Increased expression and activity of p75NTR are crucial events in azacitidine-induced cell death in prostate cancer

  • Authors:
    • Giovanni Luca Gravina
    • Francesco Marampon
    • Patrizia Sanità
    • Andrea Mancini
    • Alessandro Colapietro
    • Luca Scarsella
    • Ana Jitariuc
    • Leda Biordi
    • Corrado Ficorella
    • Claudio Festuccia
  • View Affiliations

  • Published online on: May 23, 2016     https://doi.org/10.3892/or.2016.4832
  • Pages: 125-130
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The high affinity nerve growth factor (NGF) NGF receptor, p75NTR, is a member of the tumor necrosis factor (TNF) receptor superfamily that shares a conserved intracellular death domain capable of inducing apoptosis and suppressing growth in prostate epithelial cells. Expression of this receptor is lost as prostate cancer progresses and is minimal in established prostate cancer cell lines. We aimed to verify the role of p75NTR in the azacitidine-mediated antitumor effects on 22Rv1 and PC3 androgen-independent prostate cancer cells. In the present study, we reported that the antiproliferative and pro-apoptotic effects of 5-azacytidine (azacitidine) were more marked in the presence of physiological concentrations of NGF and were reduced when a blocking p75NTR antibody or the selective p75NTR inhibitor, Ro 08-2750, were used. Azacitidine increased the expression of p75NTR without interfering with the expression of the low affinity NGF receptor TrkA and induced caspase 9-dependent caspase 3 activity. Taken together, our results suggest that the NGF network could be a candidate for future pharmacological manipulation in aggressive prostate cancer.

Introduction

Prostate cancer (PCa) is one of the most common malignancies in men (1). This tumor is androgen-dependent and is treated using surgery, radiotherapy and hormone therapy (2). However, PCa frequently progresses into a hormone-independent, highly aggressive and invasive disease (3) presenting with multiple drug resistance. This neoplasia is also the most common malignancy in which the chromosome 17q21 is segregated (46) suggesting that genes located in the immediate vicinity of 17q21 can be important in PCa development/progression (5). p75NTR is a 75-kDa glycoprotein receptor that belong to the tumor necrosis factor (TNF) receptor superfamily and binds mainly to nerve growth factor (NGF) and has structural and sequential similarity to the TNF receptor (7). Notably, the human p75NTR gene locus has been mapped distal to 17q21–q22 (6). The prostate gland is one of the most abundant sources of NGF controlling cell proliferation and apoptosis (8,9). However, p75NTR is progressively lost during the progression of PCa (1012). Loss of expression of p75NTR protein is correlated with increased Gleason's score of organ-confined pathological prostate tissues (1214), and is completely absent in prostate tumor cells derived from metastases (11). The re-expression of p75NTR was previously shown in PCa to retard cell cycle progression by inducing accumulation of cells in the G1 phase with a concomitant reduction in cells in the S phase thus inducing apoptosis (15) and reverting androgen-independent growth (16). p75NTR can be upregulated by prolonged treatment with NGF (17) or GnRH treatment (18) indicating epigenetic mechanisms of protein expression.

DNA hypermethylation plays an important role in the downregulation of genes important for cell death in PCa. Demethylating agents have been shown to prevent tumorigenesis and delay androgen-independent disease (19,20) in a TRAMP mouse model of PCa as well as to revert androgen independence (19,21,22) and chemoresistance in prostate tumor cells (23).

In the present study, we demonstrated that azacitidine treatment induced a dose-dependent increase in p75NTR expression. This was in agreement with a recent report showing that azacitidine and estrogen treatment induced p75NTR (24) in 22rv1 cells. Blocking NGF antibodies or specific p75NTR inhibitors reverted this effect. Taken together, our results suggest that the NGF network could be a candidate for future pharmacological manipulation in PCa therapy.

