Disease Markers 30 (2011) 77–87 77 DOI 10.3233/DMA-2011-0749 IOS Press

Epigenetic aspects of posttraumatic stress disorder

Ulrike Schmidt, Florian Holsboer∗ and Theo Rein Max Planck Institute of Psychiatry, Munich, Germany and Neuro Novag GmbH, Munich, Germany

Abstract. Development of psychiatric such as posttraumatic stress disorder (PTSD) invokes, as with most complex diseases, both genetic and environmental factors. The era of genome-wide high throughput technologies has sparked the initiation of genotype screenings in large cohorts of diseased and control individuals, but had limited success in identification of causing genetic variants. It has become evident that these efforts at the genomic level need to be complemented with endeavours in elucidating the proteome, transcriptome and epigenetic profiles. is attractive in particular because there is accumulating evidence that the lasting impact of adverse life events is reflected in certain covalent modifications of the chromatin. In this review, we outline the characteristics of PTSD as a stress-related disease and survey recent developments revealing epigenetic aspects of stress-related disorders in general. There is also increasing direct evidence for gene programming and epigenetic components in PTSD. Finally, we discuss treatment options in the light of recent discoveries of epigenetic mechanisms of psychotropic drugs.

1. Introduction that psychiatric disorders can be traced back to one sin- gle epigenetic modification, inasmuch as many differ- The term epigenome refers to the entirety of all ent genetic variants are conveying disease susceptibili- molecular control elements programming the genome ty. These diseases present with highly variable clinical respectively regulating gene activities. Like any molec- signs and symptoms but these diverse features cannot ular action,also epigenetic programming can go wrong. be linked to specific genetic and epigenetic variations. Accordingly, distinct epigenetic defects are reported In fact it is likely that they derive from a complex web for a variety of diseases and some of them are already of connected but different causes. known to constitute a main pathogenetic mechanism, Only a small proportion of individuals develops post- e.g. in patients suffering from Rett syndrome. traumatic PTSD after having been exposed to a trau- One of the major environmental factors established matic event [13]. PTSD only occurs if a (yet still un- as inducer of epigenetic changes is stress. Stress is known) biological predisposition coincides with a trau- known to contribute to the pathogenesis of a variety of matic stressor. This instantly forces the supposition disorders, including the majority of psychiatric disor- that the epigenome, especially in regard to its capacity ders like major depression and posttraumatic stress dis- to mediate communication between environment and order (PTSD). There is robust evidence for epigenetic genome, might grossly contribute to PTSD pathogene- contribution to the development of PTSD-like symp- sis. toms in rodents while so far there are only few studies In this review the most important molecular epige- demonstrating the connection between trauma-induced netic programming mechanisms are illustrated to allow epigenetic changes and the onset and perpetuation of for a better understanding of the following paragraphs PTSD symptoms in humans. Generally, it is unlikely where the dynamic principle of the epigenome includ- ing its reagibility to environmental factors and final- ∗Corresponding author: Dr. Florian Holsboer, E-mail: holsboer@ ly the current state of knowledge regarding epigenet- mpipsykl.mpg.de. ic modifications in stress-related diseases, in particular

ISSN 0278-0240/11/$27.50  2011 – IOS Press and the authors. All rights reserved 78 U. Schmidt et al. / Epigenetic aspects of posttraumatic stress disorder

PTSD-animalmodels and PTSD patients, are discussed which in turn elicits the release of glucocorticoidsfrom on the basis of the most recent publications. the adrenalcortex. Then, cortisol as the final peripheral molecule exerts its actions on metabolism, immunity and brain functions. Increased and prolonged produc- 2. PTSD as stress-related disease tion of CRH and cortisol explain many of the behav- ioral, circulatory, metabolic and immune manifesta- The term stress was first employed in a biological tionsof syndromesassociated with acuteand especially contextby the endocrinologistHans Selye in the 1930s. with chronic stress, such as PTSD. He later broadened and popularized his concept to in- Transient HPA-system shifts were found in a variety clude inappropriate physiological response to any de- of psychiatric diseases, e.g. it is an established find- mand. In his usage “stress” refers to the condition ing that many patients suffering from major depression and “stressor” to the stimulus triggering the unphysio- (MD) display a hyperactivity of their stress hormone logical response. It covers a wide range of phenome- system [40]. In contrast to patients with MD, many au- na, from mild irritation to drastic dysfunction that may thorsfoundan HPA-axis hypoactivity in PTSD-patients cause severe health breakdown mostly occurring in re- reflected for instance by lowered serum cortisol lev- sponse to maximal stressors. els [11,107] and an enhanced suppression of cortisol These maximal stressors are also termed traumata; in the DEX/CRH-test [101,107]. However, a recent the term trauma derives from the greek word τραµα meta-analysis of 37 studies [57] found no differences meaning wound. A traumatic event involves a single, in plasma cortisol levels between adult individuals with enduring or repeating experience that seriously threat- PTSD and controls. But interestingly, sub-group anal- ens the physical or psychic integrity of the concerned yses revealed some remarkable results: both the patient person. According to DSM-IV also witnessing a seri- cluster havingexperiencedphysicalor sexualabuse and ous threat or the death of another person can be clas- the female patient cluster exhibited significantly lower sified as a traumatic event, but in any case a horrible cortisol levels in comparison to their respective con- experience can be categorized as a traumatic incident trol subgroups. Despite the limitation of differences only if the individuals exposed in the course of the in data collection and assessment, that meta-analysis event experienced feelings of helplessness and massive clearly reveals the complexity and heterogeneity of the horror. The disability to cope or integrate the ideas neuroendocrine concomitants of PTSD [71]. and emotions involved with that experience leads to the Besides the HPA-axis, also the sympathetic nervous sense of being overwhelmed. Symptoms usually arise weeks or months, in rare system (SNS) is a key circuit in PTSD pathophysiolo- cases even decades after traumatic experiences. More- gy. Its major neurotransmitter norepinephrine (NE) is over, diagnosing PTSD includes the assumption that an importantplayer in the central as well as in the auto- without a traumatic event this psychiatric disease would nomicstressresponse. NEisproducedinneuronsofthe not have been appeared. Thus, the stressor precipitat- locus coeruleus and interacts in concert with CRH and ed the onset of PTSD which is characterized mainly vasopressin to increase fear conditioning and memory by recurrent and intrusive distressing recollections of encoding. In theperiphery,stress leadsto releaseof NE the event (e.g. flashbacks), nervous hyperarousal and and epinephrine from the adrenal medulla, resulting in trauma-stimuli associated avoidance anxiety each last- an organismic alarm reaction. Thus, NE is involved in ing more than one month, thus causing significant im- the development of two main clusters of PTSD symp- pairment in social, occupational, or other important ar- toms, namely re-experiencing symptoms and hyper- eas of functioning. arousal which is expressed inter alia with an exaggerat- Like other stress-related psychiatric diseases PTSD ed startle responseand increased heart rate [85]. Hence comesalong with a variety of neuroendocrinologicalal- it is not surprising that the most consistent findings in terations. The hypothalamus-pituitary-adrenal (HPA) PTSD neuroendocrinestudies are increased central and axis has been the main focus of neuroendocrine re- peripheral noradrenergic activities [32,104]. search in PTSD [104]. Taken together, PTSD is a stress-related disease not Perception of stress leads to synthesis and release only because PTSD symptoms are precipitated by a of corticotropin-releasing hormone (CRH) and vaso- traumatic stressor but also because this severe psychi- pressin by hypothalamic nuclei which stimulate the se- atric condition is accompanied by drastic alterations in cretion of adrenocorticotropin (ACTH) at the pituitary the two major stress hormone systems. U. Schmidt et al. / Epigenetic aspects of posttraumatic stress disorder 79

3. The epigenome lated DNA is recognised and bound by a family of methylated DNA binding proteins of which methyl- The term epigenetics has been coined even long be- CpG-binding protein2 (MeCP2) has attracted particu- fore the structure of genes has been discovered [36, lar attention. MeCP2 recruits the histone deacetylase 95]. In today’s biomedical research, epigenetics ex- (HDAC) complex mSin3A, thereby attracting HDAC-1 plores the regulatory functions of covalent chromatin and HDAC-2, thus ultimately abolishing histone acety- modificationsas well as of microRNAs in gene activity. lation and inactivating chromatin [44]. DNA methyla- These chromatin modifications do not alter the genet- tion can, in addition, impede DNA bindingof transcrip- ic information per se, but govern access to it and can tion factors [89,93]. However, DNA methylation could be maintained and stably inherited through mitosis and also be associated with increased gene transcription if meiosis. The modifications comprise in mammals pre- it interferes with DNA binding of a repressor. dominantly the post-replicative attachment of a methyl A wealth of enzymes executing the addition and re- groupatcarbonposition 5 of the nucleobasecytosine as moval of covalent chromatin modifications has been well as acetylation, methylation and phosphorylation found, albeit the exact mechanism of active cytosine of histone proteinsat defined aminoacid positions[45]. demethylation has not been elucidated yet. As key ex- More rare modificationsare sumoylation [81], ubiquiti- ecutors of diverse regulatory signals these enzymes are nation [82], ADP-ribosylation [37], biotinylation [16], target of intense drug development efforts. carbonylation [100], deimination [23], proline isomer- ization [66], and glycosylation [43] of histones. Inextensionofamodelthatwasbasedonthelinkbe- 4. Epigenetic aspects of stress-related psychiatric tween histone acetylation and gene transcription [92], diseases Strahl and Allis proposed a general “histone code” model for the combinatorial function of specific post- While epigenetic mechanisms have been under in- translational histone modifications in gene transcrip- vestigation in cancer and developmental research for tion [87]. According to this concept, the combinatorial decades [10], they recently attracted intensified atten- arrangement of distinct histone modifications forms a tion by neuroscientists as a potential mediator of gene code that is read and interpreted by nonhistone chro- environment interactions. Environmental insults im- matin associating proteins that signal the encoded in- pacting on the epigenome may represent the molecu- formation to downstream functions such as gene tran- lar programming of stable long-term effects on gene scription. Since a nucleosome comprises several his- expression in the brain, and thus on cerebral function. tones, this combination results also in a “nucleosome Mostof the experimentalevidencefor the role of epi- code”, allowing a plethora of epigenetic states. genetic mechanisms in stress-related psychiatric dis- The histone modification status, in particular his- eases originates from animal models. Psychiatric dis- tone acetylation, is intimately linked to the degree of eases pose a particular challenge for researchersaiming DNA methylation [64]. DNA can be methylated in at modelling diseases in animals [46,60]. Nevertheless, mammals at cytosines residing in CpG dinucleotides. several models have been established, based on the re- 60–80% of these dinucleotides are methylated in hu- alisation of stress as central risk factor for the develop- mans [75]. In general, CpG dinucleotides are grossly ment of psychiatric diseases. Common to the diverse underrepresented in the genome. This has been ex- animal models is the exposure of the animal to stress plained by the impossibility for the cellular repair ma- during a sensitive period in life, i.e. in the prenatal, chinery to specifically correct a deaminated methylated postnatal and adolescent phase. This is in congruence cytosine, which leads to the legitimate DNA nucleo- with the observation in humans that adults who suf- side thymine, while deamination of unmethylated cy- fered from severe early life stress such as parental loss tosine produces uracile, which can easily be recognised or neglect presented with higher basal levels of stress as a non-DNA nucleoside. CpGs are enriched in so hormone and were at greater risk of developing mood called “CpG-islands” [30], which are usually found in disorders [65]. the 5’ regulatory regions of most genes. DNA methy- Already more than half a century ago, disturbances lation usually leads to gene silencing through effects of the mother-neonate interaction in rodents have been on chromatin structure. This can involve more gener- demonstrated to alter adult phenotypes that include al mechanisms leading to hypoacetylation of histones neuroendocrine function, behaviour, metabolism and and formation of condensed chromatin [8,9]. Methy- cognition, e.g. [49]. For example, rats experiencing 80 U. Schmidt et al. / Epigenetic aspects of posttraumatic stress disorder maternal separation in the first weeks of life develop patients with depression, Moreover, individuals hav- increased anxiety and cognitive deficits later in life [72, ing experienced early life trauma have elevated corti- 73]. Programming of HPA axis (re)activity has been sol [38] secretion following the Dex/CRH test, indirect- extensively investigated as a critical juncture between ly reflecting elevation of hypothalamic vasopressin. In early life environmentand health in the adult [24,29,67, fact in mice where HPA hyperactivity and depression- 86]. These alterations of HPA axis settings frequently and anxiety-like behaviour was permanently induced go along with changes in GR expression [28,50,96], by early life trauma, a vasopressin receptor antagonist whose activation is pivotal for balancing HPA activity modulated neuroendocrine and behavioural sequelae through negative feed-back loops at the level of CRH, from stress-exposure. vasopressin, and ACTH [24]. Importantly, blunting Another common way to model the pathogenesis of the early life experience-induced differences in hip- depression-like states is chronic stress exposure of ro- pocampal GR expression blocks the effects on HPA dents [46]. Social defeat of an experimental mouse by activity in adulthood [55,56]. Moreover, loss of GR serial confrontation with aggressive mice over 10 days expression in the hippocampus leads to dysfunctional induces depression-like syndromes such as anxiety- HPA axis regulation and enhanced anxiety-related be- related symptoms, anhedonia and loss of interest in so- haviour [12], further corroborating the notion of pro- cial interaction. The aptitude of this model for investi- gramming hippocampal GR levels as the underlying gating depression-like states is further substantiated by mechanism translating early life experience into dif- the observations that antidepressants can reverse most ferential HPA reactivity and differences in the HPA- of the stress-induced symptoms [7,91]. Brain derived associated behavioural spectrum. neurotrophic factor (BDNF) was found to be stably Theearly life-induced programming of GRlevelshas downregulated after chronic social defeat along with been directly linked to epigenetic alterations of the GR methylation of lysine 27 of histone H3 (H3K27) [91], gene. More specifically, in animals experiencing dif- which constitutes a repressive epigenetic mark. Chron- ferent maternal care, disparate DNA methylation pat- ic treatment with antidepressants restored bdnf RNA terns and chromatin acetylation levels (acetylation at expressiontogetherwith reversalof the behaviouralim- lysine 9 of histone 3) were identified in the exon 17 pairments. However,H3K27 methylation remained un- promoter of GR [96], which is the relevant regulato- changed, which was suggested to represent a “molecu- ry region determining GR expression in the hippocam- lar scar” that may contribute to the risk of reappearance pus [52]. The change in DNA methylation was linked of symptoms in depressed patients after discontinuing to impaired binding of the transcription factor nerve antidepressant treatment [76]. growth factor-inducible protein A, whose response ele- Even though in humans obvious limitations prevent ment harbored differentially methylated cytosines [96]. insight to the same depth as in animal studies, sig- This example provides intriguing molecular insight in- nificant evidence has accumulated for the importance to the epigenetic mechanisms underlying experience- of epigenetic mechanisms in psychiatric diseases. For induced programming of neuroendocrine circuits and example, a recent study examining post mortem brain behaviour. tissue from suicide subjects with a history of early In a more recent study, early life stress was found childhood neglect and/or abuse revealed a significant to cause a persistent increase of vasopressin, accompa- promoter-wide hypermethylation of rRNA genes [53]. nied by sustained DNA hypomethylation of a relevant In schizophrenia, alterations in GABAergic mRNA regulatory region [62]. This hypomethylation affected expression have been identified as a key feature. Ex- CpG dinucleotides that are recognised by MeCP2. In- pression of glutamate decarboxylate 1 (GAD1) has triguingly, the ability of MeCP2 to bind to these sites is been found to be decreased in schizophrenia, in con- controlledbyneuronalactivity. Thus, in this modelear- junction with a risk haplotype, along with reduced lev- ly life stress inducesepigenetic markingof an HPA reg- elsof (tri)methylationat lysine4 ofhistone H3 (H3K4), ulatory gene via neuronal activity-directed regulation astimulatory epigeneticmark [42]. Moreover,theatyp- of protein DNA interaction of MeCP2 as the molecular ical antipsychotic clozapine increased H3K4 methyla- underpinnings of neuroendocrine and behavioral alter- tion at GAD1 and promoteroccupancywith the respec- ations that are frequent features in stress-related dis- tive methyltransferase [42]. In another study, DNA hy- eases. This finding is remarkable insofar as increased permethylation of the reelin gene promoter has been vasopressin secretion from the hypothalamic nuclei is observed in post mortem brains from schizophrenic known to induce anxiety in rodents and is elevated in patients [1]. This could explain the significantly re- U. Schmidt et al. / Epigenetic aspects of posttraumatic stress disorder 81 duced expression of this important neuronal protein in In contrast to these findings in suicide victims, dif- schizophrenia [19,27]. Although reelin promoter hy- ferential methylation of GR was not found in a very re- permethylation was not found in another study [90], centstudy focussingon majordepression [3]. Levels of the role of DNA methylation at this promoter is cor- total GR were found to be unchanged in samples orig- roborated by studies reporting its demethylation by the inating from individuals who had suffered from major antipsychotic drugs clozapine and sulpiride [25]. depression, while the transcripts derived from differ- DNA methylation changes related to schizophrenia ent transcriptional start sites displayed some variation. and bipolar disorder have recently been discovered in a GR exon 1F transcripts were downregulated in major genome-wide methylation analysis using post-mortem depression, accompanied by reduced levels of the ex- braintissue [59]. The studyidentified prominentlociof on 1F transcription factor NGFI-A [3]. However, pro- methylation changes that are involved in glutamatergic moter 1F was found unmethylated throughout in ma- and GABAergic neurotransmission, brain development jor depression and controls. Therefore, changes in GR and other processes connected to disease. In addition, expression from promoter 1F are not due to direct epi- DNA methylation at the BDNF gene has been found to genetic programming at this promoter, but more likely correlate with polymorphisms that previously had been caused by diminished NGFI-A activity [3]. Assum- associated with major psychosis [59]. A study examin- ing that the individuals tested in this study did not ex- ing the DNA methylation pattern at the X chromosome perience abuse, and in comparison with the observed in peripheral cells of female monozygotic twin pairs methylation changes in abuse victims [54], this implies that were either concordant or discordant for bipolar importantdifferencesin the pathomechanismsof major disease or schizophrenia provided further evidence for depression and abuse victims. epigenetic mechanisms operating in disease etiology of Finally, a pilot study aimed at evaluating the associa- bipolar disorder [77]. Moreover, another comparison tionof DNA methylationin buccal cells and risk for de- of twin pairs concordantor discordantfor bipolardisor- pression [68]. Themethylation analysisfocussedonthe der identified differentially methylatedgenes that could serotonin transporter gene (5HTT) in a sample drawn contribute to the pathophysiology of this disease [48]. froma longitudinalstudy of adolescenthealth. In carri- MeCP2, an essential interpreter protein of the epi- ers of the 5HTTLPR short-allele depressive symptoms genetic mark DNA methylation, has been linked to a were more abundant among those with increased buc- variety of mental dysfunctions and diseases. Rett syn- cal 5HTT methylation [68]. Larger studies are under- drome is a neurodevelopmental disorder that is most way in several laboratories to explore the possibility to frequently (95%) caused by spontaneous mutations in develop peripheral epigenetic markers in depression. the MeCP2 gene located at the X-chromosome [5,74] and is the most frequent cause of mental retardation in females [35]. Moreover, mutations in MeCP2 have 5. Evidences for epigenetic modulation in PTSD been associated with X-linked autism [74] and the An- gelman Syndrome. The animal models for psychiatric diseases as well In an effort to translate the observations of early life as the human cohorts outlined above exhibit some phe- stress-induced epigenetic programming of GR expres- notypic overlap with PTSD, and therefore, these find- sion to the situation in humans, the neuron-specific GR ings also have shaped concepts for PTSD research [79, promoter was examined for epigenetic differences be- 102]. There are also examples for specific PTSD an- tween post-mortem hippocampi from suicide subjects imal models. In a mouse model that uses inescapable with a known history of early life abuse and hippocampi electric foot shock to induce PTSD-like symptoms [83], from either suicide subjects without early life abuse or maternal inexperience was identified as a risk factor controls [54]. In suicide victims with early life abuse, for PTSD [84]. This points at the relevance of epige- the mRNA levels of GR were found to be downregulat- netic factors in PTSD-like behavior. In another PTSD ed concomitantly with elevated promoter methylation. model in rats, predator scent stress was used [17], Moreover,promoter constructsmimicking theobserved and the differences in hippocampal DNA methylation methylation pattern exhibited reduced binding of the were assessed on a genome-wide scale between ani- transcription factor nerve growth factor–inducible pro- mals not responding and responding with PTSD-like tein A [54]. These findings closely resemble the situ- symptoms [15]. The findings not only suggest changes ation observed with reduced parental care as early life in global methylation pattern involved in PTSD devel- stressor in rodents [96,97]. opment, but also present the differentially methylat- 82 U. Schmidt et al. / Epigenetic aspects of posttraumatic stress disorder ed gene Disks Large-Associated Protein (DLGP2) as a induces downstream alterations in immune function by possible target in PTSD. reducing methylation levels of immune-related genes, There is also persuasive evidence for crucial epige- although – as with many association studies of that netic processes in PTSD from clinical studies. In a type – it remains unclear to what extent the leukocyte post mortem study of patients that committed suicide epigenome represents the cerebral epigenome. mRNA levels of GR were found to be downregulated Taken together, these examples from clinical and an- in those suicide victims that had been exposed to early imal studies lead to the conclusion that during vulner- life trauma. This was associated with elevated promot- able ontogenetic episodes of different species a variety er methylation [54]. Moreover, it is known from a va- of stressors can induce epigenetic modifications with riety of studies that victims of early life stress or trau- consecutive behavioral modifications which at least to matic events like abuse or neglect exhibit significantly some extent can be transmitted transgenerationally. higher morbidity rates for a variety of disorders, espe- cially for cardiovascular and affective diseases in their later adulthood [98]. A clinical study with pregnant 6. Perspectives women revealed that massive stress during pregnancy induces changes in epigenetic HPA axis programming Considerable research efforts are underway world- in utero associated with an increased vulnerability for wide to developdrugsthat target the epigenome. These psychiatric disorders possibly transmissible to follow- may also change the treatment regimes in PTSD. To ing generations [79]. Yehuda and colleagues studied betterassess the usefulnessof“epigeneticdrugs”, a bet- mothers who were pregnant while they were exposed ter understanding of the exact epigenetic mechanisms to the terror attack on September 11, 2001: Mothers in PTSD is needed, and particular attention needs to suffering from PTSD as well as their babies exhibited significantly lower salivary cortisol levels in compari- be directed towards the specificity and mechanisms of son to the exposed but PTSD-free control group [106]. drug action, and towards the time window of opportu- Moreover, the observation that a cohort of individu- nity for treatment. als that all developed PTSD after having been exposed While it is still not proven yet that the observed epi- to the same terror attack exhibited markedly changed genetic alterations are actually causal for the observed gene expression patterns that included genes involved phenotypicalterations after stress, epigenetic actions of in HPA regulation, again points to epigenetic modula- psycho-active drugs as well as psychoactivity of drugs tion of gene activity [103]. Another clinical study ex- impacting on the epigenome have been observed. For amining the offspring of Holocaust victims also points example, psychopharmaceuticalshave also been shown to a trauma-induced, inheritable epigenetic modulation to impact on DNA methylation, as demonstrated for of the stress hormone system since serum cortisol lev- specific promoters or for broader effects through inhi- els were found to be significantly lower in healthy off- bition of DNA methyltransferases (DNMTs) [25,70]. spring of victims suffering from PTSD in comparison A large body of evidence points to the potential use to those with PTSD-free ancestors [106]. of HDAC inhibitors in psychiatric diseases. Exposure A recent clinical study on epigenetics and the patho- to chronic social stress leads, after an initial decrease, genesis of PTSD applied methylation microarrays to to a persistent increase in acetylation of histone H3 assay CpG sites in leukocyte DNA from more than in the nucleus accumbens and to a decreased level of 14,000 genes among 23 PTSD-affected and 77 PTSD- HDAC2 [21]. The changes in H3 acetylation were re- unaffected individuals [108]. This association study verted by the application of HDAC inhibitors, which revealed that immune system functions are significant- also exerted robust antidepressant-like effects. More- ly overrepresented among the annotations associated over, the alterations of the transcriptome induced by with genes uniquely unmethylated among PTSD pa- an HDAC inhibitor were very similar to those induced tients. Epigenetic variability in immune function by by the antidepressant fluoxetine [21]. A related study PTSD was corroborated using an independent biolog- investigated promoter chromatin regulation by chro- ical marker of immune response to infection, namely matin immunoprecipitation-chip assays in the nucle- the typically latent herpesvirus CMV-a, whose activity us accumbens of chronically stressed mice. Many of was significantly higher among those with PTSD. Thus, the stress-induced changes were reversed by treatment this report suggests a biologic model of PTSD etiolo- with imipramine, and the resultant pattern exhibited a gy in which an externally experienced traumatic event surprising similarity to that of stress-resilient mice [99]. U. Schmidt et al. / Epigenetic aspects of posttraumatic stress disorder 83

Fig. 1. One carbon cycle linking to epigenetic modifications of DNA and histones. Methylation of DNA, histones and other molecules in the cell is orchestrated by an array of enzymes controlling the turnover of the involved metabolites. Options for (auxiliary) epigenetic treatment focus on manipulation of the enzymatic activity of DNA- and histone-methylatransferases, as well as of the levels of folate, SAM and vitamin B12. The remethylation of homocysteine via betaine occurs in live and kidney, but not in the central nervous system (dashed arrow). THF, tetrahydrofolate; SAM, S-adenosylmethionine.

HDACIs have also been shown to impact on DNA is only short-term. It is conceivable that an immedi- methylation at schizophrenia-relevant promoters, via ate posttraumatic pharmacological intervention, some- decreasing the activity of DNMTs, very similar to the times referred to as secondary prevention, might pre- effects of direct DNMT inhibitors [47]. Yet other re- vent the onset of PTSD symptoms. In the majority of search directions explore the possibility of manipulat- patients, PTSD symptoms appear weeks after the trau- ing the available pool of methyl donors. For exam- matic experience, and early intervention prior to emer- ple, S-adenosyl-methionine (SAM, Fig. 1), an impor- gence of clinical symptoms may be an opportunity also tant methyl donor in cellular processes, has been evalu- for treatment with epigenetic drugs that interfere with ated for potential antidepressant effects with promising programming of PTSD symptoms. results [58]. In addition, folate and vitamin B12 status, There is a limited number of studies exploring the two other important components of the one carbon cy- possibility of secondary treatment in PTSD: Adrenore- cle, have been linked to the incidenceof depression and ceptor antagonists like the conventionally used anti- to the success of antidepressant treatment [18,63,69]. hypertensive propranolol have been found to prevent Moreover, genetic variants of the enzyme that cataly- or at least to mitigate the onset of PTSD-typical re- ses the conversion of 5,10 methylene tetrahydrofolate experiencing symptoms like flashbacks and intrusions to 5-methyl-tetrahydrofolate (Fig. 1) have been linked if applied in the immediate aftermath of a traumat- to depression [18,69]. Thus, genetic variation of the ic event [33,94]. Many authors have suggested that respective enzymes will have to be considered when propranolol and other anti-adrenergic drugs to impede assessing the impact of treatment regimes that target the one carbon pathway [14]. memory formation thereby preventing the onset of tan- While all these developments are encouraging, it talizing flashbacks in predisposed individuals. This no- needsto be consideredthatmostof the epigenetic drugs tion however is not unanimously accepted and some exert pleiotropic effects and are thus prone to exhibit authors have questioned the therapeutic and/or preven- severe side effects [22,34]. For psychiatric diseases in tive effectsof adrenoreceptorantagonistsin PTSD [61]. general, it will be necessary to develop and test drugs Furthermore, low dosed glucocorticoid administration directed towards more specific epigenetic mechanisms. has been reported to reduce the risk of developing PTSD Together with advances in understanding the specific after a traumatic event [78]. In addition, morphine is and probably individual epigenetic processes in dis- reported to reduce the risk of subsequent development ease, this may allow formulation of personalized epige- of PTSD after serious injury [39]. Finally, there is also netic treatment, possibly as co-medication, optimizing evidence from a mouse model that post stress preven- efficacy and minimizing side effects. tive treatment with the SSRI sertraline reduces the de- InPTSDinparticular,sideeffectsmaybetolerableor velopmentof PTSD-like behavior[51]. Taken together, even less pronounced,because treatment in many cases mounting evidence supports the concept of preventive 84 U. Schmidt et al. / Epigenetic aspects of posttraumatic stress disorder treatment of PTSD in general, and the field would ben- disorder: a systematic review, Prog Neuropsychopharmacol efit from discovery and development of drugs targeting Biol Psychiatry 33 (2009), 169–180. epigenetic mechanisms. [7] O. Berton, C.A. McClung, R.J. Dileone, V. Krishnan, W. Renthal, S.J. Russo, D. Graham, N.M. Tsankova, C.A. The epigenetic therapy in PTSD is probably not lim- Bolanos, M. Rios, L.M. Monteggia, D.W. Self and E.J. ited to preventive treatment but may also extend to Nestler, Essential role of BDNF in the mesolimbic dopamine treatment of already developed PTSD. Antidepressants pathway in social defeat stress, Science 311 (2006), 864–868. [8] A. Bird, Molecular biology, Methylation talk between his- and mood stabilizers are frequently used in PTSD ther- tones and DNA, Science 294 (2001), 2113–2115. apy [2,6]. The epigenetic mode of action of some of [9] A.P. Bird and A.P. Wolffe, Methylation-induced repression- these drugs may contribute to their overall effect [70]. belts, braces, and chromatin, Cell 99 (1999), 451–454. It should be noted, though, that while SSRIs have been [10] A.P. Bird, The relationship of DNA methylation to cancer, Cancer Surv 28 (1996), 87–101. shown to reduce PTSD symptoms, treatment results are [11] J.A. Boscarino, Posttraumatic stress disorder, exposure to not fully convincing with about 40% of SSRI -treated combat, and lower plasma cortisol among Vietnam veterans: PTSDpatientsnotrespondingatallandonly20–30%of findings and clinical implications, J Consult Clin Psychol 64 them reaching full remission [6]. Certain psychother- (1996), 191–201. [12] M.P. Boyle, B.J. Kolber, S.K. Vogt, D.F. Wozniak and apeutic strategies have been shown to be effective in L.J. Muglia, Forebrain glucocorticoid receptors modulate PTSD sufferers [26], but they are characterized by a anxiety-associated locomotor activation and adrenal respon- protracted onset of improvement. siveness, J Neurosci 26 (2006), 1971–1978. A substantial portion of individuals at all ages is [13] N. Breslau, Epidemiologic studies of trauma, posttraumatic stress disorder, and other psychiatric disorders, Can J Psy- prone to develop PTSD if exposed to traumatizing chiatry 47(10) (Dec 2002), 923–929. events. We are beginning to understand the biochem- [14] D.F. Carr, G. Whiteley, A. Alfirevic and M. Pirmohamed, ical mechanisms that are translating these experiences Investigation of inter-individual variability of the one-carbon into changes of gene activity and their physiological folate pathway: a bioinformatic and genetic review, The Pharmacogenomics Journal 9 (2009), 291–305. and behavioral consequences. We hope this better un- [15] Y. Chertkow-Deutsher, H. Cohen, E. Klein and D. Ben- derstanding of underlying pathology will ultimately re- Shachar, DNA methylation in vulnerability to post-traumatic sult in better treatment options for these patients. stress in rats: evidence for the role of the post-synaptic den- sity protein Dlgap2, Int J Neuropsychopharmacol 13 (2010), Thus, there is urgent need for the development of 347–359. PTSD-specific medication, to which epigenetic therapy [16] Y.C. Chew, G. Camporeale, N. Kothapalli, G. Sarath and could make an important contribution. J. Zempleni, Lysine residues in N-terminal and C-terminal regions of human histone H2A are targets for biotinylation by biotinidase, J Nutr Biochem 17 (2006), 225–233. [17] H. Cohen and J. Zohar, An animal model of posttraumatic References stress disorder: the use of cut-off behavioural criteria, Annals of the New York Academy of Sciences 1032 (2004), 167–178. [1] H.M. Abdolmaleky, K.H. Cheng, A. Russo, C.L. Smith, S.V. [18] A. Coppen and C. Bolander-Gouaille, Treatment of depres- Faraone, M. Wilcox, R. Shafa, S.J. Glatt, G. Nguyen, J.F. sion: time to consider folic acid and vitamin B12, Journal of Ponte, S. Thiagalingam and M.T. Tsuang, Hypermethylation Psychopharmacology 19 (2005), 59–65. of the reelin (RELN) promoter in the brain of schizophrenic [19] E. Costa, J. Davis, D.R. Grayson, A. Guidotti, G.D. Pap- patients: a preliminary report, Am J Med Genet B Neuropsy- pas and C. Pesold, Dendritic pine hypoplasticity and down- chiatr Genet 134B (2005), 60–66. regulation of reelin and GABAergic tone in schizophrenia [2] M. Adamou, S. Puchalska, W. Plummer and A.S. Hale, Val- vulnerability, Neurobiol Dis 8 (2001), 723–742. proate in the treatment of PTSD: systematic review and meta [20] E. Costa, E. Dong, D.R. Grayson, A. Guidotti, W. Ruzic- analysis, Curr Med Res Opin 23 (2007), 1285–1291. ka and M. Veldic, Reviewing the role of DNA (cytosine-5) [3] S.R. Alt, J.D. Turner, M.D. Klok, O.C. Meijer, E.A.J.F. methyltransferase overexpression in the cortical GABAergic Lakke, R.H. DeRijk and C.P. Muller, Differential expres- dysfunction associated with psychosis vulnerability, Epige- sion of glucocorticoid receptor transcripts in major depres- netics 2 (2007), 29–36. sive disorder is not epigenetically programmed, Psychoneu- [21] H.E. Covington III, I. Maze, Q.C. LaPlant, V.F. Vialou, Y.N. roendocrinology 35 (2010), 544–556. Ohnishi, O. Berton, D.M. Fass, W. Renthal, A.J. Rush III, [4] American Psychiatric Association, Diagnostic and statisti- E.Y. Wu, S. Ghose, V. Krishnan, S.J. Russo, C. Tamminga, cal manual of mental disorders: DSM-IV. Washington, DC: S.J. Haggarty and E.J. Nestler, Antidepressant Actions of American Psychiatric Association, 1994. Histone Deacetylase Inhibitors, The Journal of Neuroscience [5] R.E. Amir, I.B. Van den Veyver, M. Wan, C.Q. Tran, U. 29 (2009), 11451–11460. Francke and H.Y. Zoghbi, Rett syndrome is caused by mu- [22] H.E. Covington, 3rd, V. Vialou and E.J. Nestler, From tations in X-linked MECP2, encoding methyl-CpG-binding synapse to nucleus: novel targets for treating depression, protein 2, Nat Genet 23 (1999), 185–188. Neuropharmacology 58 (2010), 683–693. [6] W. Berger, M.V. Mendlowicz, C. Marques-Portella, G. Kin- [23] G.L. Cuthbert, S. Daujat, A.W. Snowden, H. Erdjument- rys, L.F. Fontenelle, C.R. Marmar and I. Figueira, Pharma- Bromage, T. Hagiwara, M. Yamada, R. Schneider, P.D. Gre- cologic alternatives to antidepressants in posttraumatic stress gory, P. Tempst, A.J. Bannister and T. Kouzarides, His- U. Schmidt et al. / Epigenetic aspects of posttraumatic stress disorder 85

tone deimination antagonizes arginine methylation, Cell 118 [42] H.-S. Huang, A. Matevossian, C. Whittle, S.Y.Kim, A. Schu- (2004), 545–553. macher, S.P. Baker and S. Akbarian, Prefrontal dysfunc- [24] E.R. De Kloet, M. Joels¨ and F. Holsboer, Stress and the tion in Schizophrenia Involves Mixed-Lineage Leukemia 1- brain: from adaptation to disease, Nat Rev Neurosci 6 (2005), Regulated Histone Methylation at GABAergic Gene Promot- 463–475. ers, The Journal of Neuroscience 27 (2007), 11254–1262. [25] E. Dong, M. Nelson, D.R. Grayson, E. Costa and A. Guidotti, [43] T. Jenuwein and C.D. Allis, Translating the histone code, Clozapine and sulpiride but not haloperidol or olanzapine Science 293 (2001), 1074–1080. activate brain DNA demethylation, Proc Natl Acad Sci U S [44] P.L. Jones, G.J. Veenstra, P.A. Wade, D. Vermaak, S.U. Kass, A 105 (2008), 13614–13619. N. Landsberger, J. Strouboulis and A.P. Wolffe, Methylat- [26] A. Ehlers, J. Bisson, D.M. Clark, M. Creamer, S. Pilling, D. ed DNA and MeCP2 recruit histone deacetylase to repress Richards, P.P. Schnurr, S. Turner and W. Yule, Do all psy- transcription, Nat Genet 19 (1998), 187–191. chological treatments really work the same in posttraumatic [45] T. Kouzarides, Chromatin modifications and their function, stress disorder? Clin Psychol Rev 30 (2010), 269–276. Cell 128 (2007), 693–705. [27] S.H. Fatemi, J.A. Earle and T. McMenomy, Reduction in [46] V. Krishnan and E.J. Nestler, The molecular neurobiology of Reelin immunoreactivity in hippocampus of subjects with depression, Nature 455 (2008), 894–902. schizophrenia, bipolar disorder and major depression, Mol [47] M. Kundakovic Y. Chen, A. Guidotti and D.R. Grayson, Psychiatry 5 (2000), 654–663, 571. The reelin and GAD67 promoters are activated by epige- [28] D. Francis, J. Diorio, D. Liu and M.J. Meaney, Nongenomic netic drugs that facilitate the disruption of local repressor transmission across generations of maternal behavior and complexes, Mol Pharmacol 75 (2009), 342–354. stress responses in the rat, Science 286 (1999), 1155–1158. [48] G. Kuratomi, K. Iwamoto, M. Bundo, I. Kusumi, N. Kato, [29] D.D. Francis and M.J. Meaney, Maternal care and the devel- N. Iwata, N. Ozaki and T. Kato, Aberrant DNA methylation opment of stress responses, Curr Opin Neurobiol 9 (1999), associated with bipolar disorder identified from discordant 128–134. monozygotic twins, Mol Psychiatry 13 (2008), 429–441. [30] M. Gardiner-Garden and M. Frommer, CpG islands in verte- [49] S. Levine and G.W. Lewis, Critical period for effects of brate genomes, J Mol Biol 196 (1987), 261–282. infantile experience on maturation of stress response, Science [31] D.R. Grayson, X. Jia, Y. Chen, R.P. Sharma, C.P. Mitchell, 129 (1959), 42–43. A. Guidotti and E. Costa, Reelin promoter hypermethylation [50] D. Liu, J. Diorio, B. Tannenbaum, C. Caldji, D. Francis, A. in schizophrenia, Proc Natl Acad Sci U S A 102 (2005), Freedman, S. Sharma, D. Pearson, P.M. Plotsky and M.J. 9341–9346. Meaney, Maternal care, hippocampal glucocorticoid recep- [32] T.D.Jr. Geracioti, D.G. Baker, N.N. Ekhator, S.A. West, K.K. tors, and hypothalamic- pituitary-adrenal responses to stress, Hill, A.B. Bruce, D. Schmidt, B. Rounds-Kugler, R. Yehuda, Science 277 (1997), 1659–1662. P.E. Keck, Jr. and J.W. Kasckow, CSF norepinephrine con- [51] M.A. Matar, H. Cohen, Z. Kaplan and J. Zohar, The effect centrations in posttraumatic stress disorder, Am J Psychiatry of early poststressor intervention with sertraline on behav- 158 (2001), 1227–1230. ioral responses in an animal model of post-traumatic stress [33] J. Giles, Beta-blockers tackle memories of horror, Nature disorder, Neuropsychopharmacology 31 (2006), 2610–2618. 436 (2005), 448–449. [52] J.A. McCormick, V. Lyons, M.D. Jacobson, J. Noble, J. Dio- [34] D.R. Grayson, M. Kundakovic and R.P. Sharma, Is there a rio, M. Nyirenda, S. Weaver, W. Ester, J.L. Yau, M.J. Meaney, future for histone deacetylase inhibitors in the pharmacother- J.R. Seckl and K.E. Chapman, 5’-heterogeneity of glucocor- apy of psychiatric disorders? Mol Pharmacol 77 (2010), ticoid receptor messenger RNA is tissue specific: differen- 126–135. tial regulation of variant transcripts by earlylife events, Mol [35] S. Hammer, N. Dorrani, J. Dragich, S. Kudo and C. Schanen, Endocrinol 14 (2000), 506–517. The phenotypic consequences of MECP2 mutations extend [53] P.O. McGowan, A. Sasaki, T.C. Huang, A. Unterberger, M. beyond Rett syndrome, Ment Retard Dev Disabil Res Rev 8 Suderman, C. Ernst, M.J. Meaney, G. Turecki and M. Szyf, (2002), 94–98. Promoter-wide hypermethylation of the ribosomal RNA gene [36] W. Harvey, Exercitationes de Generatione Animalium, 1651. promoter in the suicide brain, PLoS One (2008), 3:e2085. [37] P.O. Hassa, S.S. Haenni, M. Elser and M.O. Hottiger, Nuclear [54] P.O. McGowan, A. Sasaki, A.C. D’Alessio, S. Dymov, B. ADP-ribosylation reactions in mammalian cells: where are Labonte’, M. Szyf, G. Turecki, J. Michael and M.J. Meaney, we today and where are we going? Microbiol Mol Biol Rev Epigenetic regulation of the glucocorticoid receptor in human 70 (2006), 789–829. brain associates with childhood abuse, Nature Neuroscience [38] C. Heim, P.M. Plotsky and C.B. Nemeroff, Importance of 12 (2009), 342–348. studying the contributions of early adverse experience to neu- [55] M.J. Meaney, D.H. Aitken, V. Viau, S. Sharma and A. Sar- robiological findings in depression, Neuropsychopharmacol- rieau, Neonatal handling alters adrenocortical negative feed- ogy 29 (2004), 641–648. back sensitivity and hippocampal type II glucocorticoid re- [39] T.L. Holbrook, M.R. Galarneau, J.L. Dye, K. Quinn and A.L. ceptor binding in the rat, Neuroendocrinology 50 (1989), Dougherty, Morphine Use after Combat Injury in Iraq and 597–604. Post-Traumatic Stress Disorder, N Engl J Med 362 (2010), [56] M.J. Meaney, Maternal care, gene expression, and the trans- 110–117. mission of individual differences in stress reactivity across [40] F. Holsboer, The corticosteroid receptor hypothesis of de- generations, Annu Rev Neurosci 24 (2001), 1161–1192. pression, Neuropsychopharmacology 23 (2000), 477–501. [57] M.L. Meewisse, J.B. Reitsma, G.J. de Vries, B.P. Gersons [41] F. Holsboer, C.J. Lauer, W. Schreiber and J.C. Krieg, Altered and M. Olff, Cortisol and post-traumatic stress disorder in hypothalamic-pituitary-adrenocortical regulation in healthy adults: systematic review and meta-analysis, Br J Psychiatry subjects at high familial risk for affective disorders, Neuroen- 191 (2007), 387–392. docrinology 62 (1995), 340–347 86 U. Schmidt et al. / Epigenetic aspects of posttraumatic stress disorder

[58] D. Mischoulon and M. Fava, Role of S-adenosyl-L- [74] M.B. Ramocki, S.U. Peters, Y.J. Tavyev, F. Zhang, C.M. methionine in the treatment of depression: a review of the Carvalho, C.P. Schaaf, R. Richman, P. Fang, D.G. Glaze, evidence, Am J Clin Nutr 76 (2002), 1158Se1161S. J.R. Lupski and H.Y. Zoghbi, Autism and other neuropsy- [59] J. Mill, T. Tang, Z. Kaminsky, T. Khare, S. Yazdanpanah, L. chiatric symptoms are prevalent in individuals with MeCP2 Bouchard, P. Jia, A. Assadzadeh, J. Flanagan, A. Schumach- duplication syndrome, Ann Neurol 66 (2009), 771–782. er, S.C. Wang and A. Petronis, Epigenomic profiling reveals [75] A. Razin and M. Szyf, DNA methylation patterns. Formation DNA-methylation changes associated with major psychosis, and function, Biochim Biophys Acta 782 (1984), 331–342. Am J Hum Genet 82 (2008), 696–711. [76] W. Renthal and E.J. Nestler, Chromatin regulation in drug ad- [60] M.B. Muller¨ and F. Holsboer, Mice with mutations in the diction and depression, Dialogues Clin Neurosci 11 (2009), HPA-system as models for symptoms of depression, Biol 257–268. Psychiatry 59 (2006), 1104–1115. [77] A. Rosa, M.M. Picchioni, S. Kalidindi, C.S. Loat, J. Knight, [61] E.V. Muravieva and C.M. Alberini, Limited efficacy of pro- T. Toulopoulou, R. Vonk, A.C. van der Schot, W. Nolen, R.S. pranolol on the reconsolidation of fear memories, Learn Mem Kahn, P. McGuffin, R.M. Murray and I.W. Craig, Differen- 17 (2010), 306–313. tial Methylation of the X-Chromosome is a Possible Source [62] C. Murgatroyd, A.V. Patchev, Y. Wu, V. Micale, Y. of Discordance for Bipolar Disorder Female Monozygotic Bockmuhl,¨ D. Fischer, F. Holsboer, C.T. Wotjak, O.F. Almei- Twins, Am J Med Genet Part B 147B (2008), 459–462. da and D. Spengler, Dynamic DNA methylation programs [78] G. Schelling, B. Roozendaal, T. Krauseneck, M. Schmoelz, persistent adverse effects of early-life stress, Nat Neurosci D. DE Quervain and D. Briegel, Efficacy of hydrocortisone 12 (2009), 1559–1566. in preventing posttraumatic stress disorder following critical [63] F.A.J. Muskieta and R.F.J. Kemperman, Folate and long- illness and major surgery, Ann N Y Acad Sci 1071 (2006), chain polyunsaturated fatty acids in psychiatric disease, Jour- 46–53. nal of Nutritional Biochemistry 17 (2006), 717–727. [79] J.R. Seckl and M.J. Meaney, Glucocorticoid Programming [64] X. Nan, H.H. Ng, C.A. Johnson, C.D. Laherty, B.M. Turner, and PTSD Risk, Ann NY Acad Sci 1071 (2006), 351–378. R.N. Eisenman and A. Bird, Transcriptional repression by [80] M.J. Selye, The significance of the adrenals for adaptation, the methyl-CpG-binding protein MeCP2 involves a histone Science 85 (1937), 247–248. deacetylase complex, Nature 393 (1998), 386–389. [81] Y. Shiio and R.N. Eisenman, Histone sumoylation is associ- [65] G.N. Neigh, C.F. Gillespie and C.B. Nemeroff, The neuro- ated with transcriptional repression, Proc Natl Acad Sci U S biological toll of child abuse and neglect, Trauma Violence A 100 (2003), 13225–13230. Abuse 10(4) (Oct 2009), 389–410. Epub 6 Aug 2009. [82] A. Shilatifard, Chromatin modifications by methylation and [66] C.J. Nelson, H. Santos-Rosa and T. Kouzarides, Proline iso- ubiquitination:implications in the regulation of gene expres- merization of histone H3 regulates lysine methylation and sion, Annu Rev Biochem 75 (2006), 243–269. gene expression, Cell 126 (2006), 905–916. [83] A. Siegmund and C.T. Wotjak, A mouse model of posttrau- [67] C.B. Nemeroff, The corticotropin releasing factor CRF hy- matic stress disorder that distinguishes between conditioned pothesis of depression: new findings and new directions, Mol and sensitised fear, J Psychiatr Res 41 (2007), 848–860. Psychiatry 1 (1996), 336–342. [84] A. Siegmund, M. Dahlhoff, U. Habersetzer, A. Mederer, E. [68] C.A. Olsson, D.L. Foley, M. Parkinson-Bates, G. Byrnes, Wolf, F. Holsboer and C.T. Wotjak, Maternal inexperience as M. McKenzie, G.C.R. Patton, R. Morley, R.J.L. Anney, J.M. a risk factor of innate fear and PTSD-like symptoms in mice, Craig and R. Saffery, Prospects for epigenetic research with- Journal of Psychiatric Research 43 (2009), 1156–1165. in cohort studies of psychological disorder: A pilot inves- [85] S.M. Southwick, J.D. Bremner, A. Rasmusson, C.A.3rd. tigation of a peripheral cell marker of epigenetic risk for Morgan, A. Arnsten and D.S. Charney, Role of nore- depression, Biological Psychology 83 (2010), 159–165. pinephrine in the pathophysiology and treatment of posttrau- [69] R.T.P. Paul, A.P. McDonnell and C.B. Kelly, Folic acid: neu- matic stress disorder, Biol Psychiatry 46 (1999), 1192–204. rochemistry, metabolism and relationship to depression, Hum [86] L.A. Sroufe, Psychopathology as an outcome of develop- Psychopharmacol Clin Exp 19 (2004), 477–488. ment, Dev Psychopathol 9 (1997), 251–268. [70] T. Perisic, N. Zimmermann, T. Kirmeier, M. Asmus, F. Tuor- [87] B.D. Strahl and C.D. Allis, The language of covalent histone to, M. Uhr, F. Holsboer, T. Rein and J. Zschocke, Valproate modifications, Nature 403 (2000), 41–45. and amitriptyline exert common and divergent influences [88] A.I. Stone and K.L. Bales, Intergenerational Transmission of on global and gene promoter-specific chromatin modifica- the Behavioral Consequences of Early Experience in Prairie tions in rat primary astrocytes, Neuropsychopharmacology Voles, Behavioural Processes, Online-pre-publication, 2010. 35 (2010), 792–805. [89] P.H. Tate and A.P.Bird, Effects of DNA methylation on DNA- [71] P. Pervanidou and G.P. Chrousos, Neuroendocrinology of binding proteins and gene expression, Curr Opin Genet Dev post-traumatic stress disorder, Prog Brain Res 182 (2010), 3 (1993), 226–231. 149–160 [90] M. Tochigi, K. Iwamoto, M. Bundo, A. Komori, T. Sasaki, N. [72] P.M. Plotsky and M.J. Meaney, Early, postnatal experience Kato and T. Kato, Methylation status of the reelin promoter alters hypothalamic corticotropin-releasing factor CRF mR- region in the brain of schizophrenic patients, Biol Psychiatry NA, median eminence CRF content and stress-induced re- 63 (2008), 530–533. lease in adult rats, Brain Res Mol Brain Res 18 (1993), 195– [91] N.M. Tsankova, O. Berton, W. Renthal, A. Kumar, R.L. Neve 200. and E.J. Nestler, Sustained hippocampal chromatin regula- [73] P.M. Plotsky, K.V. Thrivikraman, C.B. Nemeroff, C. Cald- tion in a mouse model of depression and antidepressant ac- ji, S. Sharma and M.J. Meaney, Long-term consequences of tion, Nat Neurosci 9 (2006), 519–525. neonatal rearing on central corticotropin-releasing factor sys- [92] B.M. Turner, Histone acetylation and an epigenetic code, tems in adult male rat offspring, Neuropsychopharmacology Bioessays 22 (2000), 836–845. 30 (2005), 2192–2204. [93] F. Watt and P.L. Molloy, Cytosine methylation prevents bind- ing to DNA of a HeLa cell transcription factor required for U. Schmidt et al. / Epigenetic aspects of posttraumatic stress disorder 87

optimal expression of the adenovirus major late promoter, stress disorder, Am J Psychiatry 150 (1993), 83–86. Genes Dev 2 (1988), 1136–1143. [102] R. Yehuda and Bierer, The Relevance of Epigenetics to [94] G. Vaiva, F. Ducrocq, K. Jezequel, B. Averland, P. Lestavel, PTSD: Implications for the DSM-V, Journal of Traumatic A. Brunet and C.R. Marmar, Immediate treatment with pro- Stress 22 (2009), 427–434. pranolol decreases posttraumatic stress disorder two months [103] R. Yehuda, G. Cai, J.A. Golier, C. Sarapas, S. Galea, M. after trauma, Biol Psychiatry 54 (2003), 947–949. Ising, T. Rein, J. Schmeidler, B. Muller-Myhsok,¨ F. Holsboer [95] C.H. Waddington, Endeavour 1 (1942), 18–20. and J.D. Buxbaum, Gene Expression Patterns Associated [96] I.C. Weaver, N. Cervoni, F.A. Champagne, A.C. D’Alessio, with Posttraumatic Stress Disorder Following Exposure to S. Sharma, J.R. Seckl, S. Dymov, M. Szyf and M.J. Meaney, the World Trade Center Attacks, Biol Psychiatry 66 (2009), Epigenetic programming by maternal behavior, Nat Neurosci 708–711. 7 (2004), 847–854. [104] R. Yehuda, Biology of posttraumatic stress disorder, J Clin [97] I.C. Weaver, A.C. D’Alessio, S.E. Brown, I.C. Hellstrom, Psychiatry 62(Suppl 17) (2001), 41–46. S. Dymov, S. Sharma, M. Szyf and M.J. Meaney, The tran- [105] R. Yehuda, S.L. Halligan, J.A. Golier, R. Grossman and L.M. scription factor nerve growth factor-inducible protein a me- Bierer, Effects of trauma exposure on the cortisol response to diates epigenetic programming: altering epigenetic marks by dexamethasone administration in PTSD and major depressive immediate-early genes, J Neurosci 27 (2007), 1756–1768. disorder, Psychoneuroendocrinology 29 (2004), 389–404. [98] I.C. Weaver, Shaping adult phenotypes through early life [106] R. Yehuda., S.M. Engel, S.R. Brand, J. Seckl, S.M. Mar- environments. Birth defects research. Part C, Embryo today, cus and G.S. Berkowitz, Transgenerational effects of post- reviews 87 (2009), 314–326 traumatic stress disorder in babies of mothers exposed to [99] M.B. Wilkinson, G. Xiao, A. Kumar, Q. LaPlant, Q. Renthal, the World Trade Center attacks during pregnancy, Journal of D. Sikder, T.J. Kodadek and E.J. Nestler, Imipramine Treat- Clinical Endocrinology and Metabolism 90 (2005), 4115– ment and Resiliency Exhibit Similar Chromatin Regulation 4118. in the Mouse Nucleus Accumbens in Depression Models, [107] R. Yehuda, Status of glucocorticoid alterations in post- The Journal of Neuroscience 29 (2009), 7820–7832. traumatic stress disorder, Ann N Y Acad Sci 1179 (Oct 2009), [100] G.T. Wondrak, D. Cervantes-Laurean, E.L. Jacobson and 56–69. Review. M.K. Jacobson, Histone carbonylation in vivo and in vitro, [108] M. Uddin, A.E. Aiello, D.E. Wildman, K.C. Koenen, G. Paw- Biochem J 351Pt 3 (2000), 769–777. elec, R. de Los Santos, E. Goldmann and S. Galea, Epige- [101] R. Yehuda, S.M. Southwick, J.H. Krystal, D. Bremner, D.S. netic and immune function profiles associated with posttrau- Charney and J.W. Mason, Enhanced suppression of corti- matic stress disorder, Proc Natl Acad Sci U S A 107 (2010), sol following dexamethasone administration in posttraumatic 9470–9475. M EDIATORSof INFLAMMATION

The Scientific Gastroenterology Journal of Research and Practice Diabetes Research Disease Markers World Journal Hindawi Publishing Corporation Hindawi Publishing Corporation Hindawi Publishing Corporation http://www.hindawi.com Volume 2014 Hindawi Publishing Corporation Hindawi Publishing Corporation http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

Journal of International Journal of Immunology Research Endocrinology Hindawi Publishing Corporation Hindawi Publishing Corporation http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

Submit your manuscripts at http://www.hindawi.com

BioMed PPAR Research Research International Hindawi Publishing Corporation Hindawi Publishing Corporation http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

Journal of Obesity

Evidence-Based Journal of Stem Cells Complementary and Journal of Ophthalmology International Alternative Medicine Oncology Hindawi Publishing Corporation Hindawi Publishing Corporation Hindawi Publishing Corporation Hindawi Publishing Corporation Hindawi Publishing Corporation http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

Parkinson’s Disease

Computational and Mathematical Methods Behavioural AIDS Oxidative Medicine and in Medicine Neurology Research and Treatment Cellular Longevity Hindawi Publishing Corporation Hindawi Publishing Corporation Hindawi Publishing Corporation Hindawi Publishing Corporation Hindawi Publishing Corporation http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014