J Korean Neurosurg Soc.  2018 Jul;61(4):441-449. 10.3340/jkns.2017.0219.

Nerve Growth Factor Stimulates Glioblastoma Proliferation through Notch1 Receptor Signaling

Affiliations
  • 1Department of Neurosurgery, Hallym University Sacred Heart Hospital, Anyang, Korea. nscib71@gmail.com
  • 2Department of Neurosurgery, Dongtan Sacred Heart Hospital, Hwaseong, Korea.
  • 3Department of Obstetrics and Gynecology, Hallym University Sacred Heart Hospital, Anyang, Korea.

Abstract


OBJECTIVE
Notch receptors are heterodimeric transmembrane proteins that regulate cell fate, such as differentiation, proliferation, and apoptosis. Dysregulated Notch pathway signaling has been observed in glioblastomas, as well as in other human malignancies. Nerve growth factor (NGF) is essential for cell growth and differentiation in the nervous system. Recent reports suggest that NGF stimulates glioblastoma proliferation. However, the relationship between NGF and Notch1 in glioblastomas remains unknown. Therefore, we investigated expression of Notch1 in a glioblastoma cell line (U87-MG), and examined the relationship between NGF and Notch1 signaling.
METHODS
We evaluated expression of Notch1 in human glioblastomas and normal brain tissues by immunohistochemical staining. The effect of NGF on glioblastoma cell line (U87-MG) was evaluated by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay. To evaluate the relationship between NGF and Notch1 signaling, Notch1 and Hes1 expression were evaluated by reverse transcription polymerase chain reaction (RT-PCR) and Western blot analysis, respectively. To confirm the effects of NGF on Notch1 signaling, Notch1 and Hes1 small interfering RNAs (siRNAs) were used.
RESULTS
In immunohistochemistry, Notch1 expression was higher in glioblastoma than in normal brain tissue. MTT assay showed that NGF stimulates U87-MG cells in a dose-dependent manner. RT-PCR and Western blot analysis demonstrated that Notch1 and Hes1 expression were increased by NGF in a dose-dependent manner. After transfection with Notch1 and Hes1 siRNAs, there was no significant difference between controls and 100 nM NGF-β, which means that U87-MG cell proliferation was suppressed by Notch1 and Hes1 siRNAs.
CONCLUSION
These results indicate that NGF stimulates glioblastoma cell proliferation via Notch1 signaling through Hes 1.

Keyword

Glioblastoma; Nerve growth factor; Notch1

MeSH Terms

Apoptosis
Blotting, Western
Brain
Cell Line
Cell Proliferation
Glioblastoma*
Humans
Immunohistochemistry
Nerve Growth Factor*
Nervous System
Polymerase Chain Reaction
Receptor, Notch1*
Receptors, Notch
Reverse Transcription
RNA, Small Interfering
Transfection
Nerve Growth Factor
RNA, Small Interfering
Receptor, Notch1
Receptors, Notch

Figure

  • Fig. 1. Normal brain tissue (A) and negative control (B) show no staining, but human glioblastoma tissue (C) shows brown colored changes in the nucleus and cytoplasm by the streptavidin-biotin-peroxidase complex technique. Scale bar=200 μm.

  • Fig. 2. 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay shows that nerve growth factor (NGF) stimulates U87-MG cell proliferation in a dose-dependent manner (p=0.005).

  • Fig. 3. Expression of Notch1 in the reverse transcription polymerase chain reaction (RT-PCR) (A) and a dose-dependent effect of nerve growth factor (NGF) on Notch1 expression in the U87-MG cell line (p=0.044) (B); expression of Notch1 in the Western blot analysis (C) and a dose-dependent effect of nerve growth factor (NGF) on Notch1 expression in the U87-MG cell line (p=0.037) (D). GAPDH : glyceraldehyde-3-phosphate dehydrogenase.

  • Fig. 4. Expression of Hes1 in the reverse transcription polymerase chain reaction (RT-PCR) (A) and a dose-dependent effect of nerve growth factor (NGF) on Hes1 expression in the U87-MG cell line (p=0.020) (B); Expression of Hes1 in the Western blot analysis (C) and a dose-dependent effect of NGF on Hes1 expression in the U87-MG cell line (p=0.037) (D). GAPDH : glyceraldehyde3-phosphate dehydrogenase.

  • Fig. 5. Expression of Notch1 mall interfering RNA (siRNA) in the Western blot analysis (A). Notch1 siRNA suppresses the effects of nerve growth factor (NGF) on U87-MG cell proliferation. There was difference in without lipofectamin, in lipofectamin only, and in negative control siRNA (p<0.05), but no significant difference in Notch1 siRNA (p=0.887) (B). Expression of Hes1 siRNA in the Western blot analysis (C). Hes1 siRNA suppresses the effects of NGF on U87-MG cell proliferation. There was difference in without lipofectamin, in lipofectamin only, and in negative control siRNA (p<0.05), but no significant difference in Hes1 siRNA (p=0.075) (D).