Materials and methods

Reagents

All the materials for the tissue culture were purchased from EuroClone S.p.A. (Milan, Italy). Antibodies: Bcl-2 (sc-509), Bax (N20) (sc-493), Bad (C20) (sc-943), p-Bad ser112 (sc-7998), Bcl-xL (H2) (sc-8392), TrkA (C20) (sc-20537), p-TrkA (E6) (sc-8058), p75NTR (H92) (sc-5634), TRADD (A-5) (sc46653), RIP (H207) (sc 7881), TRAF2 (D-3) (sc-136997), DR4 (C20) (sc-6823) and DR5 (N19) (sc-7192) were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). DNMT1, DNMAT3a and DNMT3b antibodies were purchased from BioCarta LLC (San Diego, CA, USA). Plasticware was obtained from Nunc (Roskilde, Denmark). Azacitidine (Vidaza®) was obtained from Celgene Corporation (Summit, NJ, USA). The p38 MAPK inhibitor SB202190 and ibuprofen were purchased from Sigma-Aldrich Italy (Milan, Italy). Ro 08-2750, a selective p75NTR inhibitor, was purchased from Tocris Bioscience (Bristol, UK).

Cell lines

Two aggressive PCa models (PC3 and 22rv1 cell lines) were obtained from the American Tissue Culture Collection (ATCC; Rockville, MD, USA) and DSMZ (Braunschweig, Germany), respectively, and were grown as recommended.

Growth assays

Cells were seeded at a density of 2×104 cells/ml into 24-well plates. Cells were left to attach and grow in 5% FCS and Dulbecco's modified Eagle's medium (DMEM). Next, the cells were cultured under appropriate experimental conditions. Morphological controls were performed every day with an inverted phase-contrast photomicroscope (Nikon Diaphot, Tokyo, Japan) before cell trypsinization and counting by the NucleoCounter™ NC-100 automated cell counter system (Chemotec, Gydevang, Denmark). The effect on cell proliferation was measured by taking the mean cell number with respect to the controls over time for the different treatment groups.

Cell cycle and apoptosis analysis

Adherent cells were trypsinized, pooled with the culture supernatant containing the apoptotic cells already detached from the dish and centrifuged. Cells (1×106) were washed in phosphate-buffered saline (PBS) and fixed for 30 min by the addition of 1 ml of 70% ethanol. Apoptosis and cell cycle analyses were performed using the Tali Cell Cycle kit and the Tali apoptosis kit, Annexin V-Alexa Fluor 488 and propidium iodide-based staining (Life Technologies Italia, Monza, Italy). Apoptosis was further confirmed by cytofluorimetric analysis following the manufacturer's instructions. Stained cells were then measured on a Tali Image-Based Cytometer. Caspase 8 and −9 activities were evaluated using enzymatic assays from GenScript (Piscataway, NJ, USA).

Western blot analysis

Cells (1×106) were washed with cold PBS and immediately lysed with 100 μl of lysis buffer (50 mM HEPES, pH 7.5, 150 mM NaCl, 10% glycerol, 1% Triton x-100, 1 mM EDTA, 1 mM EGTA, 50 mM NaF, 1 Mm sodium orthovanadate, 30 mM p-nitrophenyl phosphate, 10 mM sodium pyrophosphate, 1 mM phenylmethylsulfonyl fluoride, 10 μg/ml aprotinin and 10 μg/ml leupeptin). Cell lysates were electrophoresed in 7% SDS-PAGE, and separated proteins were transferred to a nitrocellulose membrane and probed with the appropriate antibodies as indicated.

In vivo treatments

Male CD1 nude mice (Charles River, Milan, Italy) were maintained under the guidelines established by our institution (university of L'Aquila, Medical School and Science and Technology School Board Regulations, complying with the Italian government regulation no. 116, January 27, 1992, for the use of laboratory animals). All mice received s.c. flank injections of 1×106 PC3 and 22v1 cells. Tumor growth was assessed by a bi-weekly measurement of tumor diameters with a Vernier calliper (length × width). Tumor weight was calculated as previously indicated (22). Treatments were started when tumor volumes reached ~80 mm3 (day 0) and were stopped after 28 days. Mice were grouped as follows: group 1, 10 mice received intraperitoneal (i.p.) injections of 100 l PBS for a consecutive 7 days; group 2: 10 mice received i.p. injections of 100 l of azacitidine (Aza-CR) (0.8 mg/kg) for a consecutive 7 days. Tumors were fixed in paraformaldehyde for hystochemical and immunohystochemical analyses. Indirect immunoperoxidase staining of tumor xenografts was performed on paraffin-embedded 4-μm tissue sections. Briefly, the sections were incubated with the primary antibodies overnight at 4°C. Next avidin-biotin assays were carried out using the Vectastain Elite kit (Vector Laboratories, Burlingame, CA, USA). Mayer's hematoxylin was used as a nuclear counterstain.

