Thyroid and androgen receptor signaling are antagonized by μ-Crystallin in prostate cancer

Osman Aksoy, Jan Pencik, Markus Hartenbach, Ali A Moazzami, Michaela Schlederer, Theresa Balber, Adam Varady, Cecile Philippe, Pascal A Baltzer, Bismoy Mazumder, Jonathan B Whitchurch, Christopher J Roberts, Andrea Haitel, Merima Herac, Martin Susani, Markus Mitterhauser, Rodrig Marculescu, Judith Stangl-Kremser, Melanie R Hassler, Gero Kramer, Shahrokh F Shariat, Suzanne D Turner, Boris Tichy, Jan Oppelt, Sarka Pospisilova, Sabrina Hartenbach, Simone Tangermann, Gerda Egger, Heidi A Neubauer, Richard Moriggl, Zoran Culig, Georg Greiner, Gregor Hoermann, Marcus Hacker, David M Heery, Olaf Merkel, Lukas Kenner, Osman Aksoy, Jan Pencik, Markus Hartenbach, Ali A Moazzami, Michaela Schlederer, Theresa Balber, Adam Varady, Cecile Philippe, Pascal A Baltzer, Bismoy Mazumder, Jonathan B Whitchurch, Christopher J Roberts, Andrea Haitel, Merima Herac, Martin Susani, Markus Mitterhauser, Rodrig Marculescu, Judith Stangl-Kremser, Melanie R Hassler, Gero Kramer, Shahrokh F Shariat, Suzanne D Turner, Boris Tichy, Jan Oppelt, Sarka Pospisilova, Sabrina Hartenbach, Simone Tangermann, Gerda Egger, Heidi A Neubauer, Richard Moriggl, Zoran Culig, Georg Greiner, Gregor Hoermann, Marcus Hacker, David M Heery, Olaf Merkel, Lukas Kenner

Abstract

Androgen deprivation therapy (ADT) remains a key approach in the treatment of prostate cancer (PCa). However, PCa inevitably relapses and becomes ADT resistant. Besides androgens, there is evidence that thyroid hormone thyroxine (T4) and its active form 3,5,3'-triiodo-L-thyronine (T3) are involved in the progression of PCa. Epidemiologic evidences show a higher incidence of PCa in men with elevated thyroid hormone levels. The thyroid hormone binding protein μ-Crystallin (CRYM) mediates intracellular thyroid hormone action by sequestering T3 and blocks its binding to cognate receptors (TRα/TRβ) in target tissues. We show in our study that low CRYM expression levels in PCa patients are associated with early biochemical recurrence and poor prognosis. Moreover, we found a disease stage-specific expression of CRYM in PCa. CRYM counteracted thyroid and androgen signaling and blocked intracellular choline uptake. CRYM inversely correlated with [18F]fluoromethylcholine (FMC) levels in positron emission tomography/magnetic resonance imaging of PCa patients. Our data suggest CRYM as a novel antagonist of T3- and androgen-mediated signaling in PCa. The role of CRYM could therefore be an essential control mechanism for the prevention of aggressive PCa growth.

Trial registration: ClinicalTrials.gov NCT02659527.

Keywords: PSMA-PET; androgen receptor; prostate cancer; thyroid hormone receptor; μ-Crystallin.

Conflict of interest statement

The authors declare no potential conflict of interest.

© 2020 The Authors. International Journal of Cancer published by John Wiley & Sons Ltd on behalf of Union for International Cancer Control.