Cited by  1 articles

Propranolol Inhibits the Proliferation of Human Glioblastoma Cell Lines through Notch1 and Hes1 Signaling System
Hyun Sik Kim, Young Han Park, Heui Seung Lee, Mi Jung Kwon, Joon Ho Song, In Bok Chang
J Korean Neurosurg Soc. 2021;64(5):716-725.    doi: 10.3340/jkns.2021.0068.


Reference

References

1. Artavanis-Tsakonas S, Rand MD, Lake RJ. Notch signaling: cell fate control and signal integration in development. Science. 284:770–776. 1999.
Article
2. Aster JC, Pear WS. Notch signaling in leukemia. Curr Opin Hematol. 8:237–244. 2001.
Article
3. Bigas A, Martin DI, Milner LA. Notch1 and Notch2 inhibit myeloid differentiation in response to different cytokines. Mol Cell Biol. 18:2324–2333. 1998.
Article
4. Bolós V, Grego-Bessa J, de la Pompa JL. Notch signaling in development and cancer. Endocr Rev. 28:339–363. 2007.
Article
5. Brown MC, Staniszewska I, Lazarovici P, Tuszynski GP, Del Valle L, Marcinkiewicz C. Regulatory effect of nerve growth factor in alpha9beta1 integrin-dependent progression of glioblastoma. Neuro Oncol. 10:968–980. 2008.
Article
6. Chao MV, Hempstead BL. p75 and Trk: a two-receptor system. Trends Neurosci. 18:321–326. 1995.
Article
7. Chen J, Kesari S, Rooney C, Strack PR, Chen J, Shen H, et al. Inhibition of Notch signaling blocks growth of glioblastoma cell lines and tumor neurospheres. Genes Cancer. 1:822–835. 2010.
Article
8. Cheung HC, Corley LJ, Fuller GN, McCutcheon IE, Cote GJ. Polypyrimidine tract binding protein and notch1 are independently re-expressed in glioma. Mod Pathol. 19:1034–1041. 2006.
Article
9. Faux CH, Turnley AM, Epa R, Cappai R, Bartlett PF. Interactions between fibroblast growth factors and notch regulate neuronal differentiation. J Neurosci. 21:5587–5596. 2001.
Article
10. Giraud S, Loum E, Bessette B, Mathonnet M, Lalloué F. P75 neurotrophin receptor is sequestered in the Golgi apparatus of the U-87 MG human glioblastoma cell line. Int J Oncol. 38:391–399. 2011.
Article
11. Goretzki PE, Wahl RA, Becker R, Koller C, Branscheid D, Grussendorf M, et al. Nerve growth factor (NGF) sensitizes human medullary thyroid carcinoma (hMTC) cells for cytostatic therapy in vitro. Surgery. 102:1035–1042. 1987.
12. Greenwald I. LIN-12/Notch signaling: lessons from worms and flies. Genes Dev. 12:1751–1762. 1998.
13. Jang MS, Zlobin A, Kast WM, Miele L. Notch signaling as a target in multimodality cancer therapy. Curr Opin Mol Ther. 2:55–65. 2000.
14. Jian Q, Tao Z, Li Y, Yin ZQ. Acute retinal injury and the relationship between nerve growth factor, notch1 transcription and short-lived dedifferentiation transient changes of mammalian Müller cells. Vision Res. 110(Pt A):107–117. 2015.
Article
15. Jundt F, Anagnostopoulos I, Förster R, Mathas S, Stein H, Dörken B. Activated Notch1 signaling promotes tumor cell proliferation and survival in Hodgkin and anaplastic large cell lymphoma. Blood. 99:3398–3403. 2002.
Article
16. Kanu OO, Mehta A, Di C, Lin N, Bortoff K, Bigner DD, et al. Glioblastoma multiforme: a review of therapeutic targets. Expert Opin Ther Targets. 13:701–718. 2009.
Article
17. Kopan R, Ilagan MX. The canonical notch signaling pathway: unfolding the activation mechanism. Cell. 137:216–233. 2009.
Article
18. Lawn S, Krishna N, Pisklakova A, Qu X, Fenstermacher DA, Fournier M, et al. Neurotrophin signaling via TrkB and TrkC receptors promotes the growth of brain tumor-initiating cells. J Biol Chem. 290:3814–3824. 2015.
Article
19. Leong KG, Karsan A. Recent insights into the role of Notch signaling in tumorigenesis. Blood. 107:2223–2233. 2006.
Article
20. Li J, Cui Y, Gao G, Zhao Z, Zhang H, Wang X. Notch1 is an independent prognostic factor for patients with glioma. J Surg Oncol. 103:813–817. 2011.
Article
