insights from imaging the implanting embryo and the ... · separating lumen and glands, generation...

6
TECHNIQUES AND RESOURCES RESEARCH REPORT Insights from imaging the implanting embryo and the uterine environment in three dimensions Ripla Arora 1,2 , Adam Fries 3 , Karina Oelerich 1 , Kyle Marchuk 3 , Khalida Sabeur 1 , Linda C. Giudice 2 and Diana J. Laird 1,2, * ABSTRACT Although much is known about the embryo during implantation, the architecture of the uterine environment in which the early embryo develops is not well understood. We employed confocal imaging in combination with 3D analysis to identify and quantify dynamic changes to the luminal structure of murine uterus in preparation for implantation. When applied to mouse mutants with known implantation defects, this method detected striking peri-implantation abnormalities in uterine morphology that cannot be visualized by histology. We revealed 3D organization of uterine glands and found that they undergo a stereotypical reorientation concurrent with implantation. Furthermore, we extended this technique to generate a 3D rendering of the cycling human endometrium. Analyzing the uterine and embryo structure in 3D for different genetic mutants and pathological conditions will help uncover novel molecular pathways and global structural changes that contribute to successful implantation of an embryo. KEY WORDS: Implantation, Receptivity, Uterus, Blastocyst, Embryo, Confocal Imaging, Surface curvature, Wnt5a INTRODUCTION The uterus is the prerequisite for propagating mammalian species, as implantation in this tissue establishes pregnancy and protects the developing embryo throughout gestation. The window of uterine receptivity for embryo implantation is precisely timed (Ma et al., 2003). Rising levels of luteinizing hormone in concert with estradiol (E2) lead to ovulation and fertilization, termed gestational day (GD) 0 of murine pregnancy (reviewed by Cha et al., 2012; Wang and Dey, 2006). Formation of the corpus lutea at the site of ovulation releases progesterone (P4), which peaks at GD3 of pregnancy. Receptivity is achieved between GD3 and GD4 by the concurrence of high levels of P4 with a transient increase in E2 (Wang and Dey, 2006). Similarly, following human ovulation and corpus luteum formation, an increase in P4 during the secretory phase prepares the uterus for embryo implantation (Cha et al., 2012). In the mouse, blastocysts home and attach in the anti-mesometrial region of slit- like structures termed uterine crypts (Daikoku et al., 2011). The existence of crypts in the human uterus remains unknown. It stands to reason that ovarian hormones that coordinate uterine receptivity must also regulate uterine crypt formation; however, the underlying mechanisms are not defined. In preparation for implantation, uterine epithelial cells undergo morphological changes, termed plasma membrane transformation (PMT) (Murphy, 2004). In the presence of E2, epithelial cells possess microvilli on their apical surfaces and short tight junctions on the lateral plasma membrane, whereas they lose microvilli, acquire flattened morphology and increase their number of tight junctions upon concurrent exposure to E2 and P4. PMT is observed at the time of implantation in eutherian mammals (Murphy, 2004), viviparous squamate reptiles (Hosie et al., 2003) and marsupials (Laird et al., 2014). These dramatic changes at the cellular level are accompanied by changes in the organization of the uterine lumen, as suggested by scanning electron microscopy studies (Winkelmann and Spornitz, 1997); however, the three-dimensional uterine architectural changes remain unknown. Establishment of pregnancy also depends upon specialized uterine glands that provide nourishment to the embryo before the placenta forms (Filant and Spencer, 2014). A key factor secreted by the glands, leukemia inhibitory factor (LIF), is required for uterine receptivity (Daikoku et al., 2004; Stewart et al., 1992). The importance of uterine glands and their secretions is demonstrated by the drastic impact of their depletion on fertility (Filant et al., 2012). Studies involving uterine glands have been limited to histological analysis and currently there is no information on the 3D branching morphogenesis or structural organization of these glands during development, estrous or implantation. In the past, localizing embryos and studying their interactions with the uterine lining required generating numerous tissue sections. This process is labor intensive and generates only two-dimensional images. We now combine the strengths of confocal imaging with quantitative image analysis to enable visualization of the intact uterus in three dimensions. We observe dynamic changes in the architecture of the uterine lumen surrounding implantation. We highlight the utility of this method by studying implantation defective Wnt5a mutants, as previously characterized by molecular marker expression, defective crypt formation and embryonic lethality (Cha et al., 2014). Our method reveals defects in the folding pattern of Wnt5a mutant uteri that cannot be discerned by traditional histology. Furthermore, we define the organization of the uterine glands in three dimensions and the large-scale changes that coincide with implantation. This methodology will help to resolve long-standing conundrums in the field, such as determinants of embryo spacing, closure of the uterine lumen, and the regulation and function of the poorly studied uterine glands. RESULTS AND DISCUSSION Confocal imaging of the mouse and human endometrium We used whole-mount immunofluorescence and confocal imaging (Faire et al., 2015) to visualize and characterize changes in the Received 2 September 2016; Accepted 28 October 2016 1 Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA. 2 Department of Obstetrics, Gynecology and Reproductive Sciences and the Center for Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94143, USA. 3 Biological Imaging Development Center, University of California, San Francisco, CA 94143, USA. *Author for correspondence ([email protected]) D.J.L., 0000-0002-4930-0560 4749 © 2016. Published by The Company of Biologists Ltd | Development (2016) 143, 4749-4754 doi:10.1242/dev.144386 DEVELOPMENT

