Nutr Res Pract.  2010 Aug;4(4):276-282.

Effect of retinoic acid and delta-like 1 homologue (DLK1) on differentiation in neuroblastoma

Affiliations
  • 1Department of Nutritional Science and Food Management, Ewha Womans University, 11-1 Daehyun-dong, Seodaemun-gu, Seoul 120-750, Korea. yuri.kim@ewha.ac.kr

Abstract

The principal objective of this study was to evaluate the chemopreventive and therapeutic effects of a combination of all-trans-retinoic acid (RA) and knockdown of delta-like 1 homologue (Drosophila) (DLK1) on neuroblastoma, the most common malignant disease in children. As unfavorable neuroblastoma is poorly differentiated, neuroblastoma cell was induced differentiation by RA or DLK1 knockdown. Neuroblastoma cells showed elongated neurite growth, a hallmark of neuronal differentiation at various doses of RA, as well as by DLK1 knockdown. In order to determine whether or not a combination of RA and DLK1 knockdown exerts a greater chemotherapeutic effect on neuroblastoma, cells were incubated at 10 nM RA after being transfected with SiRNA-DLK1. Neuronal differentiation was increased more by a combination of RA and DLK1 knockdown than by single treatment. Additionally, in order to assess the signal pathway of neuroblastoma differentiation induced by RA and DLK1 knockdown, treatment with the specific MEK/ERK inhibitors, U0126 and PD 98059, was applied to differentiated neuroblastoma cells. Differentiation induced by RA and DLK1 knockdown increased ERK phosphorylation. The MEK/ERK inhibitor U0126 completely inhibited neuronal differentiation induced by both RA and DLK1 knockdown, whereas PD98059 partially blocked neuronal differentiation. After the withdrawal of inhibitors, cellular differentiation was fully recovered. This study is, to the best of our knowledge, the first to demonstrate that the specific inhibitors of the MEK/ERK pathway, U0126 and PD98059, exert differential effects on the ERK phosphorylation induced by RA or DLK1 knockdown. Based on the observations of this study, it can be concluded that a combination of RA and DLK1 knockdown increases neuronal differentiation for the control of the malignant growth of human neuroblastomas, and also that both MEK1 and MEK2 are required for the differentiation induced by RA and DLK1 knockdown.

Keyword

Retinoic acid; DLK1; differentiation; neuroblastoma

MeSH Terms

Butadienes
Child
Flavonoids
Humans
Neurites
Neuroblastoma
Neurons
Nitriles
Phosphorylation
Signal Transduction
Tretinoin
Butadienes
Flavonoids
Nitriles
Tretinoin

Figure

  • Fig. 1 All-trans-retinoic acid (RA) induces neuronal differentiation in neuroblastoma and down-regulates stem cell markers (CD133, SOX2, and DLK1). A. BE(2)C cells were treated without (w/o) or with RA (10 nM, 100 nM, or 1 µM) for 5 d. β-Tubulin III expression was detected by immunofluorescence. Magnification = ×100. B. BE(2)C cells were treated without (-) or with 1µM RA (+) for 5 d. CD133, SOX2, and DLK1 expression were detected by immunofluorescence. Magnification = ×100

  • Fig. 2 Neuronal differentiation was increased by a combination of RA and DLK1 knockdown. BE(2)C cells were transfected with DLK1 SiRNA (05), SiRNA (07), or a nontargetting SiRNA (Ctrl). Cells were treated without (w/o) or with 10 nM RA for 3 d. A. β-Tubulin III expression was detected by immunofluorescence. B. DLK1 expression was detected by immunofluorescence.

  • Fig. 3 MAPK inhibitors block ERK phosphorylation induced by RA. BE(2)C cells were treated at various concentrations (0, 1 µM, 10 µM) of RA. Cells were treated with MEK/ERK inhibitors, U0126 (5 µM) and PD98059 (PD, 5 µM) on the next day, and then maintained for 3 d. ERK phosphorylation was analyzed by Western Blotting. β-actin was used as a loading control.

