Ovarian surface epithelium at the junction area contains a cancer-prone stem cell niche

Andrea Flesken-Nikitin, Chang-Il Hwang, Chieh-Yang Cheng, Tatyana V Michurina, Grigori Enikolopov, Alexander Yu Nikitin, Andrea Flesken-Nikitin, Chang-Il Hwang, Chieh-Yang Cheng, Tatyana V Michurina, Grigori Enikolopov, Alexander Yu Nikitin

Abstract

Epithelial ovarian cancer (EOC) is the fifth leading cause of cancer deaths among women in the United States, but its pathogenesis is poorly understood. Some epithelial cancers are known to occur in transitional zones between two types of epithelium, whereas others have been shown to originate in epithelial tissue stem cells. The stem cell niche of the ovarian surface epithelium (OSE), which is ruptured and regenerates during ovulation, has not yet been defined unequivocally. Here we identify the hilum region of the mouse ovary, the transitional (or junction) area between the OSE, mesothelium and tubal (oviductal) epithelium, as a previously unrecognized stem cell niche of the OSE. We find that cells of the hilum OSE are cycling slowly and express stem and/or progenitor cell markers ALDH1, LGR5, LEF1, CD133 and CK6B. These cells display long-term stem cell properties ex vivo and in vivo, as shown by our serial sphere generation and long-term lineage-tracing assays. Importantly, the hilum cells show increased transformation potential after inactivation of tumour suppressor genes Trp53 and Rb1, whose pathways are altered frequently in the most aggressive and common type of human EOC, high-grade serous adenocarcinoma. Our study supports experimentally the idea that susceptibility of transitional zones to malignant transformation may be explained by the presence of stem cell niches in those areas. Identification of a stem cell niche for the OSE may have important implications for understanding EOC pathogenesis.

Figures

Figure 1. Identification and location of putative…
Figure 1. Identification and location of putative OSE stem cells
a, OSE sphere, Phase contrast. Bar, 20 μm. b, Confocal imaging of compact (left) and expanded (right) OSE spheres from Tgβ-actin EGFP mice at 4 and 10 days, respectively. Green fluorescence, counterstaining with DAPI, blue. Bar, both images, 50 μm. c, Frequency of sphere formation by ALDH- and ALDH+ OSE cells for 5 consecutive generations (G, dissociation/clonal formation; n = 6, mean ± s.d.). ALDH- derived cells very rarely formed spheres in G2 and did not yield any spheres in G3. d, ALDH1 (brown color) is preferentially expressed in the OSE (arrows) of the hilum region as compared to that of the antral region, corpus luteum or distal region. ALDH1 staining is also present in the theca cells (TC) of the ovary. Rectangles in top left image indicate respective location (clockwise) of the regions in the mouse ovary. Arrowhead, the junction between OSE and tubal epithelium. B, bursa; CL, corpus luteum; F, follicle; H, hilum; OV, ovary; TC, theca cells; UT, uterine tube; U, uterus. 6 weeks old mouse. ABC Elite method, hematoxylin counterstaining. Bar, top left image, 500 μm; all other images, 50 μm. e, Quantification of BrdU label retaining cells (LRCs) in the antral, corpus luteum (CL), distal and hilum regions (n = 4, mean ± s.d.). At 3 months after BrdU pulse two tailed P: hilum (*) versus antral region, 0.0005; versus CL or distal region, < 0.0001. f, Detection of BrdU LRCs (green), ALDH1 (red) and overlay (orange) in the hilum OSE (arrows) after 3 months of chase. Arrowhead, BrdU LRC in the neighboring stroma. Counterstaining with DAPI, blue. Bar, 50 μm.
Figure 2. Functional characterization of the hilum…
Figure 2. Functional characterization of the hilum OSE cells
a, Colony formation by OSE cells isolated from the anterior part (Ovary) and the hilum. 4.0 × 103 cells per plate. Giemsa staining. Bar, 500 μm. b, Quantitative analysis of frequency of large colonies (≥ 20 cells) formed by OSE cells from the anterior part (n = 8, mean ± s.d., 11.4 ± 5.68) and the hilum (n = 6, 42.7 ± 12.8). Two tailed P < 0.0001. c, Frequency of the anterior part and hilum OSE sphere forming cells (SFC) for 1 (Ovary) 1 and 7 (hilum) consecutive generations (G, n = 3, mean ± s.d.). Anterior part derived cells very rarely formed spheres in G2 and did not yield any spheres in G3. d, Gene expression profiles of 3 independent pools (10 mice each) of ALDH- and ALDH+ cells. e, Expression of stem cell markers in ALDH- and ALDH+ cells. Quantitative PCR (n =3, mean ± s.d.; all P values <0.01, except for Lgr6). f, Detection of hilum cells (arrows) expressing CD133, CK6b, Lgr5 and Lef1. Immunofluorescence (CD133, CK6b, and Lef1) or EGFP expression under the control of Lgr5 promoter in Lgr5-EGFP-IRES-creERT2 mouse. All abbreviations as in Fig. 1d. Counterstaining with DAPI, blue. Bar for all images, 50 μm. g, Expression of microRNAs in ALDH- and ALDH+ cells. Quantitative PCR (n = 3, mean ± s.d.; all P values <0.01).
Figure 3. Tracing the fate of the…
Figure 3. Tracing the fate of the Lgr5+ hilum cells
a-f, Detection of tdTomato (red, a, b, d, f) and EGFP (green, c, e) expression in the ovaries of Lgr5-EGFP-IRES-creERT2 Ai9 mice 1 day (a-e) and 1 month (f) after administration of either tamoxifen (a-c, f) or vehicle (d, e). All abbreviations as in Fig. 1d. Counterstaining with DAPI, blue. Arrows, hilum OSE. Bar, a, f, 600 μm; b-e, 130 μm.
Figure 4. Hilum cells show preferential transformation…
Figure 4. Hilum cells show preferential transformation after conditional inactivation of Trp53 and Rb1
a-c, Characterization of primary OSE cells isolated from the hilum and the remainder of the ovary (Ovary) and evaluated at passage 6 after Cre-loxP mediated inactivation of Trp53 and Rb1. Quantification of BrdU positive OSE cells (a, n = 3, mean ± s.d., P< 0.0001). Detection (b) and quantitative analysis (c) of senescence associated-β-galactosidase (blue) in the ovary and hilum cells (c, n = 3, mean ± s.d., two tailed P = 0.002). Phase contrast. Bar, 500 μm. d, Survival of mice intraperitoneally transplanted with primary ovary (n = 12) and hilum (n = 8) OSE cells deficient for Trp53 and Rb1 (log-rank P = 0.0007). e, Neoplastic masses (arrows) arisen from hilum cells transplanted into the mouse abdominal cavity. f-n, Hilum derived-neoplastic cells have marked nuclear atypia (f), express cytokeratin 8 (CK8, g), are highly proliferative (h), grow in solid, nested and gland-like patterns (f-h, j, k, arrows), invade liver (i, arrow), metastasize to the lung (l-n, arrows) and express Wilms Tumour 1 (j, m) and PAX8 (k, n). Hematoxylin and eosin (HE) and immunostainings, ABC Elite method. Counterstaining with hematoxylin (CK8) and methyl green (Ki67, Wilms Tumour 1, PAX8). Bar, f-h, j, k, 100 μm; i, l-n, 200 μm.