Upload: others

Post on 06-Aug-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Insights from imaging the implanting embryo and the ... · separating lumen and glands, generation of 3D surfacesthrough the length of the uterine horn permitted analysis of luminal

TECHNIQUES AND RESOURCES RESEARCH REPORT

Insights from imaging the implanting embryo and the uterineenvironment in three dimensionsRipla Arora1,2, Adam Fries3, Karina Oelerich1, Kyle Marchuk3, Khalida Sabeur1, Linda C. Giudice2 andDiana J. Laird1,2,*

ABSTRACTAlthough much is known about the embryo during implantation, thearchitecture of the uterine environment in which the early embryodevelops is not well understood. We employed confocal imaging incombinationwith 3Danalysis to identifyandquantify dynamic changesto the luminal structure ofmurine uterus in preparation for implantation.When applied to mousemutants with known implantation defects, thismethod detected striking peri-implantation abnormalities in uterinemorphology that cannot be visualized by histology. We revealed 3Dorganization of uterine glands and found that they undergo astereotypical reorientation concurrent with implantation. Furthermore,we extended this technique to generate a 3D rendering of the cyclinghumanendometrium.Analyzing theuterineandembryostructure in3Dfor different genetic mutants and pathological conditions will helpuncover novel molecular pathways and global structural changes thatcontribute to successful implantation of an embryo.

KEYWORDS: Implantation, Receptivity, Uterus, Blastocyst, Embryo,Confocal Imaging, Surface curvature, Wnt5a

INTRODUCTIONThe uterus is the prerequisite for propagating mammalian species, asimplantation in this tissue establishes pregnancy and protects thedeveloping embryo throughout gestation. The window of uterinereceptivity for embryo implantation is precisely timed (Ma et al.,2003). Rising levels of luteinizing hormone in concert with estradiol(E2) lead to ovulation and fertilization, termed gestational day (GD)0 of murine pregnancy (reviewed by Cha et al., 2012; Wang andDey, 2006). Formation of the corpus lutea at the site of ovulationreleases progesterone (P4), which peaks at GD3 of pregnancy.Receptivity is achieved between GD3 and GD4 by the concurrenceof high levels of P4 with a transient increase in E2 (Wang and Dey,2006). Similarly, following human ovulation and corpus luteumformation, an increase in P4 during the secretory phase prepares theuterus for embryo implantation (Cha et al., 2012). In the mouse,blastocysts home and attach in the anti-mesometrial region of slit-like structures termed uterine crypts (Daikoku et al., 2011). Theexistence of crypts in the human uterus remains unknown. It standsto reason that ovarian hormones that coordinate uterine receptivity

must also regulate uterine crypt formation; however, the underlyingmechanisms are not defined.

In preparation for implantation, uterine epithelial cells undergomorphological changes, termed plasma membrane transformation(PMT) (Murphy, 2004). In the presence of E2, epithelial cellspossess microvilli on their apical surfaces and short tight junctionson the lateral plasma membrane, whereas they lose microvilli,acquire flattened morphology and increase their number of tightjunctions upon concurrent exposure to E2 and P4. PMT is observedat the time of implantation in eutherian mammals (Murphy, 2004),viviparous squamate reptiles (Hosie et al., 2003) and marsupials(Laird et al., 2014). These dramatic changes at the cellular level areaccompanied by changes in the organization of the uterine lumen, assuggested by scanning electron microscopy studies (Winkelmannand Spornitz, 1997); however, the three-dimensional uterinearchitectural changes remain unknown.

