Suppression of Elk1 inhibits thyroid cancer progression by mediating PTEN expression

  • Authors:
    • Yakun Kong
    • Junjie Yin
    • Yun Fu
    • Yufeng Chen
    • Yanhong Zhou
    • Xiuqin Geng
  • View Affiliations

  • Published online on: July 10, 2018     https://doi.org/10.3892/or.2018.6554
  • Pages: 1769-1776
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

ETS‑domain containing protein (Elk1) is reported to be a member of the ETS oncogene family, and promotes tumorigenesis in cancer such as bladder, prostate and ovarian. Nevertheless, the role of Elk1 in thyroid cancer progression remains unclear. In the present study, we aimed to investigate the role and underlying molecular mechanism of Elk1 in thyroid cancer. The results indicated that Elk1 was significantly upregulated in thyroid cancer tissues and cells. We found that loss of Elk1 function obviously induced the expression of early growth response‑1 (Egr‑1) and PTEN, promoted apoptosis and constrained the proliferation of thyroid cancer cells. Furthermore, Egr‑1 inhibition obviously abrogated the induction of PTEN induced by Elk1 reduction. Moreover, Egr‑1 suppression prevented the promotion of apoptosis and the inhibition of cell proliferation caused by Elk1 reduction. In conclusion, Elk1 inhibition induced thyroid cancer cell apoptosis and restrained their proliferation by regulating Egr‑1/PTEN, indicating a potential role for Elk1 in thyroid cancer treatment.

Introduction

Thyroid cancer (TC) is the most common endocrine malignancy, accounting for approximately 3% of all malignant tumors (1,2). Moreover, it has a higher incidence in women, and is the most common cancer of the head and neck (3,4). In recent years, the incidence of TC has significantly increased. A better understanding of the underlying mechanism of TC would provide novel insights for the treatment of TC.

ETS-domain containing protein (Elk1), a transcription factor belonging to the ETS oncogene family, regulates the oncogene c-fos by phosphorylation through activation of the PKC/ERK pathways (58). Studies have reported that Elk1 has roles in cell proliferation, the cell cycle, apoptosis and tumorigenesis (9,10). It has been demonstrated that Elk1 expression is upregulated, and promotes cell viability in bladder cancer (10). Elk1 has also been shown to be induced, and to play a crucial role in hormone-resistant or metastatic prostate cancers (11) and is reported to play an important role in breast cancer and ovarian cancer (7,1214). However, the molecular mechanism of Elk1 in TC remains unknown. In our study, we investigated the role of Elk1 in cell proliferation and apoptosis in TC. A previous study has shown that in SH-SY5Y neuroblastomas Elk1 represses the expression of Egr-1, which is implicated in different cellular processes containing cell proliferation, differentiation and apoptosis (15).

Early growth response-1 (Egr-1), also called Zif268, NGF1-A, and Krox24, is a transcription factor containing a zinc-finger DNA binding domain, and is known as an important immediate-early gene (IEG) (1621). Egr-1 promotes quiescent cells to enter the proliferative phase, regulating cell growth and differentiation (20,22,23). Egr-1 is found in eukaryotic genomes, and is highly conserved evolutionarily (24). Many factors can activate Egr-1, and activated Egr-1 regulates target gene transcription by interacting with the binding sites of target genes. The biological function of Egr-1 is realized by upregulating or downregulating target gene expression. Egr-1 is considered to be a class II tumor suppressor gene (25). It has been demonstrated that the expression of Egr-1 is decreased in breast cancer, non-small cell lung cancer (NSCLC) and glioma (26,27). Nevertheless, other studies have demonstrated that Egr-1 expression is increased in prostate cancer, lymphoma and Wilms' tumor, among others (28,29). It has been reported that Egr-1 could directly regulate the transcription of the phosphatase and tensin homolog deleted on chromosome ten (PTEN) (30).

PTEN is also recognized as a tumor suppressor (31,32), and it was observed that PTEN is inactivated or inhibited in multiple types of cancer including thyroid carcinoma (33). Research over the past few year has shown the mechanism by which loss of PTEN function contributes to tumor development (34). It has been reported that PTEN inhibition induces cell survival and cisplatin resistance in human ovarian cancer, and promotes the risk of breast and endometrial cancers and leukemia (35,36). Furthermore, it has been demonstrated that PTEN suppression causes thyroid cancer development, progression and invasion, authenticating PTEN as a crucial tumor suppressor in thyroid carcinogenesis (37). Moreover, transient ectopic expression of PTEN promotes cell cycle arrest and cell death in thyroid cancer cell lines (38).

