Live imaging analysis of human gastric epithelial spheroids reveals spontaneous rupture, rotation and fusion events Authors: T. Andrew Sebrell, Rachel Bruns, Royce A. Wilkinson, Blake Wiedenheft, Paul J. Taylor, Brian A. Perrino, Linda C. Samuelson, James N. Wilking, and Diane Bimczok. The final publication is available at Springer via https://doi.org/10.1007/s00441-017-2726-5 Sebrell TA, B Sidar, R Bruns, RA Wilkinson, B Wiedenheft, PJ Taylor, BA Perrino, LC Samuelson, JN Wilking, D Bimczok, “Live imaging analysis of human gastric epithelial spheroids reveals spontaneous rupture, rotation and fusion events,” Cell and Tissue Research. February 2018;371(2):293-307. doi: 10.1007/s00441-017-2726-5. Made available through Montana State University’s ScholarWorks scholarworks.montana.edu Live imaging analysis of human gastric epithelial spheroids reveals spontaneous rupture, rotation and fusion events T. Andrew Sebrell , Barkan Sidar , Rachel Bruns , Royce A. Wilkinson , Blake Wiedenheft , Paul J. Taylor , Brian A. Perrino , Linda C. Samuelson , James N. Wilking , Diane Bimczok Three-dimensional cultures of primary epithelial cells including organoids, enteroids and epithelial spheroids have become increasingly popular for studies of gastrointesti-nal development, mucosal immunology and epithelial infec-tion. However, little is known about the behavior of these complex cultures in their three- dimensional culture matrix. Therefore, we performed extended time-lapse imaging analy-sis (up to 4 days) of human gastric epithelial spheroids gener-ated from adult tissue samples in order to visualize the dynam-ics of the spheroids in detail. Human gastric epithelial spher-oids cultured in our laboratory grew to an average diameter of 443.9 ± 34.6 μm after 12 days, with the largest spheroids reaching diameters of >1000 μm. Live imaging analysis re-vealed that spheroid growth was associated with cyclic rupture of the epithelial shell at a frequency of 0.32 ± 0.1/day, which led to the release of luminal contents. Spheroid rupture usually resulted in an initial collapse, followed by spontaneous re-formation of the spheres. Moreover, spheroids frequently rotated around their axes within the Matrigel matrix, possibly propelled by basolateral pseudopodia-like formations of the epithelial cells. Interestingly, adjacent spheroids occasionally underwent luminal fusion, as visualized by injection of indi-vidual spheroids with FITC–Dextran (4 kDa). In summary, our analysis revealed unexpected dynamics in human gastric spheroids that challenge our current view of cultured epithelia as static entities and that may need to be considered when performing spheroid infection experiments. In recent years, epithelial organoids have become increasingly popular as a new and powerful tool to study gastrointestinal development and disease (Dedhia et al. 2016; Hynds and Giangreco 2013; Leushacke and Barker 2014). Since the ini-tial description of epithelial organoids derived from murine intestine (Ootani et al. 2009; Sato et al. 2009), organoid cul-ture systems have been adapted to multiple epithelial organ systems and species (Powell and Behnke 2017; Sato and Clevers 2015). For the first time, primary gastrointestinal ep-ithelial cells from various species can now be propagated for extended periods of time by culturing the cells in a three-dimensional collagen matrix and by supplementing Wnt3a, noggin and R-spondin to support maintenance of the stem cell compartment. Importantly, the utilization of adult stem cells enables the generation of primary epithelial cell lines from patient tissues with the potential of performing translational studies and patient-specific analyses (Dekkers et al. 2013; VanDussen et al. 2015). The availability of established proto-cols for primary epithelial cell cultures has led to exponential growth in publications using these methods over the last 5 years, with the number of Borganoid^ publications per year increasing from <10 in 2007 to ≥450 in 2016. Thus, various forms of organoid cultures are starting to replace more tradi- tional epithelial culture methods that involve the use of trans- formed cell lines derived from gastrointestinal tumors. However, little is known about the behavior of these com- plex living spheres in their three-dimensional matrix, beyond diametrical expansion due to cell proliferation. Several publi- cations on gastrointestinal organoids or spheroids include vid- eo supplements showing growth over time (Mahe et al. 2013; Schlaermann et al. 2016; Schumacher et al. 2015; Schwank et al. 2013). Some of these videos challenge our concept of epithelial organoids as static entities that undergo diametrical growth, though the dynamic behaviors observed were not evaluated in detail. In our study, we established primary gastric epithelial cell lines from 13 healthy human adults. These lines were maintained as spheroids following the protocol published by Miyoshi and Stappenbeck (2013), which has been used by multiple groups to culture organoids from intestinal (Bradford et al. 2017; Howitt et al. 2016; Powell and Behnke 2017; Riehl et al. 2015) and gastric sites (Demitrack et al. 2017; den Hartog et al. 2016; Gifford et al. 2017; VanDussen et al. 2015). With these cultures, we per- formed live imaging analysis for up to 4 days. Under the culture conditions used, rupture events occurred cyclically in the majority of the cultures imaged and were associated with the release of luminal material and subsequent healing of the majority of spheroids. In addition, we also observed spheroids that rotated in the Matrigel and spheroids that fused with other spheroids. These observations indicate that human gastric spheroid cultures are surprisingly dynamic, at least under the specific culture conditions used here. Notably, the epithelial barrier function of the spheroids may temporarily be compro- mised because of relatively common spontaneous rupture events that are not visible using routine monitoring techniques. Materials and methods Human gastric epithelial spheroid culture Thirteen gastric tissue specimens from sleeve gastrectomy surgeries were obtained with Institutional Review Board (IRB) approval by the National Disease Research Interchange (NDRI; Philadelphia, PA, USA) or by Dr. Kent Sasse (Sasse Surgical Associates, Reno, NV, USA). Donor characteristics are listed in Table 1. Information on patholog- ical alteration of the tissues was provided by the NDRI. Gastric glands were isolated as described previously (Bimczok 2013, 2014). Briefly, we dissected the