Establishment of pregnancy also depends upon specializeduterine glands that provide nourishment to the embryo before theplacenta forms (Filant and Spencer, 2014). A key factor secreted bythe glands, leukemia inhibitory factor (LIF), is required for uterinereceptivity (Daikoku et al., 2004; Stewart et al., 1992). Theimportance of uterine glands and their secretions is demonstrated bythe drastic impact of their depletion on fertility (Filant et al., 2012).Studies involving uterine glands have been limited to histologicalanalysis and currently there is no information on the 3D branchingmorphogenesis or structural organization of these glands duringdevelopment, estrous or implantation.

In the past, localizing embryos and studying their interactionswith the uterine lining required generating numerous tissue sections.This process is labor intensive and generates only two-dimensionalimages. We now combine the strengths of confocal imaging withquantitative image analysis to enable visualization of the intactuterus in three dimensions. We observe dynamic changes in thearchitecture of the uterine lumen surrounding implantation. Wehighlight the utility of this method by studying implantationdefective Wnt5a mutants, as previously characterized by molecularmarker expression, defective crypt formation and embryoniclethality (Cha et al., 2014). Our method reveals defects in thefolding pattern of Wnt5a mutant uteri that cannot be discerned bytraditional histology. Furthermore, we define the organization of theuterine glands in three dimensions and the large-scale changes thatcoincide with implantation. This methodology will help to resolvelong-standing conundrums in the field, such as determinants ofembryo spacing, closure of the uterine lumen, and the regulation andfunction of the poorly studied uterine glands.

RESULTS AND DISCUSSIONConfocal imaging of the mouse and human endometriumWe used whole-mount immunofluorescence and confocal imaging(Faire et al., 2015) to visualize and characterize changes in theReceived 2 September 2016; Accepted 28 October 2016

1Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research,University of California San Francisco, San Francisco, CA 94143, USA.2Department of Obstetrics, Gynecologyand Reproductive Sciences and theCenterfor Reproductive Sciences, University of California, San Francisco, San Francisco,CA 94143, USA. 3Biological Imaging Development Center, University of California,San Francisco, CA 94143, USA.

*Author for correspondence ([email protected])

D.J.L., 0000-0002-4930-0560

4749

© 2016. Published by The Company of Biologists Ltd | Development (2016) 143, 4749-4754 doi:10.1242/dev.144386

DEVELO

PM

ENT

Page 2: Insights from imaging the implanting embryo and the ... · separating lumen and glands, generation of 3D surfacesthrough the length of the uterine horn permitted analysis of luminal

mouse endometrium from fertilization through implantation.Luminal and glandular epithelium in the uterus were identifiedusing the epithelial marker E-cadherin (E-CAD) (Reardon et al.,2012), whereas FOXA2 (Besnard et al., 2004) was used todistinguish glandular epithelium throughout the length and depthof the uterine horn (Fig. 1A; Movie 1). Developing embryos at theblastocyst and epiblast stages were identified within the uterinehorns using E-CAD staining along with the nuclear stain Hoechst(Fig. 1B-E). We applied similar methodology to image a fullthickness biopsy of the proliferative phase human uterus obtainedby hysterectomy (Fig. 1F; Movie 2). As expected, 3D renderings ofthe E-CAD+ endometrial lumen and glands revealed increasedglandular complexity of the human uterus compared with mouse(Fig. 1G; Movie 3).

Uterine crypts are folds in the uterine epitheliumTo visualize the structure of the mouse uterine lumen, wecomputationally subtracted the FOXA2+ glandular signal fromE-CAD+ total epithelium (Fig. 2A,B; Movies 4 and 5). Afterseparating lumen and glands, generation of 3D surfaces through thelength of the uterine horn permitted analysis of luminal structureand glandular organization (Movies 6 and 7). At GD 4.25, 2Doptical sections revealed the overlap of 3D surface luminal foldswith potential uterine crypts or chambers (Fig. 2C,D), which housethe incoming embryo (Daikoku et al., 2011).

