Lineage-specific restraint of pituitary gonadotroph cell adenoma growth

Vera Chesnokova, Svetlana Zonis, Cuiqi Zhou, Anat Ben-Shlomo, Kolja Wawrowsky, Yoel Toledano, Yunguang Tong, Kalman Kovacs, Bernd Scheithauer, Shlomo Melmed, Vera Chesnokova, Svetlana Zonis, Cuiqi Zhou, Anat Ben-Shlomo, Kolja Wawrowsky, Yoel Toledano, Yunguang Tong, Kalman Kovacs, Bernd Scheithauer, Shlomo Melmed

Abstract

Although pituitary adenomas are usually benign, unique trophic mechanisms restraining cell proliferation are unclear. As GH-secreting adenomas are associated with p53/p21-dependent senescence, we tested mechanisms constraining non-functioning pituitary adenoma growth. Thirty six gonadotroph-derived non-functioning pituitary adenomas all exhibited DNA damage, but undetectable p21 expression. However, these adenomas all expressed p16, and >90% abundantly expressed cytoplasmic clusterin associated with induction of the Cdk inhibitor p15 in 70% of gonadotroph and in 26% of somatotroph lineage adenomas (p = 0.006). Murine LβT2 and αT3 gonadotroph pituitary cells, and αGSU.PTTG transgenic mice with targeted gonadotroph cell adenomas also abundantly expressed clusterin and exhibited features of oncogene-induced senescence as evidenced by C/EBPβ and C/EBPδ induction. In turn, C/EBPs activated the clusterin promoter ∼5 fold, and elevated clusterin subsequently elicited p15 and p16 expression, acting to arrest murine gonadotroph cell proliferation. In contrast, specific clusterin suppression by RNAis enhanced gonadotroph proliferation. FOXL2, a tissue-specific gonadotroph lineage factor, also induced the clusterin promoter ∼3 fold in αT3 pituitary cells. As nine of 12 pituitary carcinomas were devoid of clusterin expression, this protein may limit proliferation of benign adenomatous pituitary cells. These results point to lineage-specific pathways restricting uncontrolled murine and human pituitary gonadotroph adenoma cell growth.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. PTTG and DNA damage in…
Figure 1. PTTG and DNA damage in human pituitary adenomas.
A) PTTG immunoreactivity (brown signal, both intranuclear and cytoplasmic)) in non-tumorous pituitary and human pituitary gonadotroph adenomas (T-1,2); B) DNA damage in human pituitary adenoma. Confocal image of human pituitary adenoma and non-tumorous pituitary tissue specimens labeled with γH2.AX or pATM antibodies (green). Both proteins are expressed in the nucleus. Specimens here and shown below were counterstained with DNA-specific dye ToPro3 (blue).
Figure 2. Cdk inhibitor expression in human…
Figure 2. Cdk inhibitor expression in human pituitary adenomas.
Confocal image of human gonadotroph pituitary adenoma specimens labeled with p21, p16 or p15 antibodies (green, intranucelar).
Figure 3. Clusterin in human pituitary adenomas.
Figure 3. Clusterin in human pituitary adenomas.
Confocal image of A) human pituitary adenoma and non-tumorous pituitary tissue specimens showing clusterin (green) expressed exclusively in the cytoplasm; B) Co-localization of clusterin with GH, PRL and αGSU in respective human pituitary adenoma specimens (clusterin green, respective hormones red).
Figure 4. Pituitary proliferation, DNA damage and…
Figure 4. Pituitary proliferation, DNA damage and senescence markers in the αGSU.PTTG pituitary gland.
A) In vivo BrdU incorporation. Mice were injected with BrdU (50 µg/g BW), and pituitary sections stained for BrdU. One thousand cells/section, 3 sections/animal, n = 3 animals/genotype were analyzed. *, p<0.05; Western blot analysis of B) proliferation markers; C) DNA damage, DNA repair and p53-dependent senescence markers, and D) oncogene-induced senescence markers; E) Confocal image showing immunofluorescent cytoplasmic clusterin, and intranuclear p15 and p16 expression (green) in WT and in pre-tumorous αGSU.PTTG pituitary glands, and in αGSU.PTTG pituitary adenomas; F) Pituitary SA-β-galactosidase enzymatic activity (blue) in WT and in pre-tumorous αGSU.PTTG pituitary gland. Three pituitary cryosections/animal were analyzed from 3 animals/genotype, and a representative image shown. Western blots here and elsewhere were repeated 3 times with similar results and representative blots shown.