References

    1. Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA Cancer J Clin. 2012;62:10–29.
    1. Lengyel E. Ovarian cancer development and metastasis. Am J Pathol. 2010;177:1053–1064.
    1. Bowtell DD. The genesis and evolution of high-grade serous ovarian cancer. Nat Rev Cancer. 2010;10:803–808.
    1. Visvader JE. Cells of origin in cancer. Nature. 2011;469:314–322.
    1. Auersperg N. The origin of ovarian carcinomas: a unifying hypothesis. Int J Gynecol Pathol. 2011;30:12–21.
    1. Medema JP, Vermeulen L. Microenvironmental regulation of stem cells in intestinal homeostasis and cancer. Nature. 2011;474:318–326.
    1. The Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474:609–615.
    1. Ahmed AA, et al. Driver mutations in TP53 are ubiquitous in high grade serous carcinoma of the ovary. J Pathol. 2010;221:49–56.
    1. Kurman RJ, Shih Ie M. The origin and pathogenesis of epithelial ovarian cancer: a proposed unifying theory. Am J Surg Pathol. 2010;34:433–443.
    1. Dubeau L. The cell of origin of ovarian epithelial tumours. Lancet Oncol. 2008;9:1191–1197.
    1. Auersperg N, Woo MM, Gilks CB. The origin of ovarian carcinomas: a developmental view. Gynecol Oncol. 2008;110:452–454.
    1. Seidman JD, Yemelyanova A, Zaino RJ, Kurman RJ. The fallopian tube-peritoneal junction: a potential site of carcinogenesis. Int J Gynecol Pathol. 2011;30:4–11.
    1. Szotek PP, et al. Normal ovarian surface epithelial label-retaining cells exhibit stem/progenitor cell characteristics. Proc Natl Acad Sci U S A. 2008;105:12469–12473.
    1. Blanpain C, Horsley V, Fuchs E. Epithelial stem cells: turning over new leaves. Cell. 2007;128:445–458.
    1. Simons BD, Clevers H. Strategies for homeostatic stem cell self-renewal in adult tissues. Cell. 2011;145:851–862.
    1. Ginestier C, et al. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell. 2007;1:555–567.
    1. Storms RW, et al. Isolation of primitive human hematopoietic progenitors on the basis of aldehyde dehydrogenase activity. Proc Natl Acad Sci U S A. 1999;96:9118–9123.
    1. Corti S, et al. Identification of a primitive brain-derived neural stem cell population based on aldehyde dehydrogenase activity. Stem Cells. 2006;24:975–985.
    1. Burger PE, et al. High aldehyde dehydrogenase activity: a novel functional marker of murine prostate stem/progenitor cells. Stem Cells. 2009;27:2220–2228.
    1. Deng S, et al. Distinct expression levels and patterns of stem cell marker, aldehyde dehydrogenase isoform 1 (ALDH1), in human epithelial cancers. PLoS One. 2010;5:e10277.
    1. Bullough WS. Oogenesis and its relation to the oestrous cycle in the adult mouse. Journal of Endocrinology. 1942;3:141–149.
    1. Ji Q, et al. MicroRNA miR-34 inhibits human pancreatic cancer tumor-initiating cells. PLoS One. 2009;4:e6816.
    1. Liu C, et al. The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nat Med. 2011;17:211–215.
    1. Choi YJ, et al. miR-34 miRNAs provide a barrier for somatic cell reprogramming. Nat Cell Biol. 2011;13:1353–1360.
    1. Barker N, et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature. 2007;449:1003–1007.
    1. Madisen L, et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat Neurosci. 2010;13:133–140.
    1. Flesken-Nikitin A, Choi KC, Eng JP, Shmidt EN, Nikitin AY. Induction of carcinogenesis by concurrent inactivation of p53 and Rb1 in the mouse ovarian surface epithelium. Cancer Res. 2003;63:3459–3463.