Uterine architecture changes dynamically from fertilizationto implantationWe observed that the murine uterine lumen undergoesdynamic changes in topology on the day preceding implantation(Fig. 3A-D). Three-dimensional imaging revealed an increase in

the number of folds at gestational day (GD) 3.5-3.75, specificallyalong the mesometrial–anti-mesometrial (M-AM) axis, whichlies perpendicular to both the oviductal-cervical axis and thedorsalventral axis of the uterine horn (Fig. 3C). Luminal closureensues at GD4.25 to lock the embryo into position (Chen et al.,2013; Yoshinaga, 2013). Concomitant with closure, we observedthat the lumen flanking the embryo, the peri-implantation region,becomes entirely devoid of folds in the M-AM axis; folds alsogrow more shallow along the oviductal-cervical axis, while foldingalong the M-AM axis is retained in the inter-implantation region(Fig. 3D,D′; Movie 6). This is consistent with uterine foldingchanges first described by scanning electron microscopy ofepithelial cells (Winkelmann and Spornitz, 1997). We developedan algorithm to quantify topological changes in the E-CAD+FOXA2– luminal epithelium by measuring curvature. The SurfaceCurvature algorithm splits the 3D surface into vertices andestimates the radius of the curve formed by connecting neighborvertices. Radii calculated with multiple neighbor vertices areaveraged to obtain curvature mean (Cmean). Topology of uterineluminal segments is expressed as the folding factor ( f ), which is

Fig. 1. Confocal imaging of pregnant mouse uterus and cycling humanendometrium. (A) Optical z slice showing two-thirds of both horns of aGD4.25mouse uterus attached at the cervix (yellow arrow) stained with nuclear markerHoechst (gray), epithelial marker E-CAD (red) and glandular marker FOXA2(green). (B-D) Identification of blastocysts (white arrows) in optical slices ofintact uteri at GD3.25 (B), GD3.75 (C) and GD4.25 (D). (E) Epiblast at GD5.75.(F,G) Three z slices (F) through a full-thickness segment of humanendometrium stained for E-CAD (red) and FOXA2 (green), and correspondingsurface rendering of the same specimen based on E-CAD staining (G). Scalebars: 500 μm in A,F,G; 50 μm in B-E. Lat, lateral; Med, medial; A, anterior; P,posterior; SM, smooth muscle.

Fig. 2. Uterine crypts are generated by folding the luminal epithelium.(A,B) Optical slices through a segment of the uterus at GD4.25 immunolabeledfor FOXA2 signal (green) and ECAD signal (red) (A), and the resultingsubtraction of FOXA2 from ECAD signal to obtain the uterine lumen (yellow)(B). (C) Optical slice showing uterine crypts (arrows) revealed by luminalepithelial staining at GD4.25. (D) Luminal folds (arrows) in the 3D surfacemodel of the uterus coincide with crypts in the optical slice. Scale bars: 500 μmin A,B; 200 μm in C,D. M, mesometrial; AM, anti-mesometrial.

4750

TECHNIQUES AND RESOURCES Development (2016) 143, 4749-4754 doi:10.1242/dev.144386

DEVELO

PM

ENT

Page 3: Insights from imaging the implanting embryo and the ... · separating lumen and glands, generation of 3D surfacesthrough the length of the uterine horn permitted analysis of luminal

the ratio of the Cmean for the most highly curved luminalepithelium (0.125<Cmean>0.3), to the least curved luminalepithelium regions (0<Cmean>0.05); (Fig. S1A-C). An increasein the depth of folds was observed from GD2.5 (Fig. 3B, f=0.55) toGD3.5-GD3.75 (Fig. 3C, f=0.75). At GD4.25, when embryoattachment has occurred, folds in the uterine lumen resolvedspecifically at implantation sites but were retained in interveninginter-implantation regions (Fig. 3D″, f=0.66; Movie 6). Thesequantitative descriptions of endometrial topology will advanceunderstanding of mutants in uterine epithelial organization and inluminal closure.

Wnt5a-deficient uteri have altered fold morphologyConditional deletion of Wnt5a (Wnt5acKO) in the uterine luminalepithelium and mesenchyme, using the postnatally expressedPgrcre, leads to defective crypt formation, aberrant geneexpression at implantation sites and mid-gestation lethality (Chaet al., 2014). When the Surface Curvature algorithm was applied totheWnt5acKO uteri at GD3.75, we observed multiple folding defects

(Fig. 3E,F). Lumens appeared flatter and the orientation of folds wasaltered, as ridges increasingly ran along the oviductal-cervical axisinstead of the M-AM axis. Owing to resolution of folds at theimplantation site at GD4.25 in wild type (Fig. 3D), we askedwhether aberrant folding was retained in inter-implantation regionsin theWnt5acKO uteri. At GD4.25, folds were still misoriented alongthe oviductal-cervical axis in the inter-implantation regions(Fig. 3G,H) of the Wnt5acKO uteri. Orientation of folding alongthe incorrect axis is reminiscent of planar cell polarity phenotypes,such as incorrect orientation of cochlear cilia in Wnt5a-deficientmutants (Qian et al., 2007). Defects in folding pattern such as theseare very difficult to visualize using 2D histology, thusdemonstrating the application of the 3D imaging technique.

