APP下载

Impact of glucocorticoid on neurogenesis

2017-08-07HarukiOdakaNaokiAdachiTadahiroNumakawa

Haruki Odaka, Naoki Adachi, Tadahiro Numakawa,,

1 Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan

2 Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan

3 Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Hyogo, Japan

4 Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan

Impact of glucocorticoid on neurogenesis

Haruki Odaka1,2, Naoki Adachi3,4, Tadahiro Numakawa2,4,*

1 Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan

2 Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan

3 Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Hyogo, Japan

4 Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan

How to cite this article:Odaka H, Adachi N, Numakawa T (2017) Impact of glucocorticoid on neurogenesis. Neural Regen Res 12(7):1028-1035.

Funding: This study was supported by grants from by Takeda Science Foundation (TN, NA), and from the Grant-in-Aid for Scientific Research(C) (JSPS KAKENHI JP16K06996) (to TN) and JSPS KAKENHI Grant Number 17J04183 (to HO) in the Ministry of Education, Culture, Sports, Science, and Technology of Japan.

Neurogenesis is currently an area of great interest in neuroscience. It is closely linked to brain diseases, including mental disorders and neurodevelopmental disease. Both embryonic and adult neurogeneses are in fl uenced by glucocorticoids secreted from the adrenal glands in response to a variety of stressors. Moreover, proliferation/di ff erentiation of the neural stem/progenitor cells (NSPCs) is a ff ected by glucocorticoids through intracellular signaling pathways such as phosphoinositide 3-kinase (PI3K)/Akt, hedgehog, and Wnt. Our review presents recent evidence of the impact of glucocorticoids on NSPC behaviors and the underlying molecular mechanisms; this provides important information for understanding the pathological role of glucocorticoids on neurogenesis-associated brain diseases.

neural progenitor cells; glucocorticoids; neurogenesis; intracellular signaling pathways

Accepted: 2017-07-07

Introduction

Glucocorticoids have been suggested to be involved in several brain diseases associated with stress such as post-traumatic stress disorder (PTSD), anxiety disorders, and major depressive disorder (MDD) (Holsboer et al., 2000; Herbert et al., 2013; Numakawa et al., 2013; Griffin et al., 2014; Raglan et al., 2017). Blood levels of glucocorticoids are increased in response to a variety of environmental stressors and are regulated by the negative feedback loop of the hypothalamic-pituitary-adrenal (HPA) axis (Ising et al., 2005; Owashi et al., 2008). Maternal stress during pregnancy has also been demonstrated to increase the risk of attention de fi cit hyperactivity disorder (ADHD) and autism spectrum disorder (ASD) in the offspring, through the elevation of glucocorticoid levels (Graignic-Philippeet et al., 2014).e impact of glucocorticoids on neuronal functions, including cell survival and synaptic plasticity in the central nervous system (CNS), has been extensively investigated throughin vitroandin vivostudies. Moreover, research has indicated that neural stem/progenitor cells (NSPCs) are an important target of excess glucocorticoids during stress.e resultant dysregulation of neurogenesis may thus be involved in the onset of the brain diseases mentioned above. Furthermore, impaired neurogenesis in the hippocampal region may disrupt the HPA-axis functions because hippocampal neurons play an essential role in the negative feedback regulation of the HPA-axis (Roozendaa et al., 2001; Furay et al., 2008).

Adult Neurogenesis and Major Depressive Disorder

Chronically elevated glucocorticoid levels under prolonged stress are the most common biological feature in MDD patients (Figure 1) (Numakawa et al., 2013). Animals chronically treated with CORT at a dose of 20 mg/kg/day for 25 days exhibited depressive behaviors and decreased neurogenesis in the hippocampal dentate gyrus (Sawamoto et al., 2016). Adrenalectomy (surgical removal of the adrenal glands) also prevents depressive behaviors and reduced neurogenesis in chronically stressed murines, suggesting that glucocorticoids are a major mediator of depressive behaviors and impaired neurogenesis under chronic stress (Lehmann et al., 2013). Importantly, the hippocampus negatively regulates HPA-axis activity in response to elevated blood glucocorticoid levels, raising the possibility that impaired hippocampal neurogenesis in fl uences the negative feedback function to regulate HPA-axis activity.

Schloesser et al. (2009) reported that suppression of hippocampal neurogenesis led to increased HPA response aer exposure to a mild stressor. In the study, NSPCs in the hippocampus were eliminated in transgenic mice using the herpes simplex virus thymidine kinase (HSV-tk) in glial fi brillary acidic protein (GFAP) positive NSPCs by administration of toxins specific to HSV-tk expressing proliferative cells. With X-irradiation of mouse hippocampi, Surget showed that ablation of hippocampal neurogenesis alone did not a ff ect the negative feedback in the HPA-axis; however, it diminished fl uoxetine-induced restoration of the hippocampal regulation of HPA-axis activity under chronic stress (Surget et al., 2011). It has also been reported that enhancing newborn neuron survival restored depressive behaviors in mice, without a ff ecting the HPA-axis regulation either at baseline or following CORT treatment (Hill et al., 2015).ese conflicting results suggest that the influence of hippocampal neurogenesis on the regulation of HPA-axis activity may vary based on the type of stress and antidepressant used.

