forkhead box o transcription factors as possible mediators in …368852/uq368852... · 2019. 10....

37
Accepted Manuscript Forkhead Box O Transcription Factors as Possible Mediators in the Development of Major Depression Haitao Wang, Rémi Quirion, Peter J. Little, Yufang Cheng, Zhong-Ping Feng, Hong- Shuo Sun, Prof. Jiangping Xu, Ph.D, MD., Prof. Wenhua Zheng, MD, PhD. PII: S0028-3908(15)30066-6 DOI: 10.1016/j.neuropharm.2015.08.020 Reference: NP 5966 To appear in: Neuropharmacology Received Date: 11 March 2015 Revised Date: 22 July 2015 Accepted Date: 12 August 2015 Please cite this article as: Wang, H., Quirion, R., Little, P.J., Cheng, Y., Feng, Z.-P., Sun, H.-S., Xu, J., Zheng, W., Forkhead Box O Transcription Factors as Possible Mediators in the Development of Major Depression, Neuropharmacology (2015), doi: 10.1016/j.neuropharm.2015.08.020. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Upload: others

Post on 14-Sep-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

Accepted Manuscript

Forkhead Box O Transcription Factors as Possible Mediators in the Development ofMajor Depression

Haitao Wang, Rémi Quirion, Peter J. Little, Yufang Cheng, Zhong-Ping Feng, Hong-Shuo Sun, Prof. Jiangping Xu, Ph.D, MD., Prof. Wenhua Zheng, MD, PhD.

PII: S0028-3908(15)30066-6

DOI: 10.1016/j.neuropharm.2015.08.020

Reference: NP 5966

To appear in: Neuropharmacology

Received Date: 11 March 2015

Revised Date: 22 July 2015

Accepted Date: 12 August 2015

Please cite this article as: Wang, H., Quirion, R., Little, P.J., Cheng, Y., Feng, Z.-P., Sun, H.-S., Xu, J.,Zheng, W., Forkhead Box O Transcription Factors as Possible Mediators in the Development of MajorDepression, Neuropharmacology (2015), doi: 10.1016/j.neuropharm.2015.08.020.

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service toour customers we are providing this early version of the manuscript. The manuscript will undergocopyediting, typesetting, and review of the resulting proof before it is published in its final form. Pleasenote that during the production process errors may be discovered which could affect the content, and alllegal disclaimers that apply to the journal pertain.

Page 2: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

1

Forkhead Box O Transcription Factors as Possible Mediators in

the Development of Major Depression

Haitao Wang1,2, Rémi Quirion3, Peter J. Little4, Yufang Cheng1, Zhong-Ping Feng5,

Hong-Shuo Sun5, Jiangping Xu1,*, Wenhua Zheng2,*

1Guangdong Provincial Key Laboratory of New Drug Screening, School of

Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China;

2 Faculty of Health Sciences, University of Macau, Taipa, Macau, China and Key

Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University,

Guangzhou 510006, China.

3Douglas Mental Health University Institute, McGill University, Montreal, Canada;

4 School of Pharmacy, The University of Queensland, QLD 4072, Australia;

5 Department of Physiology, Faculty of Medicine, University of Toronto, Toronto,

Ontario, Canada.

*Corresponding author Prof. Jiangping Xu, Ph.D, MD. Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China. Tel:/Fax:+86 20 6164 8236, E-mail address: [email protected] Prof. Wenhua Zheng, MD, PhD. Faculty of Health Sciences, University of Macau Room 4021, Building E12, Avenida de Universidade, Taipa, Macau. China Tel: +853-88224919; Email: [email protected]

Page 3: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

2

Abstract

Forkhead box O (FoxO) transcription factors play important roles in cellular

physiology and biology. Recent findings indicate that FoxOs are also involved in the

development of major depressive disorder. Alterations in the upstream molecules of

FoxOs, such as brain derived neurotrophic factor or protein kinase B, have been

linked to depression. Antidepressants, such as imipramine and venlafaxine, modify

the FoxOs phosphorylation. Furthermore, FoxOs could be regulated by serotonin and

norepinephrine receptor signaling as well as the hypothalamic-pituitary-adrenal axis,

all of which are involved in the pathogenesis of depression. FoxOs also regulate

neuronal morphology, synaptogenesis and adult hippocampal neurogenesis, which are

viewed as candidate mechanisms for the etiology of depression. In this review, we

emphasize the possible roles of FoxOs during the development of depression and

make some strategic recommendations for future research. We propose that FoxOs

and its signaling pathways may constitute potential therapeutic targets in the treatment

of depression.

Keywords: FoxO, major depression, PI3K/Akt, neuronal atrophy, neurogenesis,

antidepressants

Page 4: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

3

1. Introduction

Major depression is one of the most severe and common psychiatric disorders

with high rates of self-harm and suicide attempts (Hegerl et al., 2013). The causes of

major depressive disorder are not well understood. A diverse contribution of genetic,

neurochemical and environmental factors are involved in the onset and progression of

depression. Besides the common monoamine hypotheses, the diathesis stress model is

another attempt to explain the etiology of depressed behavior (Gold and Chrousos,

2013). Diathesis interacts with the subsequent stress response of an individual,

especially some early life experiences which are known to increase the risk for

depression (Kiejna et al., 2010). Antidepressants initially correct imbalances of

neurotransmitters in the synapse cleft (Mahar et al., 2014; Werner and Coveñas, 2013),

while the therapeutic response may result from above and the downstream adaptive

changes that occur as a consequence of increased neuroplasticity. These subcellular

events include synaptic transmission, gene expression, neuronal morphological

changes and many other molecular events in the brain (Kavalali and Monteggia, 2012;

Pilar-Cuéllar et al., 2012; Seo et al., 2014). However, some patients do not tolerate

some of the undesirable effects associated with these agents such as nervousness,

insomnia or sexual dysfunction (Sussman. 1994). Novel targets and new classes of

antidepressant agents are required to improve the therapeutic arsenal for the treatment

of depression. In addition, currently available antidepressants are prescribed for both

moderately and severely depressed patients (Fournier et al., 2010), and for severe

major depression, a combination of antidepressant medication and psychotherapy or

electroconvulsive therapy are usually considered (Oudega et al., 2011; Hollon et al.,

2014). Antidepressants have unusual temporal responses with many agents having a

delayed onset of action since clinical improvement is observed several weeks after

initiation of antidepressant drug and usually patients have to take antidepressants for

months to get a stable maximal improvement (Kirsch et al., 2008; Mitchell. 2006).

Antidepressants require a timescale of minutes to boost the synaptic concentration of

serotonin and/or norepinephrine, whereas the onset of the therapeutic action is usually

a few weeks after the initiation of therapy. The traditional view about the delayed

Page 5: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

4

onset of current antidepressant action relies on neurobiological effects that develop

slowly under chronic drug treatment, such as alterations of glucocorticoid receptor

systems in patients with depression, and changes of gene and protein expression

related to dendritic spines, production of brain derived neurotrophic factor (BDNF),

adult hippocampal neurogenesis and enhanced synaptic plasticity (Hajszan et al., 2005;

Anacker et al., 2011b; Malberg et al., 2000; Groves. 2007). The pathological

mechanisms of depression need to be further explored and the controversy about the

mechanism of current antidepressant agents to be resolved.

In recent years, the regulation of gene expression has been implicated in the

etiology and treatment of depression; several genes such as BDNF, fibroblast growth

factor receptor 1, NCAM1 neural cell adhesion molecule 1, and

Calcium/Calmodulin-dependent Protein Kinase II have been identified to be of

interest (Groves. 2007; Tochigi et al., 2008). Transcription factor DNA binding

peptides are essential for the regulation of gene expression and are themselves

regulated by phosphorylation and dephosphorylation. For example, phosphorylated

cAMP-response element binding protein (CREB) transcription factor binds to the

regulatory site on the BDNF promoter and increases BDNF protein levels, both of

which have been associated with depression (Breuillaud et al., 2012). Transcription

factors could serve as the intermediates between intracellular signaling cascades and

gene expression, and therefore be involved in the pathophysiology of depression and

represent potential targets for the pharmacotherapy.

Forkhead box O (FoxO) is a transcription factor, which plays a regulatory role in

multiple biological and pathological systems, including the central nervous system

(CNS). Recent discoveries indicate a role for FoxO in the pathogenesis of

depression and other psychiatric disorders (Polter et al., 2009; Weeks et al., 2010;

Zheng et al., 2013). Enhanced neurotrophins or serotonin neurotransmission in animal

brain phosphorylate and inactivate FoxO3a (Zheng et al., 2002; Polter et al., 2009).

BDNF, which is reduced in depression (Karege et al., 2005; Angelucci et al., 2005),

promotes phosphorylation of the FoxO protein (Mojsilovic-Petrovic et al., 209; Zhu et

al., 2004); moreover, FoxO3a-deficient mice display an antidepressant-like behavior

Page 6: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

5

(Zheng et al., 2002; Polter et al., 2009; Zheng and Quirion, 2004); Furthermore, both

the serotonin-norepinephrine reuptake inhibitor antidepressant venlafaxine and the

mood stabilizer lithium, suppress FoxO3a activity in mouse brain (Wang et al., 2013;

Mao et al., 2007). FoxOs are under the control of neurotransmitters (Liang et al., 2006)

and glucocorticoids(Qin et al., 2014). FoxOs also regulate signals for cellular atrophy

(Jaitovich et al., 2015), cellular morphology (Aranha et al., 2009) and adult

neurogenesis (Zhang et al., 2013) and all of these physiological or pathological

conditions contribute to the development of depression (Borre et al., 2014; Mahar et

al., 2014; Anacker et al., 2013). Although the role of FoxOs in the process of

depression is only beginning to be unveiled, it is reasonable at this time to

hypothesize that FoxO transcription factors will be found to be pivotal mediators in

psychiatric disorders. In this review, cellular signaling cascades responsible for

regulating FoxOs, the possible roles of FoxOs in the development of depression, the

effects of antidepressants on the activity of FoxOs as well as the potential mechanistic

pathways linking major depression and FoxOs will be discussed.

2. Overview of FoxO signaling pathway

Forkhead box (Fox) proteins are a family of transcription factors characterized

by a conserved “forkhead” or “winged helix” DNA binding motif. Foxs bind to the

regulatory sequence of the downstream target genes and play important roles in

regulating the transcription of genes involved in cell growth, proliferation,

differentiation, metabolism, apoptosis and drug resistance (Lam et al., 2013; Accili

and Arden 2004). According to the sequence homology, Fox superfamily genes can be

classified from FoxA to FoxS (Lam et al., 2013). Of the Fox subfamilies, the FoxO

group is one of the most researched subgroups. FoxOs share a conserved

DNA-binding domain, the binding sequence is 5’-TTGTTTAC-3’ (Brent et al., 2008).

In mammals, there are four FoxO homologues, FoxO1, FoxO3a, FoxO4 and FoxO6.

FoxO2 was shown to be same with FoxO3a and FoxO5 is the ortholog of FoxO3a in

the zebra fish (Carter and Brunet, 2007). The activity of FoxO is regulated by

post-translational modification, including phosphorylation, acetylation, methylation,

Page 7: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

6

ubiquitination and glycosylation (Eijkelenboom and Burgering, 2013; Zhao et al.,

2011). Phosphorylation of FoxO by protein kinases has been extensively studied.

Protein kinase B (Akt), serum/glucocorticoid inducible kinase (SGK), AMP-activated

protein kinase (AMPK), mitogen-activated protein kinases (MAPK), inhibitor of

nuclear factor kappa-B kinase (IKK), cyclin-dependent kinase (CDK) and mammalian

sterile 20-like kinase (MST) are common upstream kinases for FoxO (Boccitto and

Kalb, 2011). Phosphatidylinositol 3-kinase (PI3K)/Akt signal pathway activated by

insulin or growth factors mediates the major regulation of FoxO (woodgett, 2005).

Binding of insulin or other growth factors (such as nerve growth factor, BDNF or

insulin like growth factor 1 (IGF-1) to their receptors leads to the autophosphorylation

of the growth factor receptor and its subsequent activation (Zheng et al., 2002; Zheng

and Quirion, 2004; Zheng and Quirion, 2009). Through regulation via the PI3K/Akt

pathway, many of downstream target genes of FoxO (such as Btg-1, Bim-1, FasL,

MnSOD, p27, PEPCK) are regulated and these gene products play roles in cell

proliferation (p27), differentiation (Btg-1), metabolism (PEPCK) , apoptosis (Bim-1,

FasL) and drug resistance (Wen et al., 2012; van der Vos and Coffer, 2011; Hui et al.,

2008). For example, sustained FoxO3a activation promotes drug resistance by

activating and promoting the expression of ATP-binding cassette sub-family B

member 1, which is a plasma membrane P-glycoprotein that functions as an efflux

pump for various anticancer agents, including doxorubicin (Hui et al., 2008).

The PI3K pathway is a major regulator of FoxOs activity. Akt and SGK are two

key downstream components of the PI3K pathway and both of them recognize the

RXRXXS/T motif. Akt phosphorylates FoxO3a at Thr32, Ser253 and Ser315, while

SGK phosphorylates FoxO3a at Thr32 and Ser315. For FoxO1, Akt phosphorylates it

at Thr24, Ser256 and Ser319, while SGK phosphorylates it at Thr24 and Ser319

(Burgering and Kops, 2002). Phosphorylation of these sites leads to translocation of

FoxOs from the nucleus to cytoplasm and inhibition of transcriptional activity (Chen

et al., 2013; Huang and Tindall, 2007). What is important is that the C terminal of

FoxO6 lacks the Akt phosphorylation site and thus it is constitutively nuclear (Jacobs

et al., 2003). Extracellular regulated protein kinase (ERK) is a downstream

Page 8: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

7

component of MEK and it has been shown to phosphorylate FoxO3a at Ser294,

Ser344 and Ser425 (Yang et al., 2008), similarly, activation of IKK induces FoxO3a

phosphorylation at Ser644 (Hu et al., 2004). Phosphorylation of FoxO3a by both ERK

and IKK leads to nuclear exclusion and degradation. The serine/threonine kinases p38

and c-Jun N- terminal kinase (JNK) are other two major MAPK pathways relevant to

FoxO function. p38 phosphorylates FoxO3a at Ser7 and promotes the nuclear

translocation and activation of FoxO3a (Ho et al., 2012); JNK may repress the

PI3K/Akt signal pathway, thereby activating FoxO3a indirectly, meanwhile, it may

phosphorylayte FoxO proteins in a direct way (Sunters et al., 2006). FoxOs may also

play a critical role in the regulation of energy homeostasis when phosphorylated by

the AMP-activated protein kinase (AMPK) at six sites (Greer et al., 2009). In contrast

to the effect of the above kinases on FoxOs, MST binds to FoxO3a and

phosphorylates it at Ser207; however, the phosphorylation of this site promotes its

nuclear accumulation and subsequently the expression of proapoptotic genes (Sanphui

and Biswas, 2013). Another interesting example is the role of CDK on FoxOs. CDK1

phosphorylates FoxO1 at Ser249 and enhances the content of FoxO1 in the nucleus

and thus promotes cell death (Yuan et al., 2008), CDK2 phosphorylates FoxO1 at the

same site, however, it leads to the accumulation of FoxO1 in the cytoplasm and the

inhibition of FoxO1 (Huang et al., 2006). Figure 1 summarizes the above pathways

regulating FoxO activity and subcellular localization.

Fig.1. Effects of various upstream regulators on the activity and subcellular localization of

Page 9: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

8

FoxO. Phosphorylation is one of the most common post-translational modifications. Kinases, such

as Akt, IKK, SGK, ERK1/2 and CDK promote the export from the nucleus and inactivation of

FoxO. While under stress conditions, MST, AMPK, p38MAPK and JNK induce the nuclear

accumulation and activation of FoxO. In the nuclear, FoxO recognizes and binds to FRE (FoxO

response element), activated FoxO thereby regulates the transcription activity of its downstream

targets, which makes it possible to regulate many cellular processes, including proliferation,

differentiation, metabolism, apoptosis and drug resistance.

3. FoxO expression patterns in the brain

FoxO is one of the substrates for Akt which is a pro-survival serine/threonine

kinase. The PI3K/Akt signal pathway plays a critical role in mediating neuronal

survival. FoxO regulates the expression of genes involved in neuronal cell apoptosis

and oxidative stress, such as Bim-1, FasL, Catalase and MnSOD (Brunet et al., 2001a,

Wen et al., 2012). The pattern of protein distribution related to FoxO functions points

to the associated biological functions. Understanding the expression patterns of FoxO

in the CNS should help us to understand the relationship between FoxO and

neurodevelopment, neurodegeneration and potentially neuropsychiatric disorders.

FoxOs are highly expressed in brain areas related to the regulation of mood and stress.

FoxO1, FoxO3a and FoxO4 are each expressed during neurodevelopment and also in

the adult, with distinct patterns ranging from ubiquitous to tissue-specific (Biggs et al.,

2001). Among the FoxO subtypes expressed in brain, FoxO1 is mostly expressed in

the hippocampus and the striatum (Zemva et al., 2012), its mRNA is expressed in part

of the ventral region of the CA3 pyramidal neurons, however, no expression was

observed in the dorsal CA3 neurons (Hoekman et al., 2006). FoxO3a shows the

widest brain expression, whereas FoxO4 is expressed at very low levels in the brain

and a FoxO4 probe did not reveal detectable expression in the adult murine brain

(Salih et al., 2012). FoxO6 is mostly expressed during the developmental stage with a

nuclear-predominant distribution (Jacobs et al., 2003). For FoxO6, both the protein

and mRNA levels are highly expressed during the stage of neuronal development, and

its expression decreases during maturity but then stays relatively constant. In the adult

Page 10: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

9

brain, FoxO6 protein is expressed abundantly in hippocampus, amygdala and nucleus

accumbens (Salih et al., 2012). Converging evidences suggest that the hippocampal

complex may play a role in the pathology of major depression (Campbells and

Macqueen, 2004). FoxO isoforms also have different patterns of expression in the

hippocampus where FoxO1 is enriched in the dentate gyrus (DG) region of

hippocampus, FoxO3a is present in all hippocampal areas but more highly expressed

in the DG and CA3 regions than in the CA1 region. FoxO6 is enriched in the CA1 and

CA3 hippocampal areas (Hoekman et al., 2006; Furuyama et al., 2000; Jacobs et al.,

2003; Salih et al., 2012). The differential expression patterns suggest that FoxO

isoforms may have specific and perhaps complementary or redundant roles in

different physiological or pathological processes. In conclusion, spatial expression

patterns of FoxO gene family members in the CNS, especially regions related to stress,

implicate their biological functions in stress-related behavior regulation.

4. FoxOs regulate the behavioral manifestation of depression

The role of FoxOs in psychiatric disorders has recently drawn more attention

even though the data in this area are somewhat limited at this time. FoxOs, especially

FoxO6, are expressed abundantly in hippocampus, amygdala and nucleus accumbens

which are important brain areas involved in aversive and rewarding responses to

emotional stimuli (Accili and Arden, 2004; Hoekman et al., 2006). Thus FoxOs could

mediate the inability to experience pleasure and lack of motivation that predominate

in many patients with depression (Berton and Nestler, 2006). The subregional

distributions of FoxOs could predict their roles in the regulation of mood and emotion.

In a case-control association study in a Brazilian population, FoxO3a emerged as an

gene candidate involved in bipolar disorder and related behaviors. Three

single-nucleotide polymorphisms within the FoxO3a gene (rs1536057, rs2802292 and

rs1935952) were associated with bipolar disorder (Magno et al., 2011). Mood-related

behavioral disturbance, such as depression, is highly related to stress (Krishnan and

Nestler, 2008). In a learned helplessness animal model, which is a commonly used

model to mimic human depression (Muller et al., 2011), inescapable shocks

Page 11: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

10

significantly reduced the phosphorylation of FoxO3a and induced the nuclear location

of FoxO3a in the cerebral cortex, interestingly, learned helplessness behavior was

markedly diminished in FoxO3a-deficient mice(Zhou et al., 2012). This research

provides evidence that depression activates FoxO3a. These results was consistent with

the previous report that FoxO3a influences behavioral processes linked to anxiety and

depression and FoxO3a-deficient mice show a significant antidepressant-like behavior,

but these animals didn't exhibit changes in general locomotor activity tested by open

field test (Polter et al., 2009), suggesting that FoxO3a is a transcriptional target for

mood disorder treatment. In contrast, a study using a knockout (KO) mause model

suggested that FoxO1 KO mice displayed reduced anxiety (Polter et al., 2009).

Recently, disrupted rhythms are observed in psychiatric disorders, including

depression (Emens et al., 2009). Association of Clock gene and major depressive

disorder has also been suggested (Germain and Kupfer, 2008). Although the detailed

etiologies still need to be unraveled, it is evident that there is a strong relationship

between major depression and circadian disruption (Karatsoreos, 2014). Interestingly,

clock is a transcriptional target of FoxO3a and the insulin-FoxO3a-Clock signaling

pathway plays a key role in the modulation of circadian rhythms, for instance, knock

down of FoxO3a dampens circadian amplitude (Chaves et al., 2014; Zheng et al.,

2007). These studies revealed an important new role for FoxOs in the regulation of

depressive behavior.

BDNF is a well known regulator involved in the pathophysiology of mood

disorders and a recognized upstream effector of FoxOs. The role of BDNF in

depression is well characterized. Depression, at least in its severe form, is associated

with reduced neuronal plasticity and lower levels of BDNF, while antidepressants,

such as fluoxetine, could reactivate BDNF-mediated neuronal plasticity (Castrén and

Rantamäki, 2010). BDNF reduces FoxO transcriptional activity through activation of

TrkB and downstream kinases including Akt, which is a candidate susceptibility gene

related to schizophrenia (Emamian et al., 2004). The link between Akt and depression

was also provided by both genetic studies and postmortem data (Hsiung et al., 2003;

Pereira et al., 2014). In response to BDNF, active Akt phosphorylates and inactivates

Page 12: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

11

FoxOs (except for FoxO6, which is constitutively nuclear) by facilitating their

translocation out of the nucleus (Wen et al., 2012). In both invertebrate and vertebrate

brain, FoxOs can be phosphorylated and inactivated by serotonin selective reuptake

inhibitors or exogenous serotonin via a PI3K/Akt-dependent mechanism (Polter et al.,

2009; Liang et al., 2006). All of data suggests that downregulation or inactivation of

FoxOs are highly related to antidepressant-like effects.

5. Antidepressants regulate the activities of FoxOs

The classical view of depression is that a functional deficit in neurotransmitters,

particularly decreased levels of serotonin and noradrenaline, are associated with the

clinical symptoms of depression. Serotonin could be viewed as a regulator of the

insulin/IGF-1-FoxO pathway in stress physiology and modulation of FoxO by

serotonin represents a conserved feature of stress physiology. Acute stressful life

events can lead to the recurrence of episodes of major depression and chronic stress is

strongly associated with both onset and recurrence of depression (Kessler, 1997;

Risch et al., 2009). Studies of C. elegans showed that serotonin-deficient mutants

exhibited DAF-16 (the homologues of FoxO) nuclear accumulation in constitutive

physiological stress states, while treatment with exogenous serotonin directly or with

fluoxetine, an antidepressant known to increase synaptic levels of endogenous

serotonin, prevented DAF-16 nuclear accumulation in wild-type animals under

aversive conditions (Liang et al., 2006). This result was confirmed by experiments

showing that serotonin and fluoxetine can partially suppress DAF-16 nuclear

accumulation in wild type animals exposed to hypergravity in C elegans (Kim et al.,

2007). These observations suggest that individual behavior and physiological or

psychiatric responses to the environment in animals are likely accompanied by the

changes of the subcellular location/function of FoxO protein.

The tricyclic antidepressant agent, imipramine, is a monoamine-regulating

antidepressant. Chronic treatment with imipramine for 28 days in mice caused

appreciable increases in phosphorylated FoxO1 (Ser256), FoxO3a (Ser253) and Akt

(Thr308) in the cerebral cortex, hippocampus and striatum, but no changes in the level

Page 13: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

12

of total FoxO1, FoxO3a and Akt were observed (Polter et al., 2009). Our previous

results also indicated that the FoxO3a subtype in PC12 cells could be inactivated by

the antidepressant venlafaxine via PI3K/Akt pathway. Venlafaxine, on one hand,

induced the phosphorylation of Akt and FoxO3a by the PI3K/Akt pathway and on the

other hand reversed the reduction of phosphorylated Akt and FoxO3a and the nuclear

translocation of FoxO3a induced by corticosterone (Wang et al., 2013). Lithium,

another well known therapeutic drug in the treatment of mood disorder, reduced

FoxO3a transcriptional activity, gene expression and protein levels in both the cytosol

and nucleus, and this effect was independent of Akt (Mao et al., 2007). Moreover, this

effect of lithium was observed in SH-SY5Y neuroblastoma cells and mouse brain

(cortex and hippocampus) after relevant lithium treatments, which indicate that this

effect is likely therapeutically relevant (Mao et al., 2007). Amphetamine is a

compound which can increase the concentration of dopamine in the synaptic gap and

thus induce hyperactivity, reversal of its action by lithium is used to model mania in

bipolar disorder. A study from our lab indicated that pretreatment of animals with

lithium prevented an amphetamine-induced decrease in striatal phosphorylated Akt

and FoxO1 levels (Zheng et al., 2013). Lithium also attenuated amphetamine-induced

locomotor activity and decreased prepulse inhibition (Zheng et al., 2013). The

modulation by lithium of FoxO and its upstream kinases (such as Akt) may be

relevant to the treatment of neuropsychiatric disorders. Since FoxO3a can be

regulated by BDNF-induced Akt activation, and phosphorylated by antidepressants,

we hypothesize that FoxO3a is a candidate transcription factor that plays a role in

mood disorders, including depression. Further studies are needed to elucidate the role

of FoxO transcription factors in the neuropathology and treatment of mood disorders.

6. FoxO as a regulator of serotonin and norepinephrine signaling

Decreased or abnormally low levels of serotonin and norepinephrine in the

synaptic cleft is traditionally viewed as a common cause of mood disorders, including

depression. These neurotransmitters play their roles by binding and activating their

specific receptors. With the exception of the 5-HT3 receptor, which is a ligand-gated

Page 14: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

13

ion channel, all other 5-HT receptors and adrenergic receptors are seven

transmembrane G protein-coupled receptors (GPCRs) whose binding to serotonin or

norepinephrine activates several intracellular second messenger cascades. Some of

these cascades are related to FoxOs. For example, the serotonin 2A receptor

(5-HT2AR) is highly expressed on pyramidal neurons in the frontal cortex and has

been implicated in several psychiatric disorders, including depression. The density of

5-HT2R is higher in depressive patients than that in normal persons. Higher 5-HT2A

binding in the prefrontal cortical region has been shown to be associated with greater

gene expression in depression and youth suicide (López-Figueroa et al., 2004).

Antidepressants (such as mirtazapine and mianserin) could occupy 5-HT2AR, block

its role and augment the clinical response to SSRIs (Celada et al., 2004). Serotonin

binds to 5-HT2AR and stimulates Akt phosphorylation in the frontal cortex and in

primary cortical neurons through the activation of a PI3K/Akt cascade (Schmid and

Bohn, 2010). FoxO transcription factors are downstream targets of PI3K/Akt, which

is mainly regulated by receptor tyrosine kinases (such as IGF-1 receptor) and GPCRs.

Serotonin is a regulator of the insulin/IGF-1 pathway in stress physiology and

exogenous 5HT can suppress the nuclear accumulation of FoxO in wild-type animals

under stress (Liang et al., 2006). Moreover, in the peripheral system, serotonin also

influences the physiological actions of FoxO, for example, serotonin enhances the

proliferation of hepatocellular carcinoma cells through upregulation of FoxO3a

(Liang et al., 2013) . Duodenum-derived serotonin is a critical regulator of bone mass

during the process of bone formation, and importantly FoxO1 mediates this effect

(Kode et al., 2012). High circulating serotonin levels prevent the association of

FoxO1 with CREB, resulting in suppressed osteoblast proliferation (Kode et al., 2012).

These results demonstrate that FoxO acts as an endogenous mediator of serotonin

signaling which may have consequences for the role of FoxOs in psychiatric disorders

associated with altered serotonin metabolism.

As mentioned above, the 5-HT3 receptor is a ligand-gated ion channel whereas

other receptors for serotonin are GPCRs. Serotonergic GPCRs can be classified on the

basis of their associated Gα proteins into Gs-protein coupled receptors (5-HT4, 5-HT6,

Page 15: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

14

5-HT7), Gi-protein coupled receptors (5-HT1, 5-HT5), and Gq/G11-protein coupled

receptors (5-HT2). As for norepinephrine receptors, the adrenergic α1 receptor is a Gq

coupled receptor whereas the adrenergic α2 is a Gi coupled receptor. Activation of

Gs-protein coupled receptors leads to activation of adenylate cyclase and an increase

in cellular levels of cAMP, while activation of Gi-protein coupled receptors results in

decreased levels of cAMP. cAMP is a second messenger important in many biological

processes, including mood/emotional regulation. Downstream effectors of cAMP

include cAMP-dependent protein kinase (PKA) and exchange factor directly activated

by cAMP (Epac). Deficits in cAMP/PKA or cAMP/ERK signaling are viewed as one

of the mechanisms for the behaviorial symptoms in depression (Breuillaud et al., 2012;

Musazzi et al., 2010). There is interplay between the PI3K/Akt/FoxO and cAMP/PKA

signaling pathways (Chen et al., 2007). FoxO1 is a direct substrate for PKA and PKA

phosphorylates FoxO1 at Thr24, Ser256, and Ser319, three known Akt

phosphorylation sites (Lee et al., 2011). Epac is an exchange protein activated by

cAMP. Both Epac and PKA induce ERK phosphorylation, and ERK in turn

phosphorylates FoxO3a at Ser294, Ser344 and Ser425 (Yang et al., 2008), and FoxO1

at Ser246, Ser284, Ser295, Ser326, Ser413, Ser415, Ser429, Ser467 and Ser475

(Asada et al., 2007) resulting in degradation of FoxO proteins (Fig.2). These results

show that the binding of ligands or agonists to the serotonin or norepinephrine

receptors results in the alteration of intracellular cAMP, which affect the cAMP/PKA

or cAMP/ERK signal pathway, and might influence the biological functions of

FoxOs.

Page 16: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

15

Fig.2. FoxO as a regulator of serotonin signaling. 5-HT receptor is a G protein coupled

receptor, when 5-HT binds, the receptor activates the coupled G protein, which can diffuse along

the membrane surface and directly activate PI3K/Akt signal pathway, leading to inactivation of

FoxO proteins. On the other hand, 5-HT receptor may also stimulate adenylyl cyclase/PKA and

Epac/ERK1/2 signal transduction, both PKA and ERK1/2 enhance the phosphorylation of FoxO,

which lead to inactivation of FoxO. Catalytic subunit of PKA translocates into nuclear and

promote the activation of CREB, which is also highly related with depression.

7. FoxO as an effector of Hypothalamic-pituitary-adrenal axis

The activated hypothalamic-pituitary-adrenal (HPA) axis stimulates the secretion

of adrenocorticotrophic hormone (ACTH) from the pituitary, which ultimately

promotes the secretion of glucocorticoids from the adrenal cortex. Depression has

been associated with hyperactivity of the HPA axis and increased levels of

glucocorticoid in the serum where continuously high glucocorticoid levels attenuate

the responsiveness and number of glucocorticoid receptors on hippocampal granule

cells thus leading to impaired negative feedback suppression of glucocorticoid

secretion and an even higher level of circulating corticosteroid (Anacker et al., 2011a;

Anacker et al., 2011b; Raison and Miller, 2003). A schematic presentation of how the

amygdala and the hippocampus regulate the HPA axis and the stress response is

presented in Figure 3. Recent evidence suggests that FoxO plays a crucial role in the

Page 17: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

16

pathophysiological hyperactivity of the HPA axis (Lutzner et al., 2012; Lutzner et al.,

2012). Glucocorticoids activate FoxO signaling, and FoxO signaling is a major

contributor to the mechanism of glucocorticoid-induced muscle atrophy (Sandri et al.,

2004; Zhao et al., 2007). FoxO1 and FoxO3a expression (both mRNA and protein) are

up-regulated by glucocorticoid treatment in hippocampal neurons and muscle cells

(Anacker et al., 2011a; Poulsen et al., 2011). Induction of FoxO3a expression requires

glucocorticoid receptor-binding steroids and is reversed by concomitant treatment

with the glucocorticoid receptor antagonist RU-486 (Lutzner et al., 2012). Importantly,

recent data indicate that FoxO3a is a direct glucocorticoid receptor target, as there are

two potential glucocorticoid response elements within the promoter region of FoxO3a.

These glucocorticoid hormones may induce transcriptional activation of FoxO3a

directly (Lutzner et al., 2012). These results indicate that glucocorticoids regulate

FoxO3a on a transcriptional level. Further evidence indicates that glucocorticoids

regulate FoxO3a post-translationally. Glucocorticoids induce the expression of SGK,

a serine/threonine kinase and activation and phosphorylation of SGK (Thr-256,

Ser-422) is dependent upon PI3K activity (Buse et al., 1999). Activated SGK can

promote cell survival through phosphorylating FoxO3a at sites Thr-32 and Ser-253

(Brunet et al., 2001b). When given in relatively high doses, glucocorticoids inhibit

PI3K/Akt signaling, thereby reducing the phosphorylation of FoxO (Wang et al., 2013;

Stitt et al., 2004). Moreover, consistent with the notion that overload of

glucocorticoids is crucial to the development of depression (Anacker et al., 2013;

Anacker et al., 2011b), antidepressant treatment antagonizes the role of

glucocorticoids (Budziszewska et al., 2000; David et al., 2009) and additionally

increases the level of phosphorylated FoxO3a and PI3K/Akt (Wang et al., 2013).

Page 18: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

17

Fig. 3. Regulation of HPA axis by the amygdala and hippocampus in normal and stress

conditions. The corticotropin-releasing hormone neurons are regulated by both the amygdala and

the hippocampus. Activation of amygdala promotes the release of cortisol through HPA axis.

Mineralocorticoid receptors and glucocorticoid receptors are abundant in the hippocampus, both

of which respond to and regulate the level of cortisol in the serum by glucocorticoid negative

feedback. In stress condition, inappropriate activation of the amygdala extensively stimulates the

HPA axis, resulting in high level of cortisol in the body. Chronic exposure of hippocampus to

excessive glucocorticoid leads to neuronal loss and morphological atrophy, and thus the negative

feedback becomes weaken or disrupted, which leads to much higher level of cortisol. Chronic

stress induces nuclear localization of GR, which binds to glucocorticoid response

element (GRE) and promotes the transcription of FoxO, GR may also activate SGK

and subseqently inhibit the activity of FoxO.

8. FoxOs regulate neuronal morphology and possibly mediate neuronal atrophy

Neuroimaging studies with magnetic resonance imaging scanning demonstrated

that compared with healthy individuals, depressed patients had an increased volume

of the lateral ventricles and smaller volumes of the basal ganglia, thalamus,

hippocampus, and frontal lobe (Arnone et al., 2012). The decreased cerebral size may

be attributed to the loss and atrophy of neurons. In fact, clinical and preclinical studies

have revealed that prolonged stress and major depression are associated with

decreased neuronal synapses, neuronal atrophy of the PFC and the hippocampus

Page 19: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

18

(Duman and Aghajanian, 2012; Ota et al., 2014; Sheline et al., 2003; Sheline et al.,

1996). Compared with normal controls, depressed patients showed decreased cortical

thickness, neuronal size, and neuronal and glial densities in the prefrontal cortical

regions (Rajkowska et al., 1999), but the molecular mechanisms underlying these

morphological alterations have not yet been identified.

FoxO transcription factors are important mediators of the effects of

IGF-1/PI3K/Akt signaling. Withdrawal of IGF-1 causes reduced size of white matter

structures in brain due to a decreased numbers of axons (Beck et al., 1995), while

overexpression of IGF-1 leads to increased brain size due to an increase in cell size

and apparently in cell number (Carson et al., 1993). As FoxOs are important key

mediators of insulin and IGF-1 signaling (Kennedy et al., 2013), it is possible that

FoxOs mediates the effect of IGF-1 in this process. Since overexpression of FoxO3a

leads to reduced brain size and a decrease in neural progenitor number

(Schmidt-Strassburger et al., 2012) and consistently, decreased FoxO activity has been

reported to result in larger brain size and increased neural progenitor proliferation

(Paik et al., 2009; Renault et al., 2009). Akt, one of the pivotal upstream kinases

regulating the function of FoxO, has been shown to play a role in regulating cell size,

a response which shares mechanisms with cellular atrophy or hypertrophy.

Constitutively active Akt increases muscle fiber size and prevents denervation

atrophy in regenerating and adult rat muscle (Pallafacchina et al., 2002). In 2004, Stitt

et al., (Stitt et al., 2004) reported that the IGF-1/PI3K/Akt pathway plays an important

role in preventing skeletal muscle atrophy by inhibiting the expression of

muscle-specific ubiquitin ligases MAFbx and MuRF1. Interestingly, this effect

involves Akt-mediated inhibition of the FoxO transcription factors as mutation of

FoxO1 attenuated the activation of Akt thereby preventing Akt-mediated inhibition of

muscle atrophy(Stitt et al., 2004). This research extended the function of FoxO to the

regulation of cellular atrophy and the role of FoxO in cell atrophy and hypertrophy

has since been extensively studied. As in skeletal muscle, overexpression of FoxO3a

also caused a significant reduction in cardiomyocyte size in mouse hearts in vivo and

the induction of atrogin-1 by FoxO3a accounts for this effect (Skurk et al., 2005). The

Page 20: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

19

role of FoxO transcription factors in mediating the muscle atrophy has been widely

studied (Castets and Ruegg, 2013; Sanchez et al., 2014; Lee et al., 2012), however,

the specific role of FoxO in neuronal atrophy is largely unknown. The four FoxO

members found in humans, FoxO1, FoxO3a, FoxO4, and FoxO6, are all expressed in

the CNS (Biggs et al., 2001; Salih et al., 2012). The IGF-1/PI3K/Akt signaling

pathway is a key pathway for neuronal homeostasis (Rafalski and Brunet, 2011a).

FoxOs are the classic downstream targets of the IGF-1/Akt pathway and both

IGF-1/PI3K/Akt and FoxO have been identified in the regulation of cell atrophy.

From this perspective, it is likely that FoxO mediates neuronal atrophy in the CNS

and this effect is related to the etiology of depression. This speculation is worthy of

further investigation potentially leading to a better understanding of the precise

functions of FoxO transcription factors in the regulation of neuronal cell size and this

might lead to the development of completely novel therapeutic approaches to the

prevention or limitation of the morphological alterations that prevail in ageing and

pathological states, such as depression and schizophrenia .

9. FoxOs play a inhibitory role in neurogenesis and synaptogenesis

Preclinical evidence showed that adult hippocampal neurogenesis may contribute

to the mechanism of antidepressant drug action and BDNF is probably involved in

this effect (Malberg et al., 2000; Guilloux et al., 2013; Sairanen et al., 2005). In fact

BDNF has been implicated in regulating adult neurogenesis in the subgranular zone of

the DG and is important for the regulation of the basal level of neurogenesis in adult

mice (Lee et al., 2002; Waterhouse et al., 2012). FoxOs are downstream effector

molecules of BDNF since BDNF activates a variety of signaling cascades, including

PI3K/Akt, Ras/MAPK and cAMP/PKA pathways and activation of these pathways

phosphorylates and inhibits the functioning of FoxOs (Kwon et al., 2011; de la

Torre-Ubieta and Bonni, 2011). The regulatory effects of BDNF on FoxOs and its

target genes play a significant role in the BDNF-mediated neurogenesis, neuronal

survival and plasticity (Zhu et al., 2004).

FoxO transcription factors coordinately regulate diverse pathways to govern

Page 21: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

20

neural stem cell or progenitor cell homeostasis in the brain (Paik et al., 2009; Renault

et al., 2009). Given the importance of neural stem cells and progenitor cells for the

generation of new neurons, the role of FoxO in the regulation of neurogenesis from

neural stem cells has attracted attention. Genetic manipulation producing a transgenic

mice expressing a constitutively active FoxO3a leads to mice having smaller brains

accompanied with a loss of neurons in the olfactory bulbs, the cortex, the striatum,

thalamic regions, and most notably the DG (Schmidt-Strassburger et al., 2012). Both

the olfactory bulbs and the DG region are the sites for neurogenesis in adult, whilst

neurogenesis assessed in adult FoxO3−/− and FoxO3+/+ mice revealed that FoxO3a

deficiency in vivo led to an appreciable increase in the production of new neurons in

the olfactory bulbs (Webb et al., 2013). However, the role of FoxOs in adult

hippocampal neurogenesis remains to be investigated. FoxOs are classic downstream

substrate of Akt (Zheng et al., 2002), these findings were quite consistent with earlier

investigations showing that inhibition of 3-phosphoinositide-dependent protein kinase

1/Akt signaling reduces neurogenesis in neural progenitor cells, whereas constitutive

activation of Akt or phosphatase and tensin homolog deletion promotes neurogenesis

(Oishi et al., 2009; Gregorian et al., 2009). These results support the argument that

FoxO negatively regulates neurogenesis in the brain.

Reduced synaptogenesis could be observed in the prefrontal cortex and

hippocampus of patients with depression whereas normal synptogenesis restores the

synapse connections in the brain that may deteriorate under stress and depression

(Bambico and Belzung, 2013). Mammalian target of rapamycin (mTOR) is a

ubiquitous protein kinase involved in protein synthesis and synaptogenesis. Activation

of mTOR by PI3K/Akt promotes protein synthesis and synaptogenesis (Ayuso et al.,

2010). However, FoxOs play a role in protein degradation (White et al., 2013).

Moreover, it has been shown that FoxO1 inhibits mTOR signaling and protein

synthesis. Activation of constitutively active FoxO1 reduces the phosphorylation of

4E binding protein 1 (Thr-37/46) and by consequence the phosphorylation of the

downstream protein p70S6 kinase leading ultimately to an attenuation of protein

production (Southgate et al., 2007). This inhibition of protein synthesis affects

Page 22: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

21

proteins (such as post-synaptic density protein 95 and synaptophysin) involved in

synapse formation. On the other hand, recent studies suggest that the rapid-acting

antidepressant and synaptogenic effects of ketamine are dependent upon mToR

activity in the medial prefrontal cortex (Li et al., 2010). This finding fits our

hypothesis well. As mentioned previously, FoxOs inhibit mTOR signaling (Southgate

et al., 2007), under this condition, inactivation of FoxOs may be beneficial for the

antidepressant effect of ketamine. Microtubule architecture in neurons is an essential

element in the regulation of neuronal morphology and motility as well as

synaptogenesis (Conde and Caceres, 2009). FoxO loss-of-function models display

increased stability of microtubule and overexpression of wild-type FoxO moderately

destabilizes microtubules (Nechipurenko and Broihier, 2012). Thus, it is conceivable

that FoxOs negatively regulate synaptic microtubule stability and thus negatively

regulate synaptogenesis. The roles of FoxOs in the regulation of cellular atrophy,

neurogenesis and synaptogenesis are depicted schematically in Figure 4.

Fig. 4. The roles of FoxOs in the regulation of cellular morphology. Neurotrophic factors or

growth factors, such as BDNF or IGF-1, play a pivotal role in the maintenance of normal cellular

morphology. IGF-1 binds to its receptor and activates several signaling pathways. FoxOs are the

classic downstream targets of IGF-1/PI3K/Akt pathway. Both IGF-1/PI3K/Akt and FoxOs are

involved in the regulation of cell size. Activated Akt phosphorylates and inactivates FoxO proteins,

it also phosphorylate and activate mTOR, which subsequently phosphorylates eukaryotic

Page 23: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

22

translational initiation factor 4E binding protein 1 (4E-BP1), 70kDa ribosomal protein S6 kinase

(p70S6K), and thereby promotes protein translation. Active FoxOs suppress the phosphorylation

of 4E-BP1 and p70S6 kinase and thus limit protein synthesis. In addition, FoxO transcription

factors induce the expression of atrophy- related ubiquitin ligase atrogin-1 and cause cellular

atrophy, this effect possibly account for the neural atrophy implicated in the process of depression.

FoxO may also suppress neurogenesis through inhibiting Wnt signaling. Furthermore,

FoxOs limit the stability of microtubules, all of which contribute to the morphological changes

(such as neuronal atrophy, decreased neurogenesis and synaptogenesis) occurring in the depressed

individuals.

Down-regulated adult hippocampal neurogenesis and synaptogenesis in the

prefrontal cortex and hippocampus have been linked to major depressive disorder.

Chronic stress leads to decreased neuronal/progenitor cell proliferation within the

specific region of the brain, neuronal spines and neuronal plasticity are also reduced

in this condition (Bambico and Belzung, 2013). Interestingly, this process is reversible.

Administration of antidepressants will produce an increase in the number of

new-borne neurons within the hippocampus (Malberg et al., 2000; Guilloux et al.,

2013; Sairanen et al., 2005). The observation that current antidepressant drugs

promote adult hippocampal neurogenesis provides a much stronger evidence on this

link (Mahar et al., 2014; Zunszain et al., 2013). These findings suggest a link between

depression and adult neurogenesis in the hippocampus and, as discussed previously,

many antidepressants repress the activation of FoxO. In this context, it can be

speculated that the inactivation of FoxO3a by the PI3K/Akt signaling pathway

achieves a more potent induction of adult hippocampal neurogenesis and

synaptogenesis.

10. Conclusions and Prospective

FoxOs are recently discovered transcription factors which play a role in the onset

or duration of psychiatric disorders. Results from cellular studies and animal models

support the idea that FoxOs are potential mediators in the pathogenesis of depression

Page 24: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

23

(Polter et al., 2009; Wang et al., 2013; Liang et al., 2006). Given the considerations to

target FoxO proteins as a novel therapeutic strategy for major depression, it becomes

essential to further investigate whether specific FoxO family members should be

targeted. Activation and upregulation of upstream molecules of FoxOs, including

growth factors (such as BDNF, fibroblast growth factor and IGF-1), receptor tyrosine

kinases, Akt and ERK1/2 produce beneficial effects against depression (Shi et al.,

2012; Evans et al., 2004; Krogh et al., 2014; Mikoteit et al., 2014; Musazzi et al.,

2010). In this condition, over inhibition of FoxOs through activation of its upstream

molecules increases the risk of cancer should be taken into consideration. The roles of

FoxO transcription factors in major depression are summarized schematically (Fig. 5).

However, the possibility of FoxOs and their signaling pathways being potential

therapeutic targets in depression requires considerable experimental exploration and

validation. Their nature as transcription factors and the difficulty to target them

directly suggest that targeting the signaling pathways rather than the FoxOs

themselves may be preferred strategy towards efficacious therapeutic agents. At

present, most of the information about FoxOs is derived from cellular and animal

models. Direct data from individuals diagnosed with major depression are still in

great need. It will be desirable that future work should be carried out to study the

alteration of FoxOs in depressive individuals and post mortem brain tissues. Studies

of the genetic linkage of FoxOs and depression also provide evidence to support a role

of FoxOs in this very important area of human pathophysiology.

Fig. 5. The roles of FoxOs in major depression. Chronic stress causes imbalances of

Page 25: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

24

neurotransmitters (such as 5-HT and NE), decreased production of growth factors (such as BDNF

and IGF-1) and hyperactivation of HPA axis. The interaction of neurotransmitters with their

postsynaptic receptors and binding of growth factors to their receptor tyrosine kinases (RTKs)

inactivate FoxOs through cAMP/PKA, PKC, PI3K/Akt or MEK/ERK signaling. Hyperactive HPA

axis promotes the nuclear location and activation of FoxOs. Released glucocorticoid also bind to

the GRE in the promoter of FoxOs and enhance the production of FoxOs. As a result, chronic

stress leads to the activation of FoxO, which inhibits neurogenesis/synaptogenesis and promotes

neuronal atrophy. Consequently, all of these cause behavioral manifestations related to depression.

FoxOs have been linked to skeletal muscle atrophy and control of cardiomyocyte

size in numerous models (Sandri et al., 2004; Skurk et al., 2005) so it is reasonable to

speculate that the Akt-FoxO pathway may participate in neuronal atrophy, impairment

of synaptogenesis/neurogenesis and control of brain size in depression. Currently

there is little evidence to support this intriguing possibility, but it is worth studying the

effect of FoxOs on the regulation of neuronal cell size and exploring the mechanisms

of decreased cerebral volume in depressed individuals. However, there are always

benefits and short comings to everything, what we should keep in mind is that since

FoxOs affect cell size, there are possible unwanted side effects of targeting FoxO in

treatment of depression, such as muscle hypertrophy. Moreover, abnormalities

observed in major depression most commonly occur in the raphe nuclei, nucleus

accumbens, anterior cingulated cortex, amygdala and hippocampus (Goffer et al.,

2013; Tripp et al., 2011; Underwood et al., 1999; Andrus et al., 2012), but we still

need to elucidate whether the change of FoxOs and their related signal pathways

could be altered in region specific manner.

Despite the usefulness of currently available antidepressant medications, the

limitations of both efficacy and tolerability are nonetheless evident. A better

understanding of the underlying neurobiology and neuropathophysiology of

depression will help us to discover novel targets for pharmacological interventions. In

this review, we have outlined the involvement of FoxOs and related molecules in the

pathogenesis of depression. Targeting FoxO proteins might attenuate neuronal

Page 26: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

25

apoptosis and play a neuroprotective effects in the central nervous system, which

might be considered beneficial to block degenerative disorders. However, whether or

not this can yield therapeutic target and the generation of a novel class of agents for

the treatment of major depression will depend upon further research which reveals the

importance of FoxOs in the physiological and particular processes of major

depression.

Acknowledgements

This research was supported by National Natural Science Foundation of China

(No. 81301099, No. 81373384 and No. 31371088), Natural Science Foundation of

Guangdong Province (No. S2013040014202), China Postdoctoral Science Foundation

(No. 2013M542192), and the Science and Technology Development Fund (FDCT) of

Macao (FDCT 021/2015/A1).

Conflict of Interest Disclosures

The authors declare no conflict of interest.

Page 27: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

26

References:

Accili, D., Arden, KC., 2004. FoxOs at the crossroads of cellular metabolism, differentiation, and

transformation. Cell 117, 421-426.

Anacker, C., Cattaneo, A., Musaelyan, K., Zunszain, P.A., Horowitz, M., Molteni, R., Luoni, A.,

Calabrese, F., Tansey, K., Gennarelli, M., Thuret, S., Price, J., Uher, R., Riva, M.A., Pariante, C.M.,

2013. Role for the kinase SGK1 in stress, depression, and glucocorticoid effects on hippocampal

neurogenesis. Proc. Natl. Acad. Sci. U. S. A. 110, 8708-8713.

Anacker, C., Zunszain, P.A., Carvalho, L.A., Pariante, C.M., 2011a. The glucocorticoid receptor: pivot

of depression and of antidepressant treatment? Psychoneuroendocrino. 36, 415-425. Anacker. C., Zunszain, P.A., Cattaneo, A., Carvalho, L.A., Garabedian, M.J., Thuret, S., Price, J.,

Pariante, C.M., 2011b. Antidepressants increase human hippocampal neurogenesis by activating the

glucocorticoid receptor. Mol. Psychiatry 16, 738-750.

Andrus, B.M., Blizinsky, K., Vedell, P.T., Dennis, K., Shukla, P.K., Schaffer, D.J., Radulovic, J.,

Churchill, G.A., Redei, E.E., 2012. Gene expression patterns in the hippocampus and amygdala of

endogenous depression and chronic stress models. Mol. Psychiatry 17, 49-61.

Angelucci, F., Brenè, S., Mathé, A.A., 2005. BDNF in schizophrenia, depression and corresponding

animal models. Mol. Psychiatry 10, 345-352.

Aranha, M.M., Solá, S., Low, W.C., Steer, C.J., Rodrigues, C.M., 2009. Caspases and p53 modulate

FOXO3A/Id1 signaling during mouse neural stem cell differentiation. J. Cell Biochem. 107,

748-758.

Arnone, D., McIntosh, A.M., Ebmeier, K.P., Munafo, M.R., Anderson, I.M., 2012. Magnetic resonance

imaging studies in unipolar depression: systematic review and meta-regression analyses. Eur.

Neuropsychopharmacol. 22, 1-16.

Asada, S., Daitoku, H., Matsuzaki, H., Saito, T., Sudo, T., Mukai, H., Iwashita, S., Kako, K., Kishi, T.,

Kasuya, Y., Fukamizu, A., 2007. Mitogen-activated protein kinases, Erk and p38, phosphorylate

and regulate Foxo1. Cell. Signal. 19, 519-527.

Ayuso, M.I., Hernández-Jiménez, M., Martín, M.E., Salinas, M., Alcázar, A., 2010. New hierarchical

phosphorylation pathway of the translational repressor eIF4E-binding protein 1 (4E-BP1) in

ischemia-reperfusion stress. J. Biol. Chem. 285, 34355-34363.

Bambico, F.R., Belzung, C., 2013. Novel insights into depression and antidepressants: a synergy

between synaptogenesis and neurogenesis? Curr. Top. Behav. Neurosci. 15, 243-291.

Beck, K.D., Powell-Braxton, L., Widmer, H.R., Valverde, J., Hefti, F., 1995. Igf1 gene disruption

results in reduced brain size, CNS hypomyelination, and loss of hippocampal granule and striatal

parvalbumin-containing neurons. Neuron 14, 717-730.

Berton, O., Nestler, E.J., 2006. New approaches to antidepressant drug discovery: beyond monoamines.

Nat. Rev. Neurosci. 7, 137-151.

Biggs, W.R., Cavenee, W.K., Arden, K.C., 2001. Identification and characterization of members of the

FKHR (FOX O) subclass of winged-helix transcription factors in the mouse. Mamm. Genome 12,

416-425.

Boccitto, M., Kalb, R.G., 2011. Regulation of Foxo-dependent transcription by post-translational

modifications. Curr. Drug Targets 12, 1303-1310.

Borre, Y.E., Panagaki, T., Koelink, P.J., Morgan, M.E., Hendriksen, H., Garssen, J., Kraneveld, A.D.,

Olivier, B., Oosting, R.S., 2014. Neuroprotective and cognitive enhancing effects of a

Page 28: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

27

multi-targeted food intervention in an animal model of neurodegeneration and depression.

Neuropharmacology 79,738-749.

Brent, M.M., Anand, R., Marmorstein, R., 2008. Structural basis for DNA recognition by FoxO1 and

its regulation by posttranslational modification. Structure 6, 1407-1416.

Breuillaud, L., Rossetti, C., Meylan, E.M., Mérinat, C., Halfon, O., Magistretti, P.J., Cardinaux, J.R.,

2012. Deletion of CREB-regulated transcription coactivator 1 induces pathological aggression,

depression-related behaviors, and neuroplasticity genes dysregulation in mice. Biol. Psychiatry 72,

528-536. Brunet, A., Datta, S.R., Greenberg, M.E., 2001a. Transcription-dependent and -independent control of

neuronal survival by the PI3K-Akt signaling pathway. Curr. Opin. Neurobiol. 11, 297-305.

Brunet, A., Park, J., Tran, H., Hu, L.S., Hemmings, B.A., Greenberg, M.E., 2001b. Protein kinase SGK

mediates survival signals by phosphorylating the forkhead transcription factor FKHRL1 (FOXO3a).

Mol. Cell. Biol. 21, 952-965.

Budziszewska, B., Jaworska-Feil, L., Kajta, M., Lasoń, W., 2000. Antidepressant drugs inhibit

glucocorticoid receptor-mediated gene transcription - a possible mechanism. Br. J. Pharmacol. 130,

1385-1393.

Burgering, B.M., Kops, G.J., 2002. Cell cycle and death control: long live Forkheads. Trends Biochem.

Sci. 27,352-360.

Buse, P., Tran, S.H., Luther, E., Phu, P.T., Aponte, G.W., Firestone, G.L., 1999. Cell cycle and

hormonal control of nuclear-cytoplasmic localization of the serum- and glucocorticoid-inducible

protein kinase, Sgk, in mammary tumor cells. A novel convergence point of anti-proliferative and

proliferative cell signaling pathways. J. Biol. Chem. 274, 7253-7263.

Campbell, S., Macqueen, G., 2004. The role of the hippocampus in the pathophysiology of major

depression. J. Psychiatry Neurosci. 29, 417-426.

Carson, M.J., Behringer, R.R., Brinster, R.L., McMorris, F.A., 1993. Insulin-like growth factor I

increases brain growth and central nervous system myelination in transgenic mice. Neuron 10,

729-740.

Carter, M.E., Brunet, A., 2007. FOXO transcription factors. Curr. Biol. 17, R113-114.

Castets, P., Ruegg, M.A., 2013. MTORC1 determines autophagy through ULK1 regulation in skeletal

muscle. Autophagy 9, 1435-1437.

Castrén, E., Rantamäki, T., 2010. The role of BDNF and its receptors in depression and antidepressant

drug action: Reactivation of developmental plasticity. Dev. Neurobiol. 70, 289-297.

Celada, P., Puig, M., Amargós-Bosch, M., Adell, A., Artigas, F.. 2004. The therapeutic role of 5-HT1A

and 5-HT2A receptors in depression. J. Psychiatry Neurosci. 29, 252-265.

Chaves, I., van der, Horst, G.T., Schellevis, R., Nijman, R.M., Koerkamp, M.G., Holstege, F.C., Smidt,

M.P., Hoekman, M.F., 2014. Insulin-FOXO3 signaling modulates circadian rhythms via regulation

of clock transcription. Curr. Biol. 24, 1248-1255.

Chen, A.T., Guo, C., Dumas, K.J., Ashrafi, K., Hu, P.J., 2013. Effects of Caenorhabditis elegans sgk-1

mutations on lifespan, stress resistance, and DAF-16/FoxO regulation. Aging Cell 12, 932-940.

Chen, Y.J., Hsiao, P.W., Lee, M.T., Mason, J.I., Ke, F.C., Hwang, J.J., 2007. Interplay of PI3K and

cAMP/PKA signaling, and rapamycin-hypersensitivity in TGFbeta1 enhancement of

FSH-stimulated steroidogenesis in rat ovarian granulosa cells. J. Endocrinol. 192, 405-419.

Conde, C., Caceres, A., 2009. Microtubule assembly, organization and dynamics in axons and

dendrites. Nat. Rev. Neurosci. 10, 319-332.

Page 29: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

28

David, D.J., Samuels, B.A., Rainer, Q., Wang, J.W., Marsteller, D., Mendez, I., Drew, M., Craig, D.A.,

Guiard, B.P., Guilloux, J.P., Artymyshyn, R.P., Gardier, A.M., Gerald, C., Antonijevic, I.A.,

Leonardo, E.D., Hen, R., 2009. Neurogenesis-dependent and -independent effects of fluoxetine in

an animal model of anxiety/depression. Neuron 62, 479-493.

de la Torre-Ubieta, L., Bonni, A., 2011. Transcriptional regulation of neuronal polarity and

morphogenesis in the mammalian brain. Neuron 72, 22-40.

Duman, R.S., Aghajanian, G.K., 2012. Synaptic dysfunction in depression: potential therapeutic targets.

Science 338, 68-72.

Eijkelenboom, A., Burgering, B.M., 2013. FOXOs: signalling integrators for homeostasis maintenance.

Nat. Rev. Mol. Cell Biol. 14, 83-97.

Emamian, E.S., Hall, D., Birnbaum, M.J., Karayiorgou, M., Gogos, J.A., 2004. Convergent evidence

for impaired AKT1-GSK3beta signaling in schizophrenia. Nat. Genet. 36, 131-137.

Emens, J., Lewy, A., Kinzie, J.M., Arntz, D., Rough, J., 2009. Circadian misalignment in major

depressive disorder. Psychiatry Res. 168, 259-261.

Evans, S.J., Choudary, P.V., Neal, C.R., Li, J.Z., Vawter, M.P., Tomita, H., Lopez, J.F., Thompson,

R.C., Meng, F., Stead, J.D., Walsh, D.M., Myers, R.M., Bunney, W.E., Watson, S.J., Jones, E.G.,

Akil, H., 2004. Dysregulation of the fibroblast growth factor system in major depression. Proc. Natl.

Acad. Sci. U. S. A. 101, 15506-15511.

Fournier, J.C., DeRubeis, R.J., Hollon, S.D., Dimidjian, S., Amsterdam, J.D., Shelton, R.C., Fawcett, J.,

2010. Antidepressant drug effects and depression severity: a patient-level meta-analysis. JAMA.

303, 47-53.

Furuyama, T., Nakazawa, T., Nakano, I., Mori, N., 2000. Identification of the differential distribution

patterns of mRNAs and consensus binding sequences for mouse DAF-16 homologues. Biochem. J.

349, 629-634.

Germain, A., Kupfer, D.J., 2008. Circadian rhythm disturbances in depression. Hum. Psychopharmacol.

23, 571-585.

Goffer, Y., Xu, D., Eberle, S.E., D'amour, J., Lee, M., Tukey, D., Froemke, R.C., Ziff, E.B., Wang, J.,

2013. Calcium-permeable AMPA receptors in the nucleus accumbens regulate depression-like

behaviors in the chronic neuropathic pain state. J. Neurosci. 33, 19034-19044.

Gold, P.W., Chrousos, G.P., 2013. Melancholic and atypical subtypes of depression represent distinct

pathophysiological entities: CRH, neural circuits, and the diathesis for anxiety and depression. Mol.

Psychiatry 18, 632-634.

Greer, E.L., Banko, M.R., Brunet, A., 2009. AMP-activated protein kinase and FoxO transcription

factors in dietary restriction-induced longevity. Ann. N. Y. Acad. Sci. 1170, 688-692.

Gregorian, C., Nakashima, J., Le Belle, J., Ohab, J., Kim, R., Liu, A., Smith, K.B., Groszer, M., Garcia,

A.D., Sofroniew, M.V., Carmichael, S.T., Kornblum, H.I., Liu, X., Wu, H., 2009. Pten deletion in

adult neural stem/progenitor cells enhances constitutive neurogenesis. J. Neurosci. 29, 1874-1886. Groves, J.O., 2007. Is it time to reassess the BDNF hypothesis of depression? Mol. Psychiatry 12,

1079-1088.

Guilloux, J.P., Mendez-David, I., Pehrson, A., Guiard, B.P., Repérant, C., Orvoën, S., Gardier, A.M.,

Hen, R., Ebert, B., Miller, S., Sanchez, C., David, D.J., 2013. Antidepressant and anxiolytic

potential of the multimodal antidepressant vortioxetine (Lu AA21004) assessed by behavioural and

neurogenesis outcomes in mice. Neuropharmacology 73,147-159.

Hajszan, T., MacLusky, N.J., Leranth, C., 2005. Short-term treatment with the antidepressant

Page 30: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

29

fluoxetine triggers pyramidal dendritic spine synapse formation in rat hippocampus. Eur. J.

Neurosci. 21, 1299-1303.

Hegerl, U., Rummel-Kluge, C., Varnik, A., Arensman, E., Koburger, N., 2013. Alliances against

depression - A community based approach to target depression and to prevent suicidal behaviour.

Neurosci. Biobehav. Rev. 37, 2404-2409.

Hoekman, M.F., Jacobs, F.M., Smidt, M.P., Burbach, J.P., 2006. Spatial and temporal expression of

FoxO transcription factors in the developing and adult murine brain. Gene Expr. Patterns 6,

134-140.

Hollon, S.D., DeRubeis, R.J., Fawcett, J., Amsterdam, J.D., Shelton, R.C., Zajecka, J., Young, P.R.,

Gallop, R., 2014. Effect of cognitive therapy with antidepressant medications vs antidepressants

alone on the rate of recovery in major depressive disorder: a randomized clinical trial. JAMA.

Psychiatry 71, 1157-1164.

Ho, K.K., McGuire, V.A., Koo, C.Y., Muir, K.W., de Olano, N., Maifoshie, E., Kelly, D.J., McGovern,

U.B., Monteiro, L.J., Gomes, A.R., Nebreda, A.R., Campbell, D.G., Arthur, J.S., Lam, E.W, 2012.

Phosphorylation of FOXO3a on Ser-7 by p38 promotes its nuclear localization in response to

doxorubicin. J. Biol. Chem. 287, 1545-1555.

Hsiung, S.C., Adlersberg, M., Arango, V., Mann, J.J., Tamir, H., Liu, K.P., 2003. Attenuated 5-HT1A

receptor signaling in brains of suicide victims: involvement of adenylyl cyclase,

phosphatidylinositol 3-kinase, Akt and mitogen-activated protein kinase. J. Neurochem. 87,

182-194.

Huang, H., Regan, K.M., Lou, Z., Chen, J., Tindall, D.J., 2006. CDK2-dependent phosphorylation of

FOXO1 as an apoptotic response to DNA damage. Science 314, 294-297.

Huang, H., Tindall, D.J., 2007. Dynamic FoxO transcription factors. J. Cell Sci. 120, 2479-2487.

Hui, R.C., Francis, R.E., Guest, S.K., Costa, J.R., Gomes, A.R., Myatt, S.S., Brosens, J.J., Lam, E.W.,

2008. Doxorubicin activates FOXO3a to induce the expression of multidrug resistance gene

ABCB1 (MDR1) in K562 leukemic cells. Mol. Cancer Ther. 7, 670-678.

Hu, M.C., Lee, D.F., Xia, W., Golfman, L.S., Ou-Yang, F., Yang, J.Y., Zou, Y., Bao, S., Hanada, N.,

Saso, H., Kobayashi, R., Hung, M.C., 2004. IkappaB kinase promotes tumorigenesis through

inhibition of forkhead FOXO3a. Cell 117, 225-237.

Jacobs, .FM., van der Heide, L.P., Wijchers, P.J., Burbach, J.P., Hoekman, M.F., Smidt, M.P., 2003.

FoxO6, a novel member of the FoxO class of transcription factors with distinct shuttling dynamics.

J. Biol. Chem. 278, 35959-35967.

Jaitovich, A., Angulo, M., Lecuona, E., Dada, L.A., Welch, L.C., Cheng, Y., Gusarova, G., Ceco, E.,

Liu, C., Shigemura, M., Barreiro, E., Patterson, C., Nader, G.A., Sznajder, J.I., 2015. High CO2

levels cause skeletal muscle atrophy via AMP-activated kinase (AMPK), FoxO3a protein, and

muscle-specific Ring finger protein 1 (MuRF1). J. Biol. Chem. 290, 9183-9194.

Karatsoreos, I.N., 2014. Links between Circadian Rhythms and Psychiatric Disease. Front. Behav.

Neurosci. 8, 162.

Karege, F., Bondolfi, G., Gervasoni, N., Schwald, M., Aubry, J.M., Bertschy, G., 2005. Low

brain-derived neurotrophic factor (BDNF) levels in serum of depressed patients probably results

from lowered platelet BDNF release unrelated to platelet reactivity. Biol. Psychiatry 57, 1068-1072.

Kavalali, E.T., Monteggia, L.M., 2012. Synaptic mechanisms underlying rapid antidepressant action of

ketamine. Am. J. Psychiatry 169, 1150-1156.

Kennedy, L.M., Pham, S.C., Grishok, A., 2013. Nonautonomous regulation of neuronal migration by

Page 31: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

30

insulin signaling, DAF-16/FOXO, and PAK-1. Cell Rep. 4, 996-1009.

Kessler, R.C., 1997. The effects of stressful life events on depression. Annu. Rev. Psychol. 48,

191-214.

Kiejna, A., Pawlowski, T., Dudek, D., Lojko, D., Siwek, M., Roczeń, R., Rybakowski, J.K., 2010. The

utility of Mood Disorder Questionnaire for the detection of bipolar diathesis in treatment-resistant

depression. J. Affect. Disord. 124, 270-274.

Kim, N., Dempsey, C.M., Kuan, C.J., Zoval, J.V., O'Rourke, E., Ruvkun, G., Madou, M.J., Sze, J.Y.,

2007. Gravity force transduced by the MEC-4/MEC-10 DEG/ENaC channel modulates

DAF-16/FoxO activity in Caenorhabditis elegans. Genetics 177, 835-845.

Kirsch, I., Deacon, B.J., Huedo-Medina, T.B., Scoboria, A., Moore, T.J., Johnson, B.T., 2008. Initial

severity and antidepressant benefits: a meta-analysis of data submitted to the Food and Drug

Administration. PLoS. Med. 5, e45.

Kode, A., Mosialou, I., Silva, B.C., Rached, M.T., Zhou, B., Wang, J., Townes, T.M., Hen, R.,

DePinho, R.A., Guo, X.E., Kousteni, S., 2012. FOXO1 orchestrates the bone-suppressing function

of gut-derived serotonin. J. Clin. Invest. 122, 3490-3503.

Krishnan, V., Nestler, E.J., 2008. The molecular neurobiology of depression. Nature 455, 894-902.

Krogh, J., Rostrup. E., Thomsen, C., Elfving, B., Videbech, P., Nordentoft, M., 2014. The effect of

exercise on hippocampal volume and neurotrophines in patients with major depression--a

randomized clinical trial. J. Affect. Disord. 165, 24-30.

Kwon, M., Fernandez, J.R., Zegarek, G.F., Lo, S.B., Firestein, B.L., 2011. BDNF-promoted increases

in proximal dendrites occur via CREB-dependent transcriptional regulation of cypin. J. Neurosci.

31, 9735-9745.

Lam, E.W., Brosens, J.J., Gomes, A.R., Koo, C.Y., 2013. Forkhead box proteins: tuning forks for

transcriptional harmony. Nat. Rev. Cancer 13, 482-495.

Lee, H.K., Rocnik, E., Fu, Q., Kwon, B., Zeng, L., Walsh, K., Querfurth, H., 2012. Foxo/atrogin

induction in human and experimental myositis. Neurobiol. Dis. 46, 463-475.

Lee, J., Duan, W., Mattson, M.P., 2002. Evidence that brain-derived neurotrophic factor is required for

basal neurogenesis and mediates, in part, the enhancement of neurogenesis by dietary restriction in

the hippocampus of adult mice. J. Neurochem. 82, 1367-1375.

Lee, J.W., Chen, H., Pullikotil, P., Quon, M.J., 2011. Protein kinase A-alpha directly phosphorylates

FoxO1 in vascular endothelial cells to regulate expression of vascular cellular adhesion molecule-1

mRNA. J. Biol. Chem. 286, 6423-6432.

Li, N., Lee, B., Liu, R.J., Banasr, M., Dwyer, J.M., Iwata, M., Li, X.Y., Aghajanian, G., Duman, R.S.,

2010. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA

antagonists. Science 329, 959-964.

Liang, B., Moussaif, M., Kuan, C.J., Gargus, J.J., Sze, J.Y., 2006. Serotonin targets the DAF-16/FOXO

signaling pathway to modulate stress responses. Cell Metab. 4, 429-440.

Liang, C., Chen, W., Zhi, X., Ma, T., Xia, X., Liu, H., Zhang, Q., Hu, Q., Zhang, Y., Bai, X., Liang, T.,

2013. Serotonin promotes the proliferation of serum-deprived hepatocellular carcinoma cells via

upregulation of FOXO3a. Mol. Cancer 12,14.

López-Figueroa, A.L., Norton, C.S., López-Figueroa, M.O., Armellini-Dodel, D., Burke, S., Akil, H.,

López, J.F., Watson, S.J., 2004. Serotonin 5-HT1A, 5-HT1B, and 5-HT2A receptor mRNA

expression in subjects with major depression, bipolar disorder, and schizophrenia. Biol. Psychiatry

55, 225-233.

Page 32: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

31

Lutzner, N., Kalbacher, H., Krones-Herzig, A., Rosl, F., 2012. FOXO3 is a glucocorticoid receptor

target and regulates LKB1 and its own expression based on cellular AMP levels via a positive

autoregulatory loop. PLoS One 7, e42166.

Magno, L.A., Santana, C.V., Sacramento, E.K., Rezende, V.B., Cardoso, M.V., Maurício-da-Silva, L.,

Neves, F.S., Miranda, D.M., De Marco, L.A., Correa. H., Romano-Silva, M.A., 2011. Genetic

variations in FOXO3A are associated with Bipolar Disorder without confering vulnerability for

suicidal behavior. J. Affect. Disord 133: 633-637.

Mahar, I., Bambico, F.R., Mechawar, N., Nobrega, J.N., 2014. Stress, serotonin, and hippocampal

neurogenesis in relation to depression and antidepressant effects. Neurosci. Biobehav. Rev. 38,

173-192. Malberg, J.E., Eisch, A.J., Nestler, E.J., Duman, R.S., 2000. Chronic antidepressant treatment increases

neurogenesis in adult rat hippocampus. J. Neurosci. 20, 9104-9110.

Mao, Z., Liu, L., Zhang, R., Li, X., 2007. Lithium reduces FoxO3a transcriptional activity by

decreasing its intracellular content. Biol. Psychiatry 62, 1423-1430.

Mikoteit, T., Beck, J., Eckert, A., Hemmeter, U., Brand, S., Bischof, R., Holsboer-Trachsler, E.,

Delini-Stula, A., 2014. High baseline BDNF serum levels and early psychopathological

improvement are predictive of treatment outcome in major depression. Psychopharmacology (Berl)

231, 2955-2965.

Mitchell, A.J., 2006. Two-week delay in onset of action of antidepressants: new evidence. Br. J.

Psychiatry 188, 105-106.

Mojsilovic-Petrovic, J., Nedelsky, N., Boccitto, M., Mano, I., Georgiades, S.N., Zhou, W., Liu, Y.,

Neve, R.L., Taylor, J.P., Driscoll, M., Clardy, J., Merry, D., Kalb, R.G., 2009. FOXO3a is broadly

neuroprotective in vitro and in vivo against insults implicated in motor neuron diseases. J. Neurosci.

29, 8236-8247.

Muller, J.M., Morelli, E., Ansorge, M., Gingrich, J.A., 2011. Serotonin transporter deficient mice are

vulnerable to escape deficits following inescapable shocks. Genes Brain Behav. 10, 166-175.

Musazzi, L., Mallei, A., Tardito, D., Gruber, S.H., El Khoury, A., Racagni, G., Mathé, A.A., Popoli, M.,

2010. Early-life stress and antidepressant treatment involve synaptic signaling and Erk kinases in a

gene-environment model of depression. J. Psychiatr. Res. 44, 511-520.

Nechipurenko, I.V., Broihier, H.T., 2012. FoxO limits microtubule stability and is itself negatively

regulated by microtubule disruption. J. Cell Biol. 196, 345-362.

Oishi, K., Watatani, K., Itoh, Y., Okano, H., Guillemot, F., Nakajima, K., Gotoh, Y., 2009. Selective

induction of neocortical GABAergic neurons by the PDK1-Akt pathway through activation of

Mash1. Proc. Natl. Acad. Sci. U. S. A. 106, 13064-13069.

Oudega, M.L., van Exel, E., Wattjes, M.P., Comijs, H.C., Scheltens, P., Barkhof, F., Eikelenboom, P.,

de Craen, A.J., Beekman, A.T., Stek, M.L., 2011. White matter hyperintensities, medial temporal

lobe atrophy, cortical atrophy, and response to electroconvulsive therapy in severely depressed

elderly patients. J. Clin. Psychiatry 72, 104-112.

Ota, K.T., Liu, R.J., Voleti, B., Maldonado-Aviles, J.G., Duric, V., Iwata, M., Dutheil, S., Duman, C.,

Boikess, S., Lewis, D.A., Stockmeier, C.A., DiLeone, R.J., Rex, C., Aghajanian, G.K., Duman,

R.S., 2014. REDD1 is essential for stress-induced synaptic loss and depressive behavior. Nat. Med.

20, 531-535.

Paik, J.H., Ding, Z., Narurkar, R., Ramkissoon, S., Muller, F., Kamoun, W.S., Chae, S.S., Zheng, H.,

Ying, H., Mahoney, J., Hiller, D., Jiang, S., Protopopov, A., Wong, W.H., Chin, L., Ligon, K.L.,

Page 33: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

32

DePinho, R.A., 2009. FoxOs cooperatively regulate diverse pathways governing neural stem cell

homeostasis. Cell Stem Cell 5, 540-553. Pallafacchina, G., Calabria, E., Serrano, A.L., Kalhovde, J.M., Schiaffino, S., 2002. A protein kinase

B-dependent and rapamycin-sensitive pathway controls skeletal muscle growth but not fiber type

specification. Proc. Natl. Acad. Sci. U. S. A. 99, 9213-9218.

Pereira, P.A., Bicalho, M.A., de Moraes, E.N., Malloy-Diniz, L., Bozzi, I.C., Nicolato, R., Valadão,

D.R., Miranda, D.M., Romano-Silva, M.A., 2014. Genetic variant of AKT1 and AKTIP associated

with late-onset depression in a Brazilian population. Int. J. Geriatr. Psychiatry 29, 399-405.

Pilar-Cuéllar, F., Vidal, R., Pazos, A., 2012. Subchronic treatment with fluoxetine and ketanserin

increases hippocampal brain-derived neurotrophic factor, β-catenin and antidepressant-like effects.

Br. J. Pharmacol. 165, 1046-1057.

Polter, A., Yang, S., Zmijewska, A.A., van Groen, T., Paik, J.H., Depinho, R.A., Peng, S.L., Jope, R.S.,

Li, X., (2009) Forkhead box, class O transcription factors in brain: regulation and behavioral

manifestation. Biol. Psychiatry 65, 150-159. Poulsen, R.C., Carr, A.J., Hulley, P.A., 2011. Protection against glucocorticoid-induced damage in

human tenocytes by modulation of ERK, Akt, and forkhead signaling. Endocrinology 152,

503-514.

Qin, W., Pan, J., Qin, Y., Lee, D.N., Bauman, W.A., Cardozo, C., 2014. Identification of functional

glucocorticoid response elements in the mouse FoxO1 promoter. Biochem. Biophys. Res. Commun.

450, 979-983.

Raison, C.L., Miller, A.H., 2003. When not enough is too much: the role of insufficient glucocorticoid

signaling in the pathophysiology of stress-related disorders. Am. J. Psychiatry 160, 1554-1565.

Rafalski, V.A., Brunet, A., 2011. Energy metabolism in adult neural stem cell fate. Prog. Neurobiol.

93,182-203.

Rajkowska, G., Miguel-Hidalgo, J.J., Wei, J., Dilley, G., Pittman, S.D., Meltzer, H.Y., Overholser, J.C.,

Roth, B.L., Stockmeier, C.A., 1999. Morphometric evidence for neuronal and glial prefrontal cell

pathology in major depression. Biol. Psychiatry 45, 1085-1098.

Renault, V.M., Rafalski, V.A., Morgan, A.A., Salih, D.A., Brett, J.O., Webb, A.E., Villeda, S.A.,

Thekkat, P.U., Guillerey, C., Denko, N.C., Palmer, T.D., Butte, A.J., Brunet, A., 2009. FoxO3

regulates neural stem cell homeostasis. Cell Stem Cell 5,527-539.

Risch, N., Herrell, R., Lehner, T., Liang, K.Y., Eaves, L., Hoh, J., Griem, A., Kovacs, M., Ott, J.,

Merikangas, K.R., 2009. Interaction between the serotonin transporter gene (5-HTTLPR), stressful

life events, and risk of depression: a meta-analysis. JAMA. 301, 2462-2471.

Sairanen, M., Lucas, G., Ernfors, P., Castrén, M., Castrén, E., 2005. Brain-derived neurotrophic factor

and antidepressant drugs have different but coordinated effects on neuronal turnover, proliferation,

and survival in the adult dentate gyrus. J. Neurosci. 25, 1089-1094.

Salih, D.A., Rashid, A.J., Colas, D., et al. 2012. FoxO6 regulates memory consolidation and synaptic

function. Genes Dev. 26, 2780-2801.

Sanchez, A.M., Candau, R.B., Bernardi, H., 2014. FoxO transcription factors: their roles in the

maintenance of skeletal muscle homeostasis. Cell Mol. Life Sci.71, 1657-1671.