Uterine glands reorient towards site of implantationWhile visualizing the uterine glands using FOXA2immunofluorescence, we observed that the glandular ducts bend,elongate and coil with increasing gestational age (Fig. 4).Generating separate surfaces of each glandular tree revealed large-

Fig. 3. Dynamic folding of the mouse uterinelumen along the mesometrial–anti-mesometrialaxis and aberrant folding in Wnt5acKO uteri.(A-C) Uterine luminal surfaces are on the left andfolding heat maps are on the right. (A) Luminalstructure of non-pregnant mouse uterus.(B,C) Luminal folds appear at GD2.5 (B) and becomepredominant at GD3.75 (C). (D) Surface model of aGD4.25 luminal segment containing an embryo.(D′) Increased translucence reveals the site wherethe embryo is present (orange arrow, embryosurface). (D″) Curvature analysis in the same uterinehorn measured using Cmean shows flatness in theperi-implantation region (Cmean values are primarilyless than 0.05, blue/purple), whereas higher values,indicated by green-yellow and red, are found distantfrom the implantation site in the inter-implantationregion. (E-H) Whereas wild-type uteri show foldingalong the M-AM axis (E,G),Wnt5acKO uterine lumensfold along the oviductal-cervical axis throughout thelumen at GD 3.75 (F) and at inter-implantation sites atGD4.25 (H). Scale bars: 500 μm in A-C; 300 μm inD-D″; 200 μm in E-H. M, mesometrial; AM, anti-mesometrial; IS, implantation site.

4751

TECHNIQUES AND RESOURCES Development (2016) 143, 4749-4754 doi:10.1242/dev.144386

DEVELO

PM

ENT

Page 4: Insights from imaging the implanting embryo and the ... · separating lumen and glands, generation of 3D surfacesthrough the length of the uterine horn permitted analysis of luminal

scale changes in their morphology during early pregnancy, whichculminates in the bending of the flanking glands towards the site ofimplantation (Fig. 4A-D, Fig. S2A; Movies 6 and 7). We measuredthe angle between the ducts of the glands and luminal epithelialplane perpendicular to the M-AM axis (Fig. S2B). This revealed adrastic decrease in the ductal angle at the time of implantation (GD4.5, ∼28°) when compared with the ∼54° inclination at GD2.5 or ina non-pregnant mouse (Fig. 4E). We also observed a qualitativeincrease in coiling and ductal length when comparing non-pregnantand GD4.25 glandular trees (Fig. 4F,G; Figs S3 and S4). Spatially,the elongation of glandular ducts and the bending of the uterineglands positions the glandular epithelium in close proximity tostroma that surround the implantation site. This reorganization canhelp explain the mechanisms behind emerging evidence thatimplicates uterine glands in decidualization and placentation post-implantation (Filant and Spencer, 2014).This new methodology allows observations in an intact organ,

and the quantification of structure and morphological changes, yet

still permits optical sectioning in any desired plane comparable withtraditional histological views. Our analysis integrates surfacecurvature measurements with confocal 3D imaging. Thealgorithm developed for Surface Curvature can easily be modifiedto account for object size and complexity for broader applications inother organ systems.

We expect that this techniquewill reveal novel aspects of uterinearchitecture during development, estrus and pregnancy. Theunderstudied uterine glands that are key to the development ofthe embryo until the formation of the placenta are now accessiblefor studies of branching morphogenesis. This technique will allowfor detailed investigation of the roles of different signalingpathways in development of the uterine lining and implantation.Elucidating such contributions to the spatial uterine environmentwill aid in: (1) better understanding of pregnancy failures due toimplantation defects; (2) improving implantation rates when usingassisted reproductive technologies such as in vitro fertilization; (3)opening doors for novel contraceptive targets; and (4) better

