Review Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Psychiatr. Oct 19, 2021; 11(10): 711-735
Published online Oct 19, 2021. doi: 10.5498/wjp.v11.i10.711
Alternative models for transgenerational epigenetic inheritance: Molecular psychiatry beyond mice and man
Gary R Hime, Terence Y Pang, Department of Anatomy and Physiology, The University of Melbourne, Parkville 3010, VIC, Australia
Sophie LA Stonehouse, Terence Y Pang, Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville 3052, VIC, Australia
ORCID number: Gary R Hime (0000-0003-4732-3184); Sophie LA Stonehouse (0000-0003-2935-0963); Terence Y Pang (0000-0002-7000-5304).
Author contributions: Stonehouse SLA, Hime GR and Pang TY prepared the initial draft; Pang TY and Hime GR edited and reviewed the final manuscript.
Conflict-of-interest statement: The authors declare no conflict of interests.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Terence Y Pang, PhD, Senior Postdoctoral Fellow, Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville 3052, VIC, Australia. terence.pang@florey.edu.au
Received: February 24, 2021
Peer-review started: February 24, 2021
First decision: July 4, 2021
Revised: July 19, 2021
Accepted: August 25, 2021
Article in press: August 25, 2021
Published online: October 19, 2021

Abstract

Mental illness remains the greatest chronic health burden globally with few in-roads having been made despite significant advances in genomic knowledge in recent decades. The field of psychiatry is constantly challenged to bring new approaches and tools to address and treat the needs of vulnerable individuals and subpopulations, and that has to be supported by a continuous growth in knowledge. The majority of neuropsychiatric symptoms reflect complex gene-environment interactions, with epigenetics bridging the gap between genetic susceptibility and environmental stressors that trigger disease onset and drive the advancement of symptoms. It has more recently been demonstrated in preclinical models that epigenetics underpins the transgenerational inheritance of stress-related behavioural phenotypes in both paternal and maternal lineages, providing further supporting evidence for heritability in humans. However, unbiased prospective studies of this nature are practically impossible to conduct in humans so preclinical models remain our best option for researching the molecular pathophysiologies underlying many neuropsychiatric conditions. While rodents will remain the dominant model system for preclinical studies (especially for addressing complex behavioural phenotypes), there is scope to expand current research of the molecular and epigenetic pathologies by using invertebrate models. Here, we will discuss the utility and advantages of two alternative model organisms–Caenorhabditis elegans and Drosophila melanogaster–and summarise the compelling insights of the epigenetic regulation of transgenerational inheritance that are potentially relevant to human psychiatry.

Key Words: Transgenerational inheritance, Epigenetics, Invertebrate models, Caenorhabditis elegans, Drosophila melanogaster, Environmental stress

Core Tip: Psychiatry research is only beginning to identify the complex epigenetic pathologies across various conditions that may regulate symptomatology. Epigenetics may account for certain conditions that are highly heritable but are not fully accounted for by genetics. Preclinical animal models are a necessary tool to accelerate our understanding of molecular mechanisms and for developing new therapeutic options. Simple behavioural and neurobiological assays combined with high levels of functional gene conservation and rapid generation time in easily genetically manipulated organisms make Caenorhabditis elegans and Drosophila melanogaster excellent systems to model transgenerational epigenetic inheritance phenotypes.



INTRODUCTION

Advances in genomic technologies have led to a rapid increase in the number of known genomic variants linked to human psychiatric illnesses. However, we still know little of the molecular and genetic functions of many of these genes or their mode of inheritance. The wealth of genetic information and experimental techniques associated with laboratory model organisms that have not been traditionally utilised for analysis of psychiatric illnesses provide an untapped resource that promise to revolutionise our understanding of these conditions.

At the turn of the 18th century, the French naturalist Jean-Baptiste Lamarck proposed that environmentally adaptive traits could be acquired by an individual over a lifetime and, more importantly, inherited by their progeny. It was not until the 21st century that Lamarckian theory re-emerged from the shadows of Darwin’s theory of natural selection and the principles of genetic inheritance. This recent revival has been driven by growing evidence of unusual inheritance patterns across a wide number of species, which collectively indicate the presence of biological mechanisms that govern how the physical environment, diet and individual experiences not only influence our individual constitution, but the health of our descendants as well. In the past decade, preclinical studies of mammalian models of human disease have uncovered robust evidence of transgenerational shifts in health. However, alternative animal models should be considered as a means of conducting more time- and cost-effective transgenerational research. Here, we summarise recent advances in transgenerational epigenetic inheritance stemming from non-mammalian models that have revealed epigenetic processes potentially relevant to psychiatry. We hope to convince readers that research based on these non-mammalian organisms have the capacity to provide novel insights into the molecular pathologies of different neuropsychiatric conditions.