Embryonic Neurogenesis and Neurodevelopmental Disorder

Embryonic neurogenesis may be involved in the onset of neurodevelopmental and psychiatric diseases; as such, it is an important target for investigating the impact of stress and glucocorticoids. Growing epidemiological evidence indicates that maternal stress during pregnancy increases the risk of ADHD, depression, schizophrenia, and ASDs in the o ff spring (Figure 1) (van Os and Selten, 1998; Van den Bergh and Marcoen, 2004; Beversdorf et al., 2005; Kinney et al., 2008; Khashan et al., 2008; Van den Bergh et al., 2008; Grizenko et al., 2012; Graignic-Philippe et al., 2014). Although the mechanisms of how maternal stress affects fetal brain development are not fully understood, excessive glucocorticoid transfer from mother to fetus is proposed as a key factor (Wilcoxon and Redei, 2007; Salomon et al., 2011). Catalytic conversion of glucocorticoid to cortisone by placental 11 beta-hydroxysteroid dehydrogenase type 2 prevents maternal glucocorticoid transfer to the fetus (Reynolds, 2013). Chronic severe stress and prolonged elevation of glucocorticoid levels in maternal serum exceeds catalytic conversion capacity and a considerable quantity of glucocorticoids then reaches the fetus (Reynolds, 2013). Fetal exposure to glucocorticoids can also be induced during therapeutic administration of synthetic glucocorticoids to promote fetal lung maturation. Such treatment is routinely used in obstetrical practice, although some clinical studies have demonstrated its adverse e ff ect on childhood cognition and long-term behavior (Crowther et al., 2007; French et al., 2009; Braun et al., 2013). However, it should be noted that reports regarding the long-term e ff ect of therapeutic glucocorticoid administration are still mixed; thus, further large-scale randomized controlled trials are required (Stutch fi eld et al., 2013).

Animals exposed to prenatal stress appear to exhibit a variety of behavioral abnormalities, including reduced exploration activity, decreased spatial memory, inability to extinguish conditioned fear memory, increased anxiety, and depressive behaviors (Alonso et al., 1991; Lordi et al., 2000; Schneider et al., 2002; Sundberg et al., 2006; Salomon et al., 2011; Anacker et al., 2013a; Bingham et al., 2013). Interestingly, anxiogenic behavior observed in prenatally-stressed rats (viaa combination of restraint, forced swim and elevated platform stress) were ameliorated by maternal adrenalectomy, which was reversed by maternal administration of high-dose CORT (Salomon et al., 2011). In another report, dams receiving both adrenalectomy and CORT administration produced o ff spring with increased depressive behavior (Wilcoxon and Redei, 2007). Maternal CORT treatment also impaired the ability to extinguish conditioned fear memory, a hallmark of PTSD, in o ff spring as well as in prenatally stressed rats (Bingham et al., 2013).ese studies essentially suggest that increased maternal serum levels of glucocorticoids could cause some behavioral abnormalities in offspring.

Although the impact of glucocorticoids on embryonic neurogenesis is still being investigated, several studies have demonstrated the detrimental e ff ects of perinatal glucocorticoid exposure on neurogenesis. Decreased body weight, hippocampal volume, and number of proliferating cells in the subventricular zone of the lateral ventricle, subgranular zone of the hippocampus, and cortex were observed in rats after administration of dexamethasone (DEX), a synthetic glucocorticoid, at postnatal days 4–7 (Kanagawa et al.,2006).Furthermore, a single administration of DEX on pregnant mice at embryonic day 15.5 resulted in decreased body weight and hippocampal volume, increased apoptotic cells in the hippocampus, and reduced cell proliferation in the subgranular zone of the dentate gyrus in pups (Noorlander et al., 2014). Remarkably, the prenatally DEX-treated mice exhibited a de fi cit in spatial memory, impaired hippocampal long-term depression, decreased hippocampal neurogenesis, and shortened lifespan in the adult period, indicating the long-lasting impact of prenatal DEX exposure on the CNS aer birth (Noorlander et al., 2008).

Glucocorticoid E ff ect on Neural Stem/ Progenitor Cells

To clarify the molecular mechanisms underlying neurogenesis impairment caused by increased glucocorticoids, severalin vitrostudies have proposed various impacts of glucocorticoids on the cellular system in NSPCs. Endogenous glucocorticoids have two speci fi c receptors: the mineralocorticoid receptor (MR) and the glucocorticoid receptor (GR), both of which can function as transcription factors. As MR has a high a ffi nity for glucocorticoids, it is occupied even at the basal blood glucocorticoid levels (De Kloet et al., 1998). On the other hand, GR has a low a ffi nity to glucocorticoids and is activated in response to increased levels of stress-induced glucocorticoids (De Kloet et al., 1998). Interestingly, MR and GR differentially affect the proliferation and differentiation of NSPCs. Anacker et al. (2013a) demonstrated that a low concentration of cortisol (human endogenous glucocorticoid) enhanced proliferation of human hippocampal progenitor cells and differentiation into astroglias, and suppressed di ff erentiation into neurons, through MR function. Meanwhile, a high concentration of cortisol decreased proliferation of NSPCs and neural differentiation without a ff ecting astroglial di ff erentiation,viaactivating GR (Anacker et al., 2013a).ese results suggest that the basal level of glucocorticoids modulates neurogenesisviaMR, while an increased level of glucocorticoids inhibits neurogenesisviaGR function. Consistent with this idea, the negative impact of glucocorticoids on neurogenesisviaGR activation has been reported in various types of NSPCs (Bose et al., 2010; Samarasinghe et al., 2011; Raciti et al., 2016).

It is well known that ligand binding triggers translocation of cytosolic GR to the nucleus, and subsequently, GR directly binds to the promoter region of target genes or modi fi es activity of other transcriptional factors (Mitre-Aguilar et al., 2015). In addition to such genomic actions, plasma membrane GR (mGR) rapidly activates intracellular signaling cascades in response to ligand binding, which is known as non-genomic functions (Mitre-Aguilar et al., 2015). Although little is known about the in fl uence of non-genomic pathways of GR on NSPCs, one study has reported a possible contribution of the non-genomic pathway on glucocorticoid-induced suppression of neurogenesis in cultured NSPCs (Samarasinghe et al., 2011). Activation of mGR in cultured NSPCs rapidly stimulated ERK1/2 signaling after DEX treatment in a Caveolin-1 dependent manner, which led to phosphorylation of connexin43. Because connexin43 diminishes gap junction intercellular communication (GJIC) and pharmacological GJIC inhibition is su ffi cient to suppress the proliferation of NSPCs, the mGR-dependent GJIC inhibition would contribute to NSPCs proliferation (Samarasinghe et al., 2011). Interestingly, the inhibitory action of mGR on NSPC proliferation lasted at least 24 hours, even though DEX exposure was transient (1 hour).ese fi ndings imply that proliferative activity of NSPCs can be interrupted by a transient increase of glucocorticoids triggered by stressful events.

In cultured rat embryonic NSPCs, DEX exposure for two days upregulated negative regulators of the cell cycle (p16 and p21) and senescence-related genes (high mobility group 1 and heterochromatin protein 1), and downregulated the mitochondrial genes (NADH dehydrogenase 3 and cytochrome b) concomitant with the suppression of NSPCs proliferation. Importantly, these features were retained for 10 days aer ceasing DEX exposure, resembling the long-lasting e ff ect of GR. Because DEX-exposed NSPCs showed decreased methylation of global DNA and expression of DNA methyltransferases, GR-mediated epigenetic events may also contribute to changes in gene expression pro fi les in daughter cells as a long-lasting influence of glucocorticoid exposure (Bose et al., 2010). This group recently showed that DEX treatment on human induced pluripotent stem cell-derived neuroepithelial-like stem cells exhibited a long-lasting decrease of neural di ff erentiation, with signi fi cant downregulation of antioxidants and increased intracellular reactive oxygen species generation. Decreased neural di ff erentiation was counteracted by the antioxidant N-acetyl-cysteine, suggesting that the intracellular redox system a ff ects neural di ff erentiation (Raciti et al., 2016). Importantly, the DEX-induced neural differentiation defect was accompanied by reduced expression of TrkB, a receptor of brain-derived neurotrophic factor (BDNF), which was also reversed by antioxidant treatment. Although the relationship between TrkB expression and neural differentiation was not elucidated in the study, the interference of glucocorticoids with the BDNF/TrkB signaling pathways is of interest given that BDNF is a potent enhancer of neural di ff erentiation and maturation.

Besides transcriptional regulation, alteration of the ubiquitin proteasome system (UPS) is also involved in the inhibitory e ff ect of glucocorticoids on neurogenesis. Rat embryonic NSPCs showed a decrease in cell proliferation after DEX exposure, which was accompanied by the reduction of cyclin D1 levels, a positive regulator of the cell cycle (Sundberg et al., 2006).e increased ubiquitination of cyclin D1 and counteraction of MG132 (an inhibitor of the UPS) reduced DEX e ff ects (on both expression of cyclin D1 and cell proliferation), implying an involvement of the UPS in the DEX-inhibited cell proliferation. Moreover, the authors proposed that baculoviral inhibitor of apoptosis repeat-containing 6 gene (BRUCE/Apollon) is another possible target of DEX-induced UPS alteration in the suppression of NSPCs proliferation (Sippel et al., 2009). Further, it was revealed that DEX increased the expression of the deubiquitinating enzymeUsp8/Ubpy, which decreased BRUCE possiblyviathe stabilization of BRUCE-targeting ubiquitin-protein ligase Nrdp1. Although the molecular mechanism behind increased cyclin D1 ubiquitination is still not clear, these fi ndings suggest that the alteration of the UPS may be accountable for part of the gene expression mediated by glucocorticoids.

Figure 1 Glucocorticoid hypothesis of neurodevelopmental disease and depressive disorder.

Figure 2 Inhibition of Akt signaling by corticosterone (CORT) exposure in neural stem/progenitor cells (NSPCs).

Figure 3 Possible crosstalk of intracellular signaling pathways a ff ected by glucocorticoid exposure.

Intrinsic and extrinsic signaling cascades regulate proliferation and di ff erentiation of NSPCs. Several groups, includingthe authors, have reported impairment of various signaling cascades by glucocorticoid exposure. Gene expression microarray combined with pathway analysis showed that a high-dose of cortisol inhibited TGF-β-SMAD2/3 and hedgehog signaling in human hippocampal progenitor cell lines (Anacker et al., 2013a).ese signaling abnormalities were also observed in the hippocampus of adult rats exposed to prenatal stress. In addition, hedgehog signaling activation by smoothened agonist, purmorphamine, canceled the inhibitory e ff ect of cortisol on neuronal di ff erentiation (Anacker et al., 2013a). This supports the possible involvement of hedgehog signaling in neurogenesis regulation. Serum- and glucocorticoid-inducible kinase 1 (SGK1), a direct target gene of GR, has been proposed as a contributor to repression of hedgehog signaling (Anacker et al., 2013b). In human hippocampal progenitor cell lines, an SGK1 inhibitor reversed the cortisol-induced suppression of hedgehog signaling, proliferation and neuronal di ff erentiation. Interestingly, SGK1 potentiated and maintained the cortisol-induced phosphorylation and nuclear translocation of GR even aer withdrawal of cortisol, implying a positive feedback role of SGK1 on GR function.is study indicates that SGK1 is one of the candidate factors regulating the long-lasting e ff ects of glucocorticoids. It may mediate hedgehog signaling indirectly, as the SGK1 inhibitor would reverse various GR-mediated phenotypes through repressing its positive feedback action.

Wnt signaling pathways are also involved in self-renewal, expansion, di ff erentiation and maturation of NSPCs in both the developing and adult brain (Bengoa-Vergniory et al., 2015).us, their correlation with glucocorticoids is salient. DEX treatment of human NSPCs inhibited both proliferation and neural differentiation of NSPCs, accompanied by increased Dickkopf1 (DKK1, endogenous Wnt-signaling antagonist) levels and reduced levels of canonical Wnt target genes including cyclin D1 and inhibitor of DNA binding 2 (ID2) (Moors et al., 2012). Neutralization with anti-DKK1 antibody antagonized the DEX-induced impairment in proliferation and di ff erentiation of NSPCs.e GR was also shown to bind to the promoter region of DKK1 gene, implying direct regulation of DKK1 expression by GR. Interestingly, DEX-induced alteration of DKK1/Wnt pathway was also reported in osteoblasts in the context of DEX-induced osteoporosis, indicating the importance of glucocorticoid-mediated DKK1/Wnt pathway in a variety of cell populations (Ohnaka et al., 2005).