Fig. 4. Glandular ducts reorient towards thesite of implantation. (A-D′) 3D images andsurface renderings of luminal segments anduterine glands, with separate glandularstructures randomly pseudocolored for easyvisualization. (A-B′) View from the ventral side(A,A′) and mesometrial side (B,B′) of non-pregnant uterine segment. (C-D′) View from theventral side (C,C′) and mesometrial side (D,D′)of GD4.5 uterine segment. Boxed areas in B,Drepresent magnified regions in B′,D′,respectively. (E) The glandular ducts in non-pregnant uteri (B,B′) are at amean angle of∼54°to the uterine oviductal-cervical axis. (D,D′) Withthe introduction of the embryo (orange arrow) atthe anti-mesometrial pole at GD4.5, theglandular ducts bend drastically until they are ata mean angle of ∼28° to the oviductal-cervicalaxis towards the site of implantation. The ductangle was measured in a total of 70-80 ductsfrom two different mice and around threeembryos at GD4.5 (E). A t-test was used forstatistical analysis and significance was definedas P<0.001. ****P<10−12; ns, not significant.(F,G) Representative examples of glandularbranching in non-pregnant uteri (F), andglandular branching, coiling and duct elongationin GD4.5 uteri (G). Scale bars: 500 μm in A-D′.M, mesometrial; AM, anti-mesometrial; Ov,ovary; Cx, cervix; IS, implantation site.

4752

TECHNIQUES AND RESOURCES Development (2016) 143, 4749-4754 doi:10.1242/dev.144386

DEVELO

PM

ENT

Page 5: Insights from imaging the implanting embryo and the ... · separating lumen and glands, generation of 3D surfacesthrough the length of the uterine horn permitted analysis of luminal

understanding diseases such as uterine hyperplasia and endometrialcancer.

MATERIALS AND METHODSMiceCD1 mice were purchased from Charles River. Wnt5acKO (PgrCre;Wnt5aflox/flox, mixed 129Sv × C57BL/6 background) pregnant uteri wereobtained from S. K. Dey (Cincinnati Children’s Hospital Medical Center,Cincinnati, USA) (Cha et al., 2014). Uteri were dissected from non-pregnantadult females or from timed matings at: GD2.5, GD3.5 and GD4.5 (between12:00 and 15:00 hours); GD4.25 at 09:00 hours; and GD3.75 between 19:00and 21:00 hours on the day of dissection. The dark period was 19.00 to05.00 h and the day of mating plug was identified as gestational day (GD)0.5. All mouse work was carried out under University of CaliforniaSan Francisco Institutional Animal Care and Use Committee guidelines, inan AAALAC approved facility.

Human sample collectionHuman adult uterine samples were obtained from the NIH UCSF HumanEndometrial Tissue and DNA Bank, which contains samples of womenundergoing endometrial biopsy or hysterectomy for nonmalignantindications. The samples were collected under approved InstitutionalReview Board protocols and informed consent to participate in the studywas obtained from the patients.

Whole-mount immunofluorescenceUteri were fixed in DMSO:methanol (1:4) and blocked (using PBS+1%Triton+2% powdered milk) for 2 h at room temperature. Primary antibodiesfor mouse E-CAD (M108, Clontech), human E-CAD (ab1416, Abcam) andFOXA2 (NBP1-95426, Novus Biologicals; or ab108422, Abcam) werediluted (1:200) in block, and uteri were incubated for 5 nights at 4°C. Uteriwere washed (PBS+1% Triton) six times for 30 min each and incubated withsecondary antibodies [fluorescently conjugated Alexa Fluor IgGs(Invitrogen)] for 2 nights at 4°C. Uteri were washed (in PBS+1% Triton)six times for 30 min each, dehydrated in methanol and incubated overnight in3%H2O2 diluted in methanol. Uteri were washed in 100%methanol twice for15 min each and cleared overnight using BABB (benzyl alcohol:benzylbenzoate, 1:2). Uteri were imaged using a Leica SP5 TCS confocalmicroscope with white-light laser, using a 10× air objective with z stacksthat were 7 µm apart. Full uterine horns were imaged using 18×2 tile scans andtiles were merged using the mosaic merge function of the Leica software. Forfurther details on the immunofluorescence, see supplementary Materials andMethods.

Image analysisLIF files (Leica software) were analyzed using Imaris v8.1 (Bitplane). Usingthe channel arithmetic function, the glandular FOXA2+ signal was removedfrom the E-CAD+ signal to create lumen-only signal. Surfaces were createdin surpass 3D mode for the luminal signal and the FOXA2+ glandularsignal. Images and videos were captured using the snapshot function and theanimation function, respectively. For further details of image analysis, seesupplementary Materials and Methods.

Description of Matlab script and calculation of CmeanTheMatlab scriptwas adapted fromhttp://www.mathworks.com/matlabcentral/fileexchange/32573-patch-curvature. For further details, including equationsused to calculate Cmean, see supplementary Materials and Methods.