gastric mucosa off the muscle layer and cut the mucosa into <1-mm pieces, which were placed into RPMI-1640 medium supple- mented with 0.5 U/mL collagenase type IV, 0.2 mg/mL DNAse (Sigma-Aldrich, St. Louis, MO, USA), 0.3% BSA, 250 μg/mL amphotericin B (Fisher Scientific, Fair Lawn, NJ, USA), 100 U/mL penicillin/streptomycin, 2 mM L-gluta- mine, 1 mM HEPES (GE Healthcare Life Sciences, Logan UT, USA) and 50 μg/mL Gentamycin (IBI Scientific, Peosta, IL, USA). The tissue was incubated at 200 rpm in a 37 °C water bath for 1 h. The tube was then vortexed for 30 s to release glands from the tissue. To establish gastric spheroid cultures, the glands and remaining tissue pieces were centri- fuged at 200g at 4 °C for 5 min. The pellet was resuspended in 30mL ice-cold DPBS (Hyclone GEHealthcare Life Sciences) and vortexed again for 30 s. Tissue pieces were allowed to settle to the bottom of the tube and gastric glands in the supernatant were transferred to a new 50-mL tube, pelleted and transferred to Matrigel (Corning, Bedford, MA, USA). Glands suspended in Matrigel were pipetted into a pre- warmed 24-well plate. Gastric spheroid cultures were main- tained following the protocol of Miyoshi and Stappenbeck (2013), with minor modifications as described by Gifford et al. (2017). After polymerization, Matrigel was overlaid with 500 μL of L-WRN medium composed of Advanced DMEM/F12 (Gibco by Life Technologies, Grand Island, NY, USA) supplemented with 10 mM HEPES, 1% Pen/ Strep, 50% L-WRN conditioned medium, 10 μM ROCK- inhibitor Y-27632 (Tocris Biosciences, Bristol, UK), Amphotericin B, Gentamycin, L-Glutamine, 10 μM TGF-β inhibitor SB-431542 (Tocris), 10% FBS (Rocky Mountain Bio, Missoula, MT, USA) and 50% of cell culture supernatant from L-WRN cells, which constitutively secrete murine Wnt3a, noggin and R-spondin 3 (Miyoshi et al. 2012). L- WRN cells were kindly provided by Dr. T. Stappenbeck, Washington University, St. Louis, USA. The final concentra- tion of Wnt3a in the culture media was 6.2 ± 0.2 μg/mL as determined by testing 3 individual supernatants from L-WRN cells with the TopFlash assay (van de Wetering et al. 1991), using the Firefly Luciferase Assay Kit 2.0 (Biotium, Fremont, CA, USA) and recombinant murineWnt3a (Peprotech, Rocky Hill, NJ, USA) as a standard. Formed spheroids were main- tained in a 37 °C, 5% CO2 incubator, with fresh medium added every 2–3 days and were passaged by trypsinization and re-plating at 1:4 every 5–7 days. The conditions used in this study allowed the continuous culture of human gastric spheroids for at least 50 passages (approximately 11 months), without apparent changes in growth, morphology or viability. Immunohistochemistry and immunofluorescence analysis For paraffin-embedded sections, Matrigel plugs containing spheroids were fixed in 10% neutral buffered formalin (Richard Allen Scientific, Kalamazoo, MI, USA). Paraffin- embedded sections were prepared on a Sakura Tissue-Tek VIP1000 tissue processor and embedding center and sec- tioned at 5 μm on a Leica 2035 rotary microtome. Sections were stained with hematoxlin-eosin (HE) reagent (Richard Allen Scientific). For whole-mount staining, gastric epithelial spheroids grown on 8-well chamber slides were fixed for 30 min at 4 °C in Cytofix (BD Biosciences, San Diego, CA, USA). Samples were blocked in PBS containing 10% FBS (Rocky Mountain Bio), 0.2% Triton X-100, 0.1% BSA and 0.05% Tween-20 (Fisher Scientific) and were incubated with primary antibody overnight at 4 °C. The following primary antibodies were used: mouse anti-human cytokeratin, clone CAM5.2 (BD Biosciences, San Jose, CA, USA; recognizes Moll’s cytokeratin peptide #8 and #7) and mouse anti-human E-Cadherin, clone 67A4 (BioLegend, San Diego, CA, USA). Wells were washed with DPBS and isotype-specific Alexa488-labeled secondary antibody (SouthernBiotech, Birmingham, AL, USA) was added for 2 h at room tempera- ture. After a final wash step, samples were covered with ProLong Gold with DAPI (Fisher Scientific). Quantitative RT-PCR Total RNAwas isolated from epithelial spheroids and human gastric tissue samples using the Direct-zol RNAMiniPrep Kit (ZymoResearch, Irvine, CA, USA). Complementary DNA was generated using reverse transcription performed from 1 μg RNA using the iScript cDNA Synthesis Kit (Bio-Rad Laboratories, Hercules, CA, USA). qPCR was performed using iTaq Universal SYBR Green Supermix (Bio-Rad) on a Roche LightCyler96 (Roche, Basel, Switzerland). PCR primers sequences used are listed in Suppl. Table 1. PCR data were analyzed using the 2-ΔΔCTmethod, with gastric spheroid gene expression normalized to GAPDH and gene expression in human gastric tissue. Growth measurements and counting To determine spheroid size, cultures were imaged by phase contrast microscopy and spheroid diameters were measured on composite images of entire culture wells. To obtain cell counts, spheroid cultures were incubated with 0.25% Trypsin EDTA solution (EMD Millipore, Billerica, MA, USA) at 37 °C for 5–7 min and mixed with a pipet tip until a single cell suspension was obtained. Cells were then counted on a hemocytometer or on a flow cytometer using reference cells (Pechhold et al. 1994). Generation of EGFP- and mCherry-expressing spheroids by lentiviral transduction To produce lentiviral particles, HEK293 cells were plated at 40% confluency in DMEM/10% fetal calf serum/penicillin/ streptomycin the day before transfection. One hour before transfection, the media was replaced with pre-warmed OptiMEM (Gibco, Grand Island, NY, USA). Cells were co- transfected with pCMV-VSV-G (a gift from Bob Weinberg; Addgene plasmid # 8454; Stewart et al. 2003), psPAX2 (a gift from Didier Trono; Addgene plasmid # 12260) and either pLJM1-EGFP (a gift from David Sabatini; Addgene plasmid # 19319; Sancak et al. 2008) or pLV-mCherry (a gift from Pantelis Tsoulfas; Addgene plasmid # 36084) using Lipofectamine 3000 (ThermoFisher) according to the manu- facturer’s instructions. Six hours after transfection, the OptiMEM was removed and replaced with DMEM/10% fetal calf serum/1% BSA. The supernatant containing the lentivirus was harvested 48 h and 60 h post-transfection. The combined (48 h and 60 h) samples were centrifuged for 10 min at 1000g at 4 °C, filtered through a low-protein binding 0.45-μm filter and concentrated by ultracentrifugation (80,000g at 4 °C for 2 h). The virus was resuspended by overnight incubation at Table 1 Organoid lines and donor characteristics used in this study Organoid line Tissue Donor (age/sex) Pathology Experiments hu001 Gastric body 45/F