Horm Metab Res 2013; 45(02): 159-168
DOI: 10.1055/s-0032-1321789
Original Basic
© Georg Thieme Verlag KG Stuttgart · New York

Expression Profiles of the Nuclear Receptors and Their Transcriptional Coregulators During Differentiation of Neural Stem Cells

A. Androutsellis-Theotokis
1   Department of Medicine, University of Dresden and Center for Regenerative Therapies-Dresden, Dresden, Germany
,
G. P. Chrousos
2   First Department of Pediatrics, Athens University Medical School, Athens, Greece
,
R. D. McKay
3   Laboratory of Molecular Biology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
,
A. H. DeCherney
4   Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
,
T. Kino
4   Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
› Author Affiliations
Further Information

Publication History

received 12 April 2012

accepted 13 June 2012

Publication Date:
18 September 2012 (online)

Abstract

Neural stem cells (NSCs) are pluripotent precursors with the ability to proliferate and differentiate into 3 neural cell lineages, neurons, astrocytes and oligodendrocytes. Elucidation of the mechanisms underlying these biologic processes is essential for understanding both physiologic and pathologic neural development and regeneration after injury. Nuclear hormone receptors (NRs) and their transcriptional coregulators also play crucial roles in neural development, functions and fate. To identify key NRs and their transcriptional regulators in NSC differentiation, we examined mRNA expression of 49 NRs and many of their coregulators during differentiation (0–5 days) of mouse embryonic NSCs induced by withdrawal of fibroblast growth factor-2 (FGF2). 37 out of 49 NRs were expressed in NSCs before induction of differentiation, while receptors known to play major roles in neural development, such as THRα, RXRs, RORs, TRs, and COUP-TFs, were highly expressed. CAR, which plays important roles in xenobiotic metabolism, was also highly expressed. FGF2 withdrawal induced mRNA expression of RORγ, RXRγ, and MR by over 20-fold. Most of the transcriptional coregulators examined were expressed basally and throughout differentiation without major changes, while FGF2 withdrawal strongly induced mRNA expression of several histone deacetylases (HDACs), including HDAC11. Dexamethasone and aldosterone, respectively a synthetic glucocorticoid and natural mineralocorticoid, increased NSC numbers and induced differentiation into neurons and astrocytes. These results indicate that the NRs and their coregulators are present and/or change their expression during NSC differentiation, suggesting that they may influence development of the central nervous system in the absence or presence of their ligands.

Supporting Information

 
  • References

  • 1 Burris TP. The nuclear receptor superfamily. In: Burris TP, McCabe ERB. (eds.) Nuclear receptors and genetic disease. London: Academic Press; 2001: 1-58
  • 2 Horn S, Heuer H. Thyroid hormone action during brain development: More questions than answers. Mol Cell Endocrinol 2010; 315: 19-26
  • 3 Wolkowitz OM, Burke H, Epel ES, Reus VI. Glucocorticoids. Mood, memory, and mechanisms. Ann N Y Acad Sci 2009; 1179: 19-40
  • 4 Hughes ZA, Liu F, Marquis K, Muniz L, Pangalos MN, Ring RH, Whiteside GT, Brandon NJ. Estrogen receptor neurobiology and its potential for translation into broad spectrum therapeutics for CNS disorders. Curr Mol Pharmacol 2009; 2: 215-236
  • 5 Kino T, Chrousos GP. Glucocorticoid effect on gene expression. In: Steckler T, Kalin NH, Reul JMHM. eds. Handbook on Stress and the Brain. Amsterdam: Elsevier BV; 2005: 295-312
  • 6 McKenna NJ, Lanz RB, O’Malley BW. Nuclear receptor coregulators: cellular and molecular biology. Endocr Rev 1999; 20: 321-344
  • 7 Kino T, Hurt DE, Ichijo T, Nader N, Chrousos GP. Noncoding RNA gas5 is a growth arrest- and starvation-associated repressor of the glucocorticoid receptor. Sci Signal 2010; 3: ra8
  • 8 Androutsellis-Theotokis A, Murase S, Boyd JD, Park DM, Hoeppner DJ, Ravin R, McKay RD. Generating neurons from stem cells. Methods Mol Biol 2008; 438: 31-38
  • 9 Androutsellis-Theotokis A, Rueger MA, Park DM, Mkhikian H, Korb E, Poser SW, Walbridge S, Munasinghe J, Koretsky AP, Lonser RR, McKay RD. Targeting neural precursors in the adult brain rescues injured dopamine neurons. Proc Natl Acad Sci USA 2009; 106: 13570-13575
  • 10 Androutsellis-Theotokis A, Rubin de Celis MF, Ehrhart-Bornstein M, Bornstein SR. Common features between chromaffin and neural progenitor cells. Mol Psychiatry 2012; 17: 351
  • 11 Bornstein SR, Ehrhart-Bornstein M, Androutsellis-Theotokis A, Eisenhofer G, Vukicevic V, Licinio J, Wong ML, Calissano P, Nistico G, Preziosi P, Levi-Montalcini R. Chromaffin cells: the peripheral brain. Mol Psychiatry 2012; 17: 354-358
  • 12 Androutsellis-Theotokis A, Leker RR, Soldner F, Hoeppner DJ, Ravin R, Poser SW, Rueger MA, Bae SK, Kittappa R, McKay RD. Notch signalling regulates stem cell numbers in vitro and in vivo. Nature 2006; 442: 823-826
  • 13 Androutsellis-Theotokis A, Rueger MA, Mkhikian H, Korb E, McKay RD. Signaling pathways controlling neural stem cells slow progressive brain disease. Cold Spring Harb Symp Quant Biol 2008; 73: 403-410
  • 14 Kittappa R, Bornstein SR, Androutsellis-Theotokis A. The role of eNSCs in neurodegenerative disease. Mol Neurobiol 2012; in press
  • 15 Ng SS, Chang TH, Tailor P, Ozato K, Kino T. Virus-induced differential expression of nuclear receptors and coregulators in dendritic cells: Implication to interferon production. FEBS Lett 2011; 585: 1331-1337
  • 16 Kino T, Ichijo T, Amin ND, Kesavapany S, Wang Y, Kim N, Rao S, Player A, Zheng YL, Garabedian MJ, Kawasaki E, Pant HC, Chrousos GP. Cyclin-dependent kinase 5 differentially regulates the transcriptional activity of the glucocorticoid receptor through phosphorylation: Clinical Implications for the nervous system response to glucocorticoids and stress. Mol Endocrinol 2007; 21: 1552-1568
  • 17 van Neerven S, Kampmann E, Mey J. RAR/RXR and PPAR/RXR signaling in neurological and psychiatric diseases. Prog Neurobiol 2008; 85: 433-451
  • 18 Pereira FA, Qiu Y, Tsai MJ, Tsai SY. Chicken ovalbumin upstream promoter transcription factor (COUP-TF) expression during mouse embryogenesis. J Steroid Biochem Mol Biol 1995; 53: 503-508
  • 19 Levesque D, Rouillard C. Nur77 and retinoid X receptors: Crucial factors in dopamine-related neuroadaptation. Trends Neurosci 2007; 30: 22-30
  • 20 Hall MG, Quignodon L, Desvergne B. Peroxisome proliferator-activated receptor β/δ in the brain: Facts and hypothesis. PPAR Res 2008; 2008: 780452
  • 21 Lee YF, Lee HJ, Chang C. Recent advances in the TR2 and TR4 orphan receptors of the nuclear receptor superfamily. J Steroid Biochem Mol Biol 2002; 81: 291-308