Determination of folding using Surface CurvatureFolding measurements were generated by modifying an existing Matlabscript made available by Imaris Open (Bitplane), originally intended tocalculate localized, small area, curvature. The script was modified tocalculate the curvature in larger objects, such as the uterus (Matlab Script forSurface Curvature). The analysis was carried out using the verticesgenerated in the Surface mode in Imaris. Surface complexity is reducedby evenly reducing the number of vertices to one-tenth of the originalnumber. Using the normals in each axis at each vertex, the radius of the

curve formed by connecting neighbor vertices was calculated. Radiicalculated with six neighbor vertices are averaged to obtain curvature meanand multiplied by 10 to calculate Cmean. Cmean is displayed as a heat mapwhere the value of 0-0.05 is blue/purple and curvature values above 0.200are displayed in red. Morphology of uterine luminal segments is expressedas the folding factor ( f ), which is the ratio of the Cmean for the highlycurved luminal epithelium (0.125<Cmean>0.3), to the least curved luminalepithelium regions (0<Cmean>0.05) (Fig. S1A-C):

Folding factor f ¼ Number of vertices ðCmean 0:125� 0:3ÞNumber of vertices ðCmean 0:0� 0:05Þ :

Cmean values above 0.3 were attributed to remnants of glandular ductsafter subtraction and accordingly excluded from lumen curvature analysis.

Determination of bending angle for glandular ductsThe angle of glandular duct bending towards the implantation site wasmeasured using the Imaris function measurement points. Three planes alongthe xy/yz and zx axes can be displayed using the orthogonal slicer. The threeplanes separate along the dorsal/ventral, anterior/posterior and mesometrial/anti-mesometrial axis, respectively. Using both the confocal z slices of theuterus and glandular surfaces, the three planes can be positioned to define fourstandardized measurement points. The first point is placed at the duct of thegland (where it branches from the luminal epithelium) and the intersection ofthe anterior/posterior axis and the dorsal/ventral axis. The secondmeasurement point is placed at the farthest branched tip of the gland alongthe intersection of the dorsal/ventral axis and the mesometrial/anti-mesometrial axis. The third measurement point is placed at theintersection of the mesometrial/anti-mesometrial plane, the anterior/posterior(luminal) plane and the dorsal/ventral axis. The fourth measurement point isplaced at the starting position that creates a right-angled triangle that dictatesthe degree to which the glandular duct is bending, with the luminal epitheliumalways being the standard comparison point.

AcknowledgementsWe thank S. K. Dey and Jeeyeon Cha for the Wnt5acKO uteri, David Rowitch of theUCSF IVF core, Matthew Krummel of the Biological Imaging Development Center,and Zev Gartner for feedback on the manuscript.

Competing interestsThe authors declare no competing or financial interests.

Author contributionsR.A. and D.J.L. conceived the project; R.A., K.S. and K.O. performed theexperiments; R.A., A.F. and K.M. performed the image analysis; R.A., L.C.G. andD.J.L. analyzed the data and wrote the manuscript; K.S., A.F. and K.M. providedmanuscript feedback.

FundingWe acknowledge support from the California Institute for Regenerative Medicine(TG2-01153), the UCSF Program for Breakthrough Biomedical Research and theNational Institutes of Health (5T32HD007263-32 to R.A., P50HD055764 to L.C.G.,and DP2OD007420 and 1R21ES023297 to D.J.L.). Deposited in PMC for releaseafter 12 months.

Data availabilityTheMatlab script used to determine surface curvature, which waswasmodified froma script made available by Imaris Open (Bitplane), is provided in the supplementarydata.

Supplementary informationSupplementary information available online athttp://dev.biologists.org/lookup/doi/10.1242/dev.144386.supplemental

ReferencesBesnard, V., Wert, S. E., Hull, W. M. and Whitsett, J. A. (2004).

Immunohistochemical localization of Foxa1 and Foxa2 in mouse embryos andadult tissues. Gene Expr. Patterns 5, 193-208.

Cha, J., Sun, X. and Dey, S. K. (2012). Mechanisms of implantation: strategies forsuccessful pregnancy. Nat. Med. 18, 1754-1767.

4753

TECHNIQUES AND RESOURCES Development (2016) 143, 4749-4754 doi:10.1242/dev.144386

DEVELO

PM

ENT

Page 6: Insights from imaging the implanting embryo and the ... · separating lumen and glands, generation of 3D surfacesthrough the length of the uterine horn permitted analysis of luminal

Cha, J., Bartos, A., Park, C., Sun, X., Li, Y., Cha, S.-W., Ajima, R., Ho, H.-Y. H.,Yamaguchi, T. P. and Dey, S. K. (2014). Appropriate crypt formation in the uterus forembryo homingand implantation requiresWnt5a-RORsignaling.Cell Rep.8, 382-392.