Figure 5. Senescence markers in gonadotroph-derived LβT2…
Figure 5. Senescence markers in gonadotroph-derived LβT2 cells transfected with mPttg.
Western blot analysis of senescence markers in A) LβT2 cells transiently transfected with mPttg; B) in LβT2 cells stably transfected with mPttg; C) Percent BrdU positive cells in two selected clones stably transfected with mPttg. Duplicate samples were pulsed with BrdU for 30 min and analyzed by flow cytometry; D) Senescent morphology of LβT2 cells stably transfected with mPttg. Brown dots depict incorporated BrdU; E) Percent apoptotic cells stably transfected with mPttg. Cells were fixed, and one thousand cells/field counted in three randomly chosen visual fields; F) Percent SA-β-galactosidase positivity in cells stably transfected with Pttg was assessed in 6-well plates in triplicate. One thousand cells/field were counted in three fields/well. G) SA-β-galactosidase enzymatic activity (blue) in cells stably transfected with mPttg. *, p<0.05.
Figure 6. C/EBPs induce clusterin.
Figure 6. C/EBPs induce clusterin.
A) C/EBPβ is up-regulated in the αGSU.PTTG pituitary. Confocal image showing C/EBPβ co-localization with αGSU-positive, GH-positive and PRL-positive cells in WT and pre-tumorous αGSU.PTTG pituitary glands. (Hormones-green, cytoplsmic, C/EBPβ–red, intranuclear); B) Western blot analysis of C/EBPβ and δ isoforms induced in LβT2 cells stably transfected with mPttg; C) Effects of C/EBPs on the clusterin promoter in LβT2 and αT3 cells 24 h after transfection. Cells were co-transfected with 200 ng murine pGL3-luc-mClu reporter plasmid and 800 ng murine pCDNA3-C/EBPα, β or δ. The ratio of luciferase to co-trasfected β-galactosidase control reporter vector was normalized to pCDNA3-null expression vector. SEM was calculated from triplicate assays, and experiments repeated three times with similar results. Results of a representative experiment are shown.*, p<0.05, **,p<0.01; D) Western blot analysis of clusterin expression in gonadotroph-derived αT3 cells 24 hours after transfection with pCDNA3-C/EBPβ or E) pCDNA3-C/EBPδ; F) Western blot analysis of clusterin expression in LβT2 mPttg cells 48 hours after simultaneous transfection with siC/EBPβ and siC/EBPδ (3 nM each). Two different combinations of siRNAs were used.
Figure 7. Clusterin restrains pituitary cell proliferation…
Figure 7. Clusterin restrains pituitary cell proliferation by inducing Cdk inhibitors.
Western blot analysis of Cdk inhibitors and proliferation markers A) in LβT2 cells, B) in αT3 cells 48 h after transfection with mClu; C) Confocal images of immunofluoprescence of histone H3 methylation on lysine 9 (H3-K9M) (red) in vector and Clu-expressing αT3 cells 48 hours after transfection; D) Quantification of positive H3-K9M foci. Cells were fixed, stained with H3-K9M antibody, and one thousand cells/field counted in three randomly chosen visual fields; E) Percentage of BrdU positive cells 48 h after transfection with mClu. Triplicate samples were pulsed with BrdU for 30 min and analyzed by flow cytometry, *, p<0.05; F) αT3 cells stably overexpressing mClu or vector were synchronized in 0.1% fetal bovine serum for 18 hours, and then cultured in 10% fetal bovine serum. At the indicated times, duplicate samples were pulsed with BrdU for 30 min, analyzed by flow cytometry, and cells in S-phase identified by staining with BrdU antibodies.
Figure 8. Clusterin attenuation promotes proliferation.
Figure 8. Clusterin attenuation promotes proliferation.
Western blot analysis of Cdk inhibitors and proliferation markers A) in LβT2 cells, B) in αT3 cells; C) Percentage of BrdU positive cells 48 h after transfection with siClu. D) Upper panel, Western blot confirms p15 down-regulation, Lower panel, Percentage of BrdU positive LβT2 cells 48 h after transfection with sip15. E) Upper panel, Western blot confirms p16 down-regulation, Lower panel, Percentage of BrdU positive LβT2 cells 48 h after transfection with sip16. For BrdU detection, cells were fixed, stained with BrdU antibody and one thousand cells/field in three randomly chosen fields counted. *, p<0.05.
Figure 9. FOXL2 stimulates the clusterin promoter.
Figure 9. FOXL2 stimulates the clusterin promoter.
A) Effects of FOXL2 on the clusterin promoter in αT3 cells 24 h after transfection. Cells were co-transfected with 200 ng murine pGL3-luc-mClu reporter plasmid and indicated amounts of pcDNA3-His-mFoxl2. The ratio of luciferase to co-trasfected β-galactosidase control reporter vector was normalized to pCDNA3-null expression vector. SEM was calculated from triplicate assays, and experiments repeated three times with similar results. Results of a representative experiment are shown; **,p<0.01; B) Western blot analysis of clusterin expression in αT3 cells 24 hours after transfection with pcDNA3-His-mFoxl2; C) ChiP assay was performed in nuclear fractions derived from αT3 cell lysates. Top, schematic of the approximate location of primers used in the PCR reactions. Enrichment of specific clusterin promoter sequences was obtained with primer Set 2. FOXL2, specific antibody, IgG, nonspecific antibody, PCP, positive control primers. The experiment was repeated twice, and results of a representative assay shown.
Figure 10. Proliferation restricting pathways in the…
Figure 10. Proliferation restricting pathways in the pituitary gonadotroph cell lineage.
FOXL2 directly activates the clusterin promoter, while Pttg overexpression results in proliferation, DNA damage and stimulation of C/EBPβ and δ; C/EBPs activate the clusterin promoter. High levels of secretory clusterin trigger expression of Cdk inhibitors p15, p16 and p27, and C/EBPβ also cooperates to induce p15. Up-regulated tumor suppressor proteins likely underlie proliferation restraint preventing uncontrolled growth of benign pituitary adenomas of gonadotroph cell origin.

References

    1. Kovacs K, Horvath E, Vidal S. Classification of pituitary adenomas. J Neurooncol. 2001;54:121–127.
    1. Fernandez A, Karavitaki N, Wass JA. Prevalence of pituitary adenomas: a community-based, cross-sectional study in Banbury (Oxfordshire, UK). Clin Endocrinol (Oxf) 2010;72:377–382.
    1. Melmed S. Mechanisms for pituitary tumorigenesis: the plastic pituitary. J Clin Invest. 2003;112:1603–1618.
    1. Scheithauer BW, Gaffey TA, Lloyd RV, Sebo TJ, Kovacs KT, et al. Pathobiology of pituitary adenomas and carcinomas. Neurosurgery. 2006;59:341–353; discussion 341–353.
    1. Sharpless NE, DePinho RA. Telomeres, stem cells, senescence, and cancer. J Clin Invest. 2004;113:160–168.
    1. Serrano M, Blasco MA. Putting the stress on senescence. Curr Opin Cell Biol. 2001;13:748–753.
    1. Serrano M, Lin AW, McCurrach ME, Beach D, Lowe SW. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell. 1997;88:593–602.
    1. Michaloglou C, Vredeveld LC, Soengas MS, Denoyelle C, Kuilman T, et al. BRAFE600-associated senescence-like cell cycle arrest of human naevi. Nature. 2005;436:720–724.
    1. Collado M, Gil J, Efeyan A, Guerra C, Schuhmacher AJ, et al. Tumour biology: senescence in premalignant tumours. Nature. 2005;436:642.
    1. Braig M, Lee S, Loddenkemper C, Rudolph C, Peters AH, et al. Oncogene-induced senescence as an initial barrier in lymphoma development. Nature. 2005;436:660–665.