    1. Szabova L, et al. Perturbation of Rb, p53, and Brca1 or Brca2 Cooperate in Inducing Metastatic Serous Epithelial Ovarian Cancer. Cancer Res. 2012;72:4141–4153.
    1. Pellegrini G, et al. Location and clonal analysis of stem cells and their differentiated progeny in the human ocular surface. J Cell Biol. 1999;145:769–782.
    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. Corney DC, Flesken-Nikitin A, Godwin AK, Wang W, Nikitin AY. MicroRNA-34b and MicroRNA-34c are targets of p53 and cooperate in control of cell proliferation and adhesion-independent growth. Cancer Res. 2007;67:8433–8438.
    1. Hwang CI, et al. Wild-type p53 controls cell motility and invasion by dual regulation of MET expression. Proc Natl Acad Sci U S A. 2011;108:14240–14245.
    1. Russo JE, Hauguitz D, Hilton J. Inhibition of mouse cytosolic aldehyde dehydrogenase by 4-(diethylamino)benzaldehyde. Biochem Pharmacol. 1988;37:1639–1642.
    1. Lawson DA, Xin L, Lukacs RU, Cheng D, Witte ON. Isolation and functional characterization of murine prostate stem cells. Proc Natl Acad Sci U S A. 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. Singec I, et al. Defining the actual sensitivity and specificity of the neurosphere assay in stem cell biology. Nat Methods. 2006;3:801–806.
    1. Zhou Z, et al. Synergy of p53 and Rb deficiency in a conditional mouse model for metastatic prostate cancer. Cancer Res. 2006;66:7889–7898.
    1. Choi J, Curtis SJ, Roy DM, Flesken-Nikitin A, Nikitin AY. Local mesenchymal stem/progenitor cells are a preferential target for initiation of adult soft tissue sarcomas associated with p53 and Rb deficiency. Am J Pathol. 2010;177:2645–2658.
    1. Nikitin A, Lee WH. Early loss of the retinoblastoma gene is associated with impaired growth inhibitory innervation during melanotroph carcinogenesis in Rb+/- mice. Genes Dev. 1996;10:1870–1879.
    1. Debacq-Chainiaux F, Erusalimsky JD, Campisi J, Toussaint O. Protocols to detect senescence-associated beta-galactosidase (SA-betagal) activity, a biomarker of senescent cells in culture and in vivo. Nat Protoc. 2009;4:1798–1806.
    1. Matoso A, Zhou Z, Hayama R, Flesken-Nikitin A, Nikitin AY. Cell lineage-specific interactions between Men1 and Rb in neuroendocrine neoplasia. Carcinogenesis. 2008;29:620–628.
    1. Nikitin AY, Lee WH. Early loss of the retinoblastoma gene is associated with impaired growth inhibitory innervation during melanotroph carcinogenesis in Rb+/- mice. Genes & Development. 1996;10:1870–1879.
    1. Zhou Z, Flesken-Nikitin A, Nikitin AY. Prostate cancer associated with p53 and Rb deficiency arises from the stem/progenitor cell-enriched proximal region of prostatic ducts. Cancer Res. 2007;67:5683–5690.
    1. Zhou Z, et al. Suppression of melanotroph carcinogenesis leads to accelerated progression of pituitary anterior lobe tumors and medullary thyroid carcinomas in Rb+/- mice. Cancer Res. 2005;65:787–796.
    1. Rugh R. The Mouse Its Reproduction and Development. Oxford University Press; Oxford: 1990.
    1. Vermot J, Fraulob V, Dolle P, Niederreither K. Expression of enzymes synthesizing (aldehyde dehydrogenase 1 and reinaldehyde dehydrogenase 2) and metabolizaing (Cyp26) retinoic acid in the mouse female reproductive system. Endocrinology. 2000;141:3638–3645.

Source: PubMed

3
Předplatit