DOI QR코드

DOI QR Code

Nox4-Mediated Cell Signaling Regulates Differentiation and Survival of Neural Crest Stem Cells

  • Lee, Ji-Eun (Department of Life Science and GT5 Program, Ewha Womans University) ;
  • Cho, Kyu Eun (Department of Life Science and GT5 Program, Ewha Womans University) ;
  • Lee, Kyung Eun (Department of Life Science and GT5 Program, Ewha Womans University) ;
  • Kim, Jaesang (Department of Life Science and GT5 Program, Ewha Womans University) ;
  • Bae, Yun Soo (Department of Life Science and GT5 Program, Ewha Womans University)
  • Received : 2014.09.04
  • Accepted : 2014.09.23
  • Published : 2014.12.31

Abstract

The function of reactive oxygen species (ROS) as second messengers in cell differentiation has been demonstrated only for a limited number of cell types. Here, we used a well-established protocol for BMP2-induced neuronal differentiation of neural crest stem cells (NCSCs) to examine the function of BMP2-induced ROS during the process. We first show that BMP2 indeed induces ROS generation in NCSCs and that blocking ROS generation by pretreatment of cells with diphenyleneiodonium (DPI) as NADPH oxidase (Nox) inhibitor inhibits neuronal differentiation. Among the ROS-generating Nox isozymes, only Nox4 was expressed at a detectable level in NCSCs. Nox4 appears to be critical for survival of NCSCs at least in vitro as down-regulation by RNA interference led to apoptotic response from NCSCs. Interestingly, development of neural crest-derived peripheral neural structures in Nox4-/- mouse appears to be grossly normal, although Nox4-/- embryos were born at a sub-Mendelian ratio and showed delayed over-all development. Specifically, cranial and dorsal root ganglia, derived from NCSCs, were clearly present in Nox4-/- embryo at embryonic days (E) 9.5 and 10.5. These results suggest that Nox4-mediated ROS generation likely plays important role in fate determination and differentiation of NCSCs, but other Nox isozymes play redundant function during embryogenesis.