  • Fig. 4 MAPK inhibitors, U0126 or PD 98059 (PD) blocks neuronal differentiation induced by RA. Cells were treated without (w/o) or with RA (1 nM, 10 nM) and then treated with 5 µM of U0126 (A) or PD (B) on the next day. The cells were maintained for 3 d. Cell morphology was examined by phase contrast microscopy. Magnification = 100×

  • Fig. 5 MAPK inhibitors, U0126 or PD 98059 (PD) blocks neuronal differentiation induced by DLK1 knockdown. BE(2)C cells were infected with control or ShRNA-DLK1 and then treated with various concentrations (0, 2.5 µM, 5 µM, 10 µM) of U0126 (A) or PD (B) next day. Cells were maintained for 3 d. Cell morphology was examined by phase contrast microscopy. Magnification = 100×

  • Fig. 6 Neuronal differentiation induced by DLK1 knockdown was recovered after withdrawal of MAPK inhibitors. BE(2)C cells were infected with control or ShRNA-DLK1. Cells were treated with 5 µM U0126 (A) or PD (B) next day and maintained for 5 d (middle panel). After 3 days of treatment, U0126 or PD was withdrawn and cells were maintained for 2 d. Cell morphology was examined by phase contrast microscopy. Magnification = 100×


Reference

1. Maris JM, Matthay KK. Molecular biology of neuroblastoma. J Clin Oncol. 1999. 17:2264–2279.
Article
2. Brodeur GM. Neuroblastoma: biological insights into a clinical enigma. Nat Rev Cancer. 2003. 3:203–216.
Article
3. Kushner BH. Neuroblastoma: a disease requiring a multitude of imaging studies. J Nucl Med. 2004. 45:1172–1188.
4. De Preter K, Vandesompele J, Heimann P, Beckman S, Schramm A, Eggert A, Stallings RL, Benoit Y, Renard M, De Paepe A, Laureys G, Påhlman S, Speleman F. Human fetal neuroblast and neuroblastoma transcriptome analysis confirms neuroblast origin and highlights neuroblastoma candidate genes. Genome Biol. 2006. 7:R84.
5. Vasudevan SA, Nuchtern JG, Shohet JM. Gene profiling of high risk neuroblastoma. World J Surg. 2005. 29:317–324.
Article
6. Matthay KK, Haas-Kogan D, Constine LS. Halperin EC, Constine LS, Tarbell NJ, Kun LE, editors. Neuroblastoma. Pediatric Radiation Oncology. 2005. Philadelphia: Lippincott Williams & Wilkins;179–222.
7. Pearson AD, Pinkerton CR, Lewis IJ, Imeson J, Ellershaw C, Machin D. European Neuroblastoma Study Group. Children's Cancer and Leukaemia Group (CCLG formerly United Kingdom Children's Cancer Study Group). High-dose rapid and standard induction chemotherapy for patients aged over 1 year with stage 4 neuroblastoma: a randomised trial. Lancet Oncol. 2008. 9:247–256.
Article
8. Smith MA, Anderson B. Where to next with retinoids for cancer therapy? Clin Cancer Res. 2001. 7:2955–2957.
9. Goodman DS, Huang HS. Biosynthesis of vitamin a with rat intestinal enzymes. Science. 1965. 149:879–880.
Article
10. von Lintig J, Vogt K. Filling the gap in vitamin A research. Molecular identification of an enzyme cleaving beta-carotene to retinal. J Biol Chem. 2000. 275:11915–11920.
11. Blomhoff R, Blomhoff HK. Overview of retinoid metabolism and function. J Neurobiol. 2006. 66:606–630.
Article