In the present study, we investigated the molecular mechanism underlying Elk1 action in thyroid cancer progression in vitro. We found that Elk1 expression was upregulated in thyroid cancer cell lines and tissues. Loss of Elk1 function significantly inhibited the proliferation and induced apoptosis in thyroid cancer cell lines. Furthermore, the results also demonstrated that Elk1 inhibition induced PTEN expression by upregulating Egr-1. Therefore, this study proposes the potential role of Elk1 in preventing thyroid cancer, providing a potential novel target for treatment.

Materials and methods

Cell lines and tissues

Human thyroid cancer cell lines FTC-133 and TPC-1 were purchased from the Protection Agency Culture Collections (HPACC, Salisbury, Wiltshire, UK). The cells were cultured in Dulbecco's modified Eagle's medium (DMEM, Sigma-Aldrich, Merck KGaA, Darmstadt, Germany) containing 10% fetal bovine serum (FBS, HyClone; GE Healthcare Life Sciences, Logan, UT, USA). The normal thyroid cell line FRTL-5 was obtained and cultured according to a previously described protocol (39). The cell lines were incubated at 37°C with a 5% CO2 atmosphere. Additionally, 10 pairs (sample collection: From February 2017 to October 2017) of tumor tissue samples and matched adjacent normal tissues were obtained from Xinxiang Central Hospital along with written informed consent of patients (4 males and 6 females; 39–50 years old), and were immediately stored in liquefied ammonia. The study was approved by the Xinxiang Central Hospital Ethics Board.

Cell transfection

The cells (FTC-133 and TPC-1) were separately seeded in 12-well plates and incubated in a humid atmosphere with 5% CO2 at 37°C until 80% fusion was achieved. The transfection procedure was performed according to the manufacturer's instructions. Elk1 siRNA (5′-AACCACCCGCCACTCTTCCT-3′), Egr-1 siRNA (5′-GTAGGTTGCTGTCGTCAGGGTAAAT-3′), and non-specific siRNA were separately diluted in FBS-free DMEM medium (200 µl) with 6 µl TurboFect (Thermo Fisher Scientific, Inc., Waltham, MA, USA), and the mixtures were added in the well. The cells were then cultured under conditions of 5% CO2 at 37°C for 24 h.

Cell growth and viability

Cell growth and viability were measured using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, and the assay was performed in accordance with standard protocols. The cells (1×05 cells/well) were cultured in 96-well plates with DMEM containing 10% FBS. The medium in each well was then replaced by 18 µl MTT (5 g/l) diluted in phosphate-buffered saline (PBS) followed by incubation at 37°C for 5 h. Subsequently, a total of 150 µl dimethyl sulfoxide was added per well, in order to dissolve the crystals. Finally, the result was read using a microplate reader (Thermo Fisher Scientific) at 490 nm. The analysis was repeated three times.

Bromodeoxyuridine (BrdU) assay

The BrdU cell proliferation assay kit (Cell Signaling Technology, Danvers, MA, USA) was used to detect cell proliferation based on the manufacturer's protocol. Briefly, the cells were plated in 96-well plates and incubated with BrdU solution (10 µl per well) for 1.5 h. A total of 150 µl denaturing solution was added per well to replace the medium followed by culturing for 30 min, and cells were incubated with anti-BrdU conjugated with peroxidase. After addition of the substrate and incubation for 20 min, the optical density at 450 nm was determined at room temperature using a SpectroFluor Plus multiwell plate reader (Tecan, Research Triangle Park, NC, USA). The experiment was repeated three times.

Caspase-3 activity detection

The caspase-3 activity assay was performed as per the manufacturer's instructions using the caspase-3 activity assay kit (Beyotime Institute of Biotechnology, Nantong, China). Briefly, cells were lysed on ice for 15 min, and 10-µl cell lysate per sample in 90 µl reaction buffer [1% NP-40, 20 mM Tris-HCl (pH 7.5), 137 mM Nad and 10% glycerol] containing 12 µl caspase-3 substrate (Ac-DEVD-pNA) (2 mM) were added into 96-well microtitre plates. Lysates were incubated at 37°C for 2 h. The results were measured with an ELISA reader (Tecan) at an absorbance of 405 nm.