Keywords

References

  1. Anderson, D.J. (1997). Cellular and molecular biology of neural crest cell lineage determination. Trends Genet. 13, 276-280. https://doi.org/10.1016/S0168-9525(97)01187-6
  2. Bae, Y.S., Oh, H., Rhee, S.G., and Yoo, Y.D. (2011). Regulation of reactive oxygen species generation in cell signaling. Mol. Cells 32, 491-509. https://doi.org/10.1007/s10059-011-0276-3
  3. Birren, S.J., Lo, L., and Anderson, D.J. (1993). Sympathetic neuroblasts undergo a developmental switch in trophic dependence. Development 119, 597-610.
  4. Buggisch, M., Ateghang, B., Ruhe, C., Strobel, C., Lange, S., Wartenberg, M., and Sauer, H. (2007). Stimulation of ES-cell-derived cardiomyogenesis and neonatal cardiac cell proliferation by reactive oxygen species and NADPH oxidase. J. Cell Sci. 120, 885-894. https://doi.org/10.1242/jcs.03386
  5. Chiarugi, P., and Giannoni, E. (2008). Anoikis: a necessary death program for anchorage-dependent cells. Biochem. Pharmacol. 76, 1352-1364. https://doi.org/10.1016/j.bcp.2008.07.023
  6. Gao, X.P., Standiford, T.J., Rahman, A., Newstead, M., Holland, S.M., Dinauer, M.C., Liu, Q.H., and Malik, A.B. (2002). Role of NADPH oxidase in the mechanism of lung neutrophil sequestration and microvessel injury induced by Gram-negative sepsis: studies in p47phox-/- and gp91phox-/- mice. J. Immunol. 168, 3974-3982. https://doi.org/10.4049/jimmunol.168.8.3974
  7. Groeger, G., Quiney, C., and Cotter, T.G. (2009). Hydrogen peroxide as a cell-survival signaling molecule. Antioxid. Redox Signal. 11, 2655-2671. https://doi.org/10.1089/ars.2009.2728
  8. Kim, J., Lo, L., Dormand, E., and Anderson, D.J. (2003). SOX10 maintains multipotency and inhibits neuronal differentiation of neural crest stem cells. Neuron 38, 17-31. https://doi.org/10.1016/S0896-6273(03)00163-6
  9. Kim, K.S., Choi, H.W., Yoon, H.E., and Kim, I.Y. (2010). Reactive oxygen species generated by NADPH oxidase 2 and 4 are required for chondrogenic differentiation. J. Biol. Chem. 285, 40294-40302. https://doi.org/10.1074/jbc.M110.126821
  10. Le Douarin, N.M. (1980). The ontogeny of the neural crest in avian embryo chimaeras. Nature 286, 663-669. https://doi.org/10.1038/286663a0
  11. Lee, J.E., Hollenberg, S.M., Snider, L., Turner, D.L., Lipnick, N., and Weintraub, H. (1995). Conversion of Xenopus ectoderm into neurons by NeuroD, a basic helix-loop-helix protein. Science 268, 836-844. https://doi.org/10.1126/science.7754368
  12. Lee, K.E., Nam, S., Cho, E.A., Seong, I., Limb, J.K., Lee, S., and Kim, J. (2008). Identification of direct regulatory targets of the transcription factor Sox10 based on function and conservation. BMC Genomics 9, 408. https://doi.org/10.1186/1471-2164-9-408
  13. Lee, J.H., Joo, J.H., Kim, J., Lim, H.J., Kim, S., Curtiss, L., Seong, J.K., Cui, W., Yabe-Nishimura, C., and Bae, Y.S. (2013). Interaction of NADPH oxidase 1 with Toll-like receptor 2 induces migration of smooth muscle cells. Cardiovasc. Res. 99, 483-493. https://doi.org/10.1093/cvr/cvt107
  14. Lo, L., Dormand, E., Greenwood, A., and Anderson, D.J. (2002). Comparison of the generic neuronal differentiation and neuron subtype specification functions of mammalian achaete-scute and atonal homologs in cultured neural progenitor cells. Development 129, 1553-1567.
  15. Ma, Q., Chen, Z., del Barco Barrantes, I., de la Pompa, J.L., and Anderson, D.J. (1998). neurogenin1 is essential for the determination of neuronal precursors for proximal cranial sensory ganglia. Neuron 20, 469-482. https://doi.org/10.1016/S0896-6273(00)80988-5
  16. Madenspacher, J.H., Azzam, K.M., Gowdy, K.M., Malcolm, K.C., Nick, J.A., Dixon, D., Aloor, J.J., Draper, D.W., Guardiola, J.J., Shatz, M., et al. (2013). p53 Integrates host defense and cell fate during bacterial pneumonia. J. Exp. Med. 210, 891-904. https://doi.org/10.1084/jem.20121674
  17. Mandal, C.C., Ganapathy, S., Gorin, Y., Mahadev, K., Block, K., Abboud, H.E., Harris, S.E., Ghosh-Choudhury, G., and Ghosh-Choudhury, N. (2011). Reactive oxygen species derived from Nox4 mediate BMP2 gene transcription and osteoblast differentiation. Biochem. J. 433, 393-402. https://doi.org/10.1042/BJ20100357
  18. Matsuno, K., Yamada, H., Iwata, K., Jin, D., Katsuyama, M., Matsuki, M., Takai, S., Yamanishi, K., Miyazaki, M., Matsubara, H., et al. (2005). Nox1 is involved in angiotensin II-mediated hypertension: a study in Nox1-deficient mice. Circulation 112, 2677-2685. https://doi.org/10.1161/CIRCULATIONAHA.105.573709
  19. Milosevic, N., Bekhite, M.M., Sharifpanah, F., Ruhe, C., Wartenberg, M., and Sauer, H. (2010). Redox stimulation of cardiomyogenesis versus inhibition of vasculogenesis upon treatment of mouse embryonic stem cells with thalidomide. Antioxid. Redox. Signal. 13, 1813-1827. https://doi.org/10.1089/ars.2010.3139
  20. Morrison, S.J., White, P.M., Zock, C., and Anderson, D.J. (1999). Prospective identification, isolation by flow cytometry, and in vivo self-renewal of multipotent mammalian neural crest stem cells. Cell 96, 737-749. https://doi.org/10.1016/S0092-8674(00)80583-8
  21. Nadworny, A.S., Guruju, M.R., Poor, D., Doran, R.M., Sharma, R.V., Kotlikoff, M.I., and Davisson, R.L. (2013). Nox2 and Nox4 influence neonatal c-kit(+) cardiac precursor cell status and differentiation. Am. J. Physiol. Heart Circ. Physiol. 305, H829-842. https://doi.org/10.1152/ajpheart.00761.2012
  22. Paoli, P., Giannoni, E., and Chiarugi, P. (2013). Anoikis molecular pathways and its role in cancer progression. Biochim. Biophys. Acta 1833, 3481-3498. https://doi.org/10.1016/j.bbamcr.2013.06.026
  23. Perris, R., and Perissinotto, D. (2000). Role of the extracellular matrix during neural crest cell migration. Mech. Dev. 95, 3-21. https://doi.org/10.1016/S0925-4773(00)00365-8
  24. Rosc-Schluter, B.I., Hauselmann, S.P., Lorenz, V., Mochizuki, M., Facciotti, F., Pfister, O., and Kuster, G.M. (2012). NOX2-derived reactive oxygen species are crucial for CD29-induced prosurvival signalling in cardiomyocytes. Cardiovasc. Res. 93, 454-462. https://doi.org/10.1093/cvr/cvr348
  25. Shah, N.M., Groves, A.K., and Anderson, D.J. (1996). Alternative neural crest cell fates are instructively promoted by TGFbeta superfamily members. Cell 85, 331-343. https://doi.org/10.1016/S0092-8674(00)81112-5
  26. Simone, S., Cosola, C., Loverre, A., Cariello, M., Sallustio, F., Rascio, F., Gesualdo, L., Schena, F.P., Grandaliano, G., and Pertosa, G. (2012). BMP-2 induces a profibrotic phenotype in adult renal progenitor cells through Nox4 activation. Am. J. Physiol. Renal. Physiol. 303, F23-34. https://doi.org/10.1152/ajprenal.00328.2011
  27. Stemple, D.L., and Anderson, D.J. (1992). Isolation of a stem cell for neurons and glia from the mammalian neural crest. Cell 71, 973-985. https://doi.org/10.1016/0092-8674(92)90393-Q

