Sox2 controls neural stem cell self-renewal through a Fos-centered gene regulatory network.

Item request has been placed! ×
Item request cannot be made. ×
loading   Processing Request
  • Additional Information
    • Source:
      Publisher: Oxford University Press Country of Publication: England NLM ID: 9304532 Publication Model: Print-Electronic Cited Medium: Internet ISSN: 1549-4918 (Electronic) Linking ISSN: 10665099 NLM ISO Abbreviation: Stem Cells Subsets: MEDLINE
    • Publication Information:
      Publication: 2022- : Oxford : Oxford University Press
      Original Publication: Dayton, OH : AlphaMed Press, c1993-
    • Subject Terms:
    • Abstract:
      The Sox2 transcription factor is necessary for the long-term self-renewal of neural stem cells (NSCs). Its mechanism of action is still poorly defined. To identify molecules regulated by Sox2, and acting in mouse NSC maintenance, we transduced, into Sox2-deleted NSC, genes whose expression is strongly downregulated following Sox2 loss (Fos, Jun, Egr2), individually or in combination. Fos alone rescued long-term proliferation, as shown by in vitro cell growth and clonal analysis. Furthermore, pharmacological inhibition by T-5224 of FOS/JUN AP1 complex binding to its targets decreased cell proliferation and expression of the putative target Suppressor of cytokine signaling 3 (Socs3). Additionally, Fos requirement for efficient long-term proliferation was demonstrated by the reduction of NSC clones capable of long-term expansion following CRISPR/Cas9-mediated Fos inactivation. Previous work showed that the Socs3 gene is strongly downregulated following Sox2 deletion, and its re-expression by lentiviral transduction rescues long-term NSC proliferation. Fos appears to be an upstream regulator of Socs3, possibly together with Jun and Egr2; indeed, Sox2 re-expression in Sox2-deleted NSC progressively activates both Fos and Socs3 expression; in turn, Fos transduction activates Socs3 expression. Based on available SOX2 ChIPseq and ChIA-PET data, we propose a model whereby Sox2 is a direct activator of both Socs3 and Fos, as well as possibly Jun and Egr2; furthermore, we provide direct evidence for FOS and JUN binding on Socs3 promoter, suggesting direct transcriptional regulation. These results provide the basis for developing a model of a network of interactions, regulating critical effectors of NSC proliferation and long-term maintenance.
      (© 2021 AlphaMed Press.)
    • References:
      Bertolini J, Mercurio S, Favaro R, Mariani J, Ottolenghi S, Nicolis SK. Sox2-dependent regulation of neural stem cells and CNS development. In: Kondoh H, Lovell-Badge R, eds. Sox2, Biology and Role in Development and Disease. Amsterdam, The Netherlands: Elsevier; 2016.
      Fantes J, Ragge NK, Lynch SA, et al. Mutations in SOX2 cause anophthalmia [in English]. Nat Genet. 2003;33(4):461-463.
      Pevny LH, Nicolis SK. Sox2 roles in neural stem cells. Int J Biochem Cell Biol. 2010;42(3):421-424.
      Sisodiya SM, Ragge NK, Cavalleri GL, et al. Role of SOX2 mutations in human hippocampal malformations and epilepsy [Comparative Study Research Support, Non-U.S. Gov't] [in English]. Epilepsia. 2006;47(3):534-542.
      Favaro R, Valotta M, Ferri AL, et al. Hippocampal development and neural stem cell maintenance require Sox2-dependent regulation of Shh [Research Support, Non-U.S. Gov't] [in English]. Nat Neurosci. 2009;12(10):1248-1256.
      Amador-Arjona A, Cimadamore F, Huang CT, et al. SOX2 primes the epigenetic landscape in neural precursors enabling proper gene activation during hippocampal neurogenesis. Proc Natl Acad Sci USA. 2015;112(15):E1936-E1945.
      Andreu-Agullo C, Maurin T, Thompson CB, Lai EC. Ars2 maintains neural stem-cell identity through direct transcriptional activation of Sox2. Nature. 2011;481(7380):195-198.
      Cimadamore F, Amador-Arjona A, Chen C, Huang CT, Terskikh AV. SOX2-LIN28/let-7 pathway regulates proliferation and neurogenesis in neural precursors. Proc Natl Acad Sci USA. 2013;110(32):E3017-E3026.
      Graham V, Khudyakov J, Ellis P, Pevny L. SOX2 functions to maintain neural progenitor identity [Research Support, Non-U.S. Gov't Research Support, U.S. Gov't, P.H.S.] [in English]. Neuron. 2003;39(5):749-765.
      Mercurio S, Serra L, Nicolis SK. More than just stem cells: functional roles of the transcription factor Sox2 in differentiated glia and neurons. Int J Mol Sci. 2019;20:4540.
      Suh H, Consiglio A, Ray J, Sawai T, D'Amour KA, Gage FH. In vivo fate analysis reveals the multipotent and self-renewal capacities of Sox2+ neural stem cells in the adult hippocampus [Research Support, N.I.H., Extramural Research Support, Non-U.S. Gov't Research Support, U.S. Gov't, Non-P.H.S.] [in English]. Cell Stem Cell. 2007;1(5):515-528.
      Yu Y, Andreu-Agullo C, Liu BF, Barboza L, Toth M, Lai EC. Regulation of embryonic and adult neurogenesis by Ars2. Development. 2020;147(2):dev180018.
      Cavallaro M, Mariani J, Lancini C, et al. Impaired generation of mature neurons by neural stem cells from hypomorphic Sox2 mutants [Research Support, Non-U.S. Gov't] [in English]. Development. 2008;135(3):541-557.
