Tumstatin induces apoptosis and stimulates phosphorylation of p65NF-κB in human osteoblastic osteosarcoma Saos-2 cells

  • Authors:
    • Yang Wang
    • Ruo-Feng Yin
    • Jia-Song Teng
  • View Affiliations

  • Published online on: April 20, 2016     https://doi.org/10.3892/or.2016.4762
  • Pages: 3403-3408
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study was aimed to investigate the effect of tumstatin on inhibition of proliferation and induction of apoptosis in Saos-2 human osteosarcoma cells and to understand the mechanism involved. Inhibition of cell proliferation was analyzed by MTT assay and induction of apoptosis through nuclear fragmentation assay. Viability of Saos-2 cells was reduced to 19% on treatment with 25 µM concentration of tumstatin after 48 h. Presence of characteristic apoptotic nuclei, rounded cell shape and shrunken size were caused by tumstatin treatment at 25 µM concentration. The level of mRNA corresponding to PTEN, FasR and FasL was increased significantly in tumstatin treated Saos-2 cells compared to untreated control. Investigation of the mechanism revealed NF-κB activation by phosphorylation on serine 536. The activated NF-κB was translocated into the nucleus from the cytoplasm on treatment with tumstatin. Degradation of the IκBα by tumstatin was found to be much slower compared to that induced by treatment with TNF-α. Thus, tumstatin inhibits proliferation and induces apoptosis in Saos-2 cells through activation of NF-κB and its translocation to the nucleus. Therefore, tumstatin can play an important role in the treatment of osteosarcoma.

Introduction

Osteosarcoma alone comprises more than 50% of the primary bone tumors and is detected mostly in children. In Japan every year more than 100 cases of osteosarcoma are detected. In osteosarcoma various changes are observed in the chromosomes and the gene mutations are also extensive resulting in the development of complex tumors (1,2). Numerous studies have been performed to understand the mechanism underlying the development of osteosarcoma (16). Despite advancement in the techniques including, wide tumor excision and aggressive chemotherapy the prognosis in osteosarcoma patients is very poor (7,8). Thus, the screening of molecules for effective treatment of osteosarcoma is desired (9).

Apoptosis is the controlled and programmed death of cells and is vital for the treatment of carcinoma and removal of unwanted cells from the body (10,11). Regulation of various factors involved in the process of apoptosis is mediated through phosphorylation (12). One more factor involved in the regulation of apoptosis in various types of carcinoma cells is the nuclear factor-kappa B (NF-κB) (1317). Activation of IκBα followed by its degradation induces translocation of NF-κB into the cell nucleus from cytoplasm where it influences several genes.

Tumstatin, is a well-known anti-angiogenic agent possessing promising antitumor potential (18,19). Various reports have demonstrated that tumstatin treatment prevents tumor proliferation in glioma and melanoma tumor models (2022). In addition, tumstatin exhibits a strong tendency to suppress proliferation and angiogenesis of tumor in the head and neck carcinoma models. Furthermore, in oral squamous cancer model treatment with tumstatin inhibited metastasis of carcinoma cells to the lymph nodes (23). The present study was aimed to investigate the effect of tumstatin on tendency of proliferation and apoptosis in Saos-2 osteosarcoma cells. The results demonstrated that tumstatin treatment in Saos-2 cells inhibited cell proliferation and induced apoptosis through activation of p65NF-κB.

Materials and methods

Chemicals and reagents

Tumstatin and TNF-α were purchased from Sigma-Aldrich (St. Louis, MO, USA). The stock solution of tumstatin was prepared in dimethyl sulfoxide and stores at −15°C before use in the experiment. Fetal bovine serum (FBS) and Dulbecco's modified Eagle's minimum essential medium (D-MEM) were obtained from Gibco-BRL (Gaithersburg, MD, USA).

Cell culture

Saos-2 osteosarcoma cell line and human embryonic kidney HEK293 cells were obtained from the American Type Culture Collection (ATCC; Rockville, MD, USA). The cells were cultured in D-MEM containing 10% FBS, 2 mM glutamine, 100 U/ml penicillin and 100 µg/ml streptomycin. Both cell lines were cultured in an incubator of humidified atmosphere with 5% CO2 at 37°C.

Analysis of cell proliferation

The effect of tumstatin on proliferation of Saos-2 cells was analyzed by 3-(4,5-dimeth-ylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT; Sigma-Aldrich) assay. The cells were distributed at density of 2×106 cells/well in 2 ml DMEM medium supplemented with 10% FBS in 6-well plates (Nunc A/S Plastfabrikation, Roskilde, Denmark). Following 12-h incubation, the medium was replaced with new medium containing 5, 10, 15, 20, 25 or 30 µM concentrations of tumstatin and incubated for 48 h. Then, 50 µl of MTT (5 µg/ml) solution was added to each well and incubation was continued for 2 h. To each well of the plate, 150 µl of DMSO was added and incubated for 5 min. The well optical density (OD) was recorded at 570 nm using an EL800 Universal Microplate Reader (BioTek Instruments, Inc., Winooski, VT, USA).

DNA fragmentation assay

Saos-2 cells were seeded in T-75 flasks at a density of 2×105 cells/flask and cultured for 12 h. Then, medium was replaced with new medium containing 25 µM concentration of tumstatin for 48 h. QIAamp DNA Mini kit (Qiagen) was used for the preparation of genomic DNA in accordance with the guidelines on the user manual. Electrophoresis of the DNA samples was performed on 1.8% agarose gel at 50 V for 2 h. For gel staining ethidium bromide (Sigma-Aldrich) was used, whereas visualization was achieved by ultraviolet (UV) transilluminator (Wealtech Corp., Reno, NV, USA).

DNA isolation and agarose gel electrophoresis

Saos-2 cells after incubation with tumstatin for 48 h were rinsed three times in PBS and lysed in lysis buffer [10 mM Tris-HCl buffer (pH 7.5), 10 mM EDTA and 0.5% Triton X-100). The cell lysates were centrifuges at 12,000 × g for 45 min to remove the insoluble material. The lysate was then treated with DNase and subjected to incubation at 37°C for 45 min. The lysates were treated with proteinase K for 50 min followed by addition of 2-propanol and NaCl to precipitate the DNA. DNA was suspended in TE-buffer and then electrophoresed using agarose gel. The UV transilluminator (Vilber Lourmat, Marne la Vallee, France) was used for the analysis of apoptotic changes.

Real-time reverse transcription polymerase chain reaction (RT-PCR)

The expression of mRNA in Saos-2 osteosarcoma cells was determines by using real-time RT-PCR. For this purpose, a total of 2×106 cells were cultured in 100-mm dishes containing DMEM medium and tumstatin and incubated for 48 h. The cells were then collected to extract the RNA using an RNeasy Plus Mini kit (Qiagen, Waco, TX, USA). RT-PCR analysis was carried out using SuperScript III First-Strand Synthesis SuperMix for qRT-PCR (Invitrogen, Carlsbad, CA, USA). NanoDrop 1000 (Thermo Fisher Scientific, Wilmington, DE, USA) was used for the determination of concentration of each cDNA after adjustment to 40 ng/ml using diethylpyrocarbonate (DPEC) water. FAM-labeled TaqMan probes and TaqMan Universal Master Mix (Applied Biosystems) using Chromo4 (Bio-Rad Laboratories, Cambridge, MA, USA) were employed to carry out the real-time PCR with the primers listed in Table I. The PCR sequence involved 2-min incubation at 50°C, 10-min denaturation at 50°C followed by 15 sec 50 cycles at 95°C and then 1 min at 60°C. GAPDH was used as an internal control. Analysis of the PCR products was performed on 2% agarose gel using ethidium bromide and a UV illuminator was used for the visualized.

Table I

The primers used in RT-PCR.

Table I

The primers used in RT-PCR.