21. Liu Y, Su C, Shan Y, Yang S, Ma G. Targeting notch1 inhibits invasion and angiogenesis of human breast cancer cells via inhibition nuclear factorkappaB signaling. Am J Transl Res. 8:2681–2692. eCollection 2016.
22. Miele L, Golde T, Osborne B. Notch signaling in cancer. Curr Mol Med. 6:905–918. 2006.
Article
23. Miele L, Miao H, Nickoloff BJ. Notch signaling as a novel cancer therapeutic target. Curr Cancer Drug Targets. 6:313–323. 2006.
Article
24. Nicolas M, Wolfer A, Raj K, Kummer JA, Mill P, van Noort M, et al. Notch1 functions as a tumor suppressor in mouse skin. Nat Genet. 33:416–421. 2003.
Article
25. Oelmann E, Sreter L, Schuller I, Serve H, Koenigsmann M, Wiedenmann B, et al. Nerve growth factor stimulates clonal growth of human lung cancer cell lines and a human glioblastoma cell line expressing high-affinity nerve growth factor binding sites involving tyrosine kinase signaling. Cancer Res. 55:2212–2219. 1995.
26. Park YH, Kim SJ, Jeong BH, Herzog TJ, Wright J, Kitajewski J, et al. Follicular stimulating hormone enhances Notch 1 expression in SK-OV-3 ovarian cancer cells. J Gynecol Oncol. 21:119–124. 2010.
Article
27. Phillips HS, Kharbanda S, Chen R, Forrest WF, Soriano RH, Wu TD, et al. Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell. 9:157–173. 2006.
Article
28. Purow BW, Haque RM, Noel MW, Su Q, Burdick MJ, Lee J, et al. Expression of Notch-1 and its ligands, Delta-like-1 and Jagged-1, is critical for glioma cell survival and proliferation. Cancer Res. 65:2353–2363. 2005.
Article
29. Qian XQ, Chen LL, Cheng Q, Tian Y, Luo XF, Wan XY. Inhibition of Notch 1 receptor influenced the differentiation of Lin-CD45RA-dendritic cell precursors within ovarian carcinoma microenvironment. BMC Immunol. 17:14. 2016.
Article
30. Rakowicz-Szulczyńska EM, Herlyn M, Koprowski H. Nerve growth factor receptors in chromatin of melanoma cells, proliferating melanocytes, and colorectal carcinoma cells in vitro. Cancer Res. 48(24 Pt 1):7200–7206. 1988.
31. Rakowicz-Szulczynska EM, Reddy U, Vorbrodt A, Herlyn D, Koprowski H. Chromatin and cell surface receptors mediate melanoma cell growth response to nerve growth factor. Mol Carcinog. 4:388–396. 1991.
Article
32. Revoltella RP, Butler RH. Nerve growth factor may stimulate either division or differentiation of cloned C1300 neuroblastoma cells in serumfree cultures. J Cell Physiol. 104:27–33. 1980.
Article
33. Singer HS, Hansen B, Martinie D, Karp CL. Mitogenesis in glioblastoma multiforme cell lines: a role for NGF and its TrkA receptors. J Neurooncol. 45:1–8. 1999.
34. Sofroniew MV, Howe CL, Mobley WC. Nerve growth factor signaling, neuroprotection, and neural repair. Annu Rev Neurosci. 24:1217–1281. 2001.
Article
35. Sriuranpong V, Borges MW, Ravi RK, Arnold DR, Nelkin BD, Baylin SB, et al. Notch signaling induces cell cycle arrest in small cell lung cancer cells. Cancer Res. 61:3200–3205. 2001.
36. Ström A, Castella P, Rockwood J, Wagner J, Caudy M. Mediation of NGF signaling by post-translational inhibition of HES-1, a basic helix-loophelix repressor of neuronal differentiation. Genes Dev. 11:3168–3181. 1997.
Article
37. Takei Y, Laskey R. Interpreting crosstalk between TNF-alpha and NGF: potential implications for disease. Trends Mol Med. 14:381–388. 2008.
Article
38. Xu P, Yu S, Jiang R, Kang C, Wang G, Jiang H, et al. Differential expression of Notch family members in astrocytomas and medulloblastomas. Pathol Oncol Res. 15:703–710. 2009.
Article
39. Yavropoulou MP, Maladaki A, Yovos JG. The role of Notch and Hedgehog signaling pathways in pituitary development and pathogenesis of pituitary adenomas. Hormones (Athens). 14:5–18. 2015.
Article
40. Zlobin A, Jang M, Miele L. Toward the rational design of cell fate modifiers: Notch signaling as a target for novel biopharmaceuticals. Curr Pharm Biotechnol. 1:83–106. 2000.
Article
Full Text Links
  • JKNS
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