Single luminal epithelial progenitors can generate prostate organoids in culture

Chee Wai Chua, Maho Shibata, Ming Lei, Roxanne Toivanen, LaMont J Barlow, Sarah K Bergren, Ketan K Badani, James M McKiernan, Mitchell C Benson, Hanina Hibshoosh, Michael M Shen, Chee Wai Chua, Maho Shibata, Ming Lei, Roxanne Toivanen, LaMont J Barlow, Sarah K Bergren, Ketan K Badani, James M McKiernan, Mitchell C Benson, Hanina Hibshoosh, Michael M Shen

Abstract

The intrinsic ability to exhibit self-organizing morphogenetic properties in ex vivo culture may represent a general property of tissue stem cells. Here we show that single luminal stem/progenitor cells can generate prostate organoids in a three-dimensional culture system in the absence of stroma. Organoids generated from CARNs (castration-resistant Nkx3.1-expressing cells) or normal prostate epithelia exhibit tissue architecture containing luminal and basal cells, undergo long-term expansion in culture and exhibit functional androgen receptor signalling. Lineage-tracing demonstrates that luminal cells are favoured for organoid formation and generate basal cells in culture. Furthermore, tumour organoids can initiate from CARNs after oncogenic transformation and from mouse models of prostate cancer, and can facilitate analyses of drug response. Finally, we provide evidence supporting the feasibility of organoid studies of human prostate tissue. Our studies underscore the progenitor properties of luminal cells, and identify in vitro approaches for studying prostate biology.

Figures

Figure 1
Figure 1
Generation of prostate epithelial organoids from lineage-marked CARNs. (a) Time course of lineage-marking of CARNs in Nkx3.1CreERT2/+; R26R-YFP/+ mice. (b) Isolation of YFP-positive lineage-marked CARNs by flow cytometry. (c,d) Bright-field (c) and epifluorescent (d) views of CARN-derived organoids that are filled or hollow (arrow). (e,f) Hematoxylin-eosin (H&E) staining of CARN organoids at low-power (e) showing range of phenotypes, and high-power (f). (g) Uniform YFP expression with Ki67 immunostaining (arrows). (h) The basal marker CK5 is expressed on the exterior (arrowheads), while the luminal marker CK8 is expressed internally. (i,j) Strong nuclear expression of AR (arrows, i) and Foxa1 (j). (k–m) Renal grafts generated by tissue recombination of CARN-derived organoids with rat embryonic urogenital mesenchyme (k) display normal stratification of basal (arrowheads, l) and luminal cells (l), and uniform YFP and nuclear AR immunostaining (m); note that the slightly atypical histology in (k) likely reflects the heterozygous phenotype of Nkx3.1 mutants, . (n) Efficiency of organoid formation by lineage-marked CARNs (YFP-positive cells from tamoxifen-induced and castrated Nkx3.1CreERT2/+; R26R-YFP/+ mice; n=4 experiments) and non-CARNs (YFP-negative cells from the same mice; n=3 experiments). Source data are provided in Supplementary Table 1. Error bars represent one standard deviation; the difference between CARNs and non-CARNs is statistically significant (p = 0.002, two-tailed Student’s t-test). (o) Generation of organoids from single CARNs. Time course of paired images shown under bright-field (top) and epifluorescent (bottom) illumination shows organoid growth from isolated single CARN. Scale bars in o correspond to 25 microns, in f–j,l,m to 50 microns, and in c–e,k to 100 microns.
Figure 2
Figure 2
Growth and androgen-responsiveness of prostate organoids from normal prostate epithelium. (a) Flow-sorting strategy to eliminate EpCAM−E-cadherin− cells from dissociated prostate tissue for organoid culture. (b) Low-power view of organoids at 20 days after plating, showing heterogeneity of phenotype. (c) Higher-power view showing hollow and filled budding organoid (arrow). (d,e) H&E staining of sections from a hollow organoid (d) and a multi-layered organoid (e). (f) Many proliferating cells are detectable by Ki67 immunostaining (arrows). (g) Organoids have an outer layer that expresses the basal marker CK5 (arrowheads). (h) Outer cells express the basal marker p63 (arrowheads), while interior cells are positive for the luminal marker CK18. (i) Nuclear immunostaining of AR (arrows) in organoids cultured in standard conditions with DHT. (j) Nuclear immunostaining for Foxa1 (arrows). (k) Tissue recombination of normal organoids with rat embryonic urogenital mesenchyme followed by renal grafting results in reconstitution of prostate tissue. (l,m) Organoids at passage 4 were passaged as single-cell suspensions and plated in the presence of DHT (l) or absence of DHT (m). (n,o) Strong nuclear AR immunostaining in the presence of DHT (n) and weak cytoplasmic AR immunostaining in the absence of DHT (o). (p) qPCR analysis of expression of AR downstream genes in organoids cultured in the presence or absence of DHT. Results are from a single experiment representative of 2 independent experiments. All assays were performed using three technical replicates and normalized to GAPDH expression; Scale bars in e–j,n,o correspond to 50 microns, and in b–d,k,l,m to 100 microns.
Figure 3
Figure 3
Lineage-tracing shows that luminal cells are favored for generation of prostate organoids. (a) Strategy for lineage-marking of basal and luminal epithelial cells for organoid culture. (b) Isolation of YFP-positive luminal cells from CK8-CreERT2; R26R-YFP (CK8-trace) mice by flow cytometry. (c) CK5-trace organoid. (d) Many cells within a CK5-trace organoid are CK5-positive, including internal cells (arrowheads). (e) CK8-trace organoid. (f) CK18-trace organoid. (g) Efficiency of organoid formation from YFP-positive CK5-trace (n=4 experiments), CK8-trace (n=3 experiments), and CK18-trace (n=2 experiments) epithelial cells. The differences in efficiency between CK5-trace and CK8-trace (p=0.001) and between CK5-trace and CK18-trace (p=0.0009) are statistically significant (**) using a two-tailed Student’s t-test; error bars correspond to one standard deviation. Source data are provided in Supplementary Table 1. (h,i) Expression of the basal marker CK5 (arrowheads) in a CK8-trace organoid, shown with (h) and without (i) YFP overlay. (j,k) Expression of the basal marker p63 (arrowheads) in a CK8-trace organoid, shown with (j) and without (k) YFP overlay. (l,m) Expression of the luminal marker CK18 in a CK8-trace organoid, shown with (l) and without (m) YFP overlay. (n) Organoid generated from mixing of red CK18-trace cells and green CK5-trace cells shows green cells on the exterior, consistent with the localization of basal cells. (o) Serial passaging of CK18-trace organoids at passage 3. (p,q) CK18-trace organoids at passage 9 cultured in the presence (p) and absence (q) of DHT. (r,s) AR immunostaining is nuclear in CK18-trace organoids in the presence of DHT (r), but is weakly cytoplasmic in the absence of DHT (s). Scale bars in c–f,h–n correspond to 50 microns, and in o–s to 100 microns.
Figure 4
Figure 4
Tumor organoids can be generated from single transformed CARNs. (a) Time course for generation of transformed CARNs in Nkx3.1CreERT2/+; Ptenflox/flox; KrasLSL/+; R26R-YFP/+ (NPK) mice. (b) NPK-CARN organoid shows extensive budding. (c) H&E staining of NPK-CARN organoids. (d–f) NPK-CARN organoids display Ki67 immunostaining (arrows, d), membrane localization of phospho-Akt (e), and patchy expression of phospho-Erk (arrows, f). (g,h) Luminal phenotype of NPK-CARN organoids, with limited expression of p63 (arrowheads, g) or CK5 (h). (i) Nuclear expression of Foxa1 (arrows). (j,k) AR expression is nuclear in the presence of DHT (arrows, j), but is cytoplasmic in the absence of DHT (k). (l–p) Renal grafts generated by recombination of NPK-CARN organoids with rat embryonic urogenital mesenchyme display high-grade PIN/carcinoma histological phenotypes (l), abundant Ki67 immunostaining (arrows, m), membrane-localized pAkt (n), patchy pErk (arrows, o), and nuclear AR (arrows, p). (q) Generation of organoids from single NPK CARNs. Time course of paired images shown under bright-field (top) and epifluorescent (bottom) illumination shows organoid growth from isolated single NPK CARN. Scale bars in q correspond to 25 microns, in d–h,m–p to 50 microns, and in b,c,i–l to 100 microns.
Figure 5
Figure 5
Modeling tumor phenotypes in organoid culture. (a–j) Formation of organoids from mouse models of prostate cancer, shown in bright-field (a–e) and H&E stained sections (f–j). (a,f) Organoids generated from TRAMP mice at 22 weeks of age. (b,g) Organoids from Nkx3.1CreERT2/+; Ptenflox/flox; p53flox/flox (NPP53) mice induced with tamoxifen at two months of age and assayed at 10 months. (c,h) Organoids from Nkx3.1− − null mutant mice at 14 months of age. (d,i) Organoids from Hi-Myc transgenic mice at 9 months. (e,j) Organoids from Nkx3.1+/−; Pten+/− mice at 10 months. (k) Organoid formation efficiency from the indicated mouse models (data are from 10 technical replicates). (l–r) Induction of tumor phenotypes in culture by tamoxifen treatment of organoids derived from CK8-CreERT2; Ptenflox/flox; KrasLSL-G12D/+; R26R-CAG-YFP mice. (l) Time course of induction experiment. (m–p) Immunostaining for YFP and pAkt in control untreated organoids (m,o) or 4-hydroxy-tamoxifen (4OHT) treated organoids (n,p) at passage 1 (m,n) and at passage 4 (o,p). (q,r) H&E staining of control (q) and 4OHT-treated organoids (r) at passage 4. Scale bars in a–j correspond to 100 microns, and in m–r to 50 microns.
Figure 6
Figure 6
Modeling drug treatment response in organoid culture. (a) Efficiency of organoid formation using organoids from Nkx3.1CreERT2/+; Ptenflox/flox; R26R-YFP/+ (NP) mice. Passaged organoids were treated with the indicated compounds (n=3 samples analyzed per treatment condition); source data are provided in Supplementary Table 1. (b–g) Bright-field images of treated NP organoids. (h,i) H&E sections from control +DHT organoids (h) and enzalutamide + MK8669 treated organoids (i). (j,k) AR expression in control +DHT organoids (arrows, j) and enzalutamide treated organoids (k). (l,m) pAkt expression in control +DHT organoids (arrow, l) and enzalutamide + MK8669 treated organoids (m). Scale bars in h–m correspond to 50 microns, and in b–g correspond to 100 microns. Error bars represent one standard deviation; ** p < 0.01.

References

    1. Peehl DM. Primary cell cultures as models of prostate cancer development. Endocr Relat Cancer. 2005;12:19–47.
    1. Shen MM, Abate-Shen C. Molecular genetics of prostate cancer: new prospects for old challenges. Genes Dev. 2010;24:1967–2000.
    1. Grisanzio C, Signoretti S. p63 in prostate biology and pathology. J Cell Biochem. 2008;103:1354–1368.
    1. Humphrey PA. Diagnosis of adenocarcinoma in prostate needle biopsy tissue. J Clin Pathol. 2007;60:35–42.
    1. Lawson DA, Xin L, Lukacs RU, Cheng D, Witte ON. Isolation and functional characterization of murine prostate stem cells. Proc Natl Acad Sci USA. 2007;104:181–186.
    1. Xin L, Lukacs RU, Lawson DA, Cheng D, Witte ON. Self-renewal and multilineage differentiation in vitro from murine prostate stem cells. Stem Cells. 2007;25:2760–2769.
    1. Shi X, Gipp J, Bushman W. Anchorage-independent culture maintains prostate stem cells. Dev Biol. 2007;312:396–406.
    1. Garraway IP, et al. Human prostate sphere-forming cells represent a subset of basal epithelial cells capable of glandular regeneration in vivo. Prostate. 2010;70:491–501.
    1. Lukacs RU, Goldstein AS, Lawson DA, Cheng D, Witte ON. Isolation, cultivation and characterization of adult murine prostate stem cells. Nat Protoc. 2010;5:702–713.
    1. Guo C, Zhang B, Garraway IP. Isolation and characterization of human prostate stem/progenitor cells. Methods Mol Biol. 2012;879:315–326.
    1. Goldstein AS, et al. Trop2 identifies a subpopulation of murine and human prostate basal cells with stem cell characteristics. Proc Natl Acad Sci USA. 2008;105:20882–20887.
    1. Lawson DA, et al. Basal epithelial stem cells are efficient targets for prostate cancer initiation. Proc Natl Acad Sci USA. 2010;107:2610–2615.
    1. Wang ZA, et al. Lineage analysis of basal epithelial cells reveals their unexpected plasticity and supports a cell-of-origin model for prostate cancer heterogeneity. Nature Cell Biology. 2013;15:274–283.
    1. Lancaster MA, Knoblich JA. Organogenesis in a dish: modeling development and disease using organoid technologies. Science. 2014;345:1247125.
    1. Sato T, et al. Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature. 2011;469:415–418.
    1. Sato T, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459:262–265.
    1. Yui S, et al. Functional engraftment of colon epithelium expanded in vitro from a single adult Lgr5(+) stem cell. Nat Med. 2012;18:618–623.
    1. Sato T, et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology. 2011;141:1762–1772.
    1. Kondo J, et al. Retaining cell-cell contact enables preparation and culture of spheroids composed of pure primary cancer cells from colorectal cancer. Proc Natl Acad Sci USA. 2011;108:6235–6240.
    1. Ootani A, et al. Sustained in vitro intestinal epithelial culture within a Wnt-dependent stem cell niche. Nat Med. 2009;15:701–706.
    1. Barker N, et al. Lgr5(+ve) stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell. 2010;6:25–36.
    1. Huch M, et al. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature. 2013;494:247–250.
    1. Huch M, et al. Unlimited in vitro expansion of adult bi-potent pancreas progenitors through the Lgr5/R-spondin axis. EMBO J. 2013;32:2708–2721.
    1. Greggio C, et al. Artificial three-dimensional niches deconstruct pancreas development in vitro. Development. 2013;140:4452–4462.
    1. Wang X, et al. A luminal epithelial stem cell that is a cell of origin for prostate cancer. Nature. 2009;461:495–500.
    1. Guo W, et al. Slug and Sox9 cooperatively determine the mammary stem cell state. Cell. 2012;148:1015–1028.
    1. Lang SH, et al. Experimental prostate epithelial morphogenesis in response to stroma and three-dimensional matrigel culture. Cell Growth Differ. 2001;12:631–640.
    1. Cano P, Godoy A, Escamilla R, Dhir R, Onate SA. Stromal-epithelial cell interactions and androgen receptor-coregulator recruitment is altered in the tissue microenvironment of prostate cancer. Cancer Res. 2007;67:511–519.
    1. Liu X, et al. ROCK inhibitor and feeder cells induce the conditional reprogramming of epithelial cells. Am J Pathol. 2012;180:599–607.
    1. Zhang L, et al. ROCK inhibitor Y-27632 suppresses dissociation-induced apoptosis of murine prostate stem/progenitor cells and increases their cloning efficiency. PLoS One. 2011;6:e18271.
    1. Xu Y, et al. Revealing a core signaling regulatory mechanism for pluripotent stem cell survival and self-renewal by small molecules. Proc Natl Acad Sci USA. 2010;107:8129–8134.
    1. Gao N, et al. Forkhead box A1 regulates prostate ductal morphogenesis and promotes epithelial cell maturation. Development. 2005;132:3431–3443.
    1. Srinivas S, et al. Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC Dev Biol. 2001;1:4.
    1. Van Keymeulen A, et al. Distinct stem cells contribute to mammary gland development and maintenance. Nature. 2011;479:189–193.
    1. Ousset M, et al. Multipotent and unipotent progenitors contribute to prostate postnatal development. Nat Cell Biol. 2012;14:1131–1138.
    1. Madisen L, et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nature neuroscience. 2010;13:133–140.
    1. Choi N, Zhang B, Zhang L, Ittmann M, Xin L. Adult murine prostate basal and luminal cells are self-sustained lineages that can both serve as targets for prostate cancer initiation. Cancer Cell. 2012;21:253–265.
    1. Lu TL, et al. Conditionally ablated Pten in prostate basal cells promotes basal-to-luminal differentiation and causes invasive prostate cancer in mice. Am J Pathol. 2013;182:975–991.
    1. Aytes A, et al. ETV4 promotes metastasis in response to activation of PI3-kinase and Ras signaling in a mouse model of advanced prostate cancer. Proc Natl Acad Sci USA. 2013;110:E3506–3515.
    1. Bhatia-Gaur R, et al. Roles for Nkx3.1 in prostate development and cancer. Genes Dev. 1999;13:966–977.
    1. Kim MJ, et al. Nkx3.1 mutant mice recapitulate early stages of prostate carcinogenesis. Cancer Res. 2002;62:2999–3004.
    1. Kim MJ, et al. Cooperativity of Nkx3.1 and Pten loss of function in a mouse model of prostate carcinogenesis. Proc Natl Acad Sci USA. 2002;99:2884–2889.
    1. Greenberg NM, et al. Prostate cancer in a transgenic mouse. Proc Natl Acad Sci USA. 1995;92:3439–3443.
    1. Masumori N, et al. A probasin-large T antigen transgenic mouse line develops prostate adenocarcinoma and neuroendocrine carcinoma with metastatic potential. Cancer Res. 2001;61:2239–2249.
    1. Ellwood-Yen K, et al. Myc-driven murine prostate cancer shares molecular features with human prostate tumors. Cancer Cell. 2003;4:223–238.
    1. Aytes A, et al. Cross-species analysis of genome-wide regulatory networks identifies a synergistic interaction between FOXM1 and CENPF that drives prostate cancer malignancy. Cancer Cell. 2014;25:638–651.
    1. Irshad S, et al. A molecular signature predictive of indolent prostate cancer. Sci Transl Med. 2013;5:202ra122.
    1. Floc’h N, et al. Dual targeting of the Akt/mTOR signaling pathway inhibits castration-resistant prostate cancer in a genetically engineered mouse model. Cancer Res. 2012;72:4483–4493.
    1. Carver BS, et al. Reciprocal feedback regulation of PI3K and androgen receptor signaling in PTEN-deficient prostate cancer. Cancer Cell. 2011;19:575–586.
    1. Korenchuk S, et al. VCaP, a cell-based model system of human prostate cancer. In Vivo. 2001;15:163–168.
    1. Sasai Y. Next-generation regenerative medicine: organogenesis from stem cells in 3D culture. Cell Stem Cell. 2013;12:520–530.
    1. McKeehan WL, Adams PS, Rosser MP. Direct mitogenic effects of insulin, epidermal growth factor, glucocorticoid, cholera toxin, unknown pituitary factors and possibly prolactin, but not androgen, on normal rat prostate epithelial cells in serum-free, primary cell culture. Cancer Res. 1984;44:1998–2010.
    1. Lang SH, et al. Differentiation of prostate epithelial cell cultures by matrigel/stromal cell glandular reconstruction. In vitro cellular & developmental biology. Animal. 2006;42:273–280.
    1. Lamb LE, Knudsen BS, Miranti CK. E-cadherin-mediated survival of androgen-receptor-expressing secretory prostate epithelial cells derived from a stratified in vitro differentiation model. J Cell Sci. 2010;123:266–276.
    1. Marker PC, Donjacour AA, Dahiya R, Cunha GR. Hormonal, cellular, and molecular control of prostatic development. Dev Biol. 2003;253:165–174.
    1. Cunha GR. Mesenchymal-epithelial interactions: past, present, and future. Differentiation. 2008;76:578–586.
    1. Stange DE, et al. Differentiated Troy+ chief cells act as reserve stem cells to generate all lineages of the stomach epithelium. Cell. 2013;155:357–368.
    1. Liu J, et al. Regenerated luminal epithelial cells are derived from preexisting luminal epithelial cells in adult mouse prostate. Mol Endocrinol. 2011;25:1849–1857.
    1. Koo BK, et al. Controlled gene expression in primary Lgr5 organoid cultures. Nature methods. 2012;9:81–83.
    1. Schwank G, et al. Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. Cell Stem Cell. 2013;13:653–658.
    1. Sachs N, Clevers H. Organoid cultures for the analysis of cancer phenotypes. Curr Opin Genet Dev. 2014;24C:68–73.
    1. Ranga A, Gjorevski N, Lutolf MP. Drug discovery through stem cell-based organoid models. Advanced drug delivery reviews. 2014;69–70C:19–28.
    1. Centenera MM, Raj GV, Knudsen KE, Tilley WD, Butler LM. Ex vivo culture of human prostate tissue and drug development. Nat Rev Urol. 2013;10:483–487.
    1. Toivanen R, et al. A preclinical xenograft model identifies castration-tolerant cancer-repopulating cells in localized prostate tumors. Sci Transl Med. 2013;5:187ra171.
    1. Lin D, et al. High fidelity patient-derived xenografts for accelerating prostate cancer discovery and drug development. Cancer Res. 2014;74:1272–1283.
    1. Van Keymeulen A, et al. Epidermal progenitors give rise to Merkel cells during embryonic development and adult homeostasis. J Cell Biol. 2009;187:91–100.
    1. Lesche R, et al. Cre/loxP-mediated inactivation of the murine Pten tumor suppressor gene. Genesis. 2002;32:148–149.
    1. Jackson EL, et al. Analysis of lung tumor initiation and progression using conditional expression of oncogenic K-ras. Genes Dev. 2001;15:3243–3248.
    1. Rock JR, et al. Basal cells as stem cells of the mouse trachea and human airway epithelium. Proc Natl Acad Sci USA. 2009;106:12771–12775.
    1. Giannico GA, Ross HM, Lotan T, Epstein JI. Aberrant expression of p63 in adenocarcinoma of the prostate: a radical prostatectomy study. Am J Surg Pathol. 2013;37:1401–1406.

Source: PubMed

3
구독하다