Figures

FIGURE 1
FIGURE 1
CRYM is stage‐dependently expressed in prostate cancer at the mRNA and protein level. A, CRYM protein levels assessed by IHC of healthy prostate tissue (n = 178), PCa (n = 178) and tissues derived from PCa metastases (n = 17). Representative staining for CRYM in healthy prostate, PCa and metastatic tissue is shown, scale bar 50 μm (low magnification), 25 μm (inlet, high magnification). B, IHC analysis of CRYM protein levels in non‐neoplastic prostate tissue (n = 30) and PCa (n = 122) in an independent cohort from Tuebingen. C, IHC CRYM protein levels in correlation to time for BCR in a Kaplan‐Meier analysis (P = .029). D, CRYM mRNA expression levels in PCa patients, analyzed from primary tumor samples and metastases. Data were extracted from the Oncomine Platform from the following studies: Chandran Prostate (left, P < .0001), Grasso Prostate (middle, P < .0001) and Yu Prostate (right, P < .0001). E, Heatmaps of human patient data depicting CRYM and AR mRNA levels in primary tumors and PCa metastases (log2 median‐centered intensity). Data were extracted from the Oncomine Platform from the following studies: Chandran Prostate (upper), Grasso Prostate (middle) and Yu Prostate (lower). BCR, biochemical recurrence; CRYM, μ‐Crystallin; IHC, immunohistochemistry; PCa, prostate cancer [Color figure can be viewed at wileyonlinelibrary.com]
FIGURE 2
FIGURE 2
CRYM overexpression leads to a reduction of free T3. A, Immunoblot analysis of CRYM and TRβ in PCa cell lines RWPE‐1, LNCaP, PC3, DU145, LAPC4, VCAP. β‐Actin was used as loading control. B, CRYM and TRβ levels in PC3 and DU145 cells that were transiently transfected with EV or CRYM(+). C, Human PCa cells PC3, DU145, 22Rv1 and LAPC4 were transfected with a plasmid bearing the CRYM insert under the CMV promoter that coexpresses GFP (CRYM(+)) or empty vector (EV). T3 concentrations were determined across human PCa cells by analyzing with electrochemiluminescence immune assay (n = 3). FM (Full medium): RPMI medium supplemented with 10% FCS. D, Transiently transfected PC3 and LNCaP cells with EV or CRYM(+) were incubated with radioactively labeled T3 ([125I] T3) in hormone free medium for 48 hours, and intracellular radioactivity was determined by scintillation counting. CRYM, μ‐Crystallin; FCS, fetal calf serum; PCa, prostate cancer [Color figure can be viewed at wileyonlinelibrary.com]
FIGURE 3
FIGURE 3
CRYM overexpression represses AR signaling and invasion in PCa cells. A, RNA‐seq analysis of LNCaP cells that were transiently transfected with EV or CRYM(+). AR responsive genes KLK3 (PSA) (P = .0018), KLK2 (P = .0013), TMPRSS2 (P = .0004) and NKX3.1 (P = .0001) are shown. B, Matrigel‐coated invasion chambers were used to test the invasive capacity of PCa cell lines PC3 and DU145 with and without CRYM(+) over 24 hours. Quantification and representative images are shown; scale bar = 50 μm. C, Heatmaps depicting THRB (TRβ) and CRYM mRNA levels in primary PCa and metastatic patient samples (log2 median‐centered intensity). Data were extracted from the Oncomine Platform from the Chandran Prostate (upper) and Varambally Prostate (lower) studies. AR, androgen receptor; CRYM, μ‐Crystallin; EV, empty vector; PCa, prostate cancer [Color figure can be viewed at wileyonlinelibrary.com]
FIGURE 4
FIGURE 4
CRYM‐induced suppression of T3 and AR signaling. A, LNCaP cells expressing empty vector (EV) or CRYM(+) were analyzed by RNA‐seq and Immunoblot. B, LNCaP CRYM(+) and EV cells were compared using three biological replicates for each group. Single end 50 bp RNA‐seq was performed with an Illumina Hi‐Seq2000 platform and LNCaP CRYM(+) to EV cells were compared using three biological replicates for each group. CRYM overexpression was validated by RNA‐seq (73.7 vs 2.2 normalized counts, P < .001). Shown are enriched pathways according to IPA pathway analysis among the deregulated genes. C, Genes deregulated by androgen DHT in the same cell line LNCaP from a published dataset. Shown are enriched pathways according to IPA pathway analysis. 24 D, 843 genes overlapped among CRYM overexpression and DHT regulated genes; 70.1% of these were counter‐regulated (anti). Androgen‐responsive genes and AR‐regulated genes are enriched in LNCaP EV vs CRYM(+) and control vs DHT. E, CRYM knockdown in the LNCaP cell line generated using lentiviral‐transfected shRNAs (shCRYM) and a nontargeting control shRNA (shControl). CRYM reduction was confirmed by Immunoblot. PSA release levels were measured by a chemiluminescence immune assay. AR, androgen receptor; CRYM, μ‐Crystallin; DHT, dihydrotestosterone; EV, empty vector; IPA, ingenuity pathways analysis; PSA, prostate‐specific antigen [Color figure can be viewed at wileyonlinelibrary.com]
FIGURE 5
FIGURE 5
CRYM overexpression caused to the metabolic alterations in PCa. A, Score plot of partial least squares‐discriminant analysis (PLS‐DA) model of PC3 cell pellet extract fitted using 1H‐NMR spectral data. PC3 cells without CRYM overexpressing vector (right) were separated from PC3 cells with CRYM overexpressing vector (left) along the first component. B, Measurement of free choline in CRYM overexpression (P < .01) or EV in the presence of T3. C, Relative metabolite levels of glycine, glutamate, creatinine and taurine with and without T3 in LNCaP with overexpression of CRYM or EV measured by 1H‐NMR. D, Immunoblot analysis of AR and choline kinase α (CHKA) and β‐Actin as a control in RPMI 1640 medium with 10% FCS (FM), HFM and HFM with T3 supplementation (1 nM and 10 nM). CRYM, μ‐Crystallin; EV, empty vector; FCS, fetal calf serum; FM, full medium; PCa, prostate cancer [Color figure can be viewed at wileyonlinelibrary.com]
FIGURE 6
FIGURE 6
Noninvasive imaging using [18F]fluoromethylcholine (FMC) and PET is a surrogate marker for the activity of thyroid hormone metabolism in PCa. A, Hematoxylin Eosin (HE) whole mount sections of two different prostate cancer specimens on the left with a Gleason 3 lesion and on the right with a larger Gleason 4 lesion. Corresponding FMC PET/MRI of the same patient shows FMC uptake on the left (Gleason 3) and on the right (Gleason 4). IHC showing CRYM stainings positive in the Gleason 3 lesion on the left and TRβ stainings positive in the Gleason 4 lesion on the right (scale bar = 150 μm). B, Statistical evaluation of 42 PCa patients who underwent FMC PET/MRI prior to radical prostatectomy. CRYM and TRβ protein levels in tumors were analyzed using IHC. Choline uptake in tumor specimens with high TRβ expression (TRβ 2‐6 score; n = 30, P < .01) or no CRYM expression (n = 21, P = .041). C, Choline uptake correlated to BCR or metastases in receiver‐operating‐characteristics‐curve analysis (AUC = 0.77, P < .0001). D, 87 Patients with FMC PET/MRI and a mean follow‐up time of 508 days were divided into a group that developed BCR and/or metastasis (n = 37) and a group that did not (n = 50). The first group had significantly higher choline levels in FMC PET/MRI (P < .0001). AUC, area under the curve; BCR, biochemical recurrence; FMC, [18F]fluoromethylcholine; HE, hematoxylin eosin; IHC, immunohistochemistry; MRI, magnetic resonance imaging; PCa, prostate cancer; PET, positron emission tomography [Color figure can be viewed at wileyonlinelibrary.com]

References

    1. Huggins C, Hodges CV. Studies on prostate cancer. I. The effect of castration, of estrogen and of androgen injection on serum phosphatase in metastatic carcinoma of the prostate. Cancer Res. 1941;1:239‐297.
    1. Porkka KP, Visakorpi T. Molecular mechanisms of prostate cancer. Eur Urol. 2004;45(6):683‐691.
    1. Haile S, Sadar MD. Androgen receptor and its splice variants in prostate cancer. Cell Mol Life Sci. 2011;68(24):3971‐3981.
    1. Watson PA, Chen YF, Balbas MD, et al. Constitutively active androgen receptor splice variants expressed in castration‐resistant prostate cancer require full‐length androgen receptor. Proc Natl Acad Sci U S A. 2010;107(39):16759‐16765.
    1. Visser WE, Wong WS, van Mullem AA, Friesema EC, Geyer J, Visser TJ. Study of the transport of thyroid hormone by transporters of the SLC10 family. Mol Cell Endocrinol. 2010;315(1‐2):138‐145.
    1. Bernal J, Guadano‐Ferraz A, Morte B. Thyroid hormone transporters‐functions and clinical implications. Nat Rev Endocrinol. 2015;11(12):690.
    1. Vie MP, Evrard C, Osty J, et al. Purification, molecular cloning, and functional expression of the human nicodinamide‐adenine dinucleotide phosphate‐regulated thyroid hormone‐binding protein. Mol Endocrinol. 1997;11(11):1728‐1736.
    1. Kim RY, Gasser R, Wistow GJ. mu‐crystallin is a mammalian homologue of Agrobacterium ornithine cyclodeaminase and is expressed in human retina. Proc Natl Acad Sci U S A. 1992;89(19):9292‐9296.
    1. Shukla‐Dave A, Castillo‐Martin M, Chen M, et al. Ornithine decarboxylase is sufficient for prostate tumorigenesis via androgen receptor signaling. Am J Pathol. 2016;186(12):3131‐3145.
    1. Moeller LC, Fuhrer D. Thyroid hormone, thyroid hormone receptors, and cancer: a clinical perspective. Endocr Relat Cancer. 2013;20(2):R19‐R29.
    1. Suzuki S, Mori J, Hashizume K. mu‐crystallin, a NADPH‐dependent T(3)‐binding protein in cytosol. Trends Endocrinol Metab. 2007;18(7):286‐289.
    1. Malinowska K, Cavarretta IT, Susani M, et al. Identification of mu‐crystallin as an androgen‐regulated gene in human prostate cancer. Prostate. 2009;69(10):1109‐1118.
    1. Mousses S, Bubendorf L, Wagner U, et al. Clinical validation of candidate genes associated with prostate cancer progression in the CWR22 model system using tissue microarrays. Cancer Res. 2002;62(5):1256‐1260.
    1. Schlederer M, Mueller KM, Haybaeck J, et al. Reliable quantification of protein expression and cellular localization in histological sections. PLoS One. 2014;9(7):e100822.
    1. Pencik J, Schlederer M, Gruber W, et al. STAT3 regulated ARF expression suppresses prostate cancer metastasis. Nat Commun. 2015;6:7736.
    1. Moazzami AA, Andersson R, Kamal‐Eldin A. Changes in the metabolic profile of rat liver after alpha‐tocopherol deficiency as revealed by metabolomics analysis. NMR Biomed. 2011;24(5):499‐505.
    1. Moazzami AA, Shrestha A, Morrison DA, Poutanen K, Mykkanen H. Metabolomics reveals differences in postprandial responses to breads and fasting metabolic characteristics associated with postprandial insulin demand in postmenopausal women. J Nutr. 2014;144(6):807‐814.
    1. Proestling K, Hebar A, Pruckner N, Marton E, Vinatzer U, Schreiber M. The Pro allele of the p53 codon 72 polymorphism is associated with decreased intratumoral expression of BAX and p21, and increased breast cancer risk. PLoS One. 2012;7(10):e47325.
    1. Rhodes DR, Yu J, Shanker K, et al. ONCOMINE: a cancer microarray database and integrated data‐mining platform. Neoplasia. 2004;6(1):1‐6.
    1. Sadow PM, Chassande O, Koo EK, et al. Regulation of expression of thyroid hormone receptor isoforms and coactivators in liver and heart by thyroid hormone. Mol Cell Endocrinol. 2003;203(1‐2):65‐75.