Cited by

  1. What has passed is prolog: new cellular and physiological roles of G6PD vol.50, pp.10, 2016, https://doi.org/10.1080/10715762.2016.1223296
  2. Blue light potentiates neurogenesis induced by retinoic acid-loaded responsive nanoparticles vol.59, 2017, https://doi.org/10.1016/j.actbio.2017.06.044
  3. Hypoxia and hyperoxia differentially control proliferation of rat neural crest stem cells via distinct regulatory pathways of the HIF1α-CXCR4 and TP53-TPM1 proteins vol.246, pp.3, 2017, https://doi.org/10.1002/dvdy.24481
  4. Role of Nox4 in Neuronal Differentiation of Mouse Subventricular Zone Neural Stem Cells vol.26, pp.1, 2016, https://doi.org/10.5352/JLS.2016.26.1.8
  5. NADPH Oxidases: Insights into Selected Functions and Mechanisms of Action in Cancer and Stem Cells vol.2017, 2017, https://doi.org/10.1155/2017/9420539
  6. Redox signaling mechanisms in nervous system development 2018, https://doi.org/10.1089/ars.2017.7284
  7. Nox2 and Nox4 regulate self-renewal of murine induced-pluripotent stem cells vol.68, pp.12, 2016, https://doi.org/10.1002/iub.1574
  8. A 2-Substituted 8-Hydroxyquinoline Stimulates Neural Stem Cell Proliferation by Modulating ROS Signalling vol.74, pp.3, 2016, https://doi.org/10.1007/s12013-016-0747-4
  9. Nox, Reactive Oxygen Species and Regulation of Vascular Cell Fate vol.6, pp.4, 2017, https://doi.org/10.3390/antiox6040090
  10. Glucose-6-phosphate dehydrogenase is indispensable in embryonic development by modulation of epithelial-mesenchymal transition via the NOX/Smad3/miR-200b axis vol.9, pp.1, 2018, https://doi.org/10.1038/s41419-017-0005-8
  11. Novel crosstalk between Vps26a and Nox4 signaling during neurogenesis pp.1476-5403, 2018, https://doi.org/10.1038/s41418-018-0226-0
  12. PARP3 controls TGFβ and ROS driven epithelial-to-mesenchymal transition and stemness by stimulating a TG2-Snail-E-cadherin axis vol.7, pp.39, 2014, https://doi.org/10.18632/oncotarget.11627
  13. PARP3 comes to light as a prime target in cancer therapy vol.18, pp.12, 2014, https://doi.org/10.1080/15384101.2019.1617454
  14. Oxidative Stress, Neuroinflammation, and NADPH Oxidase: Implications in the Pathogenesis and Treatment of Alzheimer’s Disease vol.2021, pp.None, 2014, https://doi.org/10.1155/2021/7086512
  15. NADPH Oxidases: Redox Regulators of Stem Cell Fate and Function vol.10, pp.6, 2014, https://doi.org/10.3390/antiox10060973
  16. Metabolic and Redox Regulation of Cardiovascular Stem Cell Biology and Pathology vol.35, pp.3, 2014, https://doi.org/10.1089/ars.2020.8201