PrimerForwardReverse
PTEN 5′-ACCGCCAAATTTAATTGCAG-3′ 5′-GGGTCCTGAATTGGAFFAAT-3′
FasL 5′-TCTCAGACGTTTTTCGGCTT-3′ 5′-AAGACAGTCCCCCTTGAGGT-3′
FasR 5′-CAAGGGATTGGAATTGAGGA-3′ 5′-GACAAAGCCACCCCAAGTTA-3′
GAPDH 5′-ACCACAGTCCATGCCATCAC-3′ 5′-TCCACCACCCTGTTGCTGTA-3′
Western blot analysis

Tumstatin, TNF-α-treated or untreated control Saos-2 cells were suspended in DMEM medium containing 1.5 µM aprotinin, 5 µM AEBSF, 0.01 µM leupeptin, 10 µM E-64 and phosphatase inhibitors [1 mM sodium orthovanadate (Na2VO4); 1 mM sodium molybdate (Na2MoO4) 4 mM sodium tartrate dihydrate; 2 mM imidazole were obtained from Sigma-Aldrich]. The cell plates were kept for a period of 30 min on ice prior to centrifugation at 12,000 × g for 30 min to collect the supernatant. The bicinchoninic protein assay kit (Pierce, Rockford, IL, USA) was used for the analysis of the concentration of proteins. The 30 µg protein samples were isolated using 10% SDS-PAGE gel followed by transfer to polyvinylidene difluoride membrane (Bio-Rad Laboratories, Hercules, CA, USA) using electroblotting. Following incubation for 1 h in a blocking solution for phosphorylated proteins (Blocking One-P; Nacalai Tesque, Kyoto, Japan), the membranes were washed with PBS-Tween followed by overnight incubation with primary antibodies. Then the membranes were rinsed twice for 10 min each time in PBS and 0.05% Tween-20 before incubation with horseradish peroxidase-conjugated polyclonal horse anti-rabbit (1:2,000; Cell Signaling Technology, Inc., Danvers, MA, USA) for 1 h. The blot was developed using an enhanced chemiluminescence kit (Intron Biotechnology, Inc., Seongnam, Korea).

DNA construction and transfection

HEK293 cells seeded in plastic dishes were cultured in DMEM medium supplemented with FBS until attaining 80% confluence. The cells were washed with PBS and then treated with a mixture of DNA and Opti-MEM. QuickChange II Site-Directed Mutagenesis kit (Stratagene, La Jolla, CA, USA) was used for the transfection of p65NF-κB S536A carrying alanine instead of serine at 536 position. The cells were then incubated with tumstatin for 180 min at 37°C in D-MEM containing 10% FBS.

Statistical analysis

For the analysis of the data the unpaired Student's t-test (Graph Pad Prism V.4) was used and P-values of <0.05 were considered statistically significant.

Results

Inhibition of Saos-2 cell viability by tumstatin

Saos-2 cells were treated with various concentrations (5, 10, 15, 20, 25 and 30 µM) of tumstatin for 48 h and then analyzed by MTT assay. Tumstatin treatment reduced the viability of Saos-2 cells in a concentration-dependent manner after 48 h. No significant effect was observed on the viability of Saos-2 cells treated with 5 and 10 µM concentrations of tumstatin for 48 h. However, cell viability was reduced significantly by the concentration of 15 µM with maximum inhibition at 25 µM (Fig. 1). At 25 µM concentration of tumstatin after 48 h, viability of Saos-2 cells was reduced to 19% compared to 100% in the control cultures (Fig. 1).

Tumstatin induces apoptosis in Saos-2 cells

Analysis of the apoptosis induction in Saos-2 cells using Hoechst 33342 staining revealed presence of characteristic apoptotic nuclei on treatment with 25 µM concentration of tumstatin after 48 h. The cells were seen to be rounded in shape and shrunken in size. However, no apoptotic nuclei were observed in the control Saos-2 cell cultures after 48 h (Fig. 2). Increase in the concentration of tumstatin from 15 to 25 µM enhanced the proportion of apoptotic cells significantly compared to the control cells. The DNA fragmentation pattern showed formation of ladder like structures on tumstatin treatment for 48 h (Fig. 2).

