Effects of neonatal dexamethasone and CpdA on the expression of genes for apoptosis regulator proteins in the neonatal hippocampus

Авторы

  • Дмитрий Александрович Ланшаков Институт цитологии и генетики СО РАН https://orcid.org/0000-0002-8482-1302
  • Екатерина Викторовна Сухарева Институт цитологии и генетики СО РАН https://orcid.org/0000-0001-9263-9167
  • Вета Вячеславовна Булыгина Институт цитологии и генетики СО РАН
  • Тимофей Аркадьевич Лагунов Новосибирский государственный университет; Лаботатория цитометрии и биокинетики Института химической кинетики и горения
  • Татьяна Сергеевна Калинина Институт цитологии и генетики СО РАН https://orcid.org/0000-0002-2575-4621

DOI:

https://doi.org/10.33910/2687-1270-2021-2-1-41-48

Ключевые слова:

glucocorticoid receptor, brain cell type markers, development, hippocampus, neocortex, apoptosis regulator protein, proximity ligation assay (PLA)

Аннотация

Glucocorticoids (GC) are crucial regulators of homeostasis and function. Despite its negative side effects, glucocorticoid therapy in neonates is widely used antenatally for accelerating fetal lung maturation in cases of preterm birth. GC action is mediated via glucocorticoid receptors — ligand-activated transcription factors. Cell death and viability in the neonatal brain are regulated by many factors, but the glucocorticoid receptor signalling is high above them. The present work studies the changes in the expression of genes for apoptosis regulators with Bcl-2 homology (BH) domains (Bcl-xL, Bax, Bim, Bok, Bid) in the neonatal rat hippocampus after dexamethasone (DEX) and CpdA administration. CpdA is a dissociative ligand — glucocorticoid receptor modulator — that shifts glucocorticoid receptor (GR) activity toward transrepression. Ligands administration to P2 pups caused different patterns of timeline changes in the expression of the studied genes. We observed the first increase in the mRNA level of the genes which have glucocorticoid response element (GRE) (Bcl-xL, Bim) in their promoter 30 min after DEX administration. Activated GR action on cells in the neonatal hippocampus is complex and long-lasting; it could also contain receptor homo- and hetero-dimerisation. Using rat pheochromocytoma PC12 cells as a test system, we assessed GR-GR and GR-MR (mineralocorticoid receptor) dimerisation with proximity ligation assay (PLA) assay separately in the nucleus and cytoplasm after DEX and CpdA administration. An increase in GR-GR dimers in the cell nucleus was observed only after DEX administration. In the cell cytoplasm, we observed a gradual (DEX more than CpdA) increase in the number of both GR-GR and GR-MR dimers.

Библиографические ссылки

Alotaibi, M., Arrowsmith, S., Wray, S. (2015) Hypoxia-induced force increase (HIFI) is a novel mechanism underlying the strengthening of labor contractions, produced by hypoxic stresses. Proceedings of the National Academy of Sciences of the United States of America, vol. 112, no. 31, pp. 9763–9768. https://www.doi.org/10.1073/pnas.1503497112 (In English)

Barrington, K. J. (2001) The adverse neuro-developmental effects of postnatal steroids in the preterm infant: A systematic review of RCTs. BioMed Central Pediatrics, vol. 1, article 1. https://www.doi.org/10.1186/1471-2431-1-1 (In English)

Bulygina, V. V., Men’shanov, P. N., Lanshakov, D. A., Dygalo, N. N. (2014) The effects of dexamethasone and hypoxia on the content of active caspase-3 in the cerebellum and the behavior of neonatal rats. Biology Bulletin, vol. 41, no. 6, pp. 540–544. https://www.doi.org/10.1134/s1062359014060028 (In English)

Cao, J., Zhou, W., Steemers, F. et al. (2020) Sci-fate characterizes the dynamics of gene expression in single cells. Nature Biotechnology, vol. 38, no. 8, pp. 980–988. https://www.doi.org/10.1038/s41587-020-0480-9 (In English)

de Kloet, E. R., Claessens, S. E. F., Kentrop, J. (2014) Context modulates outcome of perinatal glucocorticoid action in the brain. Frontiers in Endocrinolology, vol. 5, article 100. https://doi.org/10.3389/fendo.2014.00100 (In English)

Dezitter, X., Fagart, J., Taront, S. et al. (2014) A structural explanation of the effects of dissociated glucocorticoids on glucocorticoid receptor transactivation. Molecular Pharmacology, vol. 85, no. 2, pp. 226–236. https://doi.org/10.1124/mol.113.085860 (In English)

Drozd, U. S., Lanshakov, D. A. (2020) Creation of the viral vectors for the inhibition of the serotonergic neurons using light sensitive proton pump. Integrative Physiology, vol. 1, no. 2, pp. 144–146. https://www.doi.org/10.33910/2687-1270-2020-1-2-144-146 (In English)

Du, H., Zhao, Y., He, J. et al. (2016) YTHDF2 destabilizes m6A-containing RNA through direct recruitment of the CCR4-NOT deadenylase complex. Nature Communication, vol. 7, article 12626. https://www.doi.org/10.1038/ncomms12626 (In English)

Holson, R. R., Gough, B., Sullivan, P. et al. (1995) Prenatal dexamethasone or stress but not ACTH or corticos-terone alter sexual behavior in male rats. Neurotoxicol Teratology, vol. 17, no. 4, pp. 393–401. https://www.doi.org/10.1016/0892-0362(94)00074-n (In English)

Kalinina, T., Sukhareva, E., Bulygina, V. et al. (2019) P.216 Long-term up-regulation of tyrosine hydroxylase gene expression after neonatal dexamethasone. European Neuropsychopharmacology, vol. 29, suppl. 6, pp. S166–S167. https://www.doi.org/10.1016/j.euroneuro.2019.09.259 (In English)

Kalkat, M., Garcia, J., Ebrahimi, J. et al. (2013) Placental autophagy regulation by the BOK-MCL1 rheostat. Autophagy, vol. 9, no. 12, pp. 2140–2153. https://www.doi.org/10.4161/auto.26452 (In English)

Lanshakov, D. A., Drozd, U. S., Dygalo, N. N. (2017) Optogenetic stimulation increases level of antiapoptotic protein Bcl-xL in Neurons. Biochemistry (Moscow), vol. 82, no. 3, pp. 340–344. https://www.doi.org/10.1134/S0006297917030129 (In English)

Lanshakov, D. A., Sukhareva, E. V., Kalinina, T. S., Dygalo, N. N. (2016) Dexamethasone-induced acute excitotoxic cell death in the developing brain. Neurobiology of Disease, vol. 91, pp. 1–9. https://www.doi.org/10.1016/j.nbd.2016.02.009 (In English)

Lee, M.-S., Kim, Y.-H., Park, W.-S. et al. (2016) Temporal variability of glucocorticoid receptor activity is functionally important for the therapeutic action of fluoxetine in the hippocampus. Molecular Psychiatry, vol. 21, no. 2, pp. 252–260. https://www.doi.org/10.1038/mp.2014.137 (In English)

Li, S.-X., Fujita, Y., Zhang, J.-C. et al. (2014) Role of the NMDA receptor in cognitive deficits, anxiety and depressive-like behavior in juvenile and adult mice after neonatal dexamethasone exposure. Neurobiology of Disease, vol. 62, pp. 124–134. https://www.doi.org/10.1016/j.nbd.2013.09.004 (In English)

Menshanov, P. N., Lanshakov, D. A., Dygalo, N. N. (2015) proBDNF is a major product of bdnf gene expressed in the perinatal rat cortex. Physiological Research, vol. 64, no. 6, pp. 925–934. https://www.doi.org/10.33549/physiolres.932996 (In English)

Nagano, M., Ozawa, H., Suzuki, H. (2008) Prenatal dexamethasone exposure affects anxiety-like behaviour and neuroendocrine systems in an age-dependent manner. Neuroscience Research, vol. 60, no. 4, pp. 364–371. https://www.doi.org/10.1016/j.neures.2007.12.005 (In English)

Oitzl, M. S., Champagne, D. L., van der Veen, R., de Kloet, E. R. (2010) Brain development under stress: Hypotheses of glucocorticoid actions revisited. Neuroscience & Biobehavioral Reviews, vol. 34, no. 6, pp. 853–866. https://www.doi.org/10.1016/j.neubiorev.2009.07.006 (In English)

Reddy, T. E., Pauli, F., Sprouse, R. O. et al. (2009) Genomic determination of the glucocorticoid response reveals unexpected mechanisms of gene regulation. Genome Research, vol. 19, no. 12, pp. 2163–2171. https://www.doi.org/10.1101/gr.097022.109 (In English)

Reynolds, R. M. (2013) Glucocorticoid excess and the developmental origins of disease: Two decades of testing the hypothesis – 2012 Curt Richter Award Winner. Psychoneuroendocrinology, vol. 38, no. 1, pp. 1–11. https://www.doi.org/10.1016/j.psyneuen.2012.08.012 (In English)

Shaburova, E. V., Lanshakov, D. A. (2020) Effects of ketamine and stress on the neurotrophin receptors expression. Integrativnaya fiziologiya — Integrative Physiology, vol. 1, no. 1, pp. 75–77. https://www.doi.org/10.33910/2687-1270-2020-1-1-75-77 (In English)

Shishkina, G. T., Bulygina, V. V. Dygalo, N. N. (2015a) Behavioral effects of glucocorticoids during the first exposures to the forced swim stress. Psychopharmacology, vol. 232, no. 5, pp. 851–860. https://www.doi.org/10.1007/s00213-014-3718-8 (in English)

Shishkina, G. T., Kalinina, T. S., Bulygina, V. V. et al. (2015b) Anti-apoptotic protein Bcl-xL expression in the midbrain raphe region is sensitive to stress and glucocorticoids. PLoS One, vol. 10, no. 12, article 0143978. https://www.doi.org/10.1371/journal.pone.0143978 (In English)

Surjit, M., Ganti, K. P., Mukherji, A. et al. (2011) Widespread negative response elements mediate direct repression by agonist-liganded glucocorticoid receptor. Cell, vol. 145, no. 2, pp. 224–241. https://www.doi.org/10.1016/j.cell.2011.03.027 (In English)

Tiwari, M., Oasa, S., Yamamoto, J. et al. (2017) A quantitative study of internal and external interactions of homodimeric glucocorticoid receptor using fluorescence cross-correlation spectroscopy in a live cell. Scientific Reports, vol. 7, article 4336. https://www.doi.org/10.1038/s41598-017-04499-7 (In English)

van Delft, M. F., Huang, D. C. S. (2006) How the Bcl-2 family of proteins interact to regulate apoptosis. Cell Research, vol. 16, no. 2, pp. 203–213. https://www.doi.org/10.1038/sj.cr.7310028 (In English)

Опубликован

27.05.2021

Выпуск

Раздел

Экспериментальные статьи