12. Fenaux P, Chastang C, Chevret S, Sanz M, Dombret H, Archimbaud E, Fey M, Rayon C, Huguet F, Sotto JJ, Gardin C, Makhoul PC, Travade P, Solary E, Fegueux N, Bordessoule D, Miguel JS, Link H, Desablens B, Stamatoullas A, Deconinck E, Maloisel F, Castaigne S, Preudhomme C, Degos L. A randomized comparison of all transretinoic acid (ATRA) followed by chemotherapy and ATRA plus chemotherapy and the role of maintenance therapy in newly diagnosed acute promyelocytic leukemia. The European APL Group. Blood. 1999. 94:1192–1200.
Article
13. Fenaux P, Chevret S, Guerci A, Fegueux N, Dombret H, Thomas X, Sanz M, Link H, Maloisel F, Gardin C, Bordessoule D, Stoppa AM, Sadoun A, Muus P, Wandt H, Mineur P, Whittaker JA, Fey M, Daniel MT, Castaigne S, Degos L. European APL group. Long-term follow-up confirms the benefit of all-trans retinoic acid in acute promyelocytic leukemia. Leukemia. 2000. 14:1371–1377.
Article
14. Laborda J. The role of the epidermal growth factor-like protein dlk in cell differentiation. Histol Histopathol. 2000. 15:119–129.
15. Floridon C, Jensen CH, Thorsen P, Nielsen O, Sunde L, Westergaard JG, Thomsen SG, Teisner B. Does fetal antigen 1 (FA1) identify cells with regenerative, endocrine and neuroendocrine potentials? A study of FA1 in embryonic, fetal, and placental tissue and in maternal circulation. Differentiation. 2000. 66:49–59.
Article
16. Smas CM, Sul HS. Pref-1, a protein containing EGF-like repeats, inhibits adipocyte differentiation. Cell. 1993. 73:725–734.
Article
17. Tornehave D, Jensen CH, Teisner B, Larsson LI. FA1 immunoreactivity in endocrine tumours and during development of the human fetal pancreas; negative correlation with glucagon expression. Histochem Cell Biol. 1996. 106:535–542.
Article
18. Villena JA, Kim KH, Sul HS. Pref-1 and ADSF/resistin: two secreted factors inhibiting adipose tissue development. Horm Metab Res. 2002. 34:664–670.
Article
19. Li L, Forman SJ, Bhatia R. Expression of DLK1 in hematopoietic cells results in inhibition of differentiation and proliferation. Oncogene. 2005. 24:4472–4476.
Article
20. Sakajiri S, O'Kelly J, Yin D, Miller CW, Hofmann WK, Oshimi K, Shih LY, Kim KH, Sul HS, Jensen CH, Teisner B, Kawamata N, Koeffler HP. Dlk1 in normal and abnormal hematopoiesis. Leukemia. 2005. 19:1404–1410.
Article
21. Bauer SR, Ruiz-Hidalgo MJ, Rudikoff EK, Goldstein J, Laborda J. Modulated expression of the epidermal growth factor-like homeotic protein dlk influences stromal-cell-pre-B-cell interactions, stromal cell adipogenesis, and pre-B-cell interleukin-7 requirements. Mol Cell Biol. 1998. 18:5247–5255.
Article
22. Kaneta M, Osawa M, Sudo K, Nakauchi H, Farr AG, Takahama Y. A role for pref-1 and HES-1 in thymocyte development. J Immunol. 2000. 164:256–264.
Article
23. Van Limpt VA, Chan AJ, Van Sluis PG, Caron HN, Van Noesel CJ, Versteeg R. High delta-like 1 expression in a subset of neuroblastoma cell lines corresponds to a differentiated chromaffin cell type. Int J Cancer. 2003. 105:61–69.
Article
24. Laborda J, Sausville EA, Hoffman T, Notario V. dlk, a putative mammalian homeotic gene differentially expressed in small cell lung carcinoma and neuroendocrine tumor cell line. J Biol Chem. 1993. 268:3817–3820.
Article
25. Yin D, Xie D, Sakajiri S, Miller CW, Zhu H, Popoviciu ML, Said JW, Black KL, Koeffler HP. DLK1: increased expression in gliomas and associated with oncogenic activities. Oncogene. 2006. 25:1852–1861.