Statistical analysis

Continuous data are presented as the mean ± standard deviation (SD), and were compared using an umpired Student's t-test.

Results

Azacitidine restores the expression of p75NTR in PC3 and 22rv1 cell models

In the present study, we demonstrated that the antiproliferative and pro-apoptotic effects induced by azacitidine at non-toxic concentrations (ranging between 0.1 and 0.5 μM) were enhanced in the presence of 10 ng/ml NGF. We observed that NGF-induced growth inhibition was significantly higher when compared to data observed following treatment with the demethylating agent alone both in the PC3 (Fig. 1A) and 22rv1 cells (Fig. 1B). The addition of NGF triggered a significant and early apoptosis in both cell lines (Fig. 1C and D). Next, we analyzed the molecular arrangement induced by treatments using western blotting. We observed that expression of the high affinity p75NTR, but not expression of the low affinity receptor (TrkA) was increased after in vitro administration of azacitidine in a time-dependent manner in the PC3 and 22rv1 cells (Fig. 2A and B). As a positive control we used the androgen receptor (AR) which was widely demonstrated to be induced after azacitidine treatment (19,25). Western blot analyses were confirmed by immunocytochemical analyses (Fig. 2C) in the aggressive 22rv1 cell line. The same tumor cell models were examined for expression levels of downstream components proximal to p75NTR (TRADD, RIP, DR4, DR5 and TRAF2) in both the PC3 (Fig. 2D) and 22rv1 cells (Fig. 2E). Previously, we demonstrated that an intraperitoneal administration of azacitidine (Vidaza®, 0.8 mg/kg for 7 consecutive days) in nude male mice subcutaneously inoculated with 22rv1 and PC3 cells was able to reduce tumor growth both in the PC3 and 22rv1 xenograft models (22). In the present study, we demonstrated that reduction in the tumor mass after Aza-CR treatment was associated with p75NTR re-expression in vivo (Fig. 2F). Cell death was dependent on activation of the caspase 9 pathway which we also verified in vivo by analyzing FasL and TRAIL protein expression in the control and Aza-CR-treated tumors.

Azacitidine induces p75NTR-mediated cell death

It has been previously demonstrated that p38 MAPK activation, associated with ibuprofen treatment, is related to induction of p75NTR in PCa cells (26). Therefore, considering that the antitumor effects of azacitidine are associated with increased p38 MAPK activity necessary for the induction of cell cycle arrest in the G2 phase (27,28) and apoptosis (29), we verified whether the p38 MAPK inhibitor, SB202190, was able to prevent the induction of p75NTR using as a positive control, treatment with ibuprofen (1 μM). In agreement with the literature data, we demonstrated that: i) NGF reduced ERK activation, but not p38 MAPK (Fig. 3A), and ii) ibuprofen induced p75NTR (Fig. 3B) whereas p38 MAPK inhibition was able to reduce p75NTR expression (Fig. 3B). Similar results were obtained using P38 MAPK siRNA (data not shown) indicating that p75NTR expression was under p38 MAPK-dependent epigenetic control. Thus, we analyzed the role of p75NTR in the azacitidine-induced cell death. For this aim we used a blocking antibody for p75NTR as well as the non-peptide antagonist of NGF, Ro 08-2750 [able to bind to the NGF dimer and inhibit selectively p75NTR at submicromolar concentrations whereas both p75NTR and TrkA receptors were blocked at >5 μM (29)]. Both anti-NGF agents significantly increased the cell viability (Fig. 3C) and apoptosis (Fig. 3D) induced by co-treatment with azacitidine and NGF. Conversely the p75NTR induction by Aza-CR treatment was partial after SB202190 (data not shown). Increased p75NTR observed after azacitidine treatment was related to increased expression of Bax and PARP cleavage as well as to reduced levels of Bcl2, XIAP and survivin (Fig. 4A) with caspase 8-dependent caspase 3 activation (Fig. 4B and C) in agreement with our in vivo effects (30).