Chen, Q., Zhang, Y., Elad, D., Jaffa, A. J., Cao, Y., Ye, X. and Duan, E. (2013).Navigating the site for embryo implantation: biomechanical and molecularregulation of intrauterine embryo distribution. Mol. Aspects Med. 34, 1024-1042.

Daikoku, T., Song, H., Guo, Y., Riesewijk, A., Mosselman, S., Das, S. K. andDey,S. K. (2004). Uterine Msx-1 andWnt4 signaling becomes aberrant in micewith theloss of leukemia inhibitory factor or Hoxa-10: evidence for a novel cytokine-homeobox-Wnt signaling in implantation. Mol. Endocrinol. 18, 1238-1250.

Daikoku, T., Cha, J., Sun, X., Tranguch, S., Xie, H., Fujita, T., Hirota, Y., Lydon,J., DeMayo, F., Maxson, R. et al. (2011). Conditional deletion of Msx homeoboxgenes in the uterus inhibits blastocyst implantation by altering uterine receptivity.Dev. Cell 21, 1014-1025.

Faire, M., Skillern, A., Arora, R., Nguyen, D. H., Wang, J., Chamberlain, C.,German, M. S., Fung, J. C. and Laird, D. J. (2015). Follicle dynamics and globalorganization in the intact mouse ovary. Dev. Biol. 403, 69-79.

Filant, J. and Spencer, T. E. (2014). Uterine glands: biological roles in conceptusimplantation, uterine receptivity and decidualization. Int. J. Dev. Biol. 58, 107-116.

Filant, J., Zhou, H. and Spencer, T. E. (2012). Progesterone inhibits uterine glanddevelopment in the neonatal mouse uterus. Biol. Reprod. 86, 141-149.

Hosie, M. J., Adams, S. M., Thompson, M. B. and Murphy, C. R. (2003).Viviparous lizard, Eulamprus tympanum, shows changes in the uterine surfaceepithelium during early pregnancy that are similar to the plasma membranetransformation of mammals. J. Morphol. 258, 346-357.

Laird, M. K., Thompson, M. B., Murphy, C. R. and McAllan, B. M. (2014). Uterineepithelial cell changes during pregnancy in a marsupial (Sminthopsiscrassicaudata; Dasyuridae). J. Morphol. 275, 1081-1092.

Ma, W.-G., Song, H., Das, S. K., Paria, B. C. and Dey, S. K. (2003). Estrogen is acritical determinant that specifies the duration of the window of uterine receptivityfor implantation. Proc. Natl. Acad. Sci. USA 100, 2963-2968.

Murphy, C. R. (2004). Uterine receptivity and the plasmamembrane transformation.Cell Res. 14, 259-267.

Qian, D., Jones, C., Rzadzinska, A., Mark, S., Zhang, X., Steel, K. P., Dai, X. andChen, P. (2007). Wnt5a functions in planar cell polarity regulation in mice. Dev.Biol. 306, 121-133.

Reardon, S. N., King, M. L., MacLean, J. A., II, Mann, J. L., DeMayo, F. J., Lydon,J. P. and Hayashi, K. (2012). Cdh1 Is essential for endometrial differentiation,gland development, and adult function in the mouse uterus. Biol. Reprod. 86,1-10.

Stewart, C. L., Kaspar, P., Brunet, L. J., Bhatt, H., Gadi, I., Kontgen, F. andAbbondanzo, S. J. (1992). Blastocyst implantation depends on maternalexpression of leukaemia inhibitory factor. Nature 359, 76-79.

Wang, H. and Dey, S. K. (2006). Roadmap to embryo implantation: clues frommouse models. Nat. Rev. Genet. 7, 185-199.

Winkelmann, A. and Spornitz, U. M. (1997). Alkaline phosphatase distribution inrat endometrial epithelium during early pregnancy: a scanning electron-microscopic study. Cells Tissues Organs 158, 237-246.

Yoshinaga, K. (2013). A sequence of events in the uterus prior to implantation in themouse. J. Assist. Reprod. Genet. 30, 1017-1022.

4754

TECHNIQUES AND RESOURCES Development (2016) 143, 4749-4754 doi:10.1242/dev.144386

DEVELO

PM

ENT