tissue sections were washed with PBS and fluorophore-conjugatedsecondary antibodies were diluted at 1:500 and incubated for 1 h at roomtemperature. For phospho-Smad1/5/8 staining, HRP-conjugated secondaryantibody (1:1000) and TSA detection system were used (PerkinElmer,NEL744001KT). Primary antibodies were as follows: LAMP-3/CD208(Dendritics, DDX0191, 1:200), endomucin (Santa Cruz, sc-65495, 1:250),GFP (Aves lab, GFP-1020, 1:500), HOPX (Santa Cruz, sc-398703, 1:50),RFP (Rockland, 600401379, 1:250), PDGFRβ (Cell Signaling, 3169,1:100), RAGE/AGER (R&D, MAB1179, 1:200), SFTPC (Millipore,ab3786, 1:500; Santa Cruz, SC-7706, 1:100), phospho-Smad1/5/8(Millipore, AB3848-I, 1:250; Cell Signaling, 9511). Images were obtainedusing Zeiss LSM 710, LSM 780 and Imager AxioCam microscopes.

Quantification and statisticsFor quantification, two well-separated longitudinal sections per accessarylobe were imaged and the whole areas were analyzed using ImageJ. n≥3animals/experiment. Sections of organoids (≥10 organoids/transwell) wereanalyzed after immunohistochemistry. The quantification values oftriplicate wells were averaged and plotted using Prism software. Statisticalanalysis was performed using unpaired, two-tailed, Student’s t-test betweengroups. Values on graphs are shown as mean±s.e.m.

Quantitative RT-PCRTotal RNA was extracted from FACS-sorted lineage-labeled AT2s andPdgfra-H2b:GFP cells using Direct-zol RNAMiniPrep Kit (Zymo Research).cDNAwas synthesized using SuperScript VILO kit (Invitrogen). qPCR wasperformed with iQ SYBR Green Supermix (Bio-Rad) and StepOne Plussystem (Applied Biosystems). The mRNA levels of target genes werenormalized to Gapdh. Primer sequences are in Table S1.

RNA sequencing and analysisTotal RNA was extracted from FACS-sorted lineage-labeled AT2CTRL andAT2CAB cells using Direct-zol RNA MiniPrep Kit (Zymo Research) andmRNA was enriched from 200 ng of each total RNA using NEBNextPoly(A) mRNA Magnetic Isolation Module (New England BioLabs).Libraries were prepared using NEBNext Ultra II RNA Library Prep Kit forIllumina (New England BioLabs). Paired-end sequencing (150 bp for eachread) was performed using HiSeq X with the depth of 24 million reads foreach sample. The quality of sequenced reads was assessed using FastQC(www.bioinformatics.babraham.ac.uk/projects/fastqc/). PolyA/T tails weretrimmed using PRINSEQ (Schmieder and Edwards, 2011). Adaptorsequences were trimmed and shorter reads than 24 bp were dropped usingTrimmomatic (Bolger et al., 2014). Reads were mapped to the mousereference genome (mm10) using Hisat2 (Kim et al., 2015) with defaultsetting. Duplicate reads were removed using the markdup option ofSAMtools (Li et al., 2009). Fragment numbers were counted using thefeatureCounts option of SUBREAD (Liao et al., 2014). Normalization andextraction of differentially expressed genes (DEGs) between AT2CTRL andAT2CAB were performed using an R package, DESeq2 (Love et al., 2014).Heatmaps were generated using Shinyheatmap (Khomtchouk et al., 2017).The RNA-seq data have been deposited in GEO under accession numberGSE112431.