    1. Baron A, Migita T, Tang D, Loda M. Fatty acid synthase: a metabolic oncogene in prostate cancer? J Cell Biochem. 2004;91(1):47‐53.
    1. Zadra G, Ribeiro CF, Chetta P, et al. Inhibition of de novo lipogenesis targets androgen receptor signaling in castration‐resistant prostate cancer. Proc Natl Acad Sci U S A. 2019;116(2):631‐640.
    1. Malinen M, Niskanen EA, Kaikkonen MU, Palvimo JJ. Crosstalk between androgen and pro‐inflammatory signaling remodels androgen receptor and NF‐kappaB cistrome to reprogram the prostate cancer cell transcriptome. Nucleic Acids Res. 2017;45(2):619‐630.
    1. Aoyama C, Liao H, Ishidate K. Structure and function of choline kinase isoforms in mammalian cells. Prog Lipid Res. 2004;43(3):266‐281.
    1. Morigi JJ, Stricker PD, van Leeuwen PJ, et al. Prospective comparison of 18F‐fluoromethylcholine versus 68Ga‐PSMA PET/CT in prostate cancer patients who have rising PSA after curative treatment and are being considered for targeted therapy. J Nucl Med. 2015;56(8):1185‐1190.
    1. Ohkubo Y, Sekido T, Nishio SI, et al. Loss of mu‐crystallin causes PPARγ activation and obesity in high‐fat diet‐fed mice. Biochem Biophys Res Commun. 2019;508(3):914‐920.
    1. Mishra S, Tai Q, Gu X, et al. Estrogen and estrogen receptor alpha promotes malignancy and osteoblastic tumorigenesis in prostate cancer. Oncotarget. 2015;6(42):44388‐44402.
    1. Yu L, Shi J, Cheng S, et al. Estrogen promotes prostate cancer cell migration via paracrine release of ENO1 from stromal cells. Mol Endocrinol. 2012;26(9):1521‐1530.
    1. Khan SR, Chaker L, Ruiter R, et al. Thyroid function and cancer risk: the Rotterdam study. J Clin Endocrinol Metab. 2016;101(12):5030‐5036.
    1. Suzuki S, Takei M, Nishio S, et al. Spiking expression of mu‐crystallin mRNA during treatment with methimazole in patients with graves' hyperthyroidism. Horm Metab Res. 2009;41(7):548‐553.
    1. Arlauckas SP, Popov AV, Delikatny EJ. Choline kinase alpha‐putting the ChoK‐hold on tumor metabolism. Prog Lipid Res. 2016;63:28‐40.
    1. Swanson MG, Keshari KR, Tabatabai ZL, et al. Quantification of choline‐ and ethanolamine‐containing metabolites in human prostate tissues using 1H HR‐MAS total correlation spectroscopy. Magn Reson Med. 2008;60(1):33‐40.
    1. Asim M, Massie CE, Orafidiya F, et al. Choline kinase alpha as an androgen receptor chaperone and prostate cancer therapeutic target. J Natl Cancer Inst. 2016;108(5):1‐13.
    1. Contractor K, Challapalli A, Barwick T, et al. Use of [11C]choline PET‐CT as a noninvasive method for detecting pelvic lymph node status from prostate cancer and relationship with choline kinase expression. Clin Cancer Res. 2011;17(24):7673‐7683.
    1. Hartenbach M, Hartenbach S, Bechtloff W, et al. Combined PET/MRI improves diagnostic accuracy in patients with prostate cancer: a prospective diagnostic trial. Clin Cancer Res. 2014;20(12):3244‐3253.
    1. Cristofanilli M, Yamamura Y, Kau SW, et al. Thyroid hormone and breast carcinoma. Primary hypothyroidism is associated with a reduced incidence of primary breast carcinoma. Cancer. 2005;103(6):1122‐1128.
    1. Mishkin SY, Pollack R, Yalovsky MA, Morris HP, Mishkin S. Inhibition of local and metastatic hepatoma growth and prolongation of survival after induction of hypothyroidism. Cancer Res. 1981;41(8):3040‐3045.
    1. Lim W, Nguyen NH, Yang HY, Scanlan TS, Furlow JD. A thyroid hormone antagonist that inhibits thyroid hormone action in vivo. J Biol Chem. 2002;277(38):35664‐35670.
    1. Hiroi Y, Kim HH, Ying H, et al. Rapid nongenomic actions of thyroid hormone. Proc Natl Acad Sci U S A. 2006;103(38):14104‐14109.

Source: PubMed

3
購読する