Effect of tumstatin on the expression level of PTEN, FasL and FasR mRNA in Saos-2 cells

The effect of tumstatin on the expression of PTEN, FasL and FasR mRNA following amplification of cDNA for 40 cycles revealed a significant increase after 30 min (Fig. 3). However, tumstatin exhibited no effect on the expression of mRNA corresponding to GAPDH which was constitutively expressed in Saos-2 cells (Fig. 3).

Effect of tumstatin on Iκ-Bα regulation in Saos-2 cells

The effect of tumstatin and TNF-α on the expression of IκBα in Saos-2 cells was analyzed using western blot assay. The results revealed that expression of IκBα was reduced up to 90 min by tumstatin treatment and was then increased by 180 min. However, treatment of Saos-2 cells with 10 ng/ml TNF-α as the control inhibited the expression of IκBα after 30 min (Fig. 4). The expression level of IκBα increased by 60 min and became similar to those of untreated control cells.

Effect of tumstatin on phosphorylation of NF-κB

The interaction of anti-phospho-Ser536 p65NF-κB antibody. Incubation with anti-p65NF-κB antibody also led to a strong interaction with 65-kDa band. The interaction of anti-phospho-Ser529 p65NF-κB and anti-p65NF-κB antibodies with 65-kDa band remained independent on treatment with tumstatin (Fig. 5). The interaction of anti-phospho-Ser536 p65NF-κB antibody with proteins of tumstatin treated cells showed a concentration dependent increase (Fig. 5). However, the interaction was found to be very weak in case of untreated control cells. Following removal of anti-phospho-Ser536 p65NF-κB antibody and then incubation with anti-p65NF-κB antibody, interaction was observed. Treatment of the cells with 10 ng/ml concentration of TNF-α resulted interaction of antiphospho-Ser529 p65NF-κB and anti-phospho-Ser536 antibodies with 65-kDa protein. The interaction increased after 5 min and was maximum after 15 and 60 min, respectively for antiphospho-Ser529 p65NF-κB and anti-phospho-Ser536 antibodies (Fig. 5).

Effect of tumstatin on translocation of NF-κB in Saos-2 cells

Saos-2 cells were treated with 25 µM concentration of tumstatin and 10 ng/ml concentration of TNF-α. Interaction of anti-p65NF-κB antibody and 65-kDa protein band was observed in the cytosolic fractions of both untreated and tumstatin-treated cells (Fig. 6). However, this interaction was observed at higher level in the nuclear fraction of only tumstatin-treated but not in untreated control cells. In untreated control cells none of the proteins interacted with anti-phospho-Ser536 p65NF-κB antibody (Fig. 6). In tumstatin treated cells interaction of anti-phospho-Ser536 p65NF-κB antibody with proteins was significant both in the cytosolic and nuclear fractions. The interaction of anti-phospho-Ser536 p65NF-κB antibody with the proteins of cytosolic and nuclear fractions was also observed in the TNF-α treated cells. In both tumstatin and TNF-α-treated cells interaction of anti-Eps15 antibody was found in cytosolic but not in nuclear fraction (Fig. 6). Analysis of the interaction of anti-Lamin B1 antibody with 68-kDa protein band was found in the nuclear, but not in cytosolic fraction.

In vitro dephosphorylation and phosphorylation of mutant gene products

Activation of the Ser536 of p65NF-κB by tumstatin treatment was confirmed by transfection of HEK293 cells with mutant p65NF-κB gene possessing alanine (GFP-S536A) instead of serine at 536. In both control and tumstatin treated cells transfected with GFP-S536A mutant gene, no interaction of anti-phospho-Ser536 p65NF-κB antibody was observed with any of the proteins (Fig. 7). On the contrary, interaction of anti-phospho-Ser536 p65NF-κB antibody with proteins of tumstatin treated cells increased significantly.