Discussion

The low-affinity neurotrophin receptor p75NTR, a member of the tumor necrosis factor (TNF) receptor superfamily, is expressed in prostate gland (611) and has been implicated in promoting cell apoptosis and death through a conserved intracellular death domain (3134). In the present study, we demonstrated that azacitidine was able to restore the expression of p75NTR in p75NTR-negative and aggressive prostate cancer (PCa) cell lines. This was dependent on increased p38 MAPK activation and the inhibition of DNMT actvitiy (35,36), and this was in agreement with a recent study in which estrogen and azacitidine modulated the increased expression of p75NTR and apoptosis in 22rv1 cells (24). Previously, it has been shown that treatment with nonsteroidal anti-inflammatory drugs induces p75NTR expression leading to p75NTR-mediated decreased survival through a p38 MAPK-mediated mechanism (2628). p38 MAPK could be a complementary mechanism by which prostate tumor cells re-express p75NTR. The re-expression of p75NTR tumor suppressor activity is associated with reduced tumor cell growth and activation of caspase 9-dependent caspase 3 activation. Since the re-expression of p75NTR does not alter levels of TrkA, these changes in cell cycle progression can be directly attributed to the changes in levels of the p75NTR protein. These effects were increased in the presence of NGF and indicate a TRAIL-mediated crosstalk with TRAILR (DR4) (37,38) or FAS:FASL signaling (39,40). In addition, cell cycle arrest, differentiation and apoptosis after p75NTR activation could involve further intracellular signals. It has been demonstrated that anti-inflammatory compounds modulate DNMT expression/activity (41,42) via phospholipase A2, able for example to induce cell arrest and differentiation in rat NK cells (43), and PPARγ activation (43) but not PPARβ, involved in differentiation of neuronal cells (44,45). In the absence of a ligand, p75NTR-dependent cell cycle arrest was accompanied by a rank-order increase in the apoptosis of PCa cells. NGF increased the apoptosis, via mitochondrial activation of a caspase cascade, induced by p75NTR re-expression. Taken together, it seems clear that p75NTR expression selectively alters specific cell cycle regulatory molecules that retard progression. In conclusion, the present study showed that p75NTR retards cell cycle progression and induces NGF-mediated apoptosis and suggests that the NGF network could be a candidate for future pharmacological manipulation for aggressive PCa.

References

1 

Malvezzi M, Bertuccio P, Levi F, La Vecchia C and Negri E: European cancer mortality predictions for the year 2014. Ann Oncol. 25:1650–1656. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Thoreson GR, Gayed BA, Chung PH and Raj GV: Emerging therapies in castration resistant prostate cancer. Can J Urol. 21(Supp 1): S98–S105. 2014.

3 

Matsumoto K, Hagiwara M, Tanaka N, Hayakawa N, Ishida M, Ninomiya A, Nakajima Y and Nakamura S: Survival following primary androgen deprivation therapy for localized intermediate- or high-risk prostate cancer: Comparison with the life expectancy of the age-matched normal population. Med Oncol. 31:9792014. View Article : Google Scholar : PubMed/NCBI

4 

Rettig WJ, Thomson TM, Spengler BA, Biedler JL and Old LJ: Assignment of human nerve growth factor receptor gene to chromosome 17 and regulation of receptor expression in somatic cell hybrids. Somat Cell Mol Genet. 12:441–447. 1986. View Article : Google Scholar : PubMed/NCBI

5 

Haiman CA, Chen GK, Blot WJ, Strom SS, Berndt SI, Kittles RA, Rybicki BA, Isaacs WB, Ingles SA, Stanford JL, et al: Genome-wide association study of prostate cancer in men of African ancestry identifies a susceptibility locus at 17q21. Nat Genet. 43:570–573. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Watson SK, Woolcock BW, Fee JN, Bainbridge TC, Webber D, Kinahan TJ, Lam WL and Vielkind JR: Minimum altered regions in early prostate cancer progression identified by high resolution whole genome tiling path BAC array comparative hybridization. Prostate. 69:961–975. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Johnson D, Lanahan A, Buck CR, Sehgal A, Morgan C, Mercer E, Bothwell M and Chao M: Expression and structure of the human NGF receptor. Cell. 47:545–554. 1986. View Article : Google Scholar : PubMed/NCBI

8 

Sigala S, Bodei S, Missale C, Zani D, Simeone C, Cunico SC and Spano PF: Gene expression profile of prostate cancer cell lines: Effect of nerve growth factor treatment. Mol Cell Endocrinol. 284:11–20. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Rende M, Rambotti MG, Stabile AM, Pistilli A, Montagnoli C, Chiarelli MT and Mearini E: Novel localization of low affinity NGF receptor (p75) in the stroma of prostate cancer and possible implication in neoplastic invasion: An immunohistochemical and ultracytochemical study. Prostate. 70:555–561. 2010.

10 

Arrighi N, Bodei S, Zani D, Simeone C, Cunico SC, Missale C, Spano P and Sigala S: Nerve growth factor signaling in prostate health and disease. Growth Factors. 28:191–201. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Festuccia C, Gravina GL, Muzi P, Pomante R, Ventura L, Ricevuto E, Vicentini C and Bologna M: In vitro and in vivo effects of bicalutamide on the expression of TrkA and P75 neurotrophin receptors in prostate carcinoma. Prostate. 67:1255–1264. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Bassili M, Birman E, Schor NF and Saragovi HU: Differential roles of Trk and p75 neurotrophin receptors in tumorigenesis and chemoresistance ex vivo and in vivo. Cancer Chemother Pharmacol. 65:1047–1056. 2010. View Article : Google Scholar

13 

Perez M, Regan T, Pflug B, Lynch J and Djakiew D: Loss of low-affinity nerve growth factor receptor during malignant transformation of the human prostate. Prostate. 30:274–279. 1997. View Article : Google Scholar : PubMed/NCBI

14 

Fromont G, Godet J, Pires C, Yacoub M, Dore B and Irani J: Biological significance of perineural invasion (PNI) in prostate cancer. Prostate. 72:542–548. 2012. View Article : Google Scholar

15 

Khwaja F, Tabassum A, Allen J and Djakiew D: The p75NTR tumor suppressor induces cell cycle arrest facilitating caspase mediated apoptosis in prostate tumor cells. Biochem Biophys Res Commun. 341:1184–1192. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Molloy NH, Read DE and Gorman AM: Nerve growth factor in cancer cell death and survival. Cancers. 3:510–530. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Sigala S, Tognazzi N, Rizzetti MC, Faraoni I, Missale C, Bonmassar E and Spano P: Nerve growth factor induces the re-expression of functional androgen receptors and p75NGFR in the androgen-insensitive prostate cancer cell line DU145. Eur J Endocrinol. 147:407–415. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Sánchez C, Clementi M, Benitez D, Contreras H, Huidobro C and Castellón E: Effect of GnRH analogs on the expression of TrkA and p75 neurotrophin receptors in primary cell cultures from human prostate adenocarcinoma. Prostate. 65:195–202. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Gravina GL, Marampon F, Piccolella M, Motta M, Ventura L, Pomante R, Popov VM, Zani BM, Pestell RG, Tombolini V, et al: Hormonal therapy promotes hormone-resistant phenotype by increasing DNMT activity and expression in prostate cancer models. Endocrinology. 152:4550–4561. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Gravina GL, Marampon F, Di Staso M, Bonfili P, Vitturini A, Jannini EA, Pestell RG, Tombolini V and Festuccia C: 5-Azacitidine restores and amplifies the bicalutamide response on preclinical models of androgen receptor expressing or deficient prostate tumors. Prostate. 70:1166–1178. 2010. View Article : Google Scholar : PubMed/NCBI