Article
26. Kim Y, Lin Q, Zelterman D, Yun Z. Hypoxia-regulated delta-like 1 homologue enhances cancer cell stemness and tumorigenicity. Cancer Res. 2009. 69:9271–9280.
Article
27. Hunter T. Protein kinases and phosphatases: the yin and yang of protein phosphorylation and signaling. Cell. 1995. 80:225–236.
Article
28. Berwick DC, Calissano M, Corness JD, Cook SJ, Latchman DS. Regulation of Brn-3a N-terminal transcriptional activity by MEK1/2-ERK1/2 signalling in neural differentiation. Brain Res. 2009. 1256:8–18.
Article
29. Alessi DR, Cuenda A, Cohen P, Dudley DT, Saltiel AR. PD 098059 is a specific inhibitor of the activation of mitogen-activated protein kinase kinase in vitro and in vivo. J Biol Chem. 1995. 270:27489–27494.
Article
30. Cuenda A, Alessi DR. Use of kinase inhibitors to dissect signaling pathways. Methods Mol Biol. 2000. 99:161–175.
Article
31. Ahn NG, Nahreini TS, Tolwinski NS, Resing KA. Pharmacologic inhibitors of MKK1 and MKK2. Methods Enzymol. 2001. 332:417–431.
Article
32. Yun Z, Lin Q, Giaccia AJ. Adaptive myogenesis under hypoxia. Mol Cell Biol. 2005. 25:3040–3055.
Article
33. Katayama K, Wada K, Miyoshi H, Ohashi K, Tachibana M, Furuki R, Mizuguchi H, Hayakawa T, Nakajima A, Kadowaki T, Tsutsumi Y, Nakagawa S, Kamisaki Y, Mayumi T. RNA interfering approach for clarifying the PPARgamma pathway using lentiviral vector expressing short hairpin RNA. FEBS Lett. 2004. 560:178–182.
Article
34. De los Santos M, Zambrano A, Aranda A. Combined effects of retinoic acid and histone deacetylase inhibitors on human neuroblastoma SH-SY5Y cells. Mol Cancer Ther. 2007. 6:1425–1432.
Article
35. Natale RB. Dual targeting of the vascular endothelial growth factor receptor and epidermal growth factor receptor pathways with vandetinib (ZD6474) in patients with advanced or metastatic non-small cell lung cancer. J Thorac Oncol. 2008. 3:S128–S130.
Article
36. Miller VA, Benedetti FM, Rigas JR, Verret AL, Pfister DG, Straus D, Kris MG, Crisp M, Heyman R, Loewen GR, Truglia JA, Warrell RP Jr. Initial clinical trial of a selective retinoid X receptor ligand, LGD1069. J Clin Oncol. 1997. 15:790–795.
Article
37. Shalinsky DR, Bischoff ED, Gregory ML, Lamph WW, Heyman RA, Hayes JS, Thomazy V, Davies PJ. Enhanced antitumor efficacy of cisplatin in combination with ALRT1057 (9-cis retinoic acid) in human oral squamous carcinoma xenografts in nude mice. Clin Cancer Res. 1996. 2:511–520.
38. Laborda J. The role of the epidermal growth factor-like protein dlk in cell differentiation. Histol Histopathol. 2000. 15:119–129.
39. Davies SP, Reddy H, Caivano M, Cohen P. Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem J. 2000. 351:95–105.
Article
40. Favata MF, Horiuchi KY, Manos EJ, Daulerio AJ, Stradley DA, Feeser WS, Van Dyk DE, Pitts WJ, Earl RA, Hobbs F, Copeland RA, Magolda RL, Scherle PA, Trzaskos JM. Identification of a novel inhibitor of mitogen-activated protein kinase kinase. J Biol Chem. 1998. 273:18623–18632.
Article
41. Dang ZC, Lowik CW. Differential effects of PD98059 and U0126 on osteogenesis and adipogenesis. J Cell Biochem. 2004. 92:525–533.
Article
Full Text Links
  • NRP
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr