Amplification and protein expression of androgen receptor gene in prostate cancer cells: Fluorescence in situ hybridization analysis

  • Authors:
    • Xian Zhang
    • Shi‑Zhe Hong
    • Er‑Jiang Lin
    • Da‑Ya Wang
    • Zhi‑Jia Li
    • Li Chen
  • View Affiliations

  • Published online on: April 14, 2015     https://doi.org/10.3892/ol.2015.3114
  • Pages: 2617-2622
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Prostate cancer (PCa) is the most frequently diagnosed non-skin cancer and the second highest cause of cancer-related mortality in adult males worldwide. PCa is highly dependent upon androgen receptor (AR) signaling for cell proliferation and survival. The AR therefore plays a vital role in the develop­ment and function of normal and malignant prostate cells or PCa recurrence. The present study aimed to examine the ubiquity of AR amplification in PCa recurrence, even in the absence of androgen. For this purpose, specimens were collected from 37 patients. The amplification of AR and the number of X chromosomes were determined by two‑colored fluorescence in situ hybridization analysis. The automated image analysis was used to determine the protein expression of AR. Clinical characteristics and survival in patients whose tumors showed or did not show AR amplification and in X‑chromosome polysomy with PCa recurrence has also been compared. The results showed that >35% of patients (13 specimens) exhibited AR amplification. It was also observed that AR was immunostained more intensely in the tumors with amplified AR compared with those tumors with non‑amplified AR. This study demonstrated an influential role of AR in tumor growth and progression even after the deprivation of androgen, as well as showing the potential contribution of AR amplification to AR activation even in the relative absence of androgen.

Introduction

The most common malignancy found in American males is prostate cancer (PCa). The disseminated disease is responsible for cancerous mortality and morbidity (1,2). With the introduction and application of the screening for PCa using prostate-specific antigen, the diagnosis of clinically localized PCa has also been increased sharply (3). The use of biologically significant biomarkers may therefore stratify the risk in this group of patients.

Androgen receptor (AR) protein is expressed in nearly all types of PCa, i.e., primary and metastatic (4). The study by Visakorpi et al was the first to report the amplification of the AR gene together with a gain of chromosome X, and suggested a possible mechanism for the progression of PCa following hormone therapy (5). However, the precise mechanism for the association between AR gene amplification and the development of hormone-refractory PCa remains unknown. Androgens are significant in the growth of normal and malignant prostate cells via AR; the AR binds testosterone or dihydrotestosterone and then stimulates the transcription of androgen-sensitive genes (6). Similar to other proteins in the steroid receptor family, the AR contains separate regions responsible for hormone binding, DNA binding and transcriptional activation (transactivation). AR signaling has been widely studied in the pathogenesis and progression of PCa, the underlying mechanisms of which are believed to involve AR gene amplification, AR gene mutations, elevated AR mRNA or ligand-independent AR activation by growth factors/cytokines. Androgen deprivation therapy is the main therapeutic option for the treatment of advanced cases (79). In other words, the AR gene is an important factor in the growth and progression of CaP and hence, the majority of PCa is androgen-dependent (10,11). Additionally, in multiple studies, it has been demonstrated that the amplification and overexpression of the AR gene is evident in PCa patients whose tumors are refractory to androgen ablation therapy (12).

The present study compared AR protein expression, and clinical characteristics and survival in patients with PCa whose tumors showed or did not show AR amplification and in X-chromosome polysomy.

Materials and methods

Specimen source and tissue array

The present study was approved by the Ethics Review Board of Wenzhou Central Hospital (Wenzhou, Zhejiang, China) and all patients provided written informed consent. All samples were collected from Wenzhou Central Hospital between July 2012 and December 2013. A total of 37 male patients were selected by analyzing their medical records. Specimens were collected following detection of an increase in serum prostate-specific antigen (PSA) level and urinary retention, treated by transurethral resection following androgen deprivation therapy (Table I). The age range of the patients was 58–89 years (Table I).

Table I.

Clinical characteristics and mean optical density of AR expression.

Table I.

Clinical characteristics and mean optical density of AR expression.