ERK- and PI3K/Akt-signaling are critical in neuronal events including neurogenesis, as they are pivotal to signaling aer stimulation by growth factors (Samuels et al., 2009; Wang et al., 2017). Glucocorticoids exert a negative effect on these signaling cascades in various cell types, including neurons (Sandri et al., 2004; Smith et al., 2005; Horsch et al., 2007; González et al., 2010; Kumamaru et al., 2011). Recently, the authors found that ERK and Akt activities were increased duringin vitrodifferentiation of rat embryonic NSPCs, and this activation was decreased by CORT application (Odaka et al., 2016) (Figure 2).e importance of Akt activation for proper neural di ff erentiation was demonstrated using speci fi c inhibitors for these signaling pathways. A potent PI3K/Akt signaling activator, IGF1, counteracted the CORT-induced suppression of neural differentiation, suggesting its role in impaired neurogenesis. Considering that Wnt signaling inhibition caused a defect in neural differentiation, glycogen synthase kinase-3 beta (GSK-3β) is one of the possible downstream targets of PI3K/Akt signaling. Phosphorylation of GSK-3β by Akt inhibits the enzymatic activity of GSK-3β and results in the prevention of the proteasomal degradation of β-catenin, as is the case with canonical Wnt signaling pathways (Katoh et al., 2006). Indeed, it was demonstrated that glucocorticoid suppressed PI3K/Akt/ GSK-3β/β-catenin pathways in osteoblast-like cells (Smith et al., 2005). Moreover, anin vivostudy showed that endogenous adult neurogenesis was enhanced by the activation of the PI3K/Akt/GSK-3β system after cerebral ischemia, further supporting an involvement of these pathways in neurogenesis (Kisoh et al., 2016).

Another putative downstream target of PI3K/Akt is mTOR (mammalian target of rapamycin).e stimulation of mTOR complex1 (mTORC1) by the activation of PI3K/ Akt subsequently induces activation of p70 ribosomal S6 protein kinases 1/2 and inhibition of eukaryotic initiation factor 4E-bonding proteins, triggering the translational response of the mTOR cascade (Wang et al., 2017). PI3K/ Akt/mTOR signaling is essential for normal brain development. Deletion of mTOR in NSPCs disrupted progenitor self-renewal and suppressed neural di ff erentiation, and resulted in microcephaly (Ka et al., 2014; Wang et al., 2017). It is of note that mTOR activity is negatively regulated by GSK-3, implying that Wnt or PI3K/Akt-induced GSK-3β inhibition may enhance mTOR activity (Ka et al., 2014). GSK-3 also negatively regulated hedgehog signalingviatargeting a downstream molecule of hedgehog pathway Gli (Pan et al., 2006; Wang et al., 2006). Although further studies are needed, the negative action of glucocorticoids on DKK1/Wnt, PI3K/Akt, and hedgehog signaling appear to synergistically suppress neurogenesis through their crosstalk (Figure 3).

Downregulation of both PI3K/Akt and ERK pathways by glucocorticoids implies the possible involvement of receptor tyrosine kinases (RTKs, major activators of these signaling pathways). BDNF is one of the most-studied RTK activators as a target of glucocorticoid action. BDNF has multiple functions in both the embryonic and adult brain, such as NSPCs proliferation, differentiation, survival, and synaptic plasticity. Several studies report that exposure to stress and glucocorticoids decreases expression of BDNF in the hippocampus and/or the cortex in rodents (Smith et al., 1995; Schaaf et al., 1997; Dwivedi et al., 2006). Recent studies using the BZ cell line (established by targeted oncogenesis in mouse hippocampus) showed that GR was recruited to a promoter region of the BDNF gene to repress transcription through unidentified transcription factor tethering (Chen et al., 2017). Glucocorticoids also impair BDNF-stimulated intracellular signaling pathways. Generally, BDNF binds to TrkB to activate mainly three intracellular cascades; PI3K/Akt, MAPK/ERK, and PLC-γ pathways (Begni et al., 2017). The authors previously reported that DEX exposure attenuated the interaction of TrkB with Shp2, and subsequently suppressed ERK signaling and BDNF-induced enhancement of synaptic maturation in rat cortical neurons. DEX also inhibited BDNF-induced PLC-γ activation and its regulation of neurotransmitter release in rat cortical neurons (Numakawa et al., 2009). Intracellular interaction between GR and TrkB was also revealed, which is important in the TrkBPLC-γ interaction. Although inhibitory actions of glucocorticoids on BDNF-stimulated PI3K/Akt signaling in neural cells have not been reported, upregulation of p85α monomer caused by glucocorticoids inhibited Akt activity by competing for the RTK binding site with p110/p85 heterodimers in osteoblasts and myoblasts (Kuo et al., 2012; Zou et al., 2015). Although these mechanisms might be accountable for some glucocorticoid-induced phenotypes in NSPCs, further studies are required to explore the functional crosstalk between glucocorticoids and BDNF in NSPCs.

Future Directions

In this review, we outlined recent evidence on functional interactions between stress hormone glucocorticoids and neurogenesis. Investigating altered cell fate and related intracellular signaling in NSPCs a ff ected by glucocorticoid stress is important to reduce the risk of developmental brain diseases, including mental disorders and neurodegenerative diseases. In spite of di ff erences in cell population and niche, both embryonic and adult NSPCs exhibit similar phenotypes, including suppression of proliferation and neural di ff erentiation by high levels of glucocorticoids.ese phenotypic similarities might be attributed to a common molecular mechanism in embryonic and adult NSPCs. Studies on glucocorticoid-related impairments discussed above, however, were revealed by using NSPCs originating from embryos; thus, it should be ascertained if these mechanisms can also be applied in adult NSPCs.