Western blotProtein extracts were collected from accessory lobes. Equal amounts ofproteins were separated by SDS-PAGE and transferred to polyvinylidenefluoridemembranes.Membraneswere blocked for 1 hwith 5%BSA in TBST(0.1% Tween 20) and then incubated with phospho-Smad1/5/8 antibody (Cell

8

STEM CELLS AND REGENERATION Development (2018) 145, dev163014. doi:10.1242/dev.163014

DEVELO

PM

ENT

Page 9: Niche-mediated BMP/SMAD signaling regulates lung alveolar ...STEM CELLS AND REGENERATION RESEARCH ARTICLE Niche-mediated BMP/SMAD signaling regulates lung alveolar stem cell proliferation

Signaling, 13820, 1:1000) and β-actin antibody (Abcam, ab8226, 1:2000)overnight at 4°C. Membranes were washed with TBST and then incubatedwith HRP-conjugated secondary antibodies (Jackson ImmunoResearch, 711-035-152, 715-035-151 1:10,000). Protein blots were analyzed with the ECLdetection system (FEMTOMAX-110, Rockland Immunochemicals).

AcknowledgementsY.K. is supported by Dr Purushothama Rao Tata, whom we thank for criticalcomments on the manuscript.

Competing interestsThe authors declare no competing or financial interests.

Author contributionsConceptualization: M.C., B.L.M.H.; Methodology: M.C.; Validation: M.C., M.B.;Formal analysis: M.C., M.B., Y.K.; Investigation: M.C., M.B., Y.K.; Resources: M.C.,M.B., C.E.B., Y.K.; Data curation: M.C., Y.K.; Writing - original draft: M.C.; Writing -review & editing: M.C., C.E.B., Y.K., B.L.M.H.; Visualization: M.C.; Supervision:M.C., B.L.M.H.; Project administration: M.C., B.L.M.H.; Funding acquisition:B.L.M.H.

FundingThis work was supported by the National Institutes of Health (R37HL071303 toB.L.M.H.) and by the Duke Cancer Institute Transgenic and Knockout MouseShared Resource Facility. Deposited in PMC for immediate release.

Data availabilityThe RNA-seq data have been deposited in GEO under accession numberGSE112431.

Supplementary informationSupplementary information available online athttp://dev.biologists.org/lookup/doi/10.1242/dev.163014.supplemental

ReferencesArenkiel, B. R., Hasegawa, H., Yi, J. J., Larsen, R. S., Wallace, M. L., Philpot,B. D., Wang, F. and Ehlers, M. D. (2011). Activity-induced remodeling of olfactorybulb microcircuits revealed by monosynaptic tracing. PLoS ONE 6, e29423.

Barkauskas, C. E., Cronce, M. J., Rackley, C. R., Bowie, E. J., Keene, D. R.,Stripp, B. R., Randell, S. H., Noble, P. W. and Hogan, B. L. M. (2013). Type 2alveolar cells are stem cells in adult lung. J. Clin. Invest. 123, 3025-3036.

Bolger, A. M., Lohse, M. and Usadel, B. (2014). Trimmomatic: a flexible trimmer forIllumina sequence data. Bioinformatics 30, 2114-2120.

Brody, J. S., Burki, R. and Kaplan, N. (1978). Deoxyribonucleic acid synthesis inlung cells during compensatory lung growth after pneumonectomy. Am. Rev.Respir. Dis. 117, 307-316.

Buhain, W. J. and Brody, J. S. (1973). Compensatory growth of the lung followingpneumonectomy. J. Appl. Physiol. 35, 898-902.

Butler, J. P., Loring, S. H., Patz, S., Tsuda, A., Yablonskiy, D. A. and Mentzer,S. J. (2012). Evidence for adult lung growth in humans. N. Engl. J. Med. 367,244-247.

Chen, L., Acciani, T., Le Cras, T., Lutzko, C. and Perl, A.-K. T. (2012). Dynamicregulation of platelet-derived growth factor receptor alpha expression in alveolarfibroblasts during realveolarization. Am. J. Respir. Cell Mol. Biol. 47, 517-527.

Desai, T. J., Brownfield, D. G. and Krasnow, M. A. (2014). Alveolar progenitor andstem cells in lung development, renewal and cancer. Nature 507, 190-194.

Ding, B.-S., Nolan, D. J., Guo, P., Babazadeh, A. O., Cao, Z., Rosenwaks, Z.,Crystal, R. G., Simons, M., Sato, T. N., Worgall, S. et al. (2011). Endothelial-derived angiocrine signals induce and sustain regenerative lung alveolarization.Cell 147, 539-553.

Evans, M. J., Cabral, L. J., Stephens, R. J. and Freeman, G. (1973). Renewal ofalveolar epithelium in the rat following exposure to NO2. Am. J. Pathol. 70,175-198.

Frank, D. B., Peng, T., Zepp, J. A., Snitow, M., Vincent, T. L., Penkala, I. J., Cui,Z., Herriges, M. J., Morley, M. P., Zhou, S. et al. (2016). Emergence of a wave ofWnt signaling that regulates lung alveologenesis by controlling epithelial self-renewal and differentiation. Cell Rep. 17, 2312-2325.

Genander, M., Cook, P. J., Ramskold, D., Keyes, B. E., Mertz, A. F., Sandberg, R.and Fuchs, E. (2014). BMP signaling and its pSMAD1/5 target genesdifferentially regulate hair follicle stem cell lineages. Cell Stem Cell 15, 619-633.

Green, J., Endale, M., Auer, H. andPerl, A.-K. T. (2016). Diversity of interstitial lungfibroblasts is regulated by platelet-derived growth factor receptor alpha kinaseactivity. Am. J. Respir. Cell Mol. Biol. 54, 532-545.

Hamilton, T. G., Klinghoffer, R. A., Corrin, P. D. and Soriano, P. (2003).Evolutionary divergence of platelet-derived growth factor alpha receptor signalingmechanisms. Mol. Cell. Biol. 23, 4013-4025.

Haramis, A.-P. G., Begthel, H., van den Born, M., van Es, J., Jonkheer, S.,Offerhaus, G. J. and Clevers, H. (2004). De novo crypt formation and juvenilepolyposis on BMP inhibition in mouse intestine. Science 303, 1684-1686.

He, X. C., Zhang, J., Tong, W.-G., Tawfik, O., Ross, J., Scoville, D. H., Tian, Q.,Zeng, X., He, X., Wiedemann, L. M. et al. (2004). BMP signaling inhibits intestinalstem cell self-renewal through suppression of Wnt-beta-catenin signaling. Nat.Genet. 36, 1117-1121.

Hogan, B. L. M., Barkauskas, C. E., Chapman, H. A., Epstein, J. A., Jain, R.,Hsia, C. C. W., Niklason, L., Calle, E., Le, A., Randell, S. H. et al. (2014). Repairand regeneration of the respiratory system: complexity, plasticity, andmechanisms of lung stem cell function. Cell Stem Cell 15, 123-138.

Hsia, C. C., Herazo, L. F., Fryder-Doffey, F. and Weibel, E. R. (1994).Compensatory lung growth occurs in adult dogs after right pneumonectomy.J. Clin. Invest. 94, 405-412.

Hsu, Y.-C., Li, L. and Fuchs, E. (2014). Emerging interactions between skin stemcells and their niches. Nat. Med. 20, 847-856.

Jain, R., Barkauskas, C. E., Takeda, N., Bowie, E. J., Aghajanian, H., Wang, Q.,Padmanabhan, A., Manderfield, L. J., Gupta, M., Li, D. et al. (2015a). Plasticityof Hopx(+) type I alveolar cells to regenerate type II cells in the lung.Nat. Commun.6, 6727.

Jain, R., Li, D., Gupta, M., Manderfield, L. J., Ifkovits, J. L., Wang, Q., Liu, F., Liu,Y., Poleshko, A., Padmanabhan, A. et al. (2015b). HEART DEVELOPMENT.Integration of Bmp and Wnt signaling by Hopx specifies commitment ofcardiomyoblasts. Science 348, aaa6071.

Kapanci, Y., Weibel, E. R., Kaplan, H. P. and Robinson, F. R. (1969).Pathogenesis and reversibility of the pulmonary lesions of oxygen toxicity inmonkeys. II. Ultrastructural and morphometric studies. Lab. Invest. 20, 101-118.

Khomtchouk, B. B., Hennessy, J. R. and Wahlestedt, C. (2017). shinyheatmap:ultra fast low memory heatmap web interface for big data genomics. PLoS ONE12, e0176334.

Kim, D., Langmead, B. and Salzberg, S. L. (2015). HISAT: a fast spliced alignerwith low memory requirements. Nat. Methods 12, 357-360.

Kosinski, C., Li, V. S.W., Chan, A. S. Y., Zhang, J., Ho, C., Tsui, W. Y., Chan, T. L.,Mifflin, R. C., Powell, D. W., Yuen, S. T. et al. (2007). Gene expression patternsof human colon tops and basal crypts and BMP antagonists as intestinal stem cellniche factors. Proc. Natl. Acad. Sci. USA 104, 15418-15423.

Kumar, P. A., Hu, Y., Yamamoto, Y., Hoe, N. B., Wei, T. S., Mu, D., Sun, Y., Joo,L. S., Dagher, R., Zielonka, E. M. et al. (2011). Distal airway stem cells yieldalveoli in vitro and during lung regeneration following H1N1 influenza infection.Cell 147, 525-538.

Lechner, A. J., Driver, I. H., Lee, J., Conroy, C. M., Nagle, A., Locksley, R. M. andRock, J. R. (2017). Recruited Monocytes and Type 2 Immunity Promote LungRegeneration following Pneumonectomy. Cell Stem Cell 21, 120-134.e127.

Lee, J.-H., Bhang, D. H., Beede, A., Huang, T. L., Stripp, B. R., Bloch, K. D.,Wagers, A. J., Tseng, Y.-H., Ryeom, S. and Kim, C. F. (2014). Lung stem celldifferentiation in mice directed by endothelial cells via a BMP4-NFATc1-thrombospondin-1 axis. Cell 156, 440-455.

Lewis, C. J., Mardaryev, A. N., Poterlowicz, K., Sharova, T. Y., Aziz, A., Sharpe,D. T., Botchkareva, N. V. and Sharov, A. A. (2014). Bone morphogenetic proteinsignaling suppresses wound-induced skin repair by inhibiting keratinocyteproliferation and migration. J. Invest. Dermatol. 134, 827-837.

Li, H., Handsaker, B., Wysoker, A., Fennell, T., Ruan, J., Homer, N., Marth, G.,Abecasis, G., Durbin, R. and Genome Project Data Processing Subgroup.(2009). The sequence alignment/Map format and SAMtools. Bioinformatics 25,2078-2079.

Liao, Y., Smyth, G. K. and Shi, W. (2014). featureCounts: an efficient generalpurpose program for assigning sequence reads to genomic features.Bioinformatics 30, 923-930.

Liu, Y., Kumar, V. S., Zhang, W., Rehman, J. and Malik, A. B. (2015). Activation oftype II cells into regenerative stem cell antigen-1(+) cells during alveolar repair.Am. J. Respir. Cell Mol. Biol. 53, 113-124.

Liu, Z., Wu, H., Jiang, K., Wang, Y., Zhang, W., Chu, Q., Li, J., Huang, H., Cai, T.,Ji, H. et al. (2016). MAPK-mediated YAP activation controls mechanical-tension-induced pulmonary alveolar regeneration. Cell Rep. 16, 1810-1819.

Love, M. I., Huber, W. and Anders, S. (2014). Moderated estimation of fold changeand dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550.

Mishina, Y., Hanks, M. C., Miura, S., Tallquist, M. D. and Behringer, R. R. (2002).Generation of Bmpr/Alk3 conditional knockout mice. Genesis 32, 69-72.

Nabhan, A., Brownfield, D. G., Harbury, P. B., Krasnow, M. A. and Desai, T. J.(2018). Single-cell Wnt signaling niches maintain stemness of alveolar type 2cells. Science 359, 1118-1123.

Oshimori, N. and Fuchs, E. (2012). Paracrine TGF-beta signaling counterbalancesBMP-mediated repression in hair follicle stem cell activation. Cell Stem Cell 10,63-75.

Rafii, S., Cao, Z., Lis, R., Siempos, I. I., Chavez, D., Shido, K., Rabbany, S. Y. andDing, B.-S. (2015). Platelet-derived SDF-1 primes the pulmonary capillaryvascular niche to drive lung alveolar regeneration. Nat. Cell Biol. 17, 123-136.

Rock, J. R., Barkauskas, C. E., Cronce, M. J., Xue, Y., Harris, J. R., Liang, J.,Noble, P. W. and Hogan, B. L. M. (2011). Multiple stromal populations contribute

9

STEM CELLS AND REGENERATION Development (2018) 145, dev163014. doi:10.1242/dev.163014

DEVELO

PM

ENT

Page 10: Niche-mediated BMP/SMAD signaling regulates lung alveolar ...STEM CELLS AND REGENERATION RESEARCH ARTICLE Niche-mediated BMP/SMAD signaling regulates lung alveolar stem cell proliferation

to pulmonary fibrosis without evidence for epithelial to mesenchymal transition.Proc. Natl. Acad. Sci. USA 108, E1475-E1483.

Rodriguez, P., Da Silva, S., Oxburgh, L., Wang, F., Hogan, B. L. M. and Que, J.(2010). BMP signaling in the development of the mouse esophagus andforestomach. Development 137, 4171-4176.

Schmieder, R. and Edwards, R. (2011). Quality control and preprocessing ofmetagenomic datasets. Bioinformatics 27, 863-864.

Sirianni, F. E., Chu, F. S. F. and Walker, D. C. (2003). Human alveolar wallfibroblasts directly link epithelial type 2 cells to capillary endothelium.Am. J. Respir. Crit. Care. Med. 168, 1532-1537.

Tadokoro, T., Gao, X., Hong, C. C., Hotten, D. and Hogan, B. L. M. (2016). BMPsignaling and cellular dynamics during regeneration of airway epithelium frombasal progenitors. Development 143, 764-773.

Takeda, N., Jain, R., LeBoeuf, M. R., Wang, Q., Lu, M. M. and Epstein, J. A.(2011). Interconversion between intestinal stem cell populations in distinct niches.Science 334, 1420-1424.

Thane, K., Ingenito, E. P. and Hoffman, A. M. (2014). Lung regeneration andtranslational implications of the postpneumonectomy model. Transl. Res. 163,363-376.

Toufen, C., Jr, Costa, E. L. V., Hirota, A. S., Li, H. Y., Amato, M. B. P. andCarvalho, C. R. R. (2011). Follow-up after acute respiratory distresssyndrome caused by influenza a (H1N1) virus infection. Clinics (Sao Paulo)66, 933-937.

Wang, Y., Tang, Z., Huang, H., Li, J., Wang, Z., Yu, Y., Zhang, C., Li, J., Dai, H.,Wang, F. et al. (2018). Pulmonary alveolar type I cell population consists oftwo distinct subtypes that differ in cell fate. Proc. Natl. Acad. Sci. USA 115,2407-2412.

Zacharias, W. J., Frank, D. B., Zepp, J. A., Morley, M. P., Alkhaleel, F. A., Kong,J., Zhou, S., Cantu, E. and Morrisey, E. E. (2018). Regeneration of the lungalveolus by an evolutionarily conserved epithelial progenitor. Nature 555,251-255.

Zepp, J. A., Zacharias, W. J., Frank, D. B., Cavanaugh, C. A., Zhou, S., Morley,M. P. and Morrisey, E. E. (2017). Distinct mesenchymal lineages and nichespromote epithelial self-renewal and myofibrogenesis in the lung. Cell 170,1134-1148.e1110.

Zhao, L., Yee, M. and O’Reilly, M. A. (2013). Transdifferentiation of alveolarepithelial type II to type I cells is controlled by opposing TGF-beta and BMPsignaling. Am. J. Physiol. Lung Cell. Mol. Physiol. 305, L409-L418.

10

STEM CELLS AND REGENERATION Development (2018) 145, dev163014. doi:10.1242/dev.163014

DEVELO

PM

ENT