21 

McCabe MT, Low JA, Daignault S, Imperiale MJ, Wojno KJ and Day ML: Inhibition of DNA methyltransferase activity prevents tumorigenesis in a mouse model of prostate cancer. Cancer Res. 66:385–392. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Zorn CS, Wojno KJ, McCabe MT, Kuefer R, Gschwend JE and Day ML: 5-aza-2′-deoxycytidine delays androgen-independent disease and improves survival in the transgenic adenocarcinoma of the mouse prostate mouse model of prostate cancer. Clin Cancer Res. 13:2136–2143. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Festuccia C, Gravina GL, D'Alessandro AM, Muzi P, Millimaggi D, Dolo V, Ricevuto E, Vicentini C and Bologna M: Azacitidine improves antitumor effects of docetaxel and cisplatin in aggressive prostate cancer models. Endocr Relat Cancer. 16:401–413. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Yu JD, Yang K, Mao QQ, Kong DB, Zheng XY and Xie LP: Estrogen in combination with 5-azacitidine up-regulates p75NTR expression and induces apoptosis in 22Rv1 prostate cancer cells. Mol Med Rep. 2:831–836. 2009.PubMed/NCBI

25 

Gravina GL, Festuccia C, Millimaggi D, Dolo V, Tombolini V, de Vito M, Vicentini C and Bologna M: Chronic azacitidine treatment results in differentiating effects, sensitizes against bicalutamide in androgen-independent prostate cancer cells. Prostate. 68:793–801. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Khwaja F, Allen J, Lynch J, Andrews P and Djakiew D: Ibuprofen inhibits survival of bladder cancer cells by induced expression of the p75NTR tumor suppressor protein. Cancer Res. 64:6207–6213. 2004. View Article : Google Scholar : PubMed/NCBI

27 

Uzgare AR, Kaplan PJ and Greenberg NM: Differential expression and/or activation of P38 MAPK, erk1/2, and jnk during the initiation and progression of prostate cancer. Prostate. 55:128–139. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Cho SD, Li G, Hu H, Jiang C, Kang KS, Lee YS, Kim SH and Lu J: Involvement of c-Jun N-terminal kinase in G2/M arrest and caspase-mediated apoptosis induced by sulforaphane in DU145 prostate cancer cells. Nutr Cancer. 52:213–224. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Eibl JK, Strasser BC and Ross GM: Identification of novel pyrazoloquinazolinecarboxilate analogues to inhibit nerve growth factor in vitro. Eur J Pharmacol. 708:30–37. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Festuccia C, Gravina GL, D'Alessandro AM, Millimaggi D, Di Rocco C, Dolo V, Ricevuto E, Vicentini C and Bologna M: Downmodulation of dimethyl transferase activity enhances tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis in prostate cancer cells. Int J Oncol. 33:381–388. 2008.PubMed/NCBI

31 

He W, Wang Q, Xu J, Xu X, Padilla MT, Ren G, Gou X and Lin Y: Attenuation of TNFSF10/TRAIL-induced apoptosis by an autophagic survival pathway involving TRAF2- and RIPK1/RIP1-mediated MAPK8/JNK activation. Autophagy. 8:1811–1821. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Labsch S, Liu L, Bauer N, Zhang Y, Aleksandrowicz E, Gladkich J, Schönsiegel F and Herr I: Sulforaphane and TRAIL induce a synergistic elimination of advanced prostate cancer stem-like cells. Int J oncol. 44:1470–1480. 2014.PubMed/NCBI

33 

Szliszka E, Zydowicz G, Mizgala E and Krol W: Artepillin C (3,5-diprenyl-4-hydroxycinnamic acid) sensitizes LNCaP prostate cancer cells to TRAIL-induced apoptosis. Int J Oncol. 41:818–828. 2012.PubMed/NCBI

34 

Jung YH, Lim EJ, Heo J, Kwon TK and Kim YH: Tunicamycin sensitizes human prostate cells to TRAIL-induced apoptosis by upregulation of TRAIL receptors and downregulation of cIAP2. Int J Oncol. 40:1941–1948. 2012.PubMed/NCBI

35 

Gober MD, Smith CC, Ueda K, Toretsky JA and Aurelian L: Forced expression of the H11 heat shock protein can be regulated by DNA methylation and trigger apoptosis in human cells. J Biol Chem. 278:37600–37609. 2003. View Article : Google Scholar : PubMed/NCBI