Annexin V fluorescein isothiocyanate conjugate and propidium iodide (Annexin V FITC/PI)

Apoptosis was measured using the BD Pharmingen™ Annexin V FITC/PI apoptosis detection kit (BD Biosciences, Franklin Lakes, NJ, USA) following the standard protocol. In brief, cells were precooled in cold PBS and suspended in binding buffer. Then, Annexin V FITC solution (10 µl) was added followed by incubation for 23 min. Afterwards, 10 µl PI was added and the reaction was allowed to proceed for 7 min. Cellular apoptosis was measured using a FACS analyzer (Thermo Fisher Scientific, Inc.).

Real-time quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted from cells using TRIzol (Thermo Fisher Scientific, Inc.). Total RNA extraction from tissues was performed according to a previously published method (40). RNA (5 µg) was then synthesized into cDNA using the Revert Aid First Strand cDNA Synthesis kit (Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. RT-qPCR was performed in 20 µl reaction volumes containing 10 µl Applied Biosystems® SYBR® Green PCR Master Mix (Thermo Fisher Scientific, Inc.). The genes were normalized to GAPDH. The primers used were as follows: Elk1 sense primer, 5′-CCTTGCGGTACTACTATGAC-3′ and antisense primer, 5′-GGCTGCGGCTGCAGAGACTGG-3′; Egr-1 sense primer, 5′-TTTGCCAGGAGCGATGAAC-3′ and antisense primer, 5′-CCGAAGAGGCCACAACACTT-3′; GAPDH sense primer, 5′-CGTCTTCACCACCATGGAGA-3′ and antisense primer, 5′-CGGCCATCACGCCACAGTTT-3′. The protocol: 94°C for 30 sec; 35 cycles of 95°C for 30 sec, 58°C (Elk1) or 60°C (Egr-1) for 30 sec and 72°C for 30 sec; 72°C for 10 min. The relative gene expression levels were estimated using the 2−ΔΔCt method.

Immunoblotting analysis

Proteins were extracted from the cells treated with the lysate (Beyotime Institute of Biotechnology), and quantified using the BCA kit (Beyotime). A total of 25 µg of protein was separated using 12% sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE), and transferred to a nitrocellulose membrane using a semi-dry blotting apparatus (Bio-Rad Laboratories, Inc., Hercules, CA, USA). The membranes containing the proteins were incubated in Tris-buffered saline (TBS) containing 2% non-fat dry milk at room temperature for 2 h followed by washing with TBS. The nitrocellulose membranes were then incubated overnight at 4°C with the primary antibodies against Elk1 (1:500; cat. no. ab131465), Egr1 (1:500; cat. no. ab182624), PTEN (1:800; cat. no. ab31392) and GAPDH (1:1,000; cat. no. ab37168; all from Abcam Inc., Cambridge, MA, USA), and then incubated with a horseradish peroxidase conjugated secondary antibody (1:1,000; cat. no. ab205718; Abcam) for 1 h at room temperature. Finally, proteins were visualized using Pierce enhanced chemiluminescence (Thermo Fisher Scientific, Inc.) in a Bio-Rad ChemiDoc apparatus.

Statistical analysis

Data are expressed as mean ± standard deviation (SD). Statistical significance was determined by Student's t-test for two groups or by one-way ANOVA for multiple groups. A P-value of <0.05 was considered statistically significant.

Results

Induction of Elk1 in thyroid cancer

To investigate the expression of Elk1 in thyroid cancer, we detected the expression of Elk1 in thyroid cancer tissues and thyroid cancer cells. The results showed that the mRNA (Fig. 1A) expression level of Elk1 in the thyroid cancer tissue was significantly higher than that in the normal tissue. Additionally, mRNA (Fig. 1A) and protein (Fig. 1B) were both significantly increased in the thyroid cancer cells (FTC-133 and TPC-1) compared with that in the normal thyroid cells (FRTL-5). Thus, Elk1 expression was upregulated in thyroid cancer.