Epigenetic inheritance drives the adaptation of phenotypic traits and plays a significant role in directing human health outcomes across generations. For example, the accumulation of specific epigenetic modifications is proposed to contribute to the increasing prevalence of cardiovascular and metabolic diseases[1,2]. Separately, epigenetic modifications have been demonstrated in the transgenerational transmission of risk for mental illness, and possibly contributing to the increasing prevalence of a range of psychiatric disorders[3-6]. However, non-mammalian models have also contributed by extending our understanding of the molecular pathologies in human disease. For example, studies of the nematode Caenorhabditis elegans (C. elegans) have not only provided us enlightening perspectives on the molecular regulation of aging[7,8] but also revealed how stress and nutrition are transgenerational modifiers of progeny survival[9-11].

Briefly, transgenerational inheritance broadly describes the process of a parental generation undergoing experiences and exposures that are subsequently linked to altered phenotypes and behaviours in future generations (in F2s at the very least). Note that the phrase ‘intergenerational inheritance’ describes transmission that is limited (or only studied up till) to the very next F1 generation (see Figure 1 for further patrilineal and matrilineal distinctions). While the full spectrum of biological processes underlying transgenerational inheritance is yet to be fully elucidated, a multiplex of epigenetic modifications has been implicated. Importantly, epigenetic inheritance specifically excludes the reorganisation of genome sequence through DNA mutations, and some epigenetic marks are species-specific (further emphasizing the importance of multi-species research). The most widely studied epigenetic modifications include DNA methylation, histone protein modifications (such as methylation, acetylation), as well as short and long non-coding RNAs (sncRNAs and lncRNAs, respectively) that moderate transcriptional activity. Due to space constraints, we refer readers to the following reviews that comprehensively discuss the biochemistry of epigenetic modifications relevant to the neuropsychiatric field[12-16]. The epigenome is subject to modification following exposure to stressors that challenge survival, ranging from environmental (exposure to toxic chemicals)[17] to physical (heat stress) to psychological (fear of predation)[18,19]. We now know that offspring can inherit a range of epigenetic modifications that alters their physical or behavioural traits. Over the past decade, preclinical studies of rodent models of chronic stress[20,21] and trauma[22-24] have demonstrated this phenomenon, but could alternative non-mammalian models of stress offer further insight into the relevant epigenetic pathologies? These tools offer the field of psychiatry the opportunity to clarify the extent to which the risk for mental illness may be moderated by parental or ancestral exposures to such stressors and life events, and understand the molecular mechanisms mediating such forms of transgenerational inheritance. Epidemiological studies have reported a range in heritability of neuropsychiatric disorders (although readers should note that there have been relatively few studies given the challenges of conducting such large-scale research). For example, a high degree of heritability (81%) was initially estimated for schizophrenia (SZ) based on twin studies[25], while subsequent estimates based on the Danish and Swedish populations were comparatively lower at approximately 60%[26,27]. Those latter studies also estimated that heritability of bipolar disorder (BP) was similar to SZ. However, the potential that epigenetic inheritance moderates the heritability of certain neuropsychiatric conditions has yet to be thoroughly investigated. Of course, in contrast, other psychiatric disorders such as alcohol dependence or major depression display low-moderate degrees of heritability[28] so while those disorders may involve aspects of epigenetic pathology, it is less likely that epigenetic inheritance would be a significant causal factor.

Figure 1
Figure 1  Differences between the definition of transgenerational and intergenerational inheritance through the male and female germ lines.

While studies of C. elegans and Drosophila melanogaster (D. melanogaster) may be initially dismissed as far removed from relevance to human physiology, and although most preclinical drug testing is performed with rodent models, these invertebrate model systems provide alternative approaches to conducting complementary research of common epigenetic mechanisms and biochemical processes that may be fundamental to neuropsychiatric pathologies. One should not forget that mammalian transgenerational research can trace its roots to historically rich and revealing studies of plants. Some of the earliest evidence for the phenomenon include Barbara McClintock’s ground-breaking studies of retrotransposition in maize and the transgenerational inheritance of transposon phases. While we tend to associate ‘stress’ with the notion of psychosocial stress, this term can be used to encompass any extrinsic condition that disturbs the normal function of the biological system, or a condition that decreases fitness, including thermal stress, desiccation, UV stress, starvation, chemical exposure and overcrowding. In using alternative animal models, it is crucial that etiologically relevant stressors are applied in the appropriate manner. Heat stress is well known to impact a wide range of physiological and behavioural parameters, which can result in gastrointestinal dysfunction[29], increased blood pressure and disordered metabolic function[30]. In particular, elevated temperatures cause profound disruptions to various aspects of reproduction in both mammals and invertebrates including mating behaviours[31,32], spermatogenesis and oogenesis, egg/foetal development and viability, and offspring body size[33,34]. With mounting concerns about climate change, and recent increases in unusual climate events, understanding how we adapt to such environmental changes and the implications for global population health trends have become more important than ever. A recent systematic review of the impacts of climate change on mental health reported on the complexities in attempting to consolidate the data, but highlighted more common psychopathologies such as anxiety and trauma[35]. It is unclear if and how climatic factors could influence human health outcomes through epigenetic modifications. Understandably, designing and conducting human studies of this nature would be highly challenging due to the inherent complexities e.g. having to account for geographical and ethnic diversities. However, research based in the primary production industries may be an unexpected source of early clues as to how these occur. Afterall, developing the knowledge to control the effects of heat stress has been crucial to the field of agriculture for maximising crop yield[36,37] and maintaining livestock fecundity and fitness[38,39].

There are mounting calls to recognize that ancestral health is a significant contributing factor of current day human health and phenotypes, and this would require maintaining detailed individual medical records for longitudinal epidemiological studies. On a large scale, such a perspective shift would aid us in identifying the determinants of public health issues and evaluating possible interventions and treatments. Furthermore, elucidating the mechanisms driving environmentally-induced epigenetic changes linked to specific aspects of health and disease may promote a shift towards the development of personalised treatments and drugs based on these signatures[40]. With numerous epigenetic processes conserved from invertebrates to humans, it is unsurprising that many fundamental epigenetic processes are also shared by humans and non-mammalian animals. Therefore, there is valid argument for utilising non-mammalian species as viable alternative animal models to investigate environmentally induced changes in human health, stress response and behavioural adaptations. We will now summarise recent evidence from transgenerational studies of key two non-mammalian models-C. elegans and D. melanogaster–focussing on environmental stressors and highlight their potential utility for investigating the molecular pathologies of psychiatric conditions.

EPIGENETIC MODIFICATIONS IDENTIFIED BY TRANSGENERATIONAL STUDIES OF C. ELEGANS RELEVANT TO PSYCHIATRY

In contrast to mammalian models where multigenerational studies are impeded by long generational times, logistical difficulties and confounding factors, invertebrate models breed rapidly with large progeny cohorts, making them ideal models for performing multi-generational studies. There are the obvious limitations of C. elegans as a model, primarily that it is a relatively simple organism lacking many organ systems found in vertebrates. However, the C. elegans genome possesses homologs of about two-thirds of all human disease genes. Thus, it is widely used as a model system for studying aging, age-related diseases[41] and neurogenerative conditions[42]. Transgenerational studies of C. elegans could therefore provide insight into the molecular pathologies and epigenetic modifications that could be accumulating across generations in humans. Here, we will summarise recent advances in our understanding of the transgenerational responses of C. elegans involving thermal stress and starvation and highlight their relevancy to human psychopathologies (Table 1).

Table 1 Studies of transgenerational epigenetic inheritance in Caenorhabditis elegans of relevance to neuropsychiatric conditions and mammalian preclinical models.
Type of stress (if applicable to study)
Transgenerational shifts in progeny phenotypes
Epigenetic modifications implicated in the inheritance process
Ref.
Psychiatric conditions with similar epigenetic pathology
Ref.
Elevated temperatureTemperature-induced transcriptome changes potentially up to F14 generationHeat shock reduces H3K9me3 to facilitate de-repression of endogenously repressed repeats (DNA transposons)Klosin et al[43], 2017Repetitive elements as etiological factors for schizophrenia (SZ), bipolar disorder and major depression (review)Darby and Sabunciyan 2014[44]
No difference in another repressive mark, H3K27me3Altered expression of human endogenous retroviruses associated with autism spectrum disorder and SZ (review)Misiak et al[169], 2019
Active histone marks H3K36me3 and H3K4me2 both unchangedTissue-specific repetitive elements expression differences in Parkinson’s diseaseBillingsley et al[170], 2019
Heat shockMaternal heat shock altered survival of F1 progeny through 5-HT dependent HSF-1 recruitment to heat shock protein gene promotors. Persistence of phenotypic changes not investigatedHistone H3 occupancy at hsp70 genes decreased following heat shockDas et al[9], 2020 MDD associated with increased hsp70 expression in post mortem dorsolateral prefrontal cortexMartín-Hernández et al[53], 2018
Elevated serum HSP70 levels predicted development of MDD for premenopausal women. Serum HSP70 decreased over time for women who did not develop MDDPasquali et al[54], 2018
Decreased Hsp70 expression in CA4 associated with complete seizure remission for temporal lobe epilepsyKandratavicius et al[171], 2014
NANATransgenerational inheritance of H3K36me3 is regulated by two distinct histone methyltransferases, MES-4 and MET-1Kreher et al[172], 2018 H3K36me3 implicated in SZ susceptibility SNPs. But histone lysine methyltransferases yet to be investigated in the context of SZNiu et al[65], 2019
NANALifespan regulated by the H3K9me2 methyltransferase MET-2Lee et al[49], 2019 H3K9me2 elevated in post-mortem SZ brains and peripheral blood cells. Treatment with histone methyltransferase inhibitor BIX-01294 decreased H3K9me2 levels and rescued expression of SZ risk genesChase et al[50], 2019
Reduced H3K9me2 at oxytocin and arginine vasopressin gene promotors in a rodent model of stress-induced depression. Rescued by physical exerciseKim et al[51], 2016
Cdk-5 targeted H3K9me2 attenuates cocaine-induced locomotor behaviour and conditioned place preference in a rodent model of addictionHeller et al[52], 2016
NADecline in fertilityH3K4me2 demethylase spr-5Greer et al[173], 2014 Treatment with antipsychotic drug olanzapine increased H3K4me2 binding on gene loci associated with adipogenesis and lipogenesis in a rat modelSu et al[174], 2020
KDM5C gene that encodes the H3K4me2/3 histone demethylase linked to autism and intellectual disabilityVallianatos et al[175], 2018
Heavy metal (arsenite) stressIncreased resistance to oxidative stress up to F2 generation; no change in reproduction or lifespanH3K4me3 complex components (wdr-5.1, ash-2, set-2), and transcription factors daf-16 and hsf-1Kishimoto et al[10], 2017 Increased H3K4me3 associated with three synapsin gene variants in bipolar disorder and major depressionCruceanu et al[63], 2013
SZ risk variants are over-represented in association with H3K4me3 in human frontal lobeGirdhar et al[64], 2018
H3K4me3 implicated in SZ susceptibility SNPsNiu et al[65], 2019
Increased H3K4me3 associated with increased Oxtr gene expression in a rat model of methamphetamine addictionAguilar-Valles et al[68], 2014
Hyperosmotic stressIncreased resistance to oxidative stress up to F2 generationNot further investigated in studyKishimoto et al[10], 2017Relevance to human health presently unclear
Larval starvationIncreased resistance to oxidative stress up to F2 generationNot further investigated in studyKishimoto et al[10], 2017Relevance to human health presently unclear
Larval starvationNAThirteen miRNAs up-regulated (miR-34-3p, the family of miR-35-3p to miR-41-3p, miR-39-5p, miR-41-5p, miR-240-5p, miR-246-3p and miR-4813-5p); Two miRNAs down-regulated (let-7-3p, miR-85-5p)Garcia-Segura et al[77], 2015 Eight differentially expressed blood miRNAs linked to PTSD. Four up-regulated (miR-19a-3p, miR-101-3p, miR-20a-5p, miR-20b-5p). Four down-regulated (miR-486-3p, miR-125b-5p, miR-128-3p, miR-15b-3p)Martin et al[78], 2017
Deletion of miR-34 family in mice facilitates resilience to stress-induced anxiety and extinction of fear memoryAndolina et al[84], 2016
miR-34 differentially expressed in induced pluripotent stem cells derived from schizophrenia patientsZhao et al[176], 2015
miR-34a regulates expression of p73, a p53-family member, that is implicated in neuronal differentiationAgostini et al[86], 2011
StarvationIncreased longevity of progeny up to F3 generationInheritance of small RNAs through at least 3 generations. Rechavi et al[11], 2014 miRNAs and rRNAs make up the majority of exRNAs in human plasmaDanielson et al[91], 2017
Small RNAs regulating expression of genes involved in nutrition, metabolic health and lipid transport1 specific exRNA predicted diagnosis of Alzheimer’s diseaseYan et al[94], 2020
exRNAs are potentially involved in the paternal intergenerational influence on offspring metabolic health (mouse model)van Steenwyk et al[93], 2020