      Bertolini JA, Favaro R, Zhu Y, et al. Mapping the global chromatin connectivity network for Sox2 function in neural stem cell maintenance. Cell Stem Cell. 2019;24(3):462-476.e466.
      Zhang Y, Wong CH, Birnbaum RY, et al. Chromatin connectivity maps reveal dynamic promoter-enhancer long-range associations. Nature. 2013;504(7479):306-310.
      Barbarani G, Fugazza C, Barabino SML, Ronchi AE. SOX6 blocks the proliferation of BCR-ABL1(+) and JAK2V617F(+) leukemic cells. Sci Rep. 2019;9(1):3388.
      Meers MP, Bryson TD, Henikoff JG, Henikoff S. Improved CUT&RUN chromatin profiling tools. Elife. 2019;8:46314.
      Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 2009;10(3):R25.
      Quinlan AR, Hall IM. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics. 2010;26(6):841-842.
      Meers MP, Tenenbaum D, Henikoff S. Peak calling by sparse enrichment analysis for CUT&RUN chromatin profiling. Epigenetics Chromatin. 2019;12(1):42.
      Eferl R, Wagner EF. AP-1: a double-edged sword in tumorigenesis. Nat Rev Cancer. 2003;3(11):859-868.
      Shaulian E, Karin M. AP-1 in cell proliferation and survival. Oncogene. 2001;20(19):2390-2400.
      Aikawa Y, Morimoto K, Yamamoto T, et al. Treatment of arthritis with a selective inhibitor of c-Fos/activator protein-1. Nat Biotechnol. 2008;26(7):817-823.
      Ye N, Ding Y, Wild C, Shen Q, Zhou J. Small molecule inhibitors targeting activator protein 1 (AP-1). J Med Chem. 2014;57(16):6930-6948.
      Yoshida T, Yamashita K, Watanabe M, et al. The impact of c-Fos/activator protein-1 inhibition on allogeneic pancreatic islet transplantation. Am J Transplant. 2015;15(10):2565-2575.
      Rubio A, Luoni M, Giannelli SG, et al. Rapid and efficient CRISPR/Cas9 gene inactivation in human neurons during human pluripotent stem cell differentiation and direct reprogramming. Sci Rep. 2016;6:37540.
      Wang H, Yang H, Shivalila CS, et al. One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell. 2013;153(4):910-918.
      O'Brien A, Bailey TL. GT-scan: identifying unique genomic targets. Bioinformatics. 2014;30(18):2673-2675.
      Gritti A, Parati EA, Cova L, et al. Multipotential stem cells from the adult mouse brain proliferate and self-renew in response to basic fibroblast growth factor. J Neurosci. 1996;16(3):1091-1100.
      Zappone MV, Galli R, Catena R, et al. Sox2 regulatory sequences direct expression of a (beta)-geo transgene to telencephalic neural stem cells and precursors of the mouse embryo, revealing regionalization of gene expression in CNS stem cells [Research Support, Non-U.S. Gov't] [in English]. Development. 2000;127(11):2367-2382.
      Jagga B, Edwards M, Pagin M, et al. Structural basis for nuclear import selectivity of pioneer transcription factor SOX2. Nat Commun. 2021;12(1):28.
      Barclay JL, Anderson ST, Waters MJ, Curlewis JD. Regulation of suppressor of cytokine signaling 3 (SOC3) by growth hormone in pro-B cells. Mol Endocrinol. 2007;21(10):2503-2515.
      Barclay JL, Anderson ST, Waters MJ, Curlewis JD. Characterization of the SOCS3 promoter response to prostaglandin E2 in T47D cells. Mol Endocrinol. 2007;21(10):2516-2528.
      Adepoju A, Micali N, Ogawa K, Hoeppner DJ, McKay RDG. FGF2 and insulin signaling converge to regulate cyclin D expression in multipotent neural stem cells. Stem Cells. 2014;32(3):770-778.
      Velazquez FN, Prucca CG, Etienne O, et al. Brain development is impaired in c-fos −/− mice. Oncotarget. 2015;6(19):16883-16901.
      Mercurio S, Alberti C, Serra L, et al. An early Sox2-dependent gene expression programme required for hippocampal dentate gyrus development. Open Biol. 2021;11(2):200339.
      Joo JY, Schaukowitch K, Farbiak L, Kilaru G, Kim TK. Stimulus-specific combinatorial functionality of neuronal c-fos enhancers. Nat Neurosci. 2016;19(1):75-83.
      Li S, Miao T, Sebastian M, et al. The transcription factors Egr2 and Egr3 are essential for the control of inflammation and antigen-induced proliferation of B and T cells. Immunity. 2012;37(4):685-696.
      Koo JH, Plouffe SW, Meng Z, et al. Induction of AP-1 by YAP/TAZ contributes to cell proliferation and organ growth. Genes Dev. 2020;34(1-2):72-86.
    • Contributed Indexing:
      Keywords: AP1 inhibitor T-5224; CRISPR; CUT&RUN; Fos; Socs3; Sox2; lentiviral vector; neural stem cells (NSCs); self-renewal; transcription factors
    • Accession Number:
      0 (Fos protein, mouse)
      0 (Proto-Oncogene Proteins c-fos)
      0 (SOXB1 Transcription Factors)
      0 (Suppressor of Cytokine Signaling 3 Protein)
    • Publication Date:
      Date Created: 20210319 Date Completed: 20220310 Latest Revision: 20220531
    • Publication Date:
      20231215
    • Accession Number:
      10.1002/stem.3373
    • Accession Number:
      33739574