401
Views
9
CrossRef citations to date
0
Altmetric
Review

The Potential Role of Epigenetic Drugs in the Treatment of Anxiety Disorders

Pages 597-606 | Published online: 02 Mar 2020

Abstract

There is increasing evidence that abnormalities in epigenetic mechanisms of gene expression contribute to the pathogenesis of anxiety disorders (ADs). This article discusses the role of epigenetic mechanisms of gene expression in the pathogenesis of ADs. It also discusses the data so far obtained from preclinical and clinical trials on the use of epigenetic drugs for treating ADs. Most drug trials investigating the use of epigenetic drugs for treating ADs have used histone deacetylase inhibitors (HDACi). HDACi are showing favorable results in both preclinical and clinical drug trials for treating ADs. However, at present the mode of action of HDACi in ADs is not clear. More work needs to be done to elucidate how epigenetic dysregulation contributes to the pathogenesis of ADs. More work also needs to be done on the mode of action of HDACi in alleviating the signs and symptoms of ADs.

Introduction

Anxiety disorders (ADs) are common disorders having a current global prevalence of 7.3%, with a range of 4.8% to 10.9%.Citation1 The age of onset of these disorders is usually in the mid-twenties, and women are twice as likely than men to be diagnosed to have these disorders.Citation2 In patients with ADs there is anxiety in situations where there is no external reason or cause for such distress, and the patient’s daily functioning is affected.Citation3 Such patients experience a diffuse, unpleasant, and vague feeling of apprehension, often accompanied by autonomic symptoms like headache, sweating, palpitations, tightness in the chest, mild abdominal discomfort, and restlessness.Citation4 There is a high amount of co-morbidity between ADs and depressive disordersCitation5 and drug addiction.Citation6

ADs form a group of related but distinct psychiatric disorders including panic disorder (PD), agoraphobia, specific phobia, social anxiety disorder (SAD), and generalized anxiety disorder (GAD).Citation4 PD involves acute intense attacks of anxiety with feelings of impending doom. Regarding PD, there can be many attacks in one day, or only a few attacks in a year. Agoraphobia refers to fear or anxiety about places from which escape may be difficult. Specific phobia involves an excessive fear of a specific object, circumstance, or situation. SAD (social phobia) refers to the fear of social situations which involve scrutiny or interaction with strangers. GAD involves anxiety about everything, for most days during at least a six-month period.Citation4 The different subtypes of ADs often occur together in the same patient.Citation5

Pathogenesis of Anxiety Disorders

Both psychological and biological factors are thought to contribute to the pathogenesis of ADs.Citation4 Psychological factors involved in the pathogenesis of ADs comprise traumatic and stressful life events, especially during childhood.Citation7 There is consistent evidence that many types of psychosocial stress increase the synthesis and release of cortisol.Citation4 Cortisol helps to mobilize and restore energy stores and contributes to changes like increased arousal, vigilance, focused attention, and memory formation.Citation4 However, excessive and sustained cortisol secretion can lead to serious adverse effects like hypertension, dyslipidemia, insulin resistance, and cardiovascular disease.Citation4 In addition to psychological factors, biological factors play a crucial role in the pathogenesis of ADs. For example, family and twin studies have shown that genetic factors are involved in the pathogenesis of ADs.Citation7 Segregation analysis has shown that ADs are not Mendelian disorders, but complex traits involving multiple genes interacting with environmental factors resulting in the development of the disorders in affected individuals.Citation7 However, linkage and candidate gene studies have produced inconclusive results, and genome-wide association studies (GWAS) have so far not reached genome-wide significance.Citation8 Hence, at present, the results of genetic mapping studies are equivocal and inconclusive.Citation8 The autonomic nervous system of some patients with ADs, especially those suffering from PD, shows raised sympathetic tone, slow adaptation to repeated stimuli, and excessive response to moderate stimuli.Citation4 Abnormal functioning of three neurotransmitters, noradrenaline, serotonin, and γ-aminobutyric acid (GABA) has been implicated in the pathogenesis of ADs.Citation4 Neuroimaging studies have commonly implicated the amygdala, insula, and prefrontal cortex (PFC) in the pathophysiology of ADs.Citation9

Current Pharmacotherapy of Anxiety Disorders

Many of the currently used drugs for ADs modify the functioning of noradrenaline, serotonin, and GABA by enhancing their actions. Currently used antianxiety (anxiolytic) drugs include selective serotonin reuptake inhibitors (SSRIs), serotonin-noradrenaline reuptake inhibitors (SNRIs), benzodiazepines, buspirone, and ß-adrenergic receptor blockers.Citation10 The histamine-1 receptor blocker hydroxyzine which causes short-term sedation, is used in treating patients in whom other anxiolytic drugs are not recommended.Citation10 The anticonvulsant drug pregabulin may be useful in the treatment of ADs, as in some patients with GAD, SAD, and PD.Citation4,Citation11 The monoamine oxidase inhibitor moclobemide may be helpful in treating severe cases of SAD.Citation4,Citation11

The drug of choice for treating ADs depends on the subtype of the disorder and the clinical need for acute anxiolytic effects.Citation11 Among the drugs, only the benzodiazepines and the ß-adrenergic receptor blockers are effective acutely. For the treatment of chronic anxiety SSRIs and SNRIs are used. For situational anxiety, the ß-adrenergic receptor blockers are used. For example, propranolol, in conjunction with psychological intervention, has been shown to decrease anxiety in subjects who develop situational anxiety due to spiders.Citation12 For immediate effects, the benzodiazepines are used.Citation11 In conjunction with pharmacotherapy, psychotherapy is essential for the effective control of symptoms of patients with ADs.Citation6