Polypoid fundic nodules Imaging, FACS hu002 Gastric body 49/F Moderate gastritis Imaging, FACS hu003 Gastric body 45/F Mild gastritis FACS hu004 Gastric body 52/F Mild chronic inflammation Imaging, FACS hu005 Gastric body 38/F No alterations observed FACS hu006 Gastric antrum 42/F Not available Imaging, FACS hu007 Gastric body 26/F Not available FACS hu008 Gastric body 35/F No alterations observed Imaging, FACS hu009 Gastric body 31/F No alterations observed FACS hu010 Gastric body 43/F Mild gastritis FACS hu011 Gastric body 52/M No alterations observed qRT-PCR hu013 Gastric body 57/M Not available qRT-PCR hu014 Gastric body 57/M No alterations observed qRT-PCR 4 °C in DMEM/10% FCS/1% BSA, aliquoted and stored at −80 °C. To perform lentiviral transduction of gastric spheroids, the spheroids were harvested and trypsinized to obtain a single cell suspension. Cells were suspended in spheroid L-WRN media prepared with 10 μM ROCK Inhibitor Y-27632 (Tocris Biosciences) and hexadimethrine bromide (Sigma- Aldrich). Cells were incubated with 30 μL of lentivirus in 250 μL L-WRN media and incubated at 37 °C in 5% CO2. After 6 h, suspensions were transferred to Eppendorf tubes and centrifuged at 500g for 5 min. Epithelial cells were trans- ferred to Matrigel and plated into a warmed 24-well plate for further culture and were maintained in the presence of puro- mycin (Sigma) for EGFP. Spheroids with high EGFP or mCherry expression were derived from FACS-purified sin- gle-cell suspensions. Microinjection of gastric spheroids with FITC–Dextran A Fisher stereomicroscope fitted with a Genesearch Embryo Cradle (Genesearch, Bozeman, MT, USA) and a 2 μL syringe (Hamilton, Reno, NV, USA) was used for microinjection. Injection needles with beveled tips were pulled from glass capillaries to a size of 21–23 μm. For the injection, 10-day plated spheroids in 35 mm MatTek dishes were injected with 0.2 μL of 25 μM FITC–Dextran 4 kDa (Sigma-Aldrich, St. Louis, MO, USA). Spheroids were left in the Matrigel matrix for injection. Microscopic analysis Epiflourescence and phase contrast images were acquired using a Life Technologies EVOS FL Auto system equipped with an onstage incubator. Live confocal imaging analysis was performed on an inverted Leica SP5 Confocal Scanning Laser Microscope with 405-, 488-, 561- and 633-nm laser excitation lines and a heated stage with an environmental con- trol chamber. Fluorescence, phase contrast and backscattered light images were collected at 10-min intervals over 17–112 h using a ×10 objective. Backscattered laser light images were obtained by adjusting the wavelength range of the imaging detector to capture the excitation laser wavelength. This tech- nique allows the visualization of interfaces between materials of different densities. For transmission electron microscopy analysis (Brumfield et al. 2009), epithelial spheroid cultures were fixed overnight with 3% glutaraldehyde in 0.05 M potassium sodium phos- phate buffer, pH 7.2, followed by postfixation in 2% osmium tetroxide for 4 h. Samples were dehydrated using an ethanol series (50–100%) and propylene oxide. After dehydration, epithelial spheroids were gradually infiltrated with Spurr’s resin and baked overnight at 70 °C.Then, 60- to 90-nm ultra- thin sections were cut with a Diatome diamond knife on a Reichert OM-U2 ultramicrotome, floated onto 300-mesh cop- per grids and stained with uranyl acetate and Reynold’s lead citrate. All sections were viewed with a LEO 912AB TEM and photographed with a Proscan 2048 × 2048-pixel charge- coupled-device camera. Results Phenotypic analysis of human gastric epithelial spheroids To confirm the identity and differentiation stage of our gastric epithelial spheroid lines maintained using the protocol of Miyoshi and Stappenbeck (2013), we first performed pheno- typic analyses using light microscopy, qRT-PCR and trans- mission electron microscopy (TEM). As anticipated, the epi- thelial cells formed spheroids with columnar to cuboidal epi- thelium and expressed epithelial cytokeratin and E-cadherin (Fig. 1a–c). Quantitative RT-PCR analysis of the gastric spher- oids confirmed expression of genes specific for all five major gastric epithelial cell subsets, i.e., parietal cells (ATP4B), chief cells (pepsinogen C, PGC), surface mucus cells (MUC5ac), mucus neck cells (MUC6) and enteroendocrine cells (chromogranin A, CHGA; Fig. 1d). Notably, gene expression for the enteroendocrine marker CHGA was very low. Moreover, gene expression levels of all five genes analyzed were decreased compared to the levels detected in the gastric biopsy specimens that were used as positive controls for PCR normalization. Ultrastructural analysis by TEM revealed that the gastric spheroids contained several morphologically distinct cell types (Fig. 2). The predominant cell type was a secretory- type cell with large electron-dense vesicles in the apical por- tion of the cell (Fig. 2a, a′), comparable to a mucus pit cell (Corpron 1966; Zeitoun and Lambling 1967). Cells with electron-lucent vesicles near their luminal surface were also present (Fig. 2a, a′′), similar to mucus neck cells. Some cells had large amounts of rough ER and mitochondria, consistent with active protein synthesis as typically found in chief cells (Fig. 2b) (Zeitoun and Lambling 1967). Moreover, we also detected cells with large intracellular canaliculi with projecting microvilli, similar to those described for parietal cells (Fig. 2c) (Rohrer et al. 1965). We did not detect any cells with basal vesicles typical for enteroendocrine cells. Consistent with the general morphology of the gastrointestinal epithelium, spheroid cells had basal nuclei, short apical mi- crovilli (Fig. 2d, d′) and distinct apical junctional complexes (Fig. 2e). We also observed extensive formation of interdigi- tating lateral processes (Fig. 2d, f), as shown by Necchi et al. (2009) for human gastric mucosa. Overall, gastric spheroids show typical features of the human gastric epithelium but do not appear to be fully differentiated. Fig. 2 Transmission electron microscopy of human gastric epithelial spheroids. a Cross-section of a gastric spheroid shows a simple cuboidal to columnar microvillated epithelium with vacuolated secretory cells and basal nuclei. Lm lumen; bar 5 μm. a′ Epithelial cell from (a) with large, electron-dense vacuoles (arrowheads). N nucleus; bar 2 μm. a′′ Epithelial cell from (a) with large, electron-lucent vacuoles (arrowheads). N nucleus; bar 2 μm. b Epithelial cell with copious amounts of rough endoplasmic reticulum (arrowheads) and