Suppression of Elk1 induces the expression of Egr-1 and PTEN

To detect the role of Elk1 in the regulation of Egr-1 and PTEN, we inhibited Elk1 expression using cell transfection in FTC-133 and TPC-1 cells. The results indicated that the loss-of-function experiment was successful with a significant reduction of Elk1 mRNA (Fig. 2A) and protein (Fig. 2B) expression in the Elk1 siRNA group compared with the non-specific siRNA group. Furthermore, the protein (Fig. 3) expression levels of Egr-1 and PTEN were obviously increased in the Elk1 siRNA group compared with the non-specific siRNA group.

Suppression of Elk1 promotes thyroid cancer cell apoptosis

Annexin V FITC/PI and caspase-3 activity detection were used to assess the role of Elk1 in the downregulation of thyroid cancer cell apoptosis. The results demonstrated that the caspase-3 activity in the Elk1 siRNA group was also obviously induced compared with that noted in the non-specific siRNA group (Fig. 4A). Furthermore, cellular apoptosis in the Elk1 siRNA group was markedly increased compared with the non-specific siRNA group as determined using the Annexin V FITC/PI assay (Fig. 4B).

Suppression of Elk1 constrains thyroid cancer cell proliferation

Cell proliferation was assessed using the BrdU and MTT assays to further detect the biological effect of Elk1 inhibition on thyroid cancer cells. In the BrdU assay, the results demonstrated that FTC-133 and TPC-1 cell proliferation (Fig. 5A) was markedly inhibited in the Elk1 siRNA group compared with the non-specific siRNA group. In the MTT assay, FTC-133 and TPC-1 growth and viability (Fig. 5B) were both constrained in the Elk1 siRNA group compared with the non-specific siRNA group, and the differences were significant.

Elk1 inhibition upregulates PTEN via increased Egr-1 expression

To explore the mechanism of Elk1 regulation of PTEN and Egr-1, we performed co-transfection of Elk1 siRNA and Egr-1 siRNA into FTC-133 and TPC-1 cells, and the results showed that the expression of Elk1 and Egr-1 protein (Fig. 6) was significantly suppressed in the Elk1-Egr-1 siRNA group compared with the non-specific siRNA group and with the Elk1 siRNA group, respectively. Furthermore, PTEN protein expression (Fig. 6) was significantly downregulated in the Elk1-Egr-1 siRNA group compared with the Elk1 siRNA group. The promotion of apoptosis (Fig. 7A) and inhibition of cell proliferation (Fig. 7B) caused by the suppression of Elk1 were markedly overcome by Egr-1 inhibition.

Discussion

Thyroid cancer (TC) has different histological and biological types, and the clinically significant human thyroid cancers are papillary and follicular carcinomas (41). Surgery-based treatment is the primary clinical treatment. However, specific targets for drugs to treat TC are still lacking, and the molecular mechanisms of TC remain unclear. Elk1 is reported to be a transcriptional factor that forms part of the ternary complex factor (TCF), and it can be phosphorylated by the MAPK cascade (42). Elk1 regulates different factors related to cell proliferation, differentiation and even tumorigenesis (43). Studies have demonstrated that Elk1 plays an important role in cancer progression. Kawahara et al found that Elk1 is induced in prostate cancer and promotes tumor development, whereas Elk1 inhibition suppresses tumor growth (44). Additionally, Elk1 is upregulated and promotes cell proliferation in bladder cancer and non-small cell lung cancer (NSCLC) (10). In this study, we found that Elk1 expression was significantly upregulated in TC tissues and cells. Moreover, the results showed that TC cell proliferation and apoptosis were constrained and promoted, respectively, after experimental downregulation of Elk1 by siRNA.

Egr-1 is an important immediate-early gene, and is also a tumor suppressor related to different cancers (45), as well as being implicated in cell proliferation and apoptosis (22). Additionally, studies have demonstrated that Egr-1 expression is down-regulated or absent in NSCLC and breast cancer, while it is upregulated in prostate cancer and lymphadenoma (26,28). Research has shown that Egr-1 can be regulated via Elk1, and Demir and Kurnaz demonstrated that Egr-1 could be repressed by Elk-1 expression in SH-SY5Y neuroblastomas (15). In our study, Elk1 inhibition markedly increased Egr-1 expression in the TC cell lines FTC-133 and TPC-1. Furthermore, it was previously indicated that Egr-1 positively regulates PTEN expression, and that loss of Egr-1 restrains the expression of PTEN. Thus, we investigated the role of the Elk1/Egr-1 pathway in TC.