Outline of Epigenetics

Epigenetics, which literally means above, or in addition to, genetics, is presently an active area of research in both biology and medicine. It refers to heritable changes in gene expression not involving changes in DNA sequence. It involves molecular mechanisms like DNA methylation, DNA hydoxymethylation, histone modifications, and non-coding RNA (ncRNA)-mediated regulation of gene expression.Citation13 Regarding ncRNAs, they can be considered to be a part of epigenetics, when epigenetics is considered in a broad sense.Citation13 For example, epigenetic mechanisms of gene expression like DNA methylation can regulate ncRNA expression.Citation14 Conversely, ncRNAs can regulate epigenetic mechanisms of gene expression. For example, ncRNAs can regulate the expression of genes encoding proteins like DNA methyltransferases (DNMTs) that are involved in the epigenetic regulation of gene expression.Citation14 ncRNAs can be classified based on size into long ncRNAs (lncRNAs) which are more than 200 base pairs long and short ncRNAs which are less than 200 base pairs long.Citation13 The most extensively studied short ncRNAs are microRNAs (miRNAs).Citation13

Epigenetic mechanisms of gene expression are known to interact with each other. Unlike the DNA sequence, epigenetic mechanisms of gene expression are relatively easily modified by the environment.Citation15 Epigenetic mechanisms of gene expression are known to vary between cells and tissues and also show temporal variation.Citation16,Citation17 There is increasing evidence that abnormalities in epigenetic mechanisms of gene expression contribute to the pathogenesis of disease, especially common disease. Indeed, epigenetics has been referred to as the epicenter of modern medicine.Citation18 There is also increasing evidence that epigenetic mechanisms of gene expression are dysregulated in virtually every type of psychiatric disorder and that dysregulated epigenetic mechanisms contribute to their pathogenesis.Citation16,Citation17,Citation19

Role of Epigenetics in the Pathogenesis of Anxiety Disorders

Scope of Epigenetics in Anxiety Disorders

Several lines of evidence suggest that abnormalities in epigenetic mechanisms of gene expression contribute to the pathogenesis of ADs: 1. As alluded to above, no genetic mutation or polymorphism underlying these disorders has been conclusively identified. 2. The concordance rate of identical twins for ADs is only 30 to 40%, supporting the idea that there are environmental factors involved in their pathogenesis.Citation20,Citation21 3. Environmental factors, especially psychosocial stress, are known to influence the pathogenesis of ADs.Citation22 4. There are gender differences in the epidemiology of ADs: The lifetime prevalence of ADs in women is 30.5% whereas in men it is 19.2%.Citation4 5. As given below, there is experimental evidence that epigenetic mechanisms of gene expression are dysregulated in patients with ADs.

To date, no study has investigated epigenetic changes in the post-mortem brain of patients with ADs. The reason for this is the practical difficulty of obtaining suitable post-mortem brain tissues from patients diagnosed to have ADs. However, studies have been conducted on peripheral cells and tissues obtained from patients with ADs as proxies for brain tissues.

Roles of DNA Methylation and Histone Modifications in Anxiety Disorders

Changes in DNA methylation and histone modifications in animal models of ADs are listed in and in peripheral cells in patients with ADs in . It is interesting that in two studiesCitation24,Citation25 hypermethylation of the GAD1 gene, which encodes the enzyme glutamic acid decarboxylase1, the enzyme that catalyzes the decarboxylation of glutamic acid to GABA, was found. Hypermethylation of DNA correlates with inhibition of gene expression.Citation13 Hence hypermethylation of GAD1 would lead to reduced expression of GAD1, and hence reduced formation of GABA, the major inhibitory neurotransmitter in the brain. In this context, it is known that patients with PD have been found to have lower brain concentrations of GABA than healthy controls.Citation43,Citation44 However, the data on GABA concentrations in plasma and cerebrospinal fluid of patients with anxiety is contradictory, possibly due to differences between studies in study design and methodology, tissues where sampling was done, and patient populations. Supporting a direct role for GABA in ADs is the recent finding of He et alCitation45 that repeated oral administration of GABA to emotionally stressed rats was found to reduce anxiety. These authors subjected the rats to stress using the open field and elevated plus maze models.

Table 1 Epigenetic Changes in Animal Models of Anxiety Disorders

Table 2 Epigenetic Changes in Human Peripheral Cells in Anxiety Disorders

Stressful life events, especially those during childhood, are known to increase the risk for adult-onset ADs.Citation69 Stress is known to activate many neuronal circuits, like those in the hippocampus,Citation70 and the hypothalamic-pituitary-adrenal (HPA) axis. Genes encoding proteins associated with the HPA axis like the glucocorticoid receptor (GR or NR3C1), corticotropin releasing factor (CRF), FK506 binding protein 5 (FKBP5; a co-chaperone of the glucocorticoid receptor), proopiomelanocortin (POMC), and vasopressin have been found to show abnormalities in DNA methylation by previous studies.Citation69 These studies suggest that excessive stress due to dysregulation of the HPA axis caused by changes in DNA methylation of such genes can upregulate the HPA axis and increase anxiety.Citation69 A consistent epigenetic change appears to be hypermethylation of the Nr3C1 gene, the gene encoding the GR. Such findings have been seen in animal models of ADs,Citation26 and in peripheral tissues of patients with ADs.Citation61,Citation62 Indeed, there is converging evidence that in individuals who experience stress during early life hypermethylation of the Nr3C1 gene could contribute to the development of stress during adulthood.Citation71

The GR is also known to interact with histone deacetylase 6 (HDAC6) in the brain. Lee and co-workers,Citation72 showed that acute stress, via the GR, increases glutamatergic signaling in the PFC of rats. The authors found that inhibition or knockdown of HDAC6 prevents the enhancement of glutamatergic signaling by acute stress. The same treatment of the GR chaperone protein HSP90, a substrate of HDAC6, produced similar results. These results suggested to the authors that HDAC6 is a key enzyme regulating the synaptic effects of acute stress in the PFC. Espallergues et alCitation73 showed that selective knockout of HDAC6 in the dorsal raphe neurons in mice reduces the anxiogenic effects of glucocorticoids in mice. These authors found that in mice exposed to chronic social defeat, HDAC6 depletion in serotonergic neurons prevents social avoidance. HDAC6 depletion was associated with decreased interaction between HSP90 and the GR.

FKBP5 is a chaperone protein that negatively regulates GR sensitivity by reducing binding affinity and restricting nuclear translocation.Citation69 Roberts et alCitation63 found that the FKBP5 gene in peripheral venous blood is hypermethylated in patients with agarophobia with or without PD and that this is corrected by psychotherapy. These findings support the previous data of the same groupCitation74 that children with ADs had hypermethylation in the FKBP5 gene and that this was corrected by psychotherapy.

Brain-derived neurotrophic factor (BDNF) is a member of a family of neurotrophins which also includes nerve growth factor. BDNF is a key regulator of neuronal differentiation, structure, and function.Citation75 Moreover, there is evidence that BDNF modulates neuronal activity to impact complex human phenotypes like memory, anxiety, and depression.Citation75 BDNF levels appear to be reduced in patients with ADs.Citation76 However, this is not consistent across all the different subtypes of ADs.Citation76 The gene encoding BDNF has been found to be epigenetically modified, with reports of hypermethylation in the rat amygdala,Citation28 and peripheral venous blood,Citation60 and salivaCitation64 of patients with ADs. Since hypermethylation is associated with reduced gene expression, this would lead to reduced levels of BDNF. The oxytocin neuropeptide system plays a role in social behavior and cognition.Citation69 In this regard, the gene encoding the oxytocin receptor has shown changes in DNA methylation in patients with ADs.Citation52,Citation53,Citation64

Role of Non-Coding RNAs in Anxiety Disorders

ncRNAs play a major role in neurogenesis and the regulation of normal neuronal function in the brain.Citation77 For example, they regulate neuronal plasticity, learning and memory.Citation78 There is also increasing evidence that ncRNAs, especially miRNAs, are involved in the pathogenesis of neuropsychiatric disorders including ADs.Citation78 For example, experimental up- or down-regulation of candidate miRNAs associated with neurocircuits associated with anxiety can modulate anxiety-related behavior in animal models of ADs.Citation77

Studies on animal models of ADs suggest that specific miRNAs may play various roles in the development and progression of anxiety in a brain region-specific manner.Citation78 Transposable elements (TEs) are mobile genetic elements (pieces of DNA capable of moving to new locations).Citation79 They make up to two-thirds of the human genome.Citation80 There is growing evidence of a close association between TEs and ncRNAs, and many small ncRNAs originate from TEs.Citation79 TEs have been associated with disease pathology in patients with ADs. For example, stress can interact with the epigenome to regulate the expression of TEs in a region-specific manner to alter stress responsiveness, anxiety, and brain plasticity.Citation81 Compared to changes in DNA methylation and histone modifications relatively few studies to date have investigated the role of ncRNAs in the pathogenesis of ADs ( and ).

Trials of Epigenetic Drugs in Anxiety Disorders

lists preclinical drug trials that have evaluated epigenetic drugs for treating ADs. As shown, most of the trials have evaluated the use of HDAC inhibitors (HDACi) in preclinical studies of the use of epigenetic drugs for treating ADs. All the preclinical drugs trials that evaluated the use of HDACi listed in found that these drugs alleviate stress or anxiety. The HDACi that were evaluated (MS-275, trichostatin A (TSA), vorinostat, sodium butyrate, and valproic acid) belong to different classes of HDACi. However, it is to be noted that most of them inhibit HDACs I and/or II, both of which are class I HDACs.Citation92 One studyCitation91 evaluated the use of the naturally occurring compound lactate which was found to promote resilience to stress in mice by modulating the activity of HDACs.

Table 3 Preclinical Trials of Epigenetic Drugs in Anxiety Disorders

lists the clinical trials of epigenetic drugs in the treatment of anxiety. As listed, all the clinical trials made use of valproate, which is known to functions as a HDACi. Five of the 6 trials tested the anxiolytic effect of valproate on patients with ADs, and all found valproate to give beneficial effects.Citation93Citation97 One trialCitation98 was conducted to determine whether valproate reduces anxiety in healthy subjects and it found that valproate reduces anxiety in such subjects. It must be noted that valproate in addition to acting as a HDACi, also inhibits nerve conduction, which could also contribute to its antianxiety effects.

Table 4 Clinical Trials of Epigenetic Drugs in Anxiety Disorders

miRNA-directed therapy is another option in the treatment of ADs.Citation99 miRNAs in patients with ADs can be targeted to lead to an upregulation of the targeted miRNA. Another possible therapy involves the inhibition of miRNAs in order to reduce their effects in target areas. Antagomirs and antisense oligonucleotides are being investigated for inhibiting miRNAs in preclinical drug trials.Citation99 However, miRNA-directed therapy in psychiatric disorders including ADs is in its early stages, and much more research needs to be done in order to take this field forward.Citation99

Implications of Trials of Epigenetic Drugs in Anxiety Disorders

There are several classes of drugs that are effective for the treatment of ADs. However, there are problems with the currently available drugs. For example, about one third of patients with ADs do not adequately respond to the current drugs.Citation2 A related problem is treatment- resistant ADs.Citation100 Another problem is adverse effects that include habituation, dependence, and abuse.Citation10 In this light, new drugs could be useful in the treatment of patients with ADs. One potential class of new drugs for treating ADs is epigenetic drugs. As shown in and , preclinical and clinical drug trials using HDACi for the treatment of ADs are showing favorable results.

HDACs are a group of enzymes that belong to two families and four classes.Citation101 The two families are the HDAC family and the Sir2 regulator family. In humans HDACs are divided into four classes based on sequence similarities. The class I enzymes are HDAC1, HDAC2, HDAC3, and HDAC8. The class II enzymes are HDAC4, HDAC5, HDAC6, HDAC7, HDAC9, and HDAC10. The class III enzymes have sequence similarity to the yeast Sir2 protein and are SIRT1, SIRT2, SIRT3, SIRT4, SIRT5, SIRT6, and SIRT7. Class IV HDACs has one enzyme, HDAC11.

When compared to histone acetyltransferases (HATs), the quest for specific histone substrates for HDACs has proved to be arduous. The problems encountered include very low measurable HDAC activity when HDACs are purified to homogeneity, and functional redundancy of many HDACs.Citation101 HDACs also have non-histone substrates in addition to histones. Using high-resolution mass spectrometry, it was demonstrated that there are more than 3600 acetylation sites on 1750 proteins. The acetylation sites are present on nuclear, cytoplasmic, and mitochondrial proteins involved in many different cellular processes.Citation102 The broad spectrum HDACi, SAHA and MS-275, were found to up-regulate about 10% of all the sites by at least a factor of 2, indicating that many of these acetylation reactions are regulated by classical HDACs. Computational models suggest that many more acetylation sites occur. These findings suggest that acetylation due to HDACs is common in histone and non-histone proteins. 

Clinically useful HDACi are classified according to their chemical structureCitation92 into hydroxamic acids (vorinostat or SAHA, panobinostat, belinostat, TSA, quisinostat, rocilinostat, and abexinostat), short chain fatty acids (butyrate and valproic acid), cyclic peptides (romidepsin), and benzamides (mocetinostat or MGCD0103 and entinostat or MS-275). Most currently available HDACi inhibit classes I, II, and IV of the HDACs, but not SIRT enzymes. HDACi, like HDACs, affect several targets in addition to HDACs and have mechanisms of action other than inhibition of HDAC. Many targets of HDACi are involved in anti-tumor pathways. HDACi cause accumulation of acetylated forms of histone and non-histone proteins that regulate gene expression, cell proliferation, and cell death.Citation103 Although HDACi can up-regulate global histone acetylation levels, they do not always cause histone hyperacetylation, particularly at regions near gene promoters.Citation104 These drugs up-regulate or down-regulate an equal number of genes. Hence, at present, the possible mechanism of action of HDACi in the treatment of the ADs is unclear. However, it has been shown that HDACi in addition to causing histone acetylation, also cause DNA demethylation, possibly due to increased levels of the demethylating enzyme ten-eleven translocation methylcytosine dioxygenase 1 (TET1).Citation105,Citation106 Since several genes have been shown to be hypermethylated in animal models of ADs () and in human peripheral cells in patients with ADs (), DNA demethylation of such genes by HDACi may contribute to the mechanism of action of HDACi in ADs.

As mentioned in the introduction, co-morbidity exists between ADs and depressive disorders. Since HDACi are giving favorable results in preclinical and clinical trials of depressive disorders,Citation107 these drugs could be useful for treating patients with ADs who also have depressive symptoms. Since HDACi are known to have cognition-enhancing effects, this property of these drugs may also make them suitable for treating patients with ADs some of whom can experience cognitive deficits.Citation108

Conclusions

Several lines of evidence suggest that disrupted epigenetic mechanisms of gene expression contribute to the pathogenesis of ADs. However, the study of the role of abnormal epigenetic mechanisms of gene expression in ADs is in its early stages, and more work needs to be done to clarify issues in this area. Preclinical and clinical drug trials suggest that HDACi give favorable results in the treatment of ADs. However, at present the mode of action of HDACi in ADs is not clear. More work needs to be done on elucidating the mode of action of HDACi in alleviating the signs and symptoms of ADs.

Disclosure

The author alone is responsible for the content and writing of this paper and he reports no conflicts of interest.

References

  • Stein DJ, Scott KM, de Jonge P, Kessler RC. Epidemiology of anxiety disorders: from surveys to nosology and back. Dialogues Clin Neurosci. 2017;19:127–136.28867937
  • Maron E, Nutt D. Biological predictors of pharmacological therapy in anxiety disorders. Dialogues Clin Neurosci. 2015;17:305–317.26487811
  • Feldman RS. Understanding Psychology. New York: McGraw-Hill; 2011.
  • Sadock BJ, Sadock VA, Ruiz P. Kaplan & Sadock’s Synopsis of Psychiatry. New York: Wolters-Kluwer; 2015.
  • Thibaut F. Anxiety disorders: a review of current literature. Dialogues Clin Neurosci. 2017;19:87–88.28867933
  • Lüthi A, Lüscher C. Pathological circuit function underlying addiction and anxiety disorders. Nat Neurosci. 2014;17:1635–1643. doi:10.1038/nn.384925402855
  • Schiele MA, Domschke K. Epigenetics at the crossroads between genes, environment and resilience in anxiety disorders. Genes Brain Behav. 2018;17:1–15. doi:10.1111/gbb.2018.17.issue-3
  • Shimada-Sugimoto M, Otowa T, Hettema JM. Genetics of anxiety disorders: genetic epidemiological and molecular studies in humans. Psychiatry Clin Neurosci. 2015;69:388–401. doi:10.1111/pcn.2015.69.issue-725762210
  • Taylor JM, Whalen PJ. Neuroimaging and anxiety: the neural substrates of pathological and non-pathological anxiety. Curr Psychiatry Rep. 2015;17:49. doi:10.1007/s11920-015-0586-925957101
  • O’Donnell JM, Bies RR, Shelton RC. Drug therapy of depression and anxiety disorders In: Brunton LL, Hilal-Dandan R, Knollmann BC, editors. The Pharmacological Basis of Therapeutics. New York: McGraw-Hill; 2018:267–277.
  • Bandelow B, Michaelis S, Wedekind D. Treatment of anxiety disorders. Dialogues Clin Neurosci. 2017;19:93–106.28867934
  • Soeter M, Kindt M. An abrupt transformation of phobic behavior after a post-retrieval amnesic agent. Biol Psychiatry. 2015;78:880–886. doi:10.1016/j.biopsych.2015.04.00625980916
  • Allis CD, Caparros M-L, Jenuwein T, Lachner M, Reinberg D. Overview and concepts In: Allis CD, Caparros M-L, Jenuwein T, Reinberg D, Lachner M, editors. Epigenetics. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press; 2015:47–115.
  • Chhabra R. The epigenetics of noncoding RNA In: Tollefsbol TO, editor. Handbook of Epigenetics: The New Molecular and Medical Genetics. Cambridge, MA: Elsevier; 2017:47–59.
  • Feil R, Fraga MF. Epigenetics and the environment: emerging patterns and implications. Nat Rev Genet. 2012;13:97–109. doi:10.1038/nrg314222215131
  • Peedicayil J, Grayson DR. An epigenetic basis for an omnigenic model of psychiatric disorders. J Theor Biol. 2018;443:52–55. doi:10.1016/j.jtbi.2018.01.02729378208
  • Peedicayil J, Grayson DR. Some implications of an epigenetic-based omnigenic model of psychiatric disorders. J Theor Biol. 2018;452:81–84. doi:10.1016/j.jtbi.2018.05.01429775682
  • Feinberg AP. Epigenetics at the epicenter of modern medicine. JAMA. 2008;299:1345–1350. doi:10.1001/jama.299.11.134518349095
  • Lee R, Avramopoulos D. Introduction to epigenetics in psychiatry In: Peedicayil J, Grayson DR, Avramopoulos D, editors. Epigenetics in Psychiatry. Waltham, MA, USA: Elsevier; 2014:3–25.
  • Marks IM. Genetics of fear and anxiety disorders. Br J Psychiatry. 1986;149:406–418. doi:10.1192/bjp.149.4.4063545353
  • Hettema JM, Neale MC, Kendler KS. A review and meta-analysis of the genetic epidemiology of anxiety disorders. Am J Psychiatry. 2001;158:1568–1578. doi:10.1176/appi.ajp.158.10.156811578982
  • Bartlett AA, Singh R, Hunter RG. Anxiety and epigenetics. Adv Exp Biol Med. 2017;978:145–166.
  • Simmons RK, Howard JL, Simpson DN, Akil H, Clinton SM. DNA methylation in the developing hippocampus and amygdala of anxiety-prone versus risk- taking rats. Dev Neurosci. 2012;34:58–67. doi:10.1159/00033664122572572
  • Zhou R, Chen F, Chang F, Bai Y, Chen L. Persistent overexpression of DNA methyltransferase 1 attenuating GABAergic inhibition in basolateral amygdala accounts for anxiety in rat offspring exposed perinatally to low- dose bisphenol A. J Psychiatr Res. 2013;47:1535–1544. doi:10.1016/j.jpsychires.2013.05.01323791455
  • Zhu C, Liang M, Li Y, Feng X, Hong J, Zhou R. Involvement of epigenetic modifications of GABAergic interneurons in basolateral amygdala in anxiety-like phenotype of prenatally stressed mice. Int J Neuropsychopharmacol. 2018;21:570–581. doi:10.1093/ijnp/pyy00629471396
  • Kosten TA, Huang W, Nielsen DA. Sex and litter effects on anxiety and DNA methylation levels of stress and neurotrophin genes in adolescent rats. Dev Psychobiol. 2014;56:392–406. doi:10.1002/dev.v56.323460384
  • Mccoy CR, Glover ME, Flynn LT, et al. Altered DNA methylation in the developing brains of rats genetically prone to high versus low anxiety. J Neurosci. 2019;39:3144–3158. doi:10.1523/JNEUROSCI.1157-15.201930683683
  • Sagarkar S, Bhamburkar T, Shelkar G, Choudhary A, Kokare DM, Sakharkar AJ. Minimal traumatic brain injury causes persistent changes in DNA methylation at BDNF gene promoters in rat amygdala: a possible role in anxiety-like behaviors. Neurobiol Dis. 2017;106:101–109. doi:10.1016/j.nbd.2017.06.01628663119
  • Moonat S, Sakharkar AJ, Zhang H, Tang L, Pandey SC. Aberrant HDAC2-mediated histone modifications and synaptic plasticity in the amygdala predisposes to anxiety and alcoholism. Biol Psychiatry. 2013;73:763–773. doi:10.1016/j.biopsych.2013.01.01223485013
  • Suri D, Bhattacharya A, Vaidya VA. Early stress evokes temporally distinct consequences on the hippocampal transcriptome, anxiety and cognitive behaviour. Int J Neuropsychopharmacol. 2014;17:289–301. doi:10.1017/S146114571300100424025219
  • Patki G, Solanki N, Atrooz F, et al. Novel mechanistic insights into treadmill exercise based rescue of social defeat-induced anxiety-like behavior and memory impairment in rats. Physiol Behav. 2014;130:135–144. doi:10.1016/j.physbeh.2014.04.01124732411
  • Kumari A, Singh P, Baghel MS, Thakur MK. Social isolation mediated anxiety like behavior is associated with enhanced expression and regulation of BDNF in the female mouse brain. Phyisiol Behav. 2016;158:34–42. doi:10.1016/j.physbeh.2016.02.032
  • Huang S, Dong W, Jiao Z, et al. Prenatal dexamethasone exposure induced alterations in neurobehavior and hippocampal glutamatergic system balance in female rat offspring. Toxicol Sci. 2019;171:369–384. doi:10.1093/toxsci/kfz163
  • Viana Borges J, Souza de Freitas B, Antoniazzi V, et al. Social isolation and social support at adulthood affect epigenetic mechanisms, brain-derived neurotrophic factor levels and behavior of chronically stressed rats. Behav Brain Res. 2019;366:36–44. doi:10.1016/j.bbr.2019.03.02530880220
  • Singh S, Siddiqui SA, Tripathy S, et al. Decreased level of histone acetylation in the infralimbic prefrontal cortex following immediate extinction may result in deficit of extinction memory. Brain Res Bull. 2018;140:355–364. doi:10.1016/j.brainresbull.2018.06.00429908895
  • Wang DY, Kosowan J, Samsom J, et al. Inhibition of the G9a/GLP histone methyltransferase complex modulates anxiety-related behavior in mice. Acta Pharmacol Sin. 2018;39:866–874. doi:10.1038/aps.2017.19029417943
  • Cittaro D, Lampis V, Luchetti A, et al. Histone modifications in a mouse model of early life adversities and panic disorder: role for Asic1 and neurodevelopmental genes. Sci Rep. 2016;6:25131. doi:10.1038/srep2513127121911
  • Aten S, Page CE, Kalidindi A, et al. miR-132/212 is induced by stress and its dysregulation triggers anxiety-related behavior. Neuropharmacology. 2019;144:256–270. doi:10.1016/j.neuropharm.2018.10.02030342060
  • Cohen JL, Jackson NL, Ballestas ME, Webb WM, Lubin FD, Clinton SM. Amygdalar expression of the microRNA miR-101a and its target Ezh2 contribute to rodent anxiety-like behaviour. Eur J Neurosci. 2017;46:2241–2252. doi:10.1111/ejn.2017.46.issue-728612962
  • Liu Y, Liu D, Xu J, Jiang H, Pan F. Early adolescent stress-induced changes in prefrontal cortex miRNA-135a and hippocampal miRNA-16 in male rats. Dev Psychobiol. 2017;59:958–969. doi:10.1002/dev.v59.828944448
  • Hoban AE, Stilling RM, Moloney GM, et al. Microbial regulation of microRNA expression in the amygdala and prefrontal cortex. Microbiome. 2017;5:102. doi:10.1186/s40168-017-0321-328838324
  • Spadaro PA, Flavell CR, Widagdo J, et al. Long noncoding RNA-directed epigenetic regulation of gene expression is associated with anxiety-like behavior in mice. Biol Psychiatry. 2015;78:848–859. doi:10.1016/j.biopsych.2015.02.00425792222
  • Lydiard RB. The role of GABA in anxiety disorders. J Clin Psychiatry. 2003;64 Suppl:21–27.12662130
  • Goddard AW, Mason GF, Almai A, et al. Reductions in occipital cortex GABA levels in panic disorder detected with 1H-magnetic resonance spectroscopy. Arch Gen Psychiatry. 2001;58:556–561. doi:10.1001/archpsyc.58.6.55611386984
  • He Y, Ouyang J, Hu Z, et al. Intervention mechanism of repeated oral GABA administration on anxiety-like behaviors induced by emotional stress in rats. Psychiatry Res. 2019;271:649–657. doi:10.1016/j.psychres.2018.12.02530791338
  • Alisch RS, Van Hulle C, Chopra P, et al. A multi-dimensional characterization of anxiety in monozygotic twin pairs reveals susceptibility loci in humans. Transl Psychiatry. 2017;7:1282. doi:10.1038/s41398-017-0047-929225348
  • Domschke K, Tidow N, Schrempf M, et al. Epigenetic signature of panic disorder: a role of glutamate decarboxylase 1(GAD1) DNA hypomethylation?. Prog Neuropsychopharmacol Biol Psychiatry. 2013;46:189–196. doi:10.1016/j.pnpbp.2013.07.01423906988
  • Roberts S, Lester KJ, Hudson JL, et al. Serotonin transporter methylation and response to cognitive behaviour therapy in children with anxiety disorders. Transl Psychiatry. 2014;4:e444. doi:10.1038/tp.2014.8325226553
  • Ziegler C, Richter J, Mahr M, et al. MAOA gene hypomethylation in panic disorder- reversibility of an epigenetic risk pattern by psychotherapy. Transl Psychiatry. 2016;6:e773. doi:10.1038/tp.2016.4127045843
  • Shimada-Sugimoto M, Otowa T, Miyagawa T, et al. Epigenome-wide association study of DNA methylation in panic disorder. Clin Epigenetics. 2017;9:6. doi:10.1186/s13148-016-0307-128149334
  • Melas PA, Forsell Y. Hypomethylation of MAOA’s first exon region in depression: a replication study. Psychiatry Res. 2015;226:389–391. doi:10.1016/j.psychres.2015.01.00325623016
  • Ziegler C, Dannlowski U, Braüer D, et al. Oxytocin receptor gene methylation: converging multilevel evidence for a role in social anxiety. Neuropsychopharmacology. 2015;40:1528–1538. doi:10.1038/npp.2015.225563749
  • Sanwald S, Gahr M, Widenhorn-Müller K, et al. Relation of promoter methylation of the oxytocin gene to stressful life events and depression severity. J Mol Neurosci.2020; 70:201–211. doi:10.1007/s12031-019-01446-1
  • Murphy TM, O’Donovan A, Mullins N, O’Farrelly C, McCann A, Malone K. Anxiety is associated with higher levels of global DNA methylation and altered expression of epigenetic and interleukin-6 genes. Psychiatr Genet. 2015;25:71–78. doi:10.1097/YPG.000000000000005525350786
  • Tyrka AR, Parade SH, Welch ES, et al. Methylation of the leukocyte glucocorticoid receptor gene promoter in adults: associations with early adversity and depressive, anxiety and substance-use disorders. Transl Psychiatry. 2016;6:e848. doi:10.1038/tp.2016.11227378548