  • 22 Schnegg CI, Robbins ME. Neuroprotective mechanisms of PPARδ: Modulation of oxidative stress and inflammatory processes. PPAR Res 2011; 2011: 373560
  • 23 Real MA, Heredia R, Davila JC, Guirado S. Efferent retinal projections visualized by immunohistochemical detection of the estrogen-related receptor β in the postnatal and adult mouse brain. Neurosci Lett 2008; 438: 48-53
  • 24 Grgurevic N, Tobet S, Majdic G. Widespread expression of liver receptor homolog 1 in mouse brain. Neuro Endocrinol Lett 2005; 26: 541-547
  • 25 Seckl JR, Walker BR. Minireview: 11β-hydroxysteroid dehydrogenase type 1 – a tissue-specific amplifier of glucocorticoid action. Endocrinology 2001; 142: 1371-1376
  • 26 Almad A, McTigue DM. Chronic expression of PPAR-δ by oligodendrocyte lineage cells in the injured rat spinal cord. J Comp Neurol 2010; 518: 785-799
  • 27 D’Angelo B, Benedetti E, Di Loreto S, Cristiano L, Laurenti G, Ceru MP, Cimini A. Signal transduction pathways involved in PPARβ/δ-induced neuronal differentiation. J Cell Physiol 2011; 226: 2170-2180
  • 28 Schnegg CI, Kooshki M, Hsu FC, Sui G, Robbins ME. PPARδ prevents radiation-induced proinflammatory responses in microglia via transrepression of NF-κB and inhibition of the PKCα/MEK1/2/ERK1/2/AP-1 pathway. Free Radic Biol Med 2012; 52: 1734-1743
  • 29 Siegel DA, Davies P, Dobrenis K, Huang M. Tomoregulin-2 is found extensively in plaques in Alzheimer’s disease brain. J Neurochem 2006; 98: 34-44
  • 30 Zhou XE, Suino-Powell KM, Xu Y, Chan CW, Tanabe O, Kruse SW, Reynolds R, Engel JD, Xu HE. The orphan nuclear receptor TR4 is a vitamin A-activated nuclear receptor. J Biol Chem 2011; 286: 2877-2885
  • 31 Sousa N, Almeida OF. Corticosteroids: Sculptors of the hippocampal formation. Rev Neurosci 2002; 13: 59-84
  • 32 Kino T, Chrousos GP. Glucocorticoid and mineralocorticoid resistance/hypersensitivity syndromes. J Endocrinol 2001; 169: 437-445
  • 33 Chrousos GP. Glucocorticoid therapy. In: Felig P, Frohman LA. eds. Endocrinology & Metablism. 4th ed. New York: McGraw-Hill; 2001: 609-632
  • 34 Aimone JB, Wiles J, Gage FH. Potential role for adult neurogenesis in the encoding of time in new memories. Nat Neurosci 2006; 9: 723-727
  • 35 Kukolja J, Thiel CM, Wolf OT, Fink GR. Increased cortisol levels in cognitively challenging situations are beneficial in young but not older subjects. Psychopharmacology (Berl) 2008; 201: 293-304
  • 36 Gass P, Kretz O, Wolfer DP, Berger S, Tronche F, Reichardt HM, Kellendonk C, Lipp HP, Schmid W, Schutz G. Genetic disruption of mineralocorticoid receptor leads to impaired neurogenesis and granule cell degeneration in the hippocampus of adult mice. EMBO Rep 2000; 1: 447-451
  • 37 Yu S, Yang S, Holsboer F, Sousa N, Almeida OF. Glucocorticoid regulation of astrocytic fate and function. PLoS One 2011; 6: e22419
  • 38 Yu S, Patchev AV, Wu Y, Lu J, Holsboer F, Zhang JZ, Sousa N, Almeida OF. Depletion of the neural precursor cell pool by glucocorticoids. Ann Neurol 2010; 67: 21-30
  • 39 Kazantsev AG, Thompson LM. Therapeutic application of histone deacetylase inhibitors for central nervous system disorders. Nat Rev Drug Discov 2008; 7: 854-868
  • 40 Sun G, Yu RT, Evans RM, Shi Y. Orphan nuclear receptor TLX recruits histone deacetylases to repress transcription and regulate neural stem cell proliferation. Proc Natl Acad Sci USA 2007; 104: 15282-15287
  • 41 Sun G, Fu C, Shen C, Shi Y. Histone deacetylases in neural stem cells and induced pluripotent stem cells. J Biomed Biotechnol 2011; 2011: 835968
  • 42 Chateauvieux S, Morceau F, Dicato M, Diederich M. Molecular and therapeutic potential and toxicity of valproic acid. J Biomed Biotechnol 2010; 2010: 479364
  • 43 Faraco G, Cavone L, Chiarugi A. The therapeutic potential of HDAC inhibitors in the treatment of multiple sclerosis. Mol Med 2011; 17: 442-447
  • 44 Fischer A, Sananbenesi F, Mungenast A, Tsai LH. Targeting the correct HDAC(s) to treat cognitive disorders. Trends Pharmacol Sci 2010; 31: 605-617
  • 45 Shein NA, Shohami E. Histone deacetylase inhibitors as therapeutic agents for acute central nervous system injuries. Mol Med 2011; 17: 448-456