multiple large mitochondria (Mi); bar 1 μm. c Large microvillated intracellular canaliculus (Cn) in a gastric epithelial cell. Bar 1 μm. d Columnar epithelial cells with luminal microvilli and lateral interdigitating intercellular leaflets (arrowheads)b bar 2 μm. d′ Enlarged image from (d, dashed box) shows luminal microvilli (arrowheads) and mucus (Mu); bar 500 nm. e Luminal-junctional complex (arrowhead) between two epithelial cells; bar 500 nm. (f) Lateral interdigitating intercellular leaflets; bar 1 μm Fig. 1 Microscopic analysis and gene expression analysis of human gastric epithelial spheroids. a Spheroids were fixed, paraffin-embedded and sections stained with hematoxylin-eosin. Bar 50 μm. b, c E-Cadherin (FITC) and cytokeratin (FITC) whole-mount immunofluorescence staining of human gastric spheroids; nuclei were labeled with DAPI. Bar 50 μm. d Quantitative RT-PCR analysis of gastric spheroids (n = 3; hu011, hu013 and hu014). Samples were analyzed for expression of MUC5A (mucous neck cells), PGC (pepsinogen C; chief cells), MUC6 (surface mucus cells), CHGA (chromogranin A, enteroendocrine cells) and ATP4B (Potassium-transporting ATPase subunit beta, parietal cells). Data were analyzed by the 2-ΔΔCT method, with GAPDH used as a housekeeping gene and cDNA from human gastric tissue (geometric mean of n = 3) used for normalization Growth dynamics of human gastric epithelial spheroids Next, we analyzed spheroid growth dynamics, size distribu- tion and cellularity of the spheroids (Fig. 3). Spheroids ex- panded in size over a period of around 10 days and then plateaued, with a median diameter of 398 μm (average 443.9 ± 34.6 μm) after 12 days of culture (Fig. 3a–a′′′′, b). Spheroids within one culture well varied significantly in size (Fig. 3c). To determine the size distribution of gastric spher- oids within a culture, spheroids from 4 different lines were grown until the largest ones reached a size of >600 μm. Cultures were then imaged and spheroid diameters measured on composite digital images of culture wells (Fig. 3d). Size distribution was remarkably consistent between individual spheroid lines and followed a right-skewed bell curve, with 73 ± 8% of spheroids only reaching diameters of ≤300 μm. A small subset of spheroids (0.4 ± 0.4%) reached diameters of ≥800 μm after 7–10 days of culture. Using composite images of entire culture wells, digital im- age measurements and manual cell counting of single cell suspensions derived from the spheroids, we determined the average relationship between spheroid size and cell numbers in our cultures (Fig. 3e), with n being equivalent to the num- bers of cells that can be recovered from a spheroid of a defined radius (in μm): n ¼ 0:00395 0:0007ð Þ 1 μm 4 π r These cell counts correspond to a radius of 20 μm per single epithelial cell. Similar results were obtained using FACS- based cell counting with reference cells (Pechhold et al. 1994). The number of cells per spheroid can be used to assess the recovery of DNA, RNA and protein from cultures with organoids of certain sizes and to calculate the multiplicity of infection for pathogen microinjection experiments. Spontaneous rupture and healing events of human gastric organoids Based on published and anecdotal reports of organoid rupture events (Mahe et al. 2013; Schlaermann et al. 2016; Schwank et al. 2013), we performed live imaging analysis of six distinct human gastric spheroid lines, with 9–83 spheroids per line im- aged over 17–112 h. As shown in Fig. 4a–a′′′′ and Suppl. Movie 1, spheroids frequently oscillated in diameter, consistent with rupture and healing processes. We observed an average of 0.32 ± 0.1 ruptures per spheroid per 24-h period (Fig. 4b), with significant variations between the three lines that were analyzed (P ≤ 0.01).Within a given 24-h time period, some spheroids did not rupture at all, whereas some spheroids ruptured more than once. Overall, 43 ± 10% of spheroids in each line analyzed showed rupture events. We did not observe any significant difference in rupture frequency between low passage (<10) and high passage (>10) spheroids (Fig. 4c). Interestingly, there was a trend for an increased rupture frequency in larger epithe- lial spheroids (Fig. 4d). Imaging of EGFP-expressing epithelial spheroids with a combination of fluorescence confocal imaging, backscatter confocal imaging (Mollazade et al. 2012) and bright field imaging revealed that rupture events were focal and as- sociated with the release of material with increased scattering properties—possibly mucus and cell debris—from the spher- oid lumen (Fig. 5a–c′′′′; Suppl. Movie 2). To visualize loss of material from the spheroid lumen, we performed live imaging on gastric epithelial spheroids that were microinjected with 4- kDa FITC–Dextran. As shown in Fig. 5d–e′′′′, injected FITC– Dextran remained in the spheroid lumen for >12 h, indicating that the epithelial barrier of the spheroids was intact. Upon rupture, the FITC–Dextran completely disappeared from the spheroid lumen (Fig. 5d–e′′′′; Suppl. Movie 3). Notably, epi- thelial thickness was low prior to rupture events and increased immediately following a rupture event. In summary, our data show that spontaneous spheroid rupture is a common event and involves release of luminal material. Human gastric spheroids rotate within the Matrigel matrix Surprisingly, our live imaging analysis also revealed that, even in the absence of rupture events, spheroid interactions with the Matrigel matrix were not static. In particular, spheroids fre- quently rotated around an axis, with an average of 53.6 ± 11.4% of spheroids in all cultures analyzed displaying rotational movement within the matrix (Fig. 6a–a′′′′′, b; Suppl. Movie 4). This behavior was predominantly observed in smaller spheroids (< 100 μm in diameter, data not shown) but showed a large variation between individual spheroid lines and individual cultures. Again, there was no significant differ- ence between low passage (≤ p 10) or high passage (>10) spheroids. To further investigate the dynamic interactions be- tween the spheroids and the Matrigel, we performed backscat- ter light imaging (Fig. 6c-c′′; Suppl. Movie 5). This backscat- ter light analysis revealed that spheroid epithelia form tempo- rary, pseudopod-like basolateral projections that may enable movement of the spheroids within the Matrigel capsule. Adjacent spheroids may undergo membrane fusion Live imaging analysis also revealed that, in some cases, two adjacent spheroids fused to form one larger spheroid (Fig. 7). These events were rare but seen in three out of five different lines analyzed between passages 4 and 18 (Fig. 7a–a′′′′′, b). To confirm that the observed events rep- resented true luminal fusions, spheroids that had been injected with FITC–Dextran (4 kDa) were monitored by live confocal imaging. As shown in Fig. 7c–d′′′′ and Suppl. Movie 6, spheroid fusion resulted in the transfer of green fluorescent FITC–Dextran from the lumen of one injected spheroid to a non-injected spheroid. These observations indicate that spheroid fusions lead to the formation of a common luminal compartment. Fig. 3 Gastric epithelial spheroid growth. a– a′′′′ Representative phase contrast images of one gastric epithelial spheroid imaged repeatedly between days 3 and 12 after plating (line hu001, p17). b Diameters of 65 randomly selected spheroids in one representative well (line hu001, p17) were measured repeatedly on days 3, 5, 7, 10 and 12 after plating. Box delineates 1st and 3rd quartile and median, whiskers show minimum and maximum size. c Composite image of a representative culture well containing mCherry-expressing human gastric spheroids; bar 500 μm. d Size distribution of gastric epithelial spheroids in one representative well each from 4 different lines, imaged on days 7 or 10 after seeding (see label); graph shows mean (–) and individual values for the 4 spheroid lines. e Relationship between spheroid diameter and cell count was determined by measuring all spheroids within one well followed by disruption of the spheroids using trypsin-EDTA and hemocytometer cell counting. Averages ± SEM calculated from 3 independent cultures are shown Discussion In this study, we used live imaging to demonstrate that human gastric epithelial spheroids—or gastrospheres (Stelzner et al. 2012)—cultured under the conditions described (see BMaterials and methods") display several dynamic behaviors that may be overlooked when performing end-point analyses and that may impact experimental results. Specifically, spher- oids spontaneously ruptured and sometimes fused and frequently rotated in the Matrigel matrix. Notably, there were significant differences in the behavior of distinct gastric spher- oid lines and in the behavior of individual spheroid lines analyzed after a different number of passages. Spontaneous rupture was a relatively common event, with >40% of spheroids imaged showing rupture events. Videos included in several previous publications on murine intestinal organoids and human gastric organoids also showed organoid ruptures, although these events were not addressed directly (Mahe et al. 2013; Schlaermann et al. 2016; Schwank et al. 2013), which suggests that spontaneous ruptures are not spe- cific to the specific culture conditions used here. In our study, we quantified rupture events and demonstrated that ruptures occurred in all gastric spheroid lines analyzed in our labora- tory. We also showed that ruptures were preceded by thinning of the epithelium, suggesting that the luminal pressure of the spheroids increased over time. There was a trend for larger spheroids to rupture more frequently but the reasons for this increased rupture behavior are unclear. Since the gastric epi- thelium normally lines a tubular structure rather than a closed sphere, mechanisms that prevent rupture are not likely to exist. Notably, our observation that spheroids rupture on a regular basis could theoretically impact the outcome of experiments if injected microorganisms or other luminally secreted sub- stances were to be released into theMatrigel and unexpectedly interacted with the basolateral side of the epithelium. Gastric spheroids generally underwent additional growth following rupture, indicating efficient and rapid epithelial res- titution. As previously described (Bartfeld et al. 2015; Miyoshi and Stappenbeck 2013), isolated glands spontane- ously form spheres within several hours, which represents another example of epithelial healing behavior. Moreover, we observed that adjacent spheroids occasionally fused, which may also represent a healing mechanism. The ability of organoids to contribute to epithelial healing has been pre- viously demonstrated. In a recent study by Engevik et al. (2016), gastric organoids were transplanted into the gastric submucosa of mice following chemical induction of gastric ulcers. These transplanted organoids significantly improved gastric wound healing, demonstrating that gastric organoids efficiently regenerate defective epithelia (Engevik et al. 2016). Similarly, colonic organoids improved the healing of epitheli- al defects in the murine colon (Yui et al. 2012). Such wound- healing behavior of gastrointestinal epithelia involves chemo- tactic migration of epithelial cells and epithelial flattening and lateral lamellipodia extension that lead to rapid closure of epithelial gaps independent of cell proliferation (Iizuka and Konno 2011; Silen and Ito 1985; Smith et al. 2005). In addi- tion, the efficient reformation of spheroids after rupture and the fusion of closely adjacent spheroids was possibly en- hanced by the high medium concentrations of Wnt, which promotes gastrointestinal epithelial wound healing (Miyoshi 2017). Notably, gastrointestinal epithelial cells are highly dynamic in vivo, with a high epithelial cell turnover rate and efficient closure of gaps caused by apoptotic cell extrusion (Heath 1996; Williams et al. 2015). Gastric surface mucus cells mi- grate from the isthmus, where stem cells are located, to the surface mucosa within 1–3 days (Creamer et al. 1961; Karam et al. 1997; Lipkin 1965). Intestinal epithelial cells move as much as 5–10 μm/h from the base of the crypts towards the tips of the villi and apical junctional complexes have to be constantly re-organized to close gaps left by epithelial cells that undergo apoptosis (Iizuka and Konno 2011). This con- stant reformation of intracellular junctions due to epithelial cell turnover may have played a role in the formation of cell–cell junctions in the observed fusions of intact spheroids. It remains unclear whether the formation of dynamic pseu- dopods observed on the basolateral side of human gastric spheroids was a normal process or specific to our culture con- ditions. Basal pseudopodia or lamellipodia that may promote epithelial cell movement have been identified in normal intes- tinal epithelium but they occur at low frequencies (Heath 1996). In contrast, basal pseudopodia play a major role in epithelial carcinogenesis, where pseudopodia can contribute to invasion and matrix degradation by expanding tumors (McNiven 2013). Basal pseudopod-like processes in the small intestine also appear to be more frequent during early devel- opment (Burgess 1976) and on certa in types of enteroendocrine cells, where they are thought to allow direct- ed secretion of hormones (Bohorquez et al. 2011). In our spheroid cultures, increased basal pseudopod formation might be associated with the low differentiation stage of the cells or might have been provoked by the specific characteristics of ƒFig. 4 Spontaneous spheroid rupture and healing. a– a′′′′Representative image series from a 22-h time course experiment shows epithelial spheroid expansion and rupture (indicated by arrows), followed by re- forming and continued expansion over time. Phase contrast images; bar 100 μm. b Frequency of rupture events in 3 different gastric epithelial spheroid lines analyzed at passage ≤10. Graph shows average number of rupture events per 24-h period for individual spheroids (n = 33–59) and mean ± SD of each line. **Statistically significant difference at P ≤ 0.01. c Average rupture frequency per 24 h in low passage number cultures (P ≤ 10; hu002, n = 59; hu006, n = 38; hu008, n = 33) and high passage number cultures (P > 11, hu001, n = 9; hu004, n = 9; hu006, n = 45); individual values, mean ± SD; n.s. not significant. d Relationship between rupture frequency and size. Spheroid lines hu001 (n = 33 spheroids), hu002 (n = 53) and hu006 (n = 31) were analyzed the Matrigel matrix, which differs from normal basement membrane and extracellular matrix in composition and struc- ture (Benton et al. 2009). Since pseudopodia are generally associated with cellular movement, these processes may have enabled spheroid movement including the observed rotation and fusion events within the Matrigel shell. Our detailed analysis of gastric spheroid growth under pre- viously published culture conditions (Demitrack et al. 2017; Fig. 5 Release of luminal contents from ruptured gastric epithelial spheroids. a– c′′′′ Confocal and backscatter light image series from a 14-h time course analysis shows rupture, release of optically dense material (a– a′′′′) from the inside of the GFP-expressing spheroid (b– b ′′′′) and thickening of the epithelial layer upon healing. c– c′′′′ Brightfield images. Bar 50 μm. d– e′′′′ Release of injected 4-kDa FITC–Dextran (d– d′′′′) from an mCherry-expressing gastric spheroid (e– e′′′′) upon rupture. Images show disappearance of green FITC– Dextran signal after rupture at 13:20 hours. Bar 200 μm den Hartog et al. 2016; Gifford et al. 2017; Miyoshi and Stappenbeck 2013) revealed several additional interesting properties of this model system. Thus, although some spher- oids grew to a size of >1000 μm in diameter, the percentage of these large spheroids was <0.5%, while the majority of spher- oids remained in the 100–300 μm diameter size range after 7– 10 days in culture. Also, we never observed spheroids that grew larger than 1300 μm. Interestingly, TEM analysis of the cultures revealed extensive formation of lateral interdigi- tating folds, as also observed in some gastric tissue specimens in previous studies (Necchi et al. 2009). Structurally similar Fig. 7 Gastric epithelial spheroid fusion events. a– a′′′′′ Phase contrast image series of two human gastric spheroids undergoing fusion. Spheroid line hu004. Bar 200 μm. b Percentage of spheroids with observed fusion events in 5 different human gastric spheroid lines (hu001, p10, n = 149; hu002, p8–10, n = 59; hu004, p18, n = 9; hu006, p7–16, n = 83; and hu008, p4, n = 33), normalized to a 24-h observation period. c– d′′′′ 4- kDa FITC–Dextran injected into one gastric spheroid (arrowhead) is transferred to an adjacent spheroid upon membrane fusion (arrow). Bright field (c– c′′′′) and green fluorescent images (d– d′′′′). Spheroid line hu006. Bar 500 μm ƒFig. 6 Gastric epithelial spheroids rotate within the Matrigel. Spheroids commonly rotate around their axis within the Matrigel matrix. a– a′′′′′ Representative image series of a small single spheroid observed rotating counter-clockwise as indicated by the arrows over a time course of 16 h. b Percentage of low passage (hu002, p8–10, n = 59; hu006, p7–10, n = 38; hu008, p4, n = 33) and high passage gastric spheroids (hu001, p44–45, n = 9; hu004, p18, n = 9; hu006, p11–16, n = 45) observed for 46 ± 1 h that showed rotation. Bars mean ± SEM of individual cultures (n = 1–3 per line). The percentage of rotating epithelial spheroids did not differ significantly between individual lines. c– c′′ Backscatter light imaging reveals basolateral formation of pseudopod-like extensions. Arrows indicate pseudopods. Bar 25 μm lateral folds have been detected in the gall bladder and other epithelial tissues with increased ion transport capacity and have been implicated in contributing to water retention (Diamond and Tormey 1966; Larsen et al. 2009). Notably, we did not alter the composition of the culture media during the experiments in order to induce gastric epi- thelial cell differentiation and our spheroids did not develop gland-like invaginations. Differentiation of primary gastric epithelial cell cultures has previously been achieved by with- drawal of Wnt and noggin (Sato and Clevers 2015) or Notch inhibition (Demitrack et al. 2015, 2017; VanDussen et al. 2015). Since we detected expression of genes specific for all five major epithelial cell subsets and the TEM analysis re- vealed structures very similar to the intracellular canaliculi that are considered typical for gastric parietal cells, we consid- er the epithelial cells in our cultures partially differentiated. It remains unknown whether the behaviors and growth dynam- ics described here similarly occur under culture conditions different from the ones used in our study. Ruptures and organoid movement in the Matrigel have been shown for a number of different epithelial and culture protocols (Mahe et al. 2013; Schlaermann et al. 2016; Schwank et al. 2013) but the frequency of these events had not previously been evaluated. Several different protocols have been used to gen- erate and maintain gastric organoids (Bartfeld et al. 2015; Bertaux-Skeirik et al. 2015; McCracken et al. 2014; Schlaermann et al. 2016; Schumacher et al. 2015) and varia- tions in media composition were shown to affect organoid morphology, differentiation and growth. Future studies will evaluate whether altering culture conditions or adding drugs to affect gastric secretion, e.g., omeprazole, cimetidine or naproxen, may prevent dynamic events such as ruptures that may be undesirable for certain experimental applications. Overall, our study revealed several interesting features of three-dimensional gastric spheroids, including spontaneous ruptures, fusions and rotation events. Interestingly, human gastric epithelial spheroid lines derived from different human donors and spheroids analyzed after a different number of passages showed marked, sometimes significant, differences in many of the parameters analyzed. Consequently, for future studies using human spheroids, gastroids and other organoid culture systems, a general consensus within the scientific com- munity on quality control parameters of organoids would be beneficial, so that data obtained in different laboratories with different cell lines can be compared. Acknowledgements Funding for our study was provided by the National Institutes of Health grants K01 DK097144 (DB); R03 DK107960 (DB), the National Science Foundation, DMR-1455247 (JW) and the Montana University System Research Initiative 51040- MUSRI2015-03 (DB). We greatly appreciate support from the National Institutes of Health IDeA Program grant GM110732, an equipment grant from the M.J. Murdock Charitable Trust and the Montana State University Agricultural Experimental Station for the Flow Cytometry Core Facility at Montana State University. Funding for shared facilities used in this work was also provided by the NSF under award number CBET-1039785. GeneSearch, Inc. development of the GeneSearch Embryo Cradle was funded by an SBIR grant from ORIP/NIH 5R44OD012083 (PJT). We would also like to thank Dr. K. Sasse (Sasse Surgical Associates, Reno, NV) for collecting human gastric tissue samples, Dr. T. Stappenbeck (Washington University, St. Louis) for shar- ing the L-WRN cell line with us and Dr. Seth Walk for helpful discussions. Author contributions D.B., B.W., L.C.S. and J.W. planned and over- saw the experiments; T.A.S., B.S., R.B. and R.A.W. performed the ex- periments; P.J.T. developed microinjection equipment and protocols, B.A.P. provided human gastric tissue samples; D.B., T.A.S. and R.B. analyzed the data; T.A.S. and D.B. wrote the manuscript; all authors provided critical feedback on the manuscript. Compliance with ethical standards Conflict of interest statement Dr. Paul Taylor has a potential conflict of interest, since he is the owner of GeneSearch, Inc., Bozeman, MT, which manufactures the EmbryoCradle microinjector that was used in this study. None of the other authors declare a conflict of interest. References Bartfeld S, Bayram T, van deWetering M, Huch M, Begthel H, Kujala P, Vries R, Peters PJ, Clevers H (2015) In vitro expansion of human gastric epithelial stem cells and their responses to bacterial infection. Gastroenterology 148:126–136 Benton G, George J, Kleinman HK, Arnaoutova IP (2009) Advancing science and technology via 3D culture on basement membrane ma- trix. J Cell Physiol 221:18–25 Bertaux-Skeirik N, Feng R, Schumacher MA, Li J, Mahe MM, Engevik AC, Javier JE, Peek RM Jr, Ottemann K, Orian-Rousseau V, Boivin GP, Helmrath MA, Zavros Y (2015) CD44 plays a functional role in helicobacter pylori-induced epithelial cell proliferation. PLoS Pathog 11:e1004663 Bimczok D, Smythies LE, Waites KB, Grams JM, Stahl RD, Mannon PJ, Peter S, Wilcox CM, Harris PR, Das S, Ernst PB, Smith PD (2013) Helicobacter pylori infection inhibits phagocyte clearance of apo- ptotic gastric epithelial cells. J Immunol 190(12):6626–6634 Bimczok D, Kao JY, Zhang M, Cochrun S, Mannon P, Peter S, Wilcox CM, Mönkemüller KE, Harris PR, Grams JM, Stahl RD, Smith PD, Smythies LE (2014) Human gastric epithelial cells contribute to gastric immune regulation by providing retinoic acid to dendritic cells. Mucosal Immunol 8(3):533–544 Bohorquez DV, Chandra R, Samsa LA, Vigna SR, Liddle RA (2011) Characterization of basal pseudopod-like processes in ileal and co- lonic PYY cells. J Mol Histol 42:3–13 Bradford EM, Ryu SH, Singh AP, Lee G, Goretsky T, Sinh P, Williams DB, Cloud AL, Gounaris E, Patel V, Lamping OF, Lynch EB, Moyer MP, De Plaen IG, Shealy DJ, Yang GY, Barrett TA (2017) Epithelial TNF receptor signaling promotes mucosal repair in in- flammatory bowel disease. J Immunol 199:1886–1897 Brumfield SK, Ortmann AC, Ruigrok V, Suci P, Douglas T, Young MJ (2009) Particle assembly and ultrastructural features associated with replication of the lytic archaeal virus sulfolobus turreted icosahedral virus. J Virol 83:5964–5970 Burgess DR (1976) Structure of the epithelial - mesenchymal interface during early morphogenesis of the chick duodenum. Tissue Cell 8: 147–158 CorpronRE (1966) The ultrastructure of the gastricmucosa in normal and hypophysectomized rats. Am J Anat 118:53–90 Creamer B, Shorter RG, Bamforth J (1961) The turnover and shedding of epithelial cells. I. The turnover in the gastro-intestinal tract. Gut 2: 110–118 Dedhia PH, Bertaux-Skeirik N, Zavros Y, Spence JR (2016) Organoid models of human gastrointestinal development and disease. Gastroenterology 150:1098–1112 Dekkers JF, Wiegerinck CL, de Jonge HR, Bronsveld I, Janssens HM, de Winter-de Groot KM, Brandsma AM, de Jong NW, Bijvelds MJ, Scholte BJ, Nieuwenhuis EE, van den Brink S, Clevers H, van der Ent CK, Middendorp S, Beekman JM (2013) A functional CFTR assay using primary cystic fibrosis intestinal organoids. Nat Med 19: 939–945 Demitrack ES, Gifford GB, Keeley TM, Carulli AJ, VanDussen KL, Thomas D, Giordano TJ, Liu Z, Kopan R, Samuelson LC (2015) Notch signaling regulates gastric antral LGR5 stem cell function. EMBO J 34:2522–2536 Demitrack ES, Gifford GB, Keeley TM, Horita N, Todisco A, Turgeon DK, Siebel CW, Samuelson LC (2017) NOTCH1 and NOTCH2 regulate epithelial cell proliferation in mouse and human gastric corpus. Am J Physiol Gastrointest Liver Physiol 312:G133–G144 Diamond JM, Tormey JM (1966) Role of long extracellular channels in fluid transport across epithelia. Nature 210:817–820 Engevik AC, Feng R, Choi E, White S, Bertaux-Skeirik N, Li J, Mahe MM, Aihara E, Yang L, DiPasquale B, Oh S, Engevik KA, Giraud AS, Montrose MH, Medvedovic M, Helmrath MA, Goldenring JR, Zavros Y (2016) The development of spasmolytic polypeptide/ TFF2-expressing metaplasia (SPEM) during gastric repair is absent in the aged stomach. Cell Mol Gastroenterol Hepatol 2:605–624 Gifford GB, Demitrack ES, Keeley TM, TamA, La Cunza N, Dedhia PH, Spence JR, Simeone DM, Saotome I, Louvi A, Siebel CW, Samuelson LC (2017) Notch1 and Notch2 receptors regulate mouse and human gastric antral epithelial cell homoeostasis. Gut 66:1001– 1011 den Hartog G, Chattopadhyay R, Ablack A, Hall