Discussion

The present study demonstrates the viability of inhibitory and apoptosis inducing effect of tumstatin on Saos-2 osteoblastic carcinoma cells. MTT assay was used for analysis of reduction in viability and phase-contrast microscopy for analysis of alterations in morphology of Saos-2 cells following tumstatin treatment. The results from viability assay revealed significant reduction in concentration-dependent manner on treatment with tumstatin for 48 h. The cells became round and shrunken, DNA showed fragmentation and ladder-like pattern following tumstatin treatment at 25 µM. Therefore, tumstatin treatment significantly induced apoptosis in Saos-2 cells in a dose-dependent manner. Studies have demonstrated that PTEN, FasR and FasL play a vital role in the induction of apoptosis through NF-κB pathway (24). Various cell apoptosis is induced via the NF-κB pathway by increase in the expression of PTEN, FasR and FasL (25,26). Investigation of the mRNA expression corresponding to PTEN, FasL and FasR in Saos-2 cells revealed significant increase in tumstatin-treated cells. These findings indicated that tumstatin exhibited apoptosis inducing effect through increase in the expression level of PTEN, FasL and FasR.

Results from the present study revealed that tumstatin treatment activated p65NF-κB by phosphorylation of serine on position 536. TNF-α also led to the activation of p65NF-κB by phosphorylation of serine at position 536 in Saos-2 cells. Phosphorylation of p65NF-κB at position 536 serine is well known and this study also demonstrated the same (2729). Therefore, tumstatin induced phosphorylation of p65NF-κB at position 536 serine observed in the present study is also confirmed. In most of the studies on p65NF-κB, it has been observed that serine is phosphorylated on position 536 (27,30,31). Overexpression studies with the activated Akt revealed that IKK is necessary for enhanced p65NF-κB transactivation, whereas mutation of Ser536 abolishes this effect (31). Our results revealed that tumstatin treatment also induced activation of p65NF-κB in Saos-2 cells by phosphorylation of serine present on position 536. Thus, NF-κB is activated by phosphorylation of serine on position 536 in the Saos-2 cells treated with tumstatin. The present study also demonstrated that activation of p65NF-κB induced its translocation into the nucleus in Saos-2 cells. Phosphorylation of Ser536 on p65NF-κB can be detected both in the cytoplasm and the nucleus.

Activation of NF-κB stimulates the expression of IκBα which exhibits inhibitory effect on the phosphorylation of NF-κB (32). The present study revealed that treatment of Saos-2 cells with TNF-α inhibited IκBα expression and therefore enhanced the nuclear translocation of NF-κB. Tumstatin treatment also inhibited the expression of IκBα in Saos-2 cells, but the effect was weak compared to TNF-α. Therefore, tumstatin treatment leads to the activation of NF-κB in a manner independent of IκBα.

In conclusion, the present study demonstrates that tumstatin inhibits viability and induces apoptosis in Saos-2 osteosarcoma cells through NF-κB activation pathway. Therefore, tumstatin shows promise for the treatment of osteosarcoma.

References

1 

Tarkkanen M, Karhu R, Kallioniemi A, Elomaa I, Kivioja AH, Nevalainen J, Böhling T, Karaharju E, Hyytinen E, Knuutila S, et al: Gains and losses of DNA sequences in osteosarcomas by comparative genomic hybridization. Cancer Res. 55:1334–1338. 1995.PubMed/NCBI

2 

Al-Romaih K, Bayani J, Vorobyova J, Karaskova J, Park PC, Zielenska M and Squire JA: Chromosomal instability in osteosarcoma and its association with centrosome abnormalities. Cancer Genet Cytogenet. 144:91–99. 2003. View Article : Google Scholar : PubMed/NCBI

3 

Ragland BD, Bell WC, Lopez RR and Siegal GP: Cytogenetics and molecular biology of osteosarcoma. Lab Invest. 82:365–373. 2002. View Article : Google Scholar : PubMed/NCBI

4 

He JP, Hao Y, Wang XL, Yang XJ, Shao JF, Guo FJ and Feng JX: Review of the molecular pathogenesis of osteosarcoma. Asian Pac J Cancer Prev. 15:5967–5976. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Broadhead ML, Clark JC, Myers DE, Dass CR and Choong PF: The molecular pathogenesis of osteosarcoma: A review. Sarcoma. 2011:9592482011. View Article : Google Scholar : PubMed/NCBI