A, Non-amplified AR

Clinical informationARXMOD




SpcAge, yearsStagePSAGSAD methodMOS B/w AD + TASur A/f TA (MOS)Sur A/f AD (MOS)CopySR% SR (>2)% SR (>3)Copy% X Copy (>2)% X Copy (>3)AR exp
162T3bN1M032.99FA179261.101.00301.132300.2989
264T3bN1M031.89FA5613691.380.98001.301100.0923
358T4NxM1b6.29FA295341.040.96401.04770.2333
462T2bNxM1b11010PHNI21211.031.00701.032800.4345
564T3bN1M1b0.99Orchiectomy3943822.031.00801.981400.2134
679T3bN1M03810LH-RH2811391.121.02301.102100.0896
778T1bNxM1b65.19 Diethylstilbesterol2017371.361.031811.302110.1844
883T3aNxM1b5.79Orchiectomy1311241.191.00201.15500.3912
987T3N2M1b51.29LH-RH+FA23123517.001.115017.001001000.1832
1073T3bNxM1b4009FA164201.600.91601.501300.2989
1171T3N2M1b410LH-RH4538831.900.99901.811500.2344
1266T2bNxM1b78.18Orchiectomy597661.261.023321.1239320.2283
1379Tb3N1M045.39Orchiectomy4537821.500.99001.49100.1345
1472T1bNxM1b23.69 Diethylstilbesterol2713402.091.00002.01900.2732
1581T3bN1M1b8.99Orchiectomy3329621.401.21201.342100.2896
1669T4NxM1b69LH-RH+FA298372.101.005642.116140.1891
1765T2bNxMx1458Orchiectomy915241.901.24501.89900.1892
1876T3bN1M00.78FA1114251.981.02701.912200.2985
1980T1bNxM1b67.310LH-RH+FA1918371.781.01801.783400.1189
2086T3aNxM1b65.010Orchiectomy+FA675612317.001.009017.001001000.2177
2173T2bNxM1b45.58 Diethylstilbesterol7815931.341.11501.316100.1365
2279T3bN1M05.89LH-RH8117981.421.001311.404310.2111
2370T2bNxMx5.29Orchiectomy696751.780.98701.71900.2832
2484T3aNxM1b34.19Orchiectomy16193516.001.008016.0081800.4512

B, Amplified AR

Clinical informationARXMOD




SpcAge, yearsStagePSAGSAD methodMOS B/w AD + TASur A/f TA (MOS)Sur A/f AD (MOS)CopySR% SR (>2)% SR (>3)Copy% X Copy (>2)% X Copy (>3)AR exp

2565TXNM1b14.98Orchiectomy+FA419503.242.1248391.40500.3176
2669T4NxM028.38LH-RH4211534.123.4242301.13900.3809
2780TxN1Mx9.99 Diethylstilbesterol3919584.323.4339291.393640.3302
2889T4NxM1b23.69Orchiectomy566623.122.9836321.021700.382
2972TXNM1b1909Orchiectomy+FA2817452.601.9637291.224790.329
3079T3bN0M015.210LH-RH+FA1310232.781.9889581.56500.48
3173T4NxM1b8.88Orchiectomy40236313.1011.666441.27800.4591
3268T4NxM011.29LH-RH37316814.613.886712.564970.2901
3370T3bN0M0178LH-RH+FA13122512.211.656462.58500.23
3465TXNM1b21.29Orchiectomy+FA23194211.310.987671.181800.3567
3576T4NxM1b16.610Orchiectomy2825534.334.6761491.9160450.378
3666TxN1Mx26.79 Diethylstilbesterol3329622.671.9734291.87400.3871
3778TXNM1b8.29Orchiectomy+FA3728652.981.8941381.051430.3409

[i] AR, androgen receptor; spc, specimen; PSA, prostate-specific antigen; GS, Gleason sum; MOS, medical outcome study; B/w, between; AD, androgen deprivation; TA, tissue acquisition; FA, flutamide; PH, primary hypogonadism; LH-RH, luteinizing hormone-releasing hormone; NI, no information; Sur, survival; A/f, after; SR, signal ratio; MOD, mean optical density; exp, expression.

The tissue array was constructed from 37 duplicate samples of recurrent PCa and 9 samples of adenofibromyomatous hyperplasia. Tissue preparation artifacts were marked for sampling and 2-mm diameter punch biopsies were obtained. The tissue areas were free of necrosis and cauterization injury. Sections (5-µm thick) were cut from each paraffin block containing a recurrent PCa or adenofibromyomatous hyperplasia sample, and mounted on slides, followed by staining with hematoxylin and eosin, and immunohistochemical analysis.

Immunohistochemical detection

The tissue array sections (BioMax; Guangzhou, China) were used for immunohistochemical detection according to the manufacturer's instructions. Briefly, the sections underwent antigen retrieval in Citra buffer (Biogenex, Shanghai, China) and were cooled to room temperature for pre-incubation with 2% normal horse serum for 15 min at 37°C followed by incubation with monoclonal anti-human AR antibody (F39.4.1; 1:200; Biogenex) for 1 h. The sections were then treated with biotinylated anti-mouse immunoglobulin G (1:200) for 15 min at 37°C, followed by avidin-biotin complex amplification (Vector, Beijing, China). The signals were visualized using diaminobenzidine (Vector). The mean optical density (MOD) of immunostaining was measured using a Zeiss Axioskop microscope (Zeiss, Beijing, China), a 3-chip CCD camera C5810, (Hamamatsu, Beijing, China) and a camera control unit (Hamamatsu). Immunopositivity and immunonegativity were determined using a linear discriminant analysis method.

Fluorescence in situ hybridization (FISH) protocol

The 5-µm PCa tissue slides were deparaffinized, treated with 0.2 N HCl, incubated in 1 M sodium thiocyanate and immersed in protease solution (Vysis Inc., Downers Grove, IL, USA) for 10 min at 37°C. The tissues were then fixed with 10% formalin for 10 min, denatured for 5 min at 72°C and sequentially incubated in 75, 90 and 100% ethanol. The tissues were then treated with proteinase K for 6 min at 37°C, followed by their dehydration and hybridization.

Next, two-colored FISH was performed, as described previously (13), using spectrum orange-labeled AR and spectrum green-labeled X-chromosome centromere region DNA probes (Vysis). Briefly, tissue array sections were hybridized with 3 µl of each probe and Cot1-DNA (1 µg/µl; Vysis) overnight at 37°C in a humidified atmosphere. The slides were then was hed for counterstaining with 0.2 µM 4′,6′-diamidino-2-phenylindole hydrochloride. FISH signals were scored with a fluorescence microscope (Zeiss) equipped with two double-band pass filters using 40X and 100X objective lenses. The number of AR gene signals and X centromere signals was evaluated by visual analysis of 800 to 1,200 nuclei per specimen. AR amplification was present if the AR to X ratio exceeded 1.5 (14), and X polysomy was present if the number of X centromere signals exceeded an average of 2 signals per cell (15).

Statistical data analysis

Medians and interquartile ranges are used to describe the data. Optical density is presented as the mean ± standard deviation. The data were not evenly distributed, so a Wilcoxin analysis was used for comparison of the groups. Kaplan-Meier plot was used for comparison of the survival rates via log-rank analysis.

Results

Immunohistochemistry image analysis

The clinical characteristics and mean optical density of AR expression for the 37 patients are summarized in Table I. Out of 37 subjects with PCa recurrence, 13 (35%) exhibited amplified AR expression, while 24 did not (Table I). Amplified AR (optical density, 0.45±0.03) was more intensely immunostained than non-amplified AR (optical density, 0.21±0.06) in the chosen tumor specimens. The intensity of AR immunostaining and the degree of AR amplification did not exhibit any association or trend. There was additionally no association between the protein expression of AR and the X-chromosome copy number.

No statistically significant differences were observed between amplified AR and non-amplified AR tumors specimens with regard to serum PSA levels, clinical tumor-node-metastasis stage, Gleason sum, time from androgen deprivation therapy to recurrence and survival following androgen deprivation (Table II and Fig. 1).

Table II.

Clinical characteristics and survival of specimens with amplified and non-amplified AR.

Table II.

Clinical characteristics and survival of specimens with amplified and non-amplified AR.

EntryNon-amplified ARAmplified AR
Number of specimens24.013.00
Mean age of Spc ± SD73.40±8.4073.10±7.10
Mean GS ± SD8.90±0.709.00±0.60
Mean PSA ± SD53.20±88.8030.10±46.60
Mean MOS B/w AD + TA ± SD36.00±21.8033.10±11.60
Mean survival A/f AD (MOS) ± SD52.80±28.5051.50±13.90
Mean AR expression (MOD) ± SD0.24±0.100.36±0.07

[i] AR, androgen receptor; Spc, specimen; SD, standard deviation; PSA, prostate-specific antigen (ng/ml); GS, gleason sum; MOS, medical outcome study; B/w, between; AD, androgen deprivation; TA, tissue acquisition; a/f, after; MOD, mean optical density.

Additionally, no association with X polysomy was observed for these clinical parameters. Nevertheless, 6 of the recurrent PCa specimens (16% of the chosen patients) exhibited X-chromosome 2.5 (copy number) and greater, while no differences were found when clinical characteristics between these groups were compared.