36 

Monks TJ, Xie R, Tikoo K and Lau SS: Ros-induced histone modifications and their role in cell survival and cell death. Drug Metab Rev. 38:755–767. 2006. View Article : Google Scholar : PubMed/NCBI

37 

Wang D, Lu J and Tindall DJ: Androgens regulate TRAIL-induced cell death in prostate cancer cells via multiple mechanisms. Cancer Lett. 335:136–144. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Shankar S and Srivastava RK: Enhancement of therapeutic potential of TRAIL by cancer chemotherapy and irradiation: Mechanisms and clinical implications. Drug Resist Updat. 7:139–156. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Mitsiades N, Poulaki V, Tseleni-Balafouta S, Koutras DA and Stamenkovic I: Thyroid carcinoma cells are resistant to FAS-mediated apoptosis but sensitive to tumor necrosis factor-related apoptosis-inducing ligand. Cancer Res. 60:4122–4129. 2000.PubMed/NCBI

40 

Mimouni-Rongy M, White JH, Weinstein DE, Desbarats J and Almazan G: Fas ligand acts as a counter-receptor in Schwann cells and induces the secretion of bioactive nerve growth factor. J Neuroimmunol. 230:17–25. 2011. View Article : Google Scholar

41 

Yin L and Chung WO: Epigenetic regulation of human β-defensin 2 and CC chemokine ligand 20 expression in gingival epithelial cells in response to oral bacteria. Mucosal Immunol. 4:409–419. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Javierre BM and Richardson B: A new epigenetic challenge: Systemic lupus erythematosus. Adv Exp Med Biol. 711:117–136. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Cifone MG, Botti D, Festuccia C, Napolitano T, del Grosso E, Cavallo G, Chessa MA and Santoni A: Involvement of phospholipase A2 activation and arachidonic acid metabolism in the cytotoxic functions of rat NK cells. Cell Immunol. 148:247–258. 1993. View Article : Google Scholar : PubMed/NCBI

44 

Benedetti E, Galzio R, Cinque B, Biordi L, D'Amico MA, D'Angelo B, Laurenti G, Ricci A, Festuccia C, Cifone MG, et al: Biomolecular characterization of human glioblastoma cells in primary cultures: Differentiating and antiangiogenic effects of natural and synthetic PPARgamma agonists. J Cell Physiol. 217:93–102. 2008. View Article : Google Scholar : PubMed/NCBI

45 

Di Loreto S, D'Angelo B, D'Amico MA, Benedetti E, Cristiano L, Cinque B, Cifone MG, Cerù MP, Festuccia C and Cimini A: PPARbeta agonists trigger neuronal differentiation in the human neuroblastoma cell line SH-SY5Y. J Cell Physiol. 211:837–847. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2016
Volume 36 Issue 1

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
Gravina GL, Marampon F, Sanità P, Mancini A, Colapietro A, Scarsella L, Jitariuc A, Biordi L, Ficorella C, Festuccia C, Festuccia C, et al: Increased expression and activity of p75NTR are crucial events in azacitidine-induced cell death in prostate cancer. Oncol Rep 36: 125-130, 2016
APA
Gravina, G.L., Marampon, F., Sanità, P., Mancini, A., Colapietro, A., Scarsella, L. ... Festuccia, C. (2016). Increased expression and activity of p75NTR are crucial events in azacitidine-induced cell death in prostate cancer. Oncology Reports, 36, 125-130. https://doi.org/10.3892/or.2016.4832
MLA
Gravina, G. L., Marampon, F., Sanità, P., Mancini, A., Colapietro, A., Scarsella, L., Jitariuc, A., Biordi, L., Ficorella, C., Festuccia, C."Increased expression and activity of p75NTR are crucial events in azacitidine-induced cell death in prostate cancer". Oncology Reports 36.1 (2016): 125-130.
Chicago
Gravina, G. L., Marampon, F., Sanità, P., Mancini, A., Colapietro, A., Scarsella, L., Jitariuc, A., Biordi, L., Ficorella, C., Festuccia, C."Increased expression and activity of p75NTR are crucial events in azacitidine-induced cell death in prostate cancer". Oncology Reports 36, no. 1 (2016): 125-130. https://doi.org/10.3892/or.2016.4832