PTEN is accepted as a tumor suppressor, and its suppression function is realized by inhibiting the activity of P13K (32). PTEN induction is reported to facilitate cell apoptosis in bladder cancer, lung squamous carcinoma and ovarian cancer (46). PTEN mutations are extremely common in melanoma cell lines, advanced prostate cancers, and endometrial carcinomas, and PTEN deficiency was found to accelerate the proliferation and invasion of a range of cancers such as gastric cancer, pancreatic cancer, prostate cancer, among other (4749). Most importantly, many correlative data suggest that inhibition of PTEN leads to TC in vivo, and Guigon et al demonstrated that suppression of PTEN facilitates TC tumor development in a mouse model (50). In our study, PTEN expression was induced by Elk1 inhibition, decreasing TC cell proliferation and increasing apoptosis. Additionally, loss of Egr-1 significantly reversed the effect of Elk1 inhibition on PTEN expression, TC cell proliferation and apoptosis. Thus, the results showed that Elk1 inhibition could induce the expression of PTEN via upregulation of Egr-1.

In summary, this study revealed that Elk1 is induced in TC tissues and cell lines. The loss of Elk1 can markedly increase the expression of PTEN, promoting TC cell apoptosis and inhibiting proliferation. Furthermore, Elk1 suppression upregulates PTEN expression via increased Egr-1 expression, providing a novel target for the treatment of TC.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

All authors participated in the design, interpretation of the studies and analysis of the data and review of the manuscript; YK and XG designed and prepared the experiments; YK performed the experiments; JY, YF, YC and YZ contributed to the reagents/materials/analysis tools; YK wrote the manuscript; XG modified and revised the manuscript. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

The present study was approved by the Xinxiang Central Hospital Ethics Board and written informed consent obtained from the patients.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbrevations

Abbreviations:

Elk1

ETS-domain containing protein

Egr-1

early growth response-1

TC

thyroid cancer

DMEM

Dulbecco's modified Eagle's nedium

FBS

fetal bovine serum

MTT

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide

PBS

phosphate-buffered saline

BrdU

bromodeoxyuridine

Annexin V FITC/PI

Annexin V fluorescein isothiocyanate conjugate/propidium iodide

RT-qPCR

real-time quantitative polymerase chain reaction

TBS

Tris-buffered saline

References

1 

Xing M, Haugen BR and Schlumberger M: Progress in molecular-based management of differentiated thyroid cancer. Lancet. 381:1058–1069. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Kelly LM, Barila G, Liu P, Evdokimova VN, Trivedi S, Panebianco F, Gandhi M, Carty SE, Hodak SP, Luo J, et al: Identification of the transforming STRN-ALK fusion as a potential therapeutic target in the aggressive forms of thyroid cancer. Proc Natl Acad Sci USA. 111:4233–4238. 2014. View Article : Google Scholar : PubMed/NCBI

3 

McLeod DS, Sawka AM and Cooper DS: Controversies in primary treatment of low-risk papillary thyroid cancer. Lancet. 381:1046–1057. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Van Fossen VL, Wilhelm SM, Eaton JL and McHenry CR: Association of thyroid, breast and renal cell cancer: A population-based study of the prevalence of second malignancies. Ann Surg Oncol. 20:1341–1347. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Tatler AL, Habgood A, Porte J, John AE, Stavrou A, Hodge E, Kerama-Likoko C, Violette SM, Weinreb PH, Knox AJ, et al: Reduced Ets Domain-containing protein Elk1 promotes pulmonary fibrosis via increased integrin αvβ6 expression. J Biol Chem. 291:9540–9553. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Buffet C, Catelli MG, Hecale-Perlemoine K, Bricaire L, Garcia C, Gallet-Dierick A, Rodriguez S, Cormier F and Groussin L: Dual specificity phosphatase 5, a specific negative regulator of ERK signaling, is induced by serum response factor and Elk-1 transcription factor. PLoS One. 10:e01454842015. View Article : Google Scholar : PubMed/NCBI