Sandri, M., Sandri, C., Gilbert, A., Skurk, C., Calabria, E., Picard, A., Walsh, K., Schiaffino, S., Lecker,

S.H., Goldberg, A.L., 2004. Foxo transcription factors induce the atrophy-related ubiquitin ligase

atrogin-1 and cause skeletal muscle atrophy. Cell 117, 399-412.

Sanphui, P., Biswas, S.C., 2013. FoxO3a is activated and executes neuron death via Bim in response to

Page 34: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

33

beta-amyloid. Cell Death Dis. 4: e625

Schmid, C.L., Bohn, L.M., 2010. Serotonin, but not N-methyltryptamines, activates the serotonin 2A

receptor via a ss-arrestin2/Src/Akt signaling complex in vivo. J. Neurosci. 30, 13513-13524.

Schmidt-Strassburger, U., Schips, T.G., Maier, H.J., Kloiber, K., Mannella, F., Braunstein, K.E.,

Holzmann, K., Ushmorov, A., Liebau, S., Boeckers, T.M., Wirth, T., 2012. Expression of

constitutively active FoxO3 in murine forebrain leads to a loss of neural progenitors. Faseb. J. 26,

4990-5001.

Seo, M.K., Lee, C.H., Cho, H.Y., Lee, J.G., Lee, B.J., Kim, J.E., Seol, W., Kim, Y.H., Park, S.W., 2014.

Effects of antidepressant drugs on synaptic protein levels and dendritic outgrowth in hippocampal

neuronal cultures. Neuropharmacology 79, 222-233.

Sheline, Y.I., Gado, M.H., Kraemer, H.C., 2003. Untreated depression and hippocampal volume loss.

Am. J. Psychiatry 160, 1516-1518.

Sheline, Y.I., Wang, P.W., Gado, M.H., Csernansky, J.G., Vannier, M.W., 1996. Hippocampal atrophy in

recurrent major depression. Proc. Natl. Acad. Sci. U. S. A. 93, 3908-3913.

Shi, H.S., Zhu, W.L., Liu, J.F., Luo, Y.X., Si, J.J., Wang, S.J., Xue, Y.X., Ding, Z.B., Shi, J., Lu, L.,

2012. PI3K/Akt signaling pathway in the basolateral amygdala mediates the rapid

antidepressant-like effects of trefoil factor 3. Neuropsychopharmacol. 37, 2671-2683.

Skurk, C., Izumiya, Y., Maatz, H., Razeghi, P., Shiojima, I., Sandri, M., Sato, K., Zeng, L., Schiekofer,

S., Pimentel, D., Lecker, S., Taegtmeyer, H., Goldberg, A.L., Walsh, K., 2005. The FOXO3a

transcription factor regulates cardiac myocyte size downstream of AKT signaling. J. Biol. Chem.

280, 20814-20823.

Stitt, T.N., Drujan, D., Clarke, B.A., Panaro, F., Timofeyva, Y., Kline, W.O., Gonzalez, M.,

Yancopoulos, G.D., Glass, D.J., 2004. The IGF-1/PI3K/Akt pathway prevents expression of muscle

atrophy-induced ubiquitin ligases by inhibiting FOXO transcription factors. Mol. Cell 14, 395-403.

Southgate, R.J., Neill, B., Prelovsek, O., El-Osta, A., Kamei, Y., Miura, S., Ezaki, O., McLoughlin,

T.J., Zhang, W., Unterman, T.G., Febbraio, M.A., 2007. FOXO1 regulates the expression of

4E-BP1 and inhibits mTOR signaling in mammalian skeletal muscle. J. Biol. Chem. 282,

21176-21186.

Sunters, A., Madureira, P.A., Pomeranz, K.M., Aubert, M., Brosens, J.J., Cook, S.J., Burgering, B.M.,

Coombes, R.C., Lam, E.W., 2006. Paclitaxel-induced nuclear translocation of FOXO3a in breast

cancer cells is mediated by c-Jun NH2-terminal kinase and Akt. Cancer Res. 66, 212-220.

Sussman, N., 1994. The potential benefits of serotonin receptor-specific agents. J. Clin. Psychiatry 55

Suppl, 45-51.

Tochigi, M., Iwamoto, K., Bundo, M., Sasaki, T., Kato, N., Kato, T., 2008. Gene expression profiling

of major depression and suicide in the prefrontal cortex of postmortem brains. Neurosci. Res. 60,

184-191. Tripp, A., Kota, R.S., Lewis, D.A., Sibille, E., 2011. Reduced somatostatin in subgenual anterior

cingulate cortex in major depression. Neurobiol. Dis. 42, 116-124. Underwood, M.D., Khaibulina, A.A., Ellis, S.P., Moran, A., Rice, P.M., Mann, J.J., Arango, V., 1999.

Morphometry of the dorsal raphe nucleus serotonergic neurons in suicide victims. Biol. Psychiatry

46, 473-483.

van der Vos, K.E., Coffer, P.J., 2011. The extending network of FOXO transcriptional target genes.

Antioxid. Redox Signal. 14, 579-592.

Wang, H., Zhou, X., Huang, J., Mu, N., Guo, Z., Wen, Q., Wang, R., Chen, S., Feng, Z.P., Zheng, W.,

Page 35: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

34

2013. The role of Akt/FoxO3a in the protective effect of venlafaxine against corticosterone-induced

cell death in PC12 cells. Psychopharmacology (Berl) 228, 129-141. Waterhouse, E.G., An, J.J., Orefice, L.L., Baydyuk, M., Liao, G.Y., Zheng, K., Lu, B., Xu, B., 2012.

BDNF promotes differentiation and maturation of adult-born neurons through GABAergic

transmission. J. Neurosci. 32, 14318-14330.

Webb, A.E., Pollina, E.A., Vierbuchen, T., Urbán, N., Ucar, D., Leeman, D.S., Martynoga, B., Sewak,

M., Rando, T.A., Guillemot, F., Wernig, M., Brunet, A., 2013. FOXO3 shares common targets with

ASCL1 genome-wide and inhibits ASCL1-dependent neurogenesis. Cell Rep. 4: 477-491.

Weeks, K.R., Dwyer, D.S., Aamodt, E.J., 2010. Antipsychotic drugs activate the C. elegans akt

pathway via the DAF-2 insulin/IGF-1 receptor. ACS. Chem. Neurosci. 1, 463-473.

Wen, Q., Wang, H., Little, P.J., Quirion, R., Zheng, W., 2012. Forkhead family transcription factor

FoxO and neural differentiation. Neurogenetics 13, 105-113.

Werner, F.M., Coveñas, R., 2013. Classical neurotransmitters and neuropeptides involved in major

depression in a multi-neurotransmitter system: a focus on antidepressant drugs. Curr. Med. Chem.

20, 4853-4858.

White, J.P., Gao, S., Puppa, M.J., Sato, S., Welle, S.L., Carson, J.A., 2013. Testosterone regulation of

Akt/mTORC1/FoxO3a signaling in skeletal muscle. Mol. Cell. Endocrinol. 365, 174-186.

Woodgett, J.R., 2005. Recent advances in the protein kinase B signaling pathway. Curr. Opin. Cell Biol.

17, 150-157.

Yang, J.Y., Zong, C.S., Xia, W., Yamaguchi, H., Ding, Q., Xie, X., Lang, J.Y., Lai, C.C., Chang, C.J.,

Huang, W.C., Huang, H., Kuo, H.P., Lee, D.F., Li, L.Y., Lien, H.C., Cheng, X., Chang, K.J., Hsiao,

C.D., Tsai, F.J., Tsai, C.H., Sahin, A.A., Muller, W.J., Mills, G.B., Yu, D., Hortobagyi, G.N., Hung,

M.C., 2008. ERK promotes tumorigenesis by inhibiting FOXO3a via MDM2-mediated degradation.

Nat. Cell Biol. 10, 138-148.

Yuan, Z., Becker, E.B., Merlo, P., Yamada, T., DiBacco, S., Konishi, Y., Schaefer, E.M., Bonni, A.,

2008. Activation of FOXO1 by Cdk1 in cycling cells and postmitotic neurons. Science 319,

1665-1668.

Zemva, J., Schilbach, K., Stöhr, O., Moll, L., Franko, A., Krone, W., Wiesner, R.J., Schubert, M., 2012.

Central FoxO3a and FoxO6 expression is down-regulated in obesity induced diabetes but not in

aging. Exp. Clin. Endocrinol. Diabetes 120, 340-350.

Zhang, S., Huan, W., Wei, H., Shi, J., Fan, J., Zhao, J., Shen, A., Teng, H., 2013. FOXO3a/p27kip1

expression and essential role after acute spinal cord injury in adult rat. J. Cell Biochem. 114,

354-365.

Zhao, J., Brault, J.J., Schild, A., Cao, P., Sandri, M., Schiaffino, S., Lecker, S.H., Goldberg, A.L., 2007.

FoxO3 coordinately activates protein degradation by the autophagic/lysosomal and proteasomal

pathways in atrophying muscle cells. Cell Metab. 6, 472-483.

Zhao, Y., Wang, Y., Zhu, W.G., 2011. Applications of post-translational modifications of FoxO family

proteins in biological functions. J. Mol. Cell. Biol. 3, 276-282.

Zheng, W.H., Kar, S., Quirion, R., 2002. FKHRL1 and its homologs are new targets of nerve growth

factor Trk receptor signaling. J. Neurochem. 80, 1049-1061.

Zheng, W.H., Quirion, R., 2004. Comparative signaling pathways of insulin-like growth factor-1 and

brain-derived neurotrophic factor in hippocampal neurons and the role of the PI3 kinase pathway

in cell survival. J. Neurochem. 89, 844-852.

Zheng, W.H., Quirion, R., 2009. Glutamate acting on N-methyl-D-aspartate receptors attenuates

Page 36: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

35

insulin-like growth factor-1 receptor tyrosine phosphorylation and its survival signaling properties

in rat hippocampal neurons. J. Biol. Chem. 284, 855-861.

Zheng, W., Zeng, Z., Bhardwaj, S.K., Jamali, S., Srivastava, L.K., 2013. Lithium normalizes

amphetamine-induced changes in striatal FoxO1 phosphorylation and behaviors in rats.

Neuroreport 24: 560-565.

Zheng, X., Yang, Z., Yue, Z., Alvarez, J.D., Sehgal, A., 2007. FOXO and insulin signaling regulate

sensitivity of the circadian clock to oxidative stress. Proc. Natl. Acad. Sci. U. S A.

104,15899-15904.

Zhou, W., Chen, L., Yang, S., Li, F., Li, X., 2012. Behavioral stress-induced activation of FoxO3a in

the cerebral cortex of mice. Biol. Psychiatry 71, 583-592.

Zhu, W., Bijur, G.N., Styles, N.A., Li, X., 2004. Regulation of FOXO3a by brain-derived neurotrophic

factor in differentiated human SH-SY5Y neuroblastoma cells. Brain Res. Mol. Brain Res. 126,

45-56.

Zunszain, P.A., Horowitz, M.A., Cattaneo, A., Lupi, M.M., Pariante, C.M., 2013. Ketamine:

synaptogenesis, immunomodulation and glycogen synthase kinase-3 as underlying mechanisms of

its antidepressant properties. Mol. Psychiatry 18, 1236-1241.

Page 37: Forkhead Box O Transcription Factors as Possible Mediators in …368852/UQ368852... · 2019. 10. 9. · T D ACCEPTED MANUSCRIPT 1 Forkhead Box O Transcription Factors as Possible

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

Highlights

FoxO transcription factors regulate the behavioral manifestation of depression

Antidepressants regulate the phosphorylation/activities of FoxOs

FoxOs are regulated by serotonin and norepinephrine receptor signaling and HPA axis

FoxOs induce cell atrophy and this effect may affect neuronal morphology