Although GR is believed to be a major contributor of glucocorticoid stress, MR (which have a high a ffi nity receptor for glucocorticoids) act positively on neurogenesis (Anacker et al., 2013a), indicating that the functional balance between GR and MR is critical for glucocorticoid action in neurogenesis. Although genetic manipulation of GR in animals could be a promising tool for investigation, several studies on neurogenesis in GR knockout or knockdown mice have yielded inconsistent results. GR heterozygous mice displayed reduced hippocampal neurogenesis under the stress condition but not the basal condition (Kronenberg et al., 2009). Brain-specific deletion of GR did not affect neurogenesis in hippocampal granule cell layer in the basal condition (Gass et al., 2000). It is di ffi cult to evaluate the in fl uence of GR deletion in neurogenesis using systemic or whole-brain GR-de fi cient mice, as GR deletion in hippocampal or hypothalamic neurons disrupts the negative feedback loop of the HPA-axis.is in turn causes hypercorticoidism, which can interfere with neurogenesis. Indeed, cell type-speci fi c knockdown of GRin vivoshowed different results. For example, viral-mediated knockdown of GR selectively in hippocampal newborn cells resulted in enhancement of neural di ff erentiation and maturation under the basal condition (Fitzsimons et al., 2013). In the future, NSPC-specific manipulation of GR could be a powerful tool for precisely understanding GR-mediated actions in neurogenesisin vivo.

Author contributions:HO, NA and TN wrote and edited this manuscript.

Con fl icts of interest:None declared.

Open access statement:

Contributor agreement:A statement of “Publishing Agreement” has been signed by an authorized author on behalf of all authors prior to publication.

Plagiarism check:This paper has been checked twice with duplication-checking soware ienticate.

Peer review:A double-blind and stringent peer review process has been performed to ensure the integrity, quality and signi fi cance of this paper.

Open peer reviewers:Mariagrazia Grilli, University of Piemonte Orientale, Italy; Claire-Anne Gutekunst, Emory University School of Medicine, USA.

Alonso SJ, Arevalo R, Afonso D, Rodríguez M (1991) E ff ects of maternal stress during pregnancy on forced swimming test behavior of the o ff spring. Physiol Behav 50:511-517.

Anacker C, Cattaneo A, Luoni A, Musaelyan K, Zunszain PA, Milanesi E, Rybka J, Berry A, Cirulli F,uret S, Price J, Riva MA, Gennarelli M, Pariante CM (2013a) Glucocorticoid-related molecular signaling pathways regulating hippocampal neurogenesis. Neuropsychopharmacology 38:872-883.

Anacker C, Cattaneo A, Musaelyan K, Zunszain PA, Horowitz M, Molteni R, Luoni A, Calabrese F, Tansey K, Gennarelli M,uret S, Price J, Uher R, Riva MA, Pariante CM (2013b) Role for the kinase SGK1 in stress, depression, and glucocorticoid e ff ects on hippocampal neurogenesis. Proc Natl Acad Sci U S A 110:8708-8713.

Begni V, Riva MA, Cattaneo A (2017) Cellular and molecular mechanisms of the brain-derived neurotrophic factor in physiological and pathological conditions. Clin Sci (Lond) 131:123-138.

Bengoa-Vergniory N, Kypta RM (2015) Canonical and noncanonical Wnt signaling in neural stem/progenitor cells. Cell Mol Life Sci 72:4157-4172.

Beversdorf DQ, Manning SE, Hillier A, Anderson SL, Nordgren RE, Walters SE, Nagaraja HN, Cooley WC, Gaelic SE, Bauman ML (2005) Timing of prenatal stressors and autism. J Autism Dev Disord 35:471-478.

Bingham BC, Rani CS, Frazer A, Strong R, Morilak DA (2013) Exogenous prenatal corticosterone exposure mimics the e ff ects of prenatal stress on adult brain stress response systems and fear extinction behavior. Psychoneuroendocrinology 38:2746-2757.

Boldrini M, Underwood MD, Hen R, Rosoklija GB, Dwork AJ, John Mann J, Arango V (2009) Antidepressants increase neural progenitor cells in the human hippocampus. Neuropsychopharmacology 34:2376-2389.

Boldrini M, Santiago AN, Hen R, Dwork AJ, Rosoklija GB, Tamir H, Arango V, John Mann J (2013) Hippocampal granule neuron number and dentate gyrus volume in antidepressant-treated and untreated major depression. Neuropsychopharmacology 38:1068-1077.

Boldrini M, Hen R, Underwood MD, Rosoklija GB, Dwork AJ, Mann JJ, Arango V (2012) Hippocampal angiogenesis and progenitor cell proliferation are increased with antidepressant use in major depression. Biol Psychiatry 72:562-571.

Braun T, Challis JR, Newnham JP, Sloboda DM (2013) Early-life glucocorticoid exposure: the hypothalamic-pituitary-adrenal axis, placental function, and long-term disease risk. Endocr Rev 34:885-916.

Bose R, Moors M, To fi ghi R, Cascante A, Hermanson O, Ceccatelli S (2010) Glucocorticoids induce long-lasting effects in neural stem cells resulting in senescence-related alterations. Cell Death Dis 1:e92.

Chen H, Lombès M, Le Menuet D (2017) Glucocorticoid receptor represses brain-derived neurotrophic factor expression in neuron-like cells. Mol Brain 10:12.

Crowther CA, Doyle LW, Haslam RR, Hiller JE, Harding JE, Robinson JS; ACTORDS Study Group (2007) Outcomes at 2 years of age aer repeat doses of antenatal corticosteroids. N Engl J Med 357:1179-1189.

De Kloet ER, Vreugdenhil E, Oitzl MS, Joëls M (1998) Brain corticosteroid receptor balance in health and disease. Endocr Rev 19:269-301.

Dwivedi Y, Rizavi HS, Pandey GN (2006) Antidepressants reverse corticosterone-mediated decrease in brain-derived neurotrophic factor expression: differential regulation of specific exons by antidepressants and corticosterone. Neuroscience 139:1017-1029.

Fitzsimons CP, van Hooijdonk LW, Schouten M, Zalachoras I, Brinks V, Zheng T, Schouten TG, Saaltink DJ, Dijkmans T, Steindler DA, Verhaagen J, Verbeek FJ, Lucassen PJ, de Kloet ER, Meijer OC, Karst H, Joels M, Oitzl MS, Vreugdenhil E (2013) Knockdown of the glucocorticoid receptor alters functional integration of newborn neurons in the adult hippocampus and impairs fear-motivated behavior. Mol Psychiatry 18:993-1005.

French NP, Hagan R, Evans SF, Mullan A, Newnham JP (2004) Repeated antenatal corticosteroids: e ff ects on cerebral palsy and childhood behavior. Am J Obstet Gynecol 190:588-595.

Furay AR, Bruestle AE, Herman JP (2008) The role of the forebrain glucocorticoid receptor in acute and chronic stress. Endocrinology 149:5482-5490.

Gass P, Kretz O, Wolfer DP, Berger S, Tronche F, Reichardt HM, Kellendonk C, Lipp HP, Schmid W, Schütz G (2000) Genetic disruption of mineralocorticoid receptor leads to impaired neurogenesis and granule cell degeneration in the hippocampus of adult mice. EMBO Rep 1:447-451.

González R, Ruiz-León Y, Gomendio M, Roldan ER (2010)e e ff ect of glucocorticoids on ERK-1/2 phosphorylation during maturation of lamb oocytes and their subsequent fertilization and cleavage ability in vitro. Reprod Toxicol 29:198-205.

Graignic-Philippe R, Dayan J, Chokron S, Jacquet AY, Tordjman S (2014) Effects of prenatal stress on fetal and child development: a critical literature review. Neurosci Biobehav Rev 43:137-162.

Gri ffi n GD, Charron D, Al-Daccak R (2014) Post-traumatic stress disorder: revisiting adrenergics, glucocorticoids, immune system e ff ects and homeostasis. Clin Transl Immunology 3:e27

Grizenko N, Fortier ME, Zadorozny C, Thakur G, Schmitz N, Duval R, Joober R (2012) Maternal stress during pregnancy, ADHD symptomatology in children and genotype: gene-environment interaction. J Can Acad Child Adolesc Psychiatry 21:9-15.

Han J, Wang B, Xiao Z, Gao Y, Zhao Y, Zhang J, Chen B, Wang X, Dai J (2008) Mammalian target of rapamycin (mTOR) is involved in the neuronal differentiation of neural progenitors induced by insulin. Mol Cell Neurosci 39:118-124.

Herbert J (2013) Cortisol and depression: three questions for psychiatry. Psychol Med 43:449-469.

Hill AS, Sahay A, Hen R (2015) Increasing adult hippocampal neurogenesis is su ffi cient to reduce anxiety and depression-like behaviors. Neuropsychopharmacology 40:2368-2378.

Holsboer F (2000) The corticosteroid receptor hypothesis of depression. Neuropsychopharmacology 23: 477-501.

Horsch K, de Wet H, Schuurmans MM, Allie-Reid F, Cato AC, Cunningham J, Burrin JM, Hough FS, Hulley PA (2007) Mitogen-activated protein kinase phosphatase 1/dual specificity phosphatase 1 mediates glucocorticoid inhibition of osteoblast proliferation. Mol Endocrinol 21:2929-2940.

Ising M, Künzel HE, Binder EB, Nickel T, Modell S, Holsboer F (2005) The combined dexamethasone/CRH test as a potential surrogate marker in depression Prog Neuropsychopharmacol Biol Psychiatry 29:1085-1093

Jha S, Dong B, Sakata K (2011) Enriched environment treatment reverses depression-like behavior and restores reduced hippocampal neurogenesis and protein levels of brain-derived neurotrophic factor in mice lacking its expression through promoter IV. Transl Psychiatry 1:e40.

Ka M, Condorelli G, Woodgett JR, Km WY (2014) mTOR regulates brain morphogenesis by mediating GSK3 signaling. Development 141:4076-4086.

Kanagawa T, Tomimatsu T, Hayashi S, Shioji M, Fukuda H, Shimoya K, Murata Y (2006)e e ff ects of repeated corticosteroid administration on the neurogenesis in the neonatal rat. Am J Obstet Gynecol 194:231-238.

Katoh M, Katoh M (2006) Cross-talk of WNT and FGF signaling pathways at GSK3beta to regulate beta-catenin and SNAIL signaling cascades. Cancer Bioler 5:1059-1064.

Khashan AS, Abel KM, McNamee R, Pedersen MG, Webb RT, Baker PN, Kenny LC, Mortensen PB (2008) Higher risk of o ff spring schizophrenia following antenatal maternal exposure to severe adverse life events. Arch Gen Psychiatry 65:146-152.

Kinney DK, Miller AM, Crowley DJ, Huang E, Gerber E (2008) Autism prevalence following prenatal exposure to hurricanes and tropical storms in Louisiana. J Autism Dev Disord 38:481-488.

Kisoh K, Hayashi H, Itoh T, Asada M, Arai M, Yuan B, Tanonaka K, Takagi N (2016) Involvement of GSK-3β phosphorylation through PI3-K/Akt in cerebral ischemia-induced neurogenesis in rats. Mol Neurobiol doi:10.1007/s12035-016-0290-8.

Kiuchi T, Lee H, Mikami T (2012) Regular exercise cures depression-like behavior via VEGF-Flk-1 signaling in chronically stressed mice. Neuroscience 207:208-217.

Kleiderman S, Gutbier S, Ugur Tufekci K, Ortega F, Sá JV, Teixeira AP, Brito C, Glaab E, Berninger B, Alves PM, Leist M (2016) Conversion of nonproliferating astrocytes into neurogenic neural stem cells: control by FGF2 and interferon-γ. Stem Cells 34:2861-2874.

Kronenberg G, Kirste I, Inta D, Chourbaji S, Heuser I, Endres M, Gass P (2009) Reduced hippocampal neurogenesis in the GR(+/-) genetic mouse model of depression. Eur Arch Psychiatry Clin Neurosci 259:499-504.