7 

Morris JF, Sul JY, Kim MS, Klein-Szanto AJ, Schochet T, Rustgi A and Eberwine JH: Elk-1 phosphorylated at threonine-417 is present in diverse cancers and correlates with differentiation grade of colonic adenocarcinoma. Hum Pathol. 44:766–776. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Glidewell-Kenney CA, Trang C, Shao PP, Gutierrez-Reed N, Uzo-Okereke AM, Coss D and Mellon PL: Neurokinin B induces c-fos transcription via protein kinase C and activation of serum response factor and Elk-1 in immortalized GnRH neurons. Endocrinology. 155:3909–3919. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Chen YS, Aubee J, DiVito KA, Zhou H, Zhang W, Chou FP, Simbulan-Rosenthal CM and Rosenthal DS: Id3 induces an Elk-1-caspase-8-dependent apoptotic pathway in squamous carcinoma cells. Cancer Med. 4:914–924. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Kawahara T, Shareef HK, Aljarah AK, Ide H, Li Y, Kashiwagi E, Netto GJ, Zheng Y and Miyamoto H: ELK1 is up-regulated by androgen in bladder cancer cells and promotes tumor progression. Oncotarget. 6:29860–29876. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Patki M, Chari V, Sivakumaran S, Gonit M, Trumbly R and Ratnam M: The ETS domain transcription factor ELK1 directs a critical component of growth signaling by the androgen receptor in prostate cancer cells. J Biol Chem. 288:11047–11065. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Kim HR, Lee HN, Lim K, Surh YJ and Na HK: 15-Deoxy-Δ12,14-prostaglandin J2 induces expression of 15-hydroxyprostaglandin dehydrogenase through Elk-1 activation in human breast cancer MDA-MB-231 cells. Mutat Res. 768:6–15. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Hsu YL, Hou MF, Kuo PL, Huang YF and Tsai EM: Breast tumor-associated osteoblast-derived CXCL5 increases cancer progression by ERK/MSK1/Elk-1/snail signaling pathway. Oncogene. 32:4436–4447. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Goncharenko-Khaider N, Matte I, Lane D, Rancourt C and Piche A: Ovarian cancer ascites increase Mcl-1 expression in tumor cells through ERK1/2-Elk-1 signaling to attenuate TRAIL-induced apoptosis. Mol Cancer. 11:842012. View Article : Google Scholar : PubMed/NCBI

15 

Demir O and Kurnaz IA: Wildtype Elk-1, but not a SUMOylation mutant, represses egr-1 expression in SH-SY5Y neuroblastomas. Neurosci Lett. 437:20–24. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Pacini L, Suffredini S, Ponti D, Coppini R, Frati G, Ragona G, Cerbai E and Calogero A: Altered calcium regulation in isolated cardiomyocytes from Egr-1 knock-out mice. Can J Physiol Pharmacol. 91:1135–1142. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Zalcman G, Federman N, de la Fuente V and Romano A: Nuclear factor kappa B-dependent Zif268 expression in hippocampus is required for recognition memory in mice. Neurobiol Learn Mem. 119:10–17. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Fan YY, Ye GH, Lin KZ, Yu LS, Wu SZ, Dong MW, Han JG, Feng XP and Li XB: Time-dependent expression and distribution of Egr-1 during skeletal muscle wound healing in rats. J Mol Histol. 44:75–81. 2013.PubMed/NCBI

19 

Klenke S, Rump K, Buschkamp K, Engler A, Peters J, Siffert W and Frey UH: Characterization of the PLCB1 promoter and regulation by early growth response transcription factor EGR-1. Eur J Pharmacol. 742:8–14. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Bhattacharyya S, Fang F, Tourtellotte W and Varga J: Egr-1: New conductor for the tissue repair orchestra directs harmony (regeneration) or cacophony (fibrosis). J Pathol. 229:286–297. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Witham EA, Meadows JD, Hoffmann HM, Shojaei S, Coss D, Kauffman AS and Mellon PL: Kisspeptin regulates gonadotropin genes via immediate early gene induction in pituitary gonadotropes. Mol Endocrinol. 27:1283–1294. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Sysol JR, Natarajan V and Machado RF: PDGF induces SphK1 expression via Egr-1 to promote pulmonary artery smooth muscle cell proliferation. Am J Physiol Cell Physiol. 310:C983–C992. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Liu QF, Yu HW, You L, Liu MX, Li KY and Tao GZ: Apelin-13-induced proliferation and migration induced of rat vascular smooth muscle cells is mediated by the upregulation of Egr-1. Biochem Biophys Res Commun. 439:235–240. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Fang F, Shangguan AJ, Kelly K, Wei J, Gruner K, Ye B, Wang W, Bhattacharyya S, Hinchcliff ME, Tourtellotte WG and Varga J: Early growth response 3 (Egr-3) is induced by transforming growth factor-β and regulates fibrogenic responses. Am J Pathol. 183:1197–1208. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Kim K, Jutooru I, Chadalapaka G, Johnson G, Frank J, Burghardt R, Kim S and Safe S: HOTAIR is a negative prognostic factor and exhibits pro-oncogenic activity in pancreatic cancer. Oncogene. 32:1616–1625. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Kim JH, Choi DS, Lee OH, Oh SH, Lippman SM and Lee HY: Antiangiogenic antitumor activities of IGFBP-3 are mediated by IGF-independent suppression of Erk1/2 activation and Egr-1-mediated transcriptional events. Blood. 118:2622–2631. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Calogero A, Arcella A, De Gregorio G, Porcellini A, Mercola D, Liu C, Lombari V, Zani M, Giannini G, Gagliardi FM, et al: The early growth response gene EGR-1 behaves as a suppressor gene that is down-regulated independent of ARF/Mdm2 but not p53 alterations in fresh human gliomas. Clin Cancer Res. 7:2788–2796. 2001.PubMed/NCBI

28 

Vockerodt M, Wei W, Nagy E, Prouzova Z, Schrader A, Kube D, Rowe M, Woodman CB and Murray PG: Suppression of the LMP2A target gene, EGR-1, protects Hodgkin's lymphoma cells from entry to the EBV lytic cycle. J Pathol. 230:399–409. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Scharnhorst V, Menke AL, Attema J, Haneveld JK, Riteco N, van Steenbrugge GJ, van der Eb AJ and Jochemsen AG: EGR-1 enhances tumor growth and modulates the effect of the Wilms' tumor 1 gene products on tumorigenicity. Oncogene. 19:791–800. 2000. View Article : Google Scholar : PubMed/NCBI

30 

Virolle T, Adamson ED, Baron V, Birle D, Mercola D, Mustelin T and de Belle I: The Egr-1 transcription factor directly activates PTEN during irradiation-induced signalling. Nat Cell Biol. 3:1124–1128. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Salmena L: PTEN: History of a tumor suppressor. Methods Mol Biol. 1388:3–11. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Jing X, Cheng W, Wang S, Li P and He L: Resveratrol induces cell cycle arrest in human gastric cancer MGC803 cells via the PTEN-regulated PI3K/Akt signaling pathway. Oncol Rep. 35:472–478. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Jolly LA, Novitskiy S, Owens P, Massoll N, Cheng N, Fang W, Moses HL and Franco AT: Fibroblast-mediated collagen remodeling within the tumor microenvironment facilitates progression of thyroid cancers driven by BrafV600E and pten loss. Cancer Res. 76:1804–1813. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Yang Z, Yuan XG, Chen J, Luo SW, Luo ZJ and Lu NH: Reduced expression of PTEN and increased PTEN phosphorylation at residue Ser380 in gastric cancer tissues: A novel mechanism of PTEN inactivation. Clin Res Hepatol Gastroenterol. 37:72–79. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Govatati S, Kodati VL, Deenadayal M, Chakravarty B, Shivaji S and Bhanoori M: Mutations in the PTEN tumor gene and risk of endometriosis: A case-control study. Hum Reprod. 29:324–336. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Fortin J, Bassi C and Mak TW: PTEN enables the development of pre-B acute lymphoblastic leukemia. Nat Med. 22:339–340. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Herranz D, Maraver A, Cañamero M, Gómez-López G, Inglada-Pérez L, Robledo M, Castelblanco E, Matias-Guiu X and Serrano M: SIRT1 promotes thyroid carcinogenesis driven by PTEN deficiency. Oncogene. 32:4052–4056. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Weng LP, Gimm O, Kum JB, Smith WM, Zhou XP, Wynford-Thomas D, Leone G and Eng C: Transient ectopic expression of PTEN in thyroid cancer cell lines induces cell cycle arrest and cell type-dependent cell death. Hum Mol Genet. 10:251–258. 2001. View Article : Google Scholar : PubMed/NCBI

39 

Curcio F, Ambesi-Impiombato FS, Perrella G and Coon HG: Long-term culture and functional characterization of follicular cells from adult normal human thyroids. Proc Natl Acad Sci USA. 91:9004–9008. 1994. View Article : Google Scholar : PubMed/NCBI

40 

Welsh JB, Sapinoso LM, Su AI, Kern SG, Wang-Rodriguez J, Moskaluk CA, Frierson HF Jr and Hampton GM: Analysis of gene expression identifies candidate markers and pharmacological targets in prostate cancer. Cancer Res. 61:5974–5978. 2001.PubMed/NCBI

41 

Jankovic B, Le KT and Hershman JM: Clinical review: Hashimoto's thyroiditis and papillary thyroid carcinoma: Is there a correlation? J Clin Endocrinol Metab. 98:474–482. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Wozniak MA, Cheng CQ, Shen CJ, Gao L, Olarerin-George AO, Won KJ, Hogenesch JB and Chen CS: Adhesion regulates MAP kinase/ternary complex factor exchange to control a proliferative transcriptional switch. Curr Biol. 22:2017–2026. 2012. View Article : Google Scholar : PubMed/NCBI

43 

Doma E, Rupp C, Varga A, Kern F, Riegler B and Baccarini M: Skin tumorigenesis stimulated by Raf inhibitors relies upon Raf functions that are dependent and independent of ERK. Cancer Res. 73:6926–6937. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Kawahara T, Aljarah AK, Shareef HK, Inoue S, Ide H, Patterson JD, Kashiwagi E, Han B, Li Y, Zheng Y and Miyamoto H: Silodosin inhibits prostate cancer cell growth via ELK1 inactivation and enhances the cytotoxic activity of gemcitabine. Prostate. 76:744–756. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Chakraborty T, Asok A, Stanton ME and Rosen JB: Variants of contextual fear conditioning induce differential patterns of Egr-1 activity within the young adult prefrontal cortex. Behav Brain Res. 302:122–130. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Peralta-Zaragoza O, Deas J, Meneses-Acosta A, De la O-Gómez F, Fernández-Tilapa G, Gómez-Cerón C, Benítez-Boijseauneau O, Burguete-García A, Torres-Poveda K, Bermúdez-Morales VH, et al: Relevance of miR-21 in regulation of tumor suppressor gene PTEN in human cervical cancer cells. BMC Cancer. 16:2152016. View Article : Google Scholar : PubMed/NCBI

47 

Yang SM, Huang C, Li XF, Yu MZ, He Y and Li J: miR-21 confers cisplatin resistance in gastric cancer cells by regulating PTEN. Toxicology. 306:162–168. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Soubani O, Ali AS, Logna F, Ali S, Philip PA and Sarkar FH: Re-expression of miR-200 by novel approaches regulates the expression of PTEN and MT1-MMP in pancreatic cancer. Carcinogenesis. 33:1563–1571. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Wu Z, He B, He J and Mao X: Upregulation of miR-153 promotes cell proliferation via downregulation of the PTEN tumor suppressor gene in human prostate cancer. Prostate. 73:596–604. 2013. View Article : Google Scholar : PubMed/NCBI

50 

Guigon CJ, Zhao L, Willingham MC and Cheng SY: PTEN deficiency accelerates tumour progression in a mouse model of thyroid cancer. Oncogene. 28:509–517. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2018
Volume 40 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kong Y, Yin J, Fu Y, Chen Y, Zhou Y and Geng X: Suppression of Elk1 inhibits thyroid cancer progression by mediating PTEN expression. Oncol Rep 40: 1769-1776, 2018
APA
Kong, Y., Yin, J., Fu, Y., Chen, Y., Zhou, Y., & Geng, X. (2018). Suppression of Elk1 inhibits thyroid cancer progression by mediating PTEN expression. Oncology Reports, 40, 1769-1776. https://doi.org/10.3892/or.2018.6554
MLA
Kong, Y., Yin, J., Fu, Y., Chen, Y., Zhou, Y., Geng, X."Suppression of Elk1 inhibits thyroid cancer progression by mediating PTEN expression". Oncology Reports 40.3 (2018): 1769-1776.
Chicago
Kong, Y., Yin, J., Fu, Y., Chen, Y., Zhou, Y., Geng, X."Suppression of Elk1 inhibits thyroid cancer progression by mediating PTEN expression". Oncology Reports 40, no. 3 (2018): 1769-1776. https://doi.org/10.3892/or.2018.6554