    1. Kuilman T, Michaloglou C, Mooi WJ, Peeper DS. The essence of senescence. Genes Dev. 2010;24:2463–2479.
    1. Jenne DE, Tschopp J. Molecular structure and functional characterization of a human complement cytolysis inhibitor found in blood and seminal plasma: identity to sulfated glycoprotein 2, a constituent of rat testis fluid. Proc Natl Acad Sci U S A. 1989;86:7123–7127.
    1. Kirszbaum L, Bozas SE, Walker ID. SP-40,40, a protein involved in the control of the complement pathway, possesses a unique array of disulphide bridges. FEBS Lett. 1992;297:70–76.
    1. Trougakos IP, Djeu JY, Gonos ES, Boothman DA. Advances and challenges in basic and translational research on clusterin. Cancer Res. 2009;69:403–406.
    1. Chen T, Turner J, McCarthy S, Scaltriti M, Bettuzzi S, et al. Clusterin-mediated apoptosis is regulated by adenomatous polyposis coli and is p21 dependent but p53 independent. Cancer Res. 2004;64:7412–7419.
    1. Rizzi F, Bettuzzi S. The clusterin paradigm in prostate and breast carcinogenesis. Endocr Relat Cancer. 2010;17:R1–17.
    1. Bettuzzi S, Davalli P, Davoli S, Chayka O, Rizzi F, et al. Genetic inactivation of ApoJ/clusterin: effects on prostate tumourigenesis and metastatic spread. Oncogene. 2009;28:4344–4352.
    1. Chayka O, Corvetta D, Dews M, Caccamo AE, Piotrowska I, et al. Clusterin, a haploinsufficient tumor suppressor gene in neuroblastomas. J Natl Cancer Inst. 2009;101:663–677.
    1. Kim HJ, Yoo EK, Kim JY, Choi YK, Lee HJ, et al. Protective role of clusterin/apolipoprotein J against neointimal hyperplasia via antiproliferative effect on vascular smooth muscle cells and cytoprotective effect on endothelial cells. Arterioscler Thromb Vasc Biol. 2009;29:1558–1564.
    1. Sivamurthy N, Stone DH, Logerfo FW, Quist WC. Apolipoprotein J inhibits the migration, adhesion, and proliferation of vascular smooth muscle cells. J Vasc Surg. 2001;34:716–723.
    1. Thomas-Tikhonenko A, Viard-Leveugle I, Dews M, Wehrli P, Sevignani C, et al. Myc-transformed epithelial cells down-regulate clusterin, which inhibits their growth in vitro and carcinogenesis in vivo. Cancer Res. 2004;64:3126–3136.
    1. Zhang X, Horwitz GA, Heaney AP, Nakashima M, Prezant TR, et al. Pituitary tumor transforming gene (PTTG) expression in pituitary adenomas. J Clin Endocrinol Metab. 1999;84:761–767.
    1. Salehi F, Kovacs K, Scheithauer BW, Lloyd RV, Cusimano M. Pituitary tumor-transforming gene in endocrine and other neoplasms: a review and update. Endocr Relat Cancer. 2008;15:721–743.
    1. Filippella M, Galland F, Kujas M, Young J, Faggiano A, et al. Pituitary tumour transforming gene (PTTG) expression correlates with the proliferative activity and recurrence status of pituitary adenomas: a clinical and immunohistochemical study. Clin Endocrinol (Oxf) 2006;65:536–543.
    1. Pei L, Melmed S. Isolation and characterization of a pituitary tumor-transforming gene (PTTG). Mol Endocrinol. 1997;11:433–441.
    1. Vlotides G, Eigler T, Melmed S. Pituitary tumor-transforming gene: physiology and implications for tumorigenesis. Endocr Rev. 2007;28:165–186.
    1. Heaney AP, Horwitz GA, Wang Z, Singson R, Melmed S. Early involvement of estrogen-induced pituitary tumor transforming gene and fibroblast growth factor expression in prolactinoma pathogenesis. Nat Med. 1999;5:1317–1321.