Kumamaru E, Numakawa T, Adachi N, Kunugi H (2011) Glucocorticoid suppresses BDNF-stimulated MAPK/ERK pathway via inhibiting interaction of Shp2 with TrkB. FEBS Lett 585:3224-3228.

Kuo T, Lew MJ, Mayba O, Harris CA, Speed TP, Wang JC (2012) Genome-wide analysis of glucocorticoid receptor-binding sites in myotubes identifies gene networks modulating insulin signaling. Proc Natl Acad Sci U S A 109:11160-11165.

Lehmann ML, Brachman RA, Martinowich K, Schloesser RJ, Herkenham M (2013) Glucocorticoids orchestrate divergent e ff ects on mood through adult neurogenesis. J Neurosci 33:2961-2972.

Lenze EJ, Mantella RC, Shi P, Goate AM, Nowotny P, Butters MA, Andreescu C, Thompson PA, Rollman BL (2011) Elevated cortisol in older adults with generalized anxiety disorder is reduced by treatment: a placebo-controlled evaluation of escitalopram. Am J Geriatr Psychiatry 19:482-490.

Levone BR, Cryan JF, O’Leary OF (2014) Role of adult hippocampal neurogenesis in stress resilience. Neurobiol Stress 1:147-155.

Lordi B, Patin V, Protais P, Mellier D, Caston J (2000) Chronic stress in pregnant rats: e ff ects on growth rate, learning, and memory capabilities of the o ff spring. Int J Psychophysiol 37:195-205.

Madsen TM, Treschow A, Bengzon J, Bolwig TG, Lindvall O, Tingström A (2000) Increased neurogenesis in a model of electroconvulsive therapy. Biol Psychiatry 47:1043-1049.

Mitre-Aguilar IB, Cabrera-Quintero AJ, Zentella-Dehesa A (2015) Genomic and non-genomic e ff ects of glucocorticoids: implications for breast cancer. Int J Clin Exp Pathol 8:1-10.

Moors M, Bose R, Johansson-Haque K, Edo ff K, Okret S, Ceccatelli S (2012) Dickkopf 1 mediates glucocorticoid-induced changes in human neural progenitor cell proliferation and di ff erentiation. Toxicol Sci 125:488-495.

Noorlander CW, Visser GH, Ramakers GM, Nikkels PG, de Graan PN (2008) Prenatal corticosteroid exposure a ff ects hippocampal plasticity and reduces lifespan. Dev Neurobiol 68:237-246.

Noorlander CW, Tijsseling D, Hessel EV, de Vries WB, Derks JB, Visser GH, de Graan PN (2014). Antenatal glucocorticoid treatment a ff ects hippocampal development in mice. PLoS One 9:e85671.

Numakawa T, Adachi N, Richards M, Chiba S, Kunugi H (2013) Brain-derived neurotrophic factor and glucocorticoids: reciprocal influence on the central nervous system. Neuroscience 239:157-172.

Numakawa T, Kumamaru E, Adachi N, Yagasaki Y, Izumi A, Kunugi H (2009) Glucocorticoid receptor interaction with TrkB promotes BDNF-triggered PLC-gamma signaling for glutamate release via a glutamate transporter. Proc Natl Acad Sci U S A 106:647-652.

Odaka H, Numakawa T, Yoshimura A, Nakajima S, Adachi N, Ooshima Y, Inoue T, Kunugi H (2016) Chronic glucocorticoid exposure suppressed the di ff erentiation and survival of embryonic neural stem/ progenitor cells: Possible involvement of ERK and PI3K/Akt signaling in the neuronal di ff erentiation. Neurosci Res 113:28-36.

Ohnaka K, Tanabe M, Kawate H, Nawata H, Takayanagi R (2005) Glucocorticoid suppresses the canonical Wnt signal in cultured human osteoblasts. Biochem Biophys Res Commun 329:177-181.

Owashi T, Otsubo T, Oshima A, Nakagome K, Higuchi T, Kamijima K (2008) Longitudinal neuroendocrine changes assessed by dexamethasone/CRH and growth hormone releasing hormone tests in psychotic depression. Psychoneuroendocrinology 33:152-161.

Pan Y, Bai CB, Joyner AL, Wang B (2006) Sonic hedgehog signaling regulates Gli2 transcriptional activity by suppressing its processing and degradation. Mol Cell Biol 26:3365-3377.

Raciti M, Ong J, Weis L, Edo ff K, Battagli C, Falk A, Ceccatelli S (2016) Glucocorticoids alter neuronal differentiation of human neuroepithelial-like cells by inducing long-lasting changes in the reactive oxygen species balance. Neuropharmacology 107: 422-431.

Raglan GB, Schmidt LA, Schulkin J (2017)e role of glucocorticoids and corticotropin-releasing hormone regulation on anxiety symptoms and response to treatment. Endocr Connect 6:R1-R7.

Reynolds RM (2013) Glucocorticoid excess and the developmental origins of disease: two decades of testing the hypothesis – 2012 Curt Richter Award Winner. Psychoneuroendocrinology 38:1-11.

Roozendaal B, Phillips RG, Power AE, Brooke SM, Sapolsky RM, Mc-Gaugh JL (2001) Memory retrieval impairment induced by hippocampal CA3 lesions is blocked by adrenocortical suppression. Nat Neurosci 4:1169-1171.

Salomon S, Bejar C, Schorer-Apelbaum D, Weinstock M (2011) Corticosterone mediates some but not other behavioural changes induced by prenatal stress in rats. J Neuroendocrinol 23:118-128

Samarasinghe RA, Di Maio R, Volonte D, Galbiati F, Lewis M, Romero G, DeFranco DB (2011) Nongenomic glucocorticoid receptor action regulates gap junction intercellular communication and neural progenitor cell proliferation. Proc Natl Acad Sci U S A 108:16657-16662.

Samuels IS, Saitta SC, Landreth GE (2009) MAP’ing CNS development and cognition: an ERKsome process. Neuron 61: 160-167.

Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, Picard A, Walsh K, Schia ffi no S, Lecker SH, Goldberg AL (2004) Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell 117:399-412.