6 

Kansara M and Thomas DM: Molecular pathogenesis of osteosarcoma. DNA Cell Biol. 26:1–18. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Tabone MD, Kalifa C, Rodary C, Raquin M, Valteau-Couanet D and Lemerle J: Osteosarcoma recurrences in pediatric patients previously treated with intensive chemotherapy. J Clin Oncol. 12:2614–2620. 1994.PubMed/NCBI

8 

Kempf-Bielack B, Bielack SS, Jurgens H, Branscheid D, Berdel WE, Exner GU, Göbel U, Helmke K, Jundt G, Kabisch SF, et al: Osteosarcoma relapse after combined modality therapy: An analysis of unselected patients in the Cooperative Osteosarcoma Study Group (COSS). J Clin Oncol. 23:559–568. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Davis AM, Bell RS and Goodwin PJ: Prognostic factors in osteosarcoma: A critical review. J Clin Oncol. 12:423–431. 1994.PubMed/NCBI

10 

Arends MJ and Wyllie AH: Apoptosis: mechanisms and roles in pathology. Int Rev Exp Pathol. 32:223–254. 1991. View Article : Google Scholar : PubMed/NCBI

11 

Jacobson MD, Weil M and Raff MC: Programmed cell death in animal development. Cell. 88:347–354. 1997. View Article : Google Scholar : PubMed/NCBI

12 

Cross TG, Scheel-Toellner D, Henriquez NV, Deacon E, Salmon M and Lord JM: Serine/threonine protein kinases and apoptosis. Exp Cell Res. 256:34–41. 2000. View Article : Google Scholar : PubMed/NCBI

13 

Aggarwal BB and Takada Y: Pro-apoptotic and anti-apoptotic effects of tumor necrosis factor in tumor cells. Role of nuclear transcription factor NF-κB. Cancer Treat Res. 126:103–127. 2005. View Article : Google Scholar

14 

Graham B and Gibson SB: The two faces of NF-κB in cell survival responses. Cell Cycle. 4:1342–1345. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Lamkanfi M, Declercq W, Van den Berghe T and Van den Abeele P: Caspases leave the beaten track: caspase-mediated activation of NF-κB. J Cell Biol. 173:165–171. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Piva R, Belardo G and Santoro MG: NF-κB: a stress-regulated switch for cell survival. Antioxid Redox Signal. 8:478–486. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Radhakrishnan SK and Kamalakaran S: Pro-apoptotic role of NF-κB: implications for cancer therapy. Biochim Biophys Acta. 1766:53–62. 2006.PubMed/NCBI

18 

Maeshima Y, Colorado PC, Torre A, Holthaus KA, Grunkemeyer JA, Ericksen MB, Hopfer H, Xiao Y, Stillman IE and Kalluri R: Distinct antitumor properties of a type IV collagen domain derived from basement membrane. J Biol Chem. 275:21340–21348. 2000. View Article : Google Scholar : PubMed/NCBI

19 

Hamano Y and Kalluri R: Tumstatin, the NC1 domain of α3 chain of type IV collagen, is an endogenous inhibitor of pathological angiogenesis and suppresses tumor growth. Biochem Biophys Res Commun. 333:292–298. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Pasco S, Brassart B, Ramont L, Maquart FX and Monboisse JC: Control of melanoma cell invasion by type IV collagen. Cancer Detect Prev. 29:260–266. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Pasco S, Ramont L, Venteo L, Pluot M, Maquart FX and Monboisse JC: In vivo overexpression of tumstatin domains by tumor cells inhibits their invasive properties in a mouse melanoma model. Exp Cell Res. 301:251–265. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Kawaguchi T, Yamashita Y, Kanamori M, Endersby R, Bankiewicz KS, Baker SJ, Bergers G and Pieper RO: The PTEN/Akt pathway dictates the direct alphaVbeta3-dependent growth-inhibitory action of an active fragment of tumstatin in glioma cells in vitro and in vivo. Cancer Res. 66:11331–11340. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Chung IS, Son YI, Ko YJ, Baek CH, Cho JK and Jeong HS: Peritumor injections of purified tumstatin delay tumor growth and lymphatic metastasis in an orthotopic oral squamous cell carcinoma model. Oral Oncol. 44:1118–1126. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Tanaka Y, Singh S and Aggarwal BB: Identification of a p65 peptide that selectively inhibits NF-κB activation induced by various inflammatory stimuli and its role in down-regulation of NF-κB-mediated gene expression and up-regulation of apoptosis. J Biol Chem. 279:15096–15104. 2004. View Article : Google Scholar

25 

Fujita M, Goto K, Yoshida K, Okamura H, Morimoto H, Kito S, Fukuda J and Haneji T: Okadaic acid stimulates expression of Fas receptor and Fas ligand by activation of nuclear factor kappa-B in human oral squamous carcinoma cells. Oral Oncol. 40:199–206. 2004. View Article : Google Scholar

26 

Bertram J, Peacock JW, Tan C, Mui AL, Chung SW, Gleave ME, Dedhar S, Cox ME and Ong CJ: Inhibition of the phosphatidylinositol 3′-kinase pathway promotes autocrine Fas-induced death of phosphatase and tensin homologue-deficient prostate cancer cells. Cancer Res. 66:4781–4788. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Yang F, Tang E, Guan K and Wang CY: IKKβ plays an essential role in the phosphorylation of RelA/p65 on serine 536 induced by lipopolysaccharide. J Immunol. 170:5630–5635. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Doyle SL, Jefferies CA and O'Neill LA: Bruton's tyrosine kinase is involved in p65-mediated transactivation and phosphorylation of p65 on serine 536 during NFκB activation by lipopolysaccharide. J Biol Chem. 280:23496–23501. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Shiraki K, Yamanaka T, Inoue H, Kawakita T, Enokimura N, Okano H, Sugimoto K, Murata K and Nakano T: Expression of TNF-related apoptosis-inducing ligand in human hepatocellular carcinoma. Int J Oncol. 26:1273–1281. 2005.PubMed/NCBI

30 

Sakurai H, Chiba H, Miyoshi H, Sugita T and Toriumi W: IκB kinases phosphorylate NF-κB p65 subunit on serine 536 in the transactivation domain. J Biol Chem. 274:30353–30356. 1999. View Article : Google Scholar : PubMed/NCBI

31 

Madrid LV, Mayo MW, Reuther JY and Baldwin AS Jr: Akt stimulates the transactivation potential of the RelA/p65 subunit of NF-κB through utilization of the IκB kinase and activation of the mitogen-activated protein kinase p38. J Biol Chem. 276:18934–18940. 2001. View Article : Google Scholar : PubMed/NCBI

32 

Sasaki CY, Barberi TJ, Ghosh P and Longo DL: Phosphorylation of RelA/p65 on serine 536 defines an IκBα-independent NF-κB pathway. J Biol Chem. 280:34538–34547. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2016
Volume 35 Issue 6

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
Wang Y, Yin R and Teng J: Tumstatin induces apoptosis and stimulates phosphorylation of p65NF-κB in human osteoblastic osteosarcoma Saos-2 cells. Oncol Rep 35: 3403-3408, 2016
APA
Wang, Y., Yin, R., & Teng, J. (2016). Tumstatin induces apoptosis and stimulates phosphorylation of p65NF-κB in human osteoblastic osteosarcoma Saos-2 cells. Oncology Reports, 35, 3403-3408. https://doi.org/10.3892/or.2016.4762
MLA
Wang, Y., Yin, R., Teng, J."Tumstatin induces apoptosis and stimulates phosphorylation of p65NF-κB in human osteoblastic osteosarcoma Saos-2 cells". Oncology Reports 35.6 (2016): 3403-3408.
Chicago
Wang, Y., Yin, R., Teng, J."Tumstatin induces apoptosis and stimulates phosphorylation of p65NF-κB in human osteoblastic osteosarcoma Saos-2 cells". Oncology Reports 35, no. 6 (2016): 3403-3408. https://doi.org/10.3892/or.2016.4762