EH, Butcher LD, Bhattacharyya A, Eckmann L, Harris PR, Das S, Ernst PB, Crowe SE (2016) Regulation of Rac1 and reactive oxygen species produc- tion in response to infection of gastrointestinal epithelia. PLoS Pathog 12:e1005382 Heath JP (1996) Epithelial cell migration in the intestine. Cell Biol Int 20: 139–146 Howitt MR, Lavoie S, Michaud M, Blum AM, Tran SV, Weinstock JV, Gallini CA, Redding K, Margolskee RF, Osborne LC, Artis D, Garrett WS (2016) Tuft cells, taste-chemosensory cells, orchestrate parasite type 2 immunity in the gut. Science 351:1329–1333 Hynds RE, Giangreco A (2013) Concise review: the relevance of human stem cell-derived organoid models for epithelial translational medi- cine. Stem Cells 31:417–422 Iizuka M, Konno S (2011) Wound healing of intestinal epithelial cells. World J Gastroenterol 17:2161–2171 Karam SM, Li Q, Gordon JI (1997) Gastric epithelial morphogenesis in normal and transgenic mice. Am J Phys 272:G1209–G1220 Larsen EH, Willumsen NJ, Mobjerg N, Sorensen JN (2009) The lateral intercellular space as osmotic coupling compartment in isotonic transport. Acta Physiol (Oxford) 195:171–186 Leushacke M, Barker N (2014) Ex vivo culture of the intestinal epitheli- um: strategies and applications. Gut 63:1345–1354 Lipkin M (1965) Cell replication in the gastrointestinal tract of man. Gastroenterology 48:616–624 Mahe MM, Aihara E, Schumacher MA, Zavros Y, Montrose MH, Helmrath MA, Sato T, Shroyer NF (2013) Establishment of gastro- intestinal epithelial organoids. Curr Protoc Mouse Biol 3:217–240 McCracken KW, Cata EM, Crawford CM, Sinagoga KL, Schumacher M, Rockich BE, Tsai YH,MayhewCN, Spence JR, Zavros Y,Wells JM (2014) Modelling human development and disease in pluripotent stem-cell-derived gastric organoids. Nature 516:400–404 McNiven MA (2013) Breaking away: matrix remodeling from the lead- ing edge. Trends Cell Biol 23:16–21 Miyoshi H (2017) Wnt-expressing cells in the intestines: guides for tissue remodeling. J Biochem 161:19–25 Miyoshi H, Stappenbeck TS (2013) In vitro expansion and genetic mod- ification of gastrointestinal stem cells in spheroid culture. Nat Protoc 8:2471–2482 Miyoshi H, Ajima R, Luo CT, Yamaguchi TP, Stappenbeck TS (2012) Wnt5a potentiates TGF-beta signaling to promote colonic crypt re- generation after tissue injury. Science 338:108–113 Mollazade K, Omid M, Tab FA, Mohtasebi SS (2012) Principles and applications of light backscattering imaging in quality evaluation of agro-food products: a review. Food Bioprocess Technol 5: 1465–1485 Necchi V, Manca R, Ricci V, Solcia E (2009) Evidence for transepithelial dendritic cells in human H. Pylori active gastritis. Helicobacter 14: 208–222 Ootani A, Li X, Sangiorgi E, Ho QT, Ueno H, Toda S, Sugihara H, Fujimoto K, Weissman IL, Capecchi MR, Kuo CJ (2009) Sustained in vitro intestinal epithelial culture within a Wnt- dependent stem cell niche. Nat Med 15:701–706 Pechhold K, Pohl T, Kabelitz D (1994) Rapid quantification of lympho- cyte subsets in heterogeneous cell populations by flow cytometry. Cytometry 16:152–159 Powell RH, Behnke MS (2017) WRN conditioned media is sufficient for in vitro propagation of intestinal organoids from large farm and small companion animals. Biol Open 6:698–705 Riehl TE, Santhanam S, Foster L, Ciorba M, Stenson WF (2015) CD44 and TLR4 mediate hyaluronic acid regulation of Lgr5+ stem cell proliferation, crypt fission, and intestinal growth in postnatal and adult mice. Am J Physiol Gastrointest Liver Physiol 309:G874– G887 Rohrer GV, Scott JR, Joel W, Wolf S (1965) The fine structure of human gastric parietal cells. Am J Dig Dis 10:13–21 Sancak Y, Peterson TR, Shaul YD, Lindquist RA, Thoreen CC, Bar-Peled L, Sabatini DM (2008) The rag GTPases bind raptor and mediate amino acid signaling to mTORC1. Science 320:1496–1501 Sato T, Clevers H (2015) SnapShot: growing organoids from stem cells. Cell 161:1700-1700 e1701 Sato T, Vries RG, Snippert HJ, van deWetering M, Barker N, Stange DE, van Es JH, AboA,Kujala P, Peters PJ, Clevers H (2009) Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchy- mal niche. Nature 459:262–265 Schlaermann P, Toelle B, Berger H, Schmidt SC, Glanemann M, Ordemann J, Bartfeld S, Mollenkopf HJ, Meyer TF (2016) A novel human gastric primary cell culture system for modelling helicobacter pylori infection in vitro. Gut 65:202–213 Schumacher MA, Aihara E, Feng R, Engevik A, Shroyer NF, Ottemann KM, Worrell RT, Montrose MH, Shivdasani RA, Zavros Y (2015) The use of murine-derived fundic organoids in studies of gastric physiology. J Physiol 593:1809–1827 Schwank G, Andersson-Rolf A, Koo BK, Sasaki N, Clevers H (2013) Generation of BAC transgenic epithelial organoids. PLoS ONE 8: e76871 Silen W, Ito S (1985) Mechanisms for rapid re-epithelialization of the gastric mucosal surface. Annu Rev Physiol 47:217–229 Smith JM, Johanesen PA, Wendt MK, Binion DG, Dwinell MB (2005) CXCL12 activation of CXCR4 regulates mucosal host defense through stimulation of epithelial cell migration and promotion of intestinal barrier integrity. Am J Physiol Gastrointest Liver Physiol 288:G316–G326 Stelzner M, Helmrath M, Dunn JC, Henning SJ, Houchen CW, Kuo C, Lynch J, Li L, Magness ST, Martin MG, Wong MH, Yu J, Consortium NIHISC (2012) A nomenclature for intestinal in vitro cultures. Am J Physiol Gastrointest Liver Physiol 302:G1359– G1363 Stewart SA, Dykxhoorn DM, Palliser D, Mizuno H, Yu EY, An DS, Sabatini DM, Chen IS, Hahn WC, Sharp PA, Weinberg RA, Novina CD (2003) Lentivirus-delivered stable gene silencing by RNAi in primary cells. RNA 9:493–501 VanDussen KL, Marinshaw JM, Shaikh N, Miyoshi H, Moon C, Tarr PI, Ciorba MA, Stappenbeck TS (2015) Development of an enhanced human gastrointestinal epithelial culture system to facilitate patient- based assays. Gut 64:911–920 van de Wetering M, Oosterwegel M, Dooijes D, Clevers H (1991) Identification and cloning of TCF-1, a T lymphocyte-specific transcription factor containing a sequence-specific HMG box. EMBO J 10:123–132 Williams JM, Duckworth CA, Burkitt MD, Watson AJ, Campbell BJ, Pritchard DM (2015) Epithelial cell shedding and barrier function: a matter of life and death at the small intestinal villus tip. Vet Pathol 52:445–455 Yui S, Nakamura T, Sato T, Nemoto Y, Mizutani T, Zheng X, Ichinose S, Nagaishi T, Okamoto R, Tsuchiya K, Clevers H, Watanabe M (2012) Functional engraftment of colon epithelium expanded in vitro from a single adult Lgr5(+) stem cell. Nat Med 18:618–623 Zeitoun P, Lambling A (1967) Ultrastructure of the gastric mucosa in human hemochromatosis. Scand J Gastroenterol 2:222–234