Santarelli L, Saxe M, Gross C, Surget A, Battaglia F, Dulawa S, Weisstaub N, Lee J, Duman R, Arancio O, Belzung C, Hen R (2003) Requirement of hippocampal neurogenesis for the behavioral e ff ects of antidepressants. Science 301:805-809.

Sawamoto A, Okuyama S, Yamamoto K, Amakura Y, Yoshimura M, Nakajima M, Furukawa Y (2016) 3,5,6,7,8,3′,4′-heptamethoxy fl avone, a citrus fl avonoid, ameliorates corticosterone-induced depression-like behavior and restores brain-derived neurotrophic factor expression, neurogenesis, and neuroplasticity in the hippocampus. Molecules 21:541.

Schaaf MJ, Hoetelmans RW, de Kloet ER, Vreugdenhil E (1997) Corticosterone regulates expression of BDNF and trkB but not NT-3 and trkC mRNA in the rat hippocampus. J Neurosci Res 48:334-341.

Schloesser RJ, Manji HK, Martinowich K (2009) Suppression of adult neurogenesis leads to an increased hypothalamo-pituitary-adrenal axis response. Neuroreport 20:553-557.

Schneider ML, Moore CF, Kraemer GW, Roberts AD, DeJesus OT (2002)e impact of prenatal stress, fetal alcohol exposure, or both on development: perspectives from a primate model. Psychoneuroendocrinology 27:285-298.

Schoenfeld TJ, Cameron HA (2015) Adult neurogenesis and mental illness. Neuropsychopharmacology 40:113-128.

Smith E, Frenkel B (2005) Glucocorticoids inhibit the transcriptional activity of LEF/TCF in differentiating osteoblasts in a glycogen synthase kinase-3beta-dependent and -independent manner. J Biol Chem 280:2388-2394.

Smith MA, Makino S, Kvetnansky R, Post RM (1995) Stress and glucocorticoids a ff ect the expression of brain-derived neurotrophic factor and neurotrophin-3 mRNAs in the hippocampus. J Neurosci (3 Pt 1):1768-1777.

Stutch fi eld PR, Whitaker R, Gliddon AE, Hobson L, Kotecha S, Doull IJ (2013) Behavioural, educational and respiratory outcomes of antenatal betamethasone for term caesarean section (ASTECS trial). Arch Dis Child Fetal Neonatal Ed 98:F195-200.

Sundberg M, Savola S, Hienola A, Korhonen L, Lindholm D (2006) Glucocorticoid hormones decrease proliferation of embryonic neural stem cells through ubiquitin-mediated degradation of cyclin D1. J Neurosci 26:5402-5410.

Surget A, Saxe M, Leman S, Ibarguen-Vargas Y, Chalon S, Griebel G, Hen R, Belzung C (2008) Drug-dependent requirement of hippocampal neurogenesis in a model of depression and of antidepressant reversal. Biol Psychiatry 64:293-301.

Surget A, Tanti A, Leonardo ED, Laugeray A, Rainer Q, Touma C, Palme R, Griebel G, Ibarguen-Vargas Y, Hen R, Belzung C (2011) Antidepressants recruit new neurons to improve stress response regulation. Mol Psychiatry 16:1177-1188.

Tang MM, Lin WJ, Pan YQ, Guan XT, Li YC (2016) Hippocampal neurogenesis dysfunction linked to depressive-like behaviors in a neuroinflammation induced model of depression. Physiol Behav 161:166-173.

Van den Bergh BR, Marcoen A (2004) High antenatal maternal anxiety is related to ADHD symptoms, externalizing problems, and anxiety in 8- and 9-year-olds. Child Dev 75:1085-1097.

Van den Bergh BR, Van Calster B, Smits T, Van Hu ff el S, Lagae L (2008) Antenatal maternal anxiety is related to HPA-axis dysregulation and self-reported depressive symptoms in adolescence: a prospective study on the fetal origins of depressed mood. Neuropsychopharmacology 33:536-545.

van Os J, Selten JP (1998) Prenatal exposure to maternal stress and subsequent schizophrenia.e May 1940 invasion ofe Netherlands. Br J Psychiatry 172:324-326.

Wang B, Li Y (2006) Evidence for the direct involvement of {beta}TrCP in Gli3 protein processing. Proc Natl Acad Sci U S A 103:33-38.

Wang L, Zhou K, Fu Z, Yu D, Huang H, Zang X, Mo X (2017) Brain development and Akt signaling: the crossroads of signaling pathway and neurodevelopmental diseases. J Mol Neurosci 61:379-384.

Wilcoxon JS, Redei EE (2007) Maternal glucocorticoid deficit affects hypothalamic-pituitary-adrenal function and behavior of rat offspring. Horm Behav 51:321-327.

Wong ML, Kling MA, Munson PJ, Listwak S, Licinio J, Prolo P, Karp B, McCutcheon IE, Geracioti TD Jr, DeBellis MD, Rice KC, Goldstein DS, Veldhuis JD, Chrousos GP, Old fi eld EH, McCann SM, Gold PW (2000) Pronounced and sustained central hypernoradrenergic function in major depression with melancholic features: relation to hypercortisolism and corticotropin-releasing hormone. Proc Natl Acad Sci U S A 97:325-330.

Yun S, Reynolds RP, Masiulis I, Eisch AJ (2016) Re-evaluating the link between neuropsychiatric disorders and dysregulated adult neurogenesis. Nat Med 22:1239-1247.

Zou W, Yang S, Zhang T, Sun H, Wang Y, Xue H, Zhou D (2015) Hypoxia enhances glucocorticoid-induced apoptosis and cell cycle arrest via the PI3K/Akt signaling pathway in osteoblastic cells. J Bone Miner Metab 33:615-624.

Tadahiro Numakawa, Ph.D., numakawa.yyrmk@gmail.com.

10.4103/1673-5374.211174

*< class="emphasis_italic">Correspondence to: Tadahiro Numakawa, Ph.D., numakawa.yyrmk@gmail.com.