    1. Abbud RA, Takumi I, Barker EM, Ren SG, Chen DY, et al. Early multipotential pituitary focal hyperplasia in the alpha-subunit of glycoprotein hormone-driven pituitary tumor-transforming gene transgenic mice. Mol Endocrinol. 2005;19:1383–1391.
    1. Chesnokova V, Zonis S, Rubinek T, Yu R, Ben-Shlomo A, et al. Senescence mediates pituitary hypoplasia and restrains pituitary tumor growth. Cancer Res. 2007;67:10564–10572.
    1. Zou H, McGarry TJ, Bernal T, Kirschner MW. Identification of a vertebrate sister-chromatid separation inhibitor involved in transformation and tumorigenesis. Science. 1999;285:418–422.
    1. Wang Z, Yu R, Melmed S. Mice lacking pituitary tumor transforming gene show testicular and splenic hypoplasia, thymic hyperplasia, thrombocytopenia, aberrant cell cycle progression, and premature centromere division. Mol Endocrinol. 2001;15:1870–1879.
    1. Hsu YH, Liao LJ, Yu CH, Chiang CP, Jhan JR, et al. Overexpression of the pituitary tumor transforming gene induces p53-dependent senescence through activating DNA damage response pathway in normal human fibroblasts. J Biol Chem. 2010;285:22630–22638.
    1. Bernal JA, Roche M, Mendez-Vidal C, Espina A, Tortolero M, et al. Proliferative potential after DNA damage and non-homologous end joining are affected by loss of securin. Cell Death Differ. 2008;15:202–212.
    1. Chesnokova V, Zonis S, Kovacs K, Ben-Shlomo A, Wawrowsky K, et al. p21(Cip1) restrains pituitary tumor growth. Proc Natl Acad Sci U S A. 2008;105:17498–17503.
    1. Ellsworth BS, Egashira N, Haller JL, Butts DL, Cocquet J, et al. FOXL2 in the pituitary: molecular, genetic, and developmental analysis. Mol Endocrinol. 2006;20:2796–2805.
    1. Sedelnikova OA, Horikawa I, Zimonjic DB, Popescu NC, Bonner WM, et al. Senescing human cells and ageing mice accumulate DNA lesions with unrepairable double-strand breaks. Nat Cell Biol. 2004;6:168–170.
    1. Shen KC, Heng H, Wang Y, Lu S, Liu G, et al. ATM and p21 cooperate to suppress aneuploidy and subsequent tumor development. Cancer Res. 2005;65:8747–8753.
    1. Donangelo I, Gutman S, Horvath E, Kovacs K, Wawrowsky K, et al. Pituitary tumor transforming gene overexpression facilitates pituitary tumor development. Endocrinology. 2006;147:4781–4791.
    1. Cuadrado M, Gutierrez-Martinez P, Swat A, Nebreda AR, Fernandez-Capetillo O. p27Kip1 stabilization is essential for the maintenance of cell cycle arrest in response to DNA damage. Cancer Res. 2009;69:8726–8732.
    1. Collado M, Blasco MA, Serrano M. Cellular senescence in cancer and aging. Cell. 2007;130:223–233.
    1. Ramji DP, Foka P. CCAAT/enhancer-binding proteins: structure, function and regulation. Biochem J. 2002;365:561–575.
    1. Nerlov C. The C/EBP family of transcription factors: a paradigm for interaction between gene expression and proliferation control. Trends Cell Biol. 2007;17:318–324.
    1. Gery S, Tanosaki S, Hofmann WK, Koppel A, Koeffler HP. C/EBPdelta expression in a BCR-ABL-positive cell line induces growth arrest and myeloid differentiation. Oncogene. 2005;24:1589–1597.
    1. Narita M, Nunez S, Heard E, Lin AW, Hearn SA, et al. Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell. 2003;113:703–716.
    1. Egashira N, Takedoshi S, Takei M, Teramoto A, Osamura R. Expression of FOXL2 in human normal pituitaries and pituitary adenomas. Modern Pathology. 2010:1–9.
    1. Swanson SM, Kopchick JJ. Nuclear localization of growth hormone receptor: another age of discovery for cytokine action? Sci STKE. 2007;2007:pe69.