  • Prelog M, Hilligardt D, Schmidt CA, et al. Hypermethylation of FOXP3 promoter and premature aging of the immune system in female patients with panic disorder?. PLoS One. 2016;11:e0157930. doi:10.1371/journal.pone.015793027362416
  • Iurato S, Carrillo-Roa T, Arloth J, et al. DNA methylation signatures in panic disorder. Transl Psychiatry. 2017;7:1287. doi:10.1038/s41398-017-0026-129249830
  • Bortoluzzi A, Salum GA, da Rosa ED, Chagas VD, Castro MAA, Manfro GG. DNA methylation in adolescents with anxiety disorder: a longitudinal study. Sci Rep. 2018;8:13800.30218003
  • Ciuculete DM, Boström AE, Tuunainen A-K, et al. Changes in methylation within the STK32B promoter are associated with an increased risk for generalized anxiety disorder in adolescents. J Psychiatr Res. 2018;102:44–51. doi:10.1016/j.jpsychires.2018.03.00829604450
  • Kang H-J, Kim K-O, Kim J-W, et al. A longitudinal study of the associations of BDNF genotype and methylation with poststroke anxiety. Int J Geriatr Psychiatry. 2019;34:1706–1715. doi:10.1002/gps.v34.1131368178
  • Palma-Gudiel H, Cordova-Palomera A, Tornador C, et al. Increased methylation at an unexplored glucocorticoid responsive element within exon 1D of NR3C1 gene is related to anxious-depressive disorders and decreased hippocampal connectivity. Eur Neuropsychopharmacol. 2018;28:579–588. doi:10.1016/j.euroneuro.2018.03.01529650294
  • Bosmans G, Young JF, Hankin BL. NR3C1 methylation as a moderator of the effects of maternal support and stress on insecure attachment development. Dev Psychol. 2018;54:29–38. doi:10.1037/dev000042229058930
  • Roberts S, Keers R, Breen G, et al. DNA methylation of FKPB5 and response to exposure-based psychological therapy. Am J Med Genet B. 2019;180:150–158. doi:10.1002/ajmg.b.32650
  • Chagnon YC, Potvin O, Hudon C, Préville M. DNA methylation and single nucleotide variants in the brain-derived neurotrophic factor (BDNF) and oxytocin receptor (OXTR) genes are associated with anxiety/depression in older women. Front Genet. 2015;30(6):230.
  • Hommers LG, Richter J, Yang Y, et al. A functional genetic variation of SLC6A2 repressor has-miR-579-3p upregulates sympathetic noradrenergic processes of fear and anxiety. Transl Psychiatry. 2018;8:226. doi:10.1038/s41398-018-0278-430341278
  • Chen SD, Sun XY, Niu W, et al. Correlation between the level of microRNA expression in peripheral blood mononuclear cells and symptomatology in patients with generalized anxiety disorder. Compr Psychiatry. 2016;69:216–224. doi:10.1016/j.comppsych.2016.05.00627423364
  • Dickson DA, Paulus JK, Mensah V, et al. Reduced levels of miRNAs 449 and 34 in sperm of mice and men exposed to early life stress. Transl Psychiatry. 2018;8:101. doi:10.1038/s41398-018-0146-229795112
  • Jacobsen DP, Eriksen MB, Rajalingam D, et al. Exposure to workplace bullying, microRNAs and pain; Evidence of a moderating effect of miR-30c rs928508 and miR-223 rs3848900. Stress. 2020;23:77–86. doi:10.1080/10253890.2019.164232031339402
  • Hettema JM, Otowa T. Genetics of anxiety disorders In: Sadock BJ, Sadock VA, Ruiz P, editors. Kaplan & Sadock’s Comprehensive Textbook of Psychiatry. Philadelphia, PA: Wolters Kluwer; 2017:1737–1741.
  • Bartlett AA, Lapp HE, Hunter RG. Epigenetic mechanisms of the glucocorticoid receptor. Trends Endocrinol Metab. 2019;30:807–818. doi:10.1016/j.tem.2019.07.00331699238
  • Champagne FA. Beyond the maternal epigenetic legacy. Nat Neurosci. 2018;21:773–774. doi:10.1038/s41593-018-0157-629786084
  • Lee JB, Wei J, Liu W, Cheng J, Feng J, Yan Z. Histone deacetylase 6 gates the synaptic action of acute stress in prefrontal cortex. J Physiol. 2012;590:1535–1546. doi:10.1113/jphysiol.2011.22490722331421
  • Espallergues J, Teegarden SL, Veerakumar A, et al. HDAC6 regulates glucocorticoid receptor signaling in serotonin pathways with critical impact on stress resilience. J Neurosci. 2012;32:4400–4416. doi:10.1523/JNEUROSCI.5634-11.201222457490
  • Roberts S, Keers R, Lester KJ, et al. HPA axis related genes and response to psychological therapies: genetics and epigenetics. Depress Anxiety. 2015;32:861–870. doi:10.1002/da.2243026647360
  • Hempstead BL. Brain-derived neurotrophic factor: three ligands, many actions. Trans Am Clin Climatol Assoc. 2015;126:9–19.26330656
  • Suliman S, Hemmings SM, Seedat S. Brain-derived neurotrophic factor (BDNF) protein levels in anxiety disorders: systematic review and meta-regression analysis. Front Integr Neurosci. 2013;7:55. doi:10.3389/fnint.2013.0005523908608
  • Varela MA, Roberts TC, Wood MJA. Epigenetics and ncRNAs in brain function and disease: mechanisms and prospects for therapy. Neurotherapeutics. 2013;10:621–631. doi:10.1007/s13311-013-0212-724068583
  • Murphy CP, Singewald N. Role of microRNAs in anxiety and anxiety-related disorders. Curr Top Behav Neurosci. 2019;42:185–220.31485988
  • Hadjiargyrao M, Delihas N. The intertwining of transposable elements and non-coding RNAs. Int J Mol Sci. 2013;14:13307–13328. doi:10.3390/ijms14071330723803660
  • Guffanti G, Bartlett A, DeCrescenzo P, Macciardi F, Hunter R. Transposable elements. Curr Top Behav Neurosci. 2019;42:221–246.31605305
  • Hunter RG, McEwen BS. Stress and anxiety across the lifespan: structural plasticity and epigenetic regulation. Epigenomics. 2013;5:177–194. doi:10.2217/epi.13.823566095
  • Whittle N, Schmuckermair C, Gunduz Cinar O, et al. Deep brain stimulation, histone deacetylase inhibitors and glutamatergic drugs rescue resistance to fear extinction in a genetic mouse model. Neuropharmacology. 2013;64:414–423. doi:10.1016/j.neuropharm.2012.06.00122722028
  • Sakharkar AJ, Zhang H, Tang L, et al. Effects of histone deacetylase inhibitors on amygdaloid histone acetylation and neuropeptide Y expression: a role in anxiety-like and alcohol-drinking behaviours. Int J Neuropsychopharmacol. 2014;17:1207–1220. doi:10.1017/S146114571400005424528596
  • You C, Zhang H, Sakharkar AJ, Teppen T, Pandey SC. Reversal of deficits in dendritic spines, BDNF and Arc expression in the amygdala during alcohol dependence by HDAC inhibitor treatment. Int J Neuropsychopharmacol. 2014;17:313–322. doi:10.1017/S146114571300114424103311
  • Tran L, Schulkin J, Ligon CO, Greenwood-van Meerveld B. Epigenetic modulation of chronic anxiety and pain by histone deacetylation. Mol Psychiatry. 2015;20:1219–1231. doi:10.1038/mp.2014.122
  • Moloney RD, Stilling RM, Dinan TG, Cryan JF. Early-life stress-induced visceral hypersensitivity and anxiety behavior is reversed by histone deacetylase inhibition. Neurogastroenterol Motil. 2015;27:1831–1836. doi:10.1111/nmo.1267526403543
  • Sah A, Sotnikov S, Kharitonova M, et al. Epigenetic mechanisms within the cingulate cortex regulate innate anxiety-like behavior. Int J Neuropsychopharmacol. 2019;22:317–328. doi:10.1093/ijnp/pyz00430668714
  • Montagud-Romero S, Cantacorps L, Valverde O. Histone deacetylases inhibitor trichostatin A reverses anxiety-like symptoms and memory impairments induced by maternal binge alcohol drinking in mice. J Psychopharmacol. 2019;33:1573–1587. doi:10.1177/026988111985720831294671
  • Hayase T. Putative epigenetic involvement of the endocannabinoid system in anxiety-and depression-related behaviors caused by nicotine as a stressor. PLoS One. 2016;11:e0158950. doi:10.1371/journal.pone.015895027404492
  • Kv A, Madhana RM, Js IC, Lahkar M, Sinha S, Naidu VGM. Antidepressant activity of vorinostat is associated with amelioration of oxidative stress and inflammation in a corticosterone-induced chronic stress model in mice. Behav Brain Res. 2018;344:73–84. doi:10.1016/j.bbr.2018.02.00929452193
  • Karnib N, El-Ghandour R, El Hayek L, et al. Lactate is an antidepressant that mediates resilience to stress by modulating the hippocampal levels and activity of histone deacetylases. Neuropsychopharmacology. 2019;44:1152–1162. doi:10.1038/s41386-019-0313-z30647450
  • Park J, Terranova-Barberio M, Zhong AY, Thomas S, Munster PN. Clinical applications of histone deacetylase inhibitors In: Tollefsbol TO, editor. Handbook of Epigenetics: The New Molecular and Medical Genetics. Cambridge, MA: Elsevier; 2017:605–621.
  • Primeau F, Fontaine R, Beauclair L. Valproic acid and panic disorder. Can J Psychiatry. 1990;35:248–250. doi:10.1177/0706743790035003092111204
  • Keck PE, Taylor VE, Tugrul KC, McElroy SL, Bennett JA. Valproate treatment of panic disorder and lactate-induced panic attacks. Biol Psychiatry. 1993;33:542–546. doi:10.1016/0006-3223(93)90010-B8513040
  • Woodman CL, Noyes R. Panic disorder: treatment with valproate. J Clin Psychiatry. 1994;55:134–136.8071256
  • Kinrys G, Pollack MH, Simon NM, Worthington JJ, Nardi AE, Versiani M. Valproic acid for the treatment of social anxiety disorder. Int Clin Psychopharmacol. 2003;18:169–172. doi:10.1097/01.yic.0000064261.66765.9f12702897
  • Aliyev NA, Aliyev ZN. Valproate (depakine-chrono) in the acute treatment of outpatients with generalized anxiety disorder without psychiatric comorbidity: randomized, double-blind placebo-controlled study. Eur Psychiatry. 2008;23:109–114. doi:10.1016/j.eurpsy.2007.08.00117945470
  • Bach DR, Korn CW, Vunder J, Bantel A. Effect of valproate and pregabalin on human anxiety-like behaviour in a randomized controlled trial. Transl Psychiatry. 2018;8:157. doi:10.1038/s41398-018-0206-730115911
  • Scott KA, Hoban AE, Clarke G, Moloney GM, Dinan TG, Cryan JF. Thinking small: towards microRNA-based therapeutics for anxiety disorders. Expert Opin Investig Drugs. 2015;24:529–542. doi:10.1517/13543784.2014.997873
  • Roy-Byrne P. Treatment-refractory anxiety; Definition, risk factors, and treatment challenges. Dialogues Clin Neurosci. 2015;17:191–206.26246793
  • Seto E, Yoshida M. Erasers of histone acetylation: the histone deacetylase enzymes In: Allis CD, Caparros M-L, Jenuwein T, Reinberg D, Lachner M, editors. Epigenetics. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press; 2015:143–168.
  • Choudhary C, Kumar C, Gnad F, et al. Lysine acetylation targets protein complexes and co-regulates major cellular functions. Science. 2009;325:834–840. doi:10.1126/science.117537119608861
  • Dokmanovic M, Marks PA. Prospects: histone deacetylase inhibitors. J Cell Biochem. 2005;96:293–304. doi:10.1002/(ISSN)1097-464416088937
  • Li W, Sun Z. Mechanism of action for HDAC inhibitors – insights from omics approaches. Int J Mol Sci. 2019;20(7):1616. doi:10.3390/ijms20071616
  • Guidotti A, Grayson DR. DNA methylation and demethylation as targets for antipsychotic therapy. Dialogues Clin Neurosci. 2014;16:419–429.25364290
  • Wei Y, Melas PA, Wegener G, Mathé AA, Lavebratt C. Antidepressant-like effect of sodium butyrate is associated with an increase in TET1 and in 5-hydroxymethylation levels in the BDNF gene. Int J Neuropsychopharmacol. 2014;18(2).
  • Peedicayil J, Kumar A. Epigenetic drugs for mood disorders. Prog Mol Biol Transl Sci. 2018;157:151–174.29933949
  • Whittle N, Singewald N. HDAC inhibitors as cognitive enhancers in fear, anxiety and trauma therapy: where do we stand?. Biochem Soc Trans. 2014;42:569–581. doi:10.1042/BST2013023324646280