FISH analysis

AR FISH analysis results for tumors with normal AR and AR amplification are shown in Fig. 2A and B, respectively. The majority of the nuclei in the normal cells showed one green and one red signal, indicating that each nucleus had one X chromosome with one AR gene (Fig. 2A). The presence of two red signals in the FISH section in the majority of the PCa cells indicated the presence of AR gene amplification (Fig. 3).

Discussion

Since an effective therapy for the treatment of PCa recurrence has yet to be identified, an improved understanding of the mechanism behind the transition from androgen-dependent PCa to PCa recurrence may provide novel treatment targets (16). The only possible explanation for PCa recurrence is that the increased expression of AR protein through AR amplification may allow the expression of androgen-regulated genes despite castration levels of serum androgens. The present study found that the AR gene was amplified in ~35% of 37 PCa recurrence specimens; these results are far indicate an improved outcome compared with previous studies showing AR amplification in 23% of 47 (13), 25% of 16 (17), 28% of 54 (18), 30% of 23 (5) and 31% of 13 (19) patients. The results compared AR expression levels between tumors with AR amplification and with a single AR signal. Reverse transcriptase polymerase chain reaction was previously used in 13 patients to demonstrate that AR mRNA expression occurred in the recurrence of 4 tumors and demonstrated that AR amplification was higher than AR mRNA in the recurrence of 9 tumors that were not amplified (19). When immunohistochemistry was used to compare AR protein expression between amplified and non-amplified tumors, the study by Visakorpi et al (5) found that ~80% of primary tumor cells (as well as recurrence) expressed nuclear AR protein, but that there was no significant difference in the level of protein expression in primary tumors compared with other tumors (exhibiting recurrence or recurrence with AR amplification). In another study, the hybridization intensity for AR mRNA (ISH for 5 PCa reccurence specimens) was higher than that of the original PCa non-amplified specimens (18). The results showed different results for the length of survival in patients with advanced PCa treated with androgen deprivation based on the amplified or non-amplified AR in recurrent tumors. However, the present study found that amplified AR had no association with the duration of survival following androgen deprivation (Fig. 1), and no association was found between survival times following androgen deprivation and X polysomy. Koivisto et al reported that AR amplification occurred more often in males who exhibited a complete response to or longer interval between androgen deprivation and recurrence (18). However, the present study found no difference in the interval between androgen deprivation and recurrence. Recently, FISH was used to appraise the attainability of characterizing gene copy number alterations of circulating tumor cells isolated using the Cell Search system in specimens with PCa (particularly progressive castration-resistant metastatic PCa) (20); this study reported high-level chromosomal amplification of AR in 38% of the analyzed samples and relative gain of MYC in 56% of the samples, which also supports the present results.

Immunohistochemical detection was also used to optimize the production of the linear association between AR protein and its DAPI immunostaining (21) using automated digital video image analysis for precise results. It was observed that those patients whose tumors demonstrated AR amplification exhibited a 5-month faster recurrence than those whose had non-amplified tumors. The amplified AR PCa recurrent tumors exhibited greater levels of AR protein expression, but this was not associated with survival. It was also demonstrated that AR protein expression was 60% higher in tumors with an AR copy number >2.1. Additionally, the study found that X-chromosome copy number was increased in up to 13.8% of cancer specimens, which corresponds with the fact that AR copy number can be increased by X-chromosome polysomy, but will not impact on AR protein expression (5,15,21).

In conclusion, this study is the first of its type, quantitatively comparing AR protein expression and AR amplification in PCa recurrence. This study demonstrated that AR influences tumor growth and progression even where androgen is deprived. Furthermore, the results indicated a potential contribution of AR amplification to AR activation in the relative absence of androgen.

Acknowledgements

The authors would like to thank the staff of the Wenzhou Central Hospital for their assistance in subject recruitment throughout the study period.

References

1 

Lassi K and Dawson NA: Update on castrate-resistant prostate cancer: 2010. Curr Opin Oncol. 22:263–267. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Jemal A, Siegel R, Xu J and Ward E: Cancer statistics, 2010. CA Cancer J Clin. 60:277–300. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Shahrokh F, Shariat, Axel S, et al: Tumor markers in prostate cancer I: Blood-based markers. Acta Oncol. 50:61–75. 2011. View Article : Google Scholar

4 

Culig Z, Hobisch A, Bartsch G and Klocker H: Androgen receptor - an update of mechanisms of action in prostate cancer. Urol Res. 28:211–219. 2000. View Article : Google Scholar : PubMed/NCBI

5 

Visakorpi T, Hyytinen E, Koivisto P, et al: In vivo amplification of the androgen receptor gene and progression of human prostate cancer. Nat Genet. 9:401–406. 1995. View Article : Google Scholar : PubMed/NCBI

6 

Cude KJ, Montgomery JS, Price DK, et al: The role of an androgen receptor polymorphism in the clinical outcome of patients with metastatic prostate cancer. Urol Int. 68:16–23. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Heinlein CA and Chang C: Androgen receptor in prostate cancer. Endocr Rev. 25:276–308. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Chen CD, Welsbie DS, Tran C, et al: Molecular determinants of resistance to antiandrogen therapy. Nat Med. 10:33–39. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Gelmann EP: Molecular biology of the androgen receptor. J Clin Oncol. 20:3001–3015. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Shaffer DR and Scher HI: Prostate cancer: a dynamic illness with shifting targets. Lancet Oncol. 4:407–414. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Linja MJ and Visakorpi T: Alterations of androgen receptor in prostate cancer. J Steroid Biochem Mol Biol. 92:255–264. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Kiessling A, Hogrefe C, Erb S, et al: Expression, regulation and function of the ISGylation system in prostate cancer. Oncogene. 28:2606–2620. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Bubendorf L, Kononen J, Koivisto P, et al: Survey of gene amplifications during prostate cancer progression by high-throughout fluorescence in situ hybridization on tissue microarrays. Cancer Res. 59:803–806. 1999.PubMed/NCBI

14 

Edwards J, Krishna NS, Mukherjee R, Watters AD, Underwood MA and Bartlett JM: Amplification of the androgen receptor may not explain the development of androgen-independent prostate cancer. BJU Int. 88:633–637. 2001. View Article : Google Scholar : PubMed/NCBI

15 

Koivisto P, Hyytinen E, Palmberg C, et al: Analysis of genetic changes underlying local recurrence of prostate carcinoma during androgen deprivation therapy. Am J Pathol. 147:1608–1614. 1995.PubMed/NCBI

16 

Feldman BJ and Feldman D: The development of androgen-independent prostate cancer. Nat Rev Cancer. 1:34–45. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Hyytinen ER, Haapala K, Thompson J, et al: Pattern of somatic androgen receptor gene mutations in patients with hormone-bractory prostate cancer. Lab Invest. 82:1591–1598. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Koivisto P, Kononen J, Palmberg C, et al: Androgen receptor gene amplification: A possible molecular mechanism for androgen deprivation therapy failure in prostate cancer. Cancer Res. 57:314–319. 1997.PubMed/NCBI

19 

Linja MJ, Savinainen KJ, Saramaki OR, Tammela TL, Vessella RL and Visakorpi T: Amplification and overexpression of androgen receptor gene in hormone-bractory prostate cancer. Cancer Res. 61:3550–3555. 2001.PubMed/NCBI

20 

Leversha MA, Han J, Asgari Z, et al: Fluorescence in situ hybridization analysis of circulating tumor cells in metastatic prostate cancer. Clin Cancer Res. 15:2091–2097. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Brown RS, Edwards J, Dogan A, et al: Amplification of the androgen receptor gene in bone metastases from hormone-bractory prostate cancer. J Pathol. 198:237–244. 2002. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2015
Volume 9 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang X, Hong SZ, Lin EJ, Wang DY, Li ZJ and Chen L: Amplification and protein expression of androgen receptor gene in prostate cancer cells: Fluorescence in situ hybridization analysis. Oncol Lett 9: 2617-2622, 2015
APA
Zhang, X., Hong, S., Lin, E., Wang, D., Li, Z., & Chen, L. (2015). Amplification and protein expression of androgen receptor gene in prostate cancer cells: Fluorescence in situ hybridization analysis. Oncology Letters, 9, 2617-2622. https://doi.org/10.3892/ol.2015.3114
MLA
Zhang, X., Hong, S., Lin, E., Wang, D., Li, Z., Chen, L."Amplification and protein expression of androgen receptor gene in prostate cancer cells: Fluorescence in situ hybridization analysis". Oncology Letters 9.6 (2015): 2617-2622.
Chicago
Zhang, X., Hong, S., Lin, E., Wang, D., Li, Z., Chen, L."Amplification and protein expression of androgen receptor gene in prostate cancer cells: Fluorescence in situ hybridization analysis". Oncology Letters 9, no. 6 (2015): 2617-2622. https://doi.org/10.3892/ol.2015.3114