    1. Ciccone NA, Xu S, Lacza CT, Carroll RS, Kaiser UB. Frequency-dependent regulation of follicle-stimulating hormone beta by pulsatile gonadotropin-releasing hormone is mediated by functional antagonism of bZIP transcription factors. Mol Cell Biol. 2010;30:1028–1040.
    1. Kleinberg DL, Wood TL, Furth PA, Lee AV. Growth hormone and insulin-like growth factor-I in the transition from normal mammary development to preneoplastic mammary lesions. Endocr Rev. 2009;30:51–74.
    1. Goffin V, Binart N, Touraine P, Kelly PA. Prolactin: the new biology of an old hormone. Annu Rev Physiol. 2002;64:47–67.
    1. Janovick JA, Conn PM. Salt bridge integrates GPCR activation with protein trafficking. Proc Natl Acad Sci U S A. 2010;107:4454–4458.
    1. Kuilman T, Peeper DS. Senescence-messaging secretome: SMS-ing cellular stress. Nat Rev Cancer. 2009;9:81–94.
    1. Thangaraju M, Rudelius M, Bierie B, Raffeld M, Sharan S, et al. C/EBPdelta is a crucial regulator of pro-apoptotic gene expression during mammary gland involution. Development. 2005;132:4675–4685.
    1. Cao Z, Umek RM, McKnight SL. Regulated expression of three C/EBP isoforms during adipose conversion of 3T3-L1 cells. Genes Dev. 1991;5:1538–1552.
    1. Rizzi F, Caccamo AE, Belloni L, Bettuzzi S. Clusterin is a short half-life, poly-ubiquitinated protein, which controls the fate of prostate cancer cells. J Cell Physiol. 2009;219:314–323.
    1. Frost SJ, Simpson DJ, Farrell WE. Decreased proliferation and cell cycle arrest in neoplastic rat pituitary cells is associated with transforming growth factor-beta1-induced expression of p15/INK4B. Mol Cell Endocrinol. 2001;176:29–37.
    1. Chi KN, Zoubeidi A, Gleave ME. Custirsen (OGX-011): a second-generation antisense inhibitor of clusterin for the treatment of cancer. Expert Opin Investig Drugs. 2008;17:1955–1962.
    1. Flanagan L, Whyte L, Chatterjee N, Tenniswood M. Effects of clusterin over-expression on metastatic progression and therapy in breast cancer. BMC Cancer. 2010;10:107.
    1. Chou TY, Chen WC, Lee AC, Hung SM, Shih NY, et al. Clusterin silencing in human lung adenocarcinoma cells induces a mesenchymal-to-epithelial transition through modulating the ERK/Slug pathway. Cell Signal. 2009;21:704–711.
    1. Criswell T, Beman M, Araki S, Leskov K, Cataldo E, et al. Delayed activation of insulin-like growth factor-1 receptor/Src/MAPK/Egr-1 signaling regulates clusterin expression, a pro-survival factor. J Biol Chem. 2005;280:14212–14221.
    1. Bettuzzi S, Scorcioni F, Astancolle S, Davalli P, Scaltriti M, et al. Clusterin (SGP-2) transient overexpression decreases proliferation rate of SV40-immortalized human prostate epithelial cells by slowing down cell cycle progression. Oncogene. 2002;21:4328–4334.
    1. Panico F, Rizzi F, Fabbri LM, Bettuzzi S, Luppi F. Clusterin (CLU) and lung cancer. Adv Cancer Res. 2009;105:63–76.
    1. Wakefield LM, Roberts AB. TGF-beta signaling: positive and negative effects on tumorigenesis. Curr Opin Genet Dev. 2002;12:22–29.
    1. Nam JS, Terabe M, Kang MJ, Chae H, Voong N, et al. Transforming growth factor beta subverts the immune system into directly promoting tumor growth through interleukin-17. Cancer Res. 2008;68:3915–3923.
    1. Alarid ET, Windle JJ, Whyte DB, Mellon PL. Immortalization of pituitary cells at discrete stages of development by directed oncogenesis in transgenic mice. Development. 1996;122:3319–3329.

Source: PubMed

3
Iratkozz fel