The most impressive finding to-date was that exposure of a single progenitor generation to an elevated rearing temperature (25 °C instead of 20 °C) caused transcriptome-wide expression changes that persisted for a further seven generations after temperature normalisation[43]. Importantly, it was identified that the ancestral exposure to a higher temperature was associated with a reduction in the repressive histone modification H3K9me3 (trimethylation of lysine 9 residue in histone H3) in both oocytes and sperm, before onset of zygotic transcription. What could be of importance to the psychiatry field was the revelation that there was de-repression of endogenously repressed repeat sequences, and increased expression of two DNA transposons remained for up to five generations. The role of repetitive elements in human health and disease is still unclear but they have been speculated to be potential etiological factors for SZ, BP and major depressive disorder (MDD)[44], despite a present lack of consistent evidence. For example, there has only been a single report of a repetitive element insertion in three monozygotic twin pairs discordant for SZ[45] but similar observations have not been detected in other studies. However, subsequent studies have reported elevated levels of Class I retrotransposon RNA in cerebrospinal fluid, whole blood and serum samples from SZ patients[46-48]. It should be noted that these latter studies were conducted by the same research group and further independent verification is still required. At the present time, there are also no available rodent models of abnormal repetitive element expression so determining its relevance to neuropsychiatric pathologies is impossible. C. elegans would therefore be a prime model organism to investigate environmental factors associated with the aforementioned psychiatric conditions, with the dysregulation of repetitive element expression as a primary outcome measurable. Such studies would either cement their causal roles or establish them as secondary molecular pathologies.

Separately, another repressive histone mark linked to C. elegans lifespan[49], dimethylation of lysine 9 residue in histone H3 (H3K9me2), has also been implicated in various psychiatric conditions. Increased levels of H3K9me2 were found in post mortem SZ brains and in peripheral blood cells[50]. However, the directionality of this change in expression may vary depending on the specific psychopathology, according to evidence from rodent studies. For example, stress-induced depression was associated with reduced H3K9me2 occupancy at the oxytocin and arginine vasopressin gene promotors, both of which were normalised by physical exercise[51]. Thus, the outcomes linked to the manipulation of H3K9me2 levels are also gene specific. This is further exemplified by the capacity for Cdk-5 targeted H3K9me2 to attenuate cocaine-induced locomotor behaviour and conditioned place preference[52]. These clearly showcase the complexity to epigenetic regulation of gene transcription and the significant challenges faced when attempting to treat psychiatric conditions by targeting a single histone modification. However, armed with precise knowledge of the molecular pathologies, aiming to modify negative behaviours in addiction through gene-targeted histone modification could be an intriguing prospect for the future.

A recent study examined a more severe temperature perturbation through acute heat shock (34 °C for 5 min) and discovered that this caused maternal neurons to release the neurotransmitter 5-HT, which facilitated transcription factor heat shock factor 1 (HSF-1)-mediated mRNA production in soon-to-be fertilized germ cells[9]. The authors proposed that this timely activation of HSF-1 in germ cells ensures viability and future stress tolerance since embryos that arose from heat-shocked mothers contained an excess of protective mRNA and their F1 progeny were more resilient to subsequent temperature insults. It was found that HSF-1 recruited the histone chaperone FAcilitates Chromatin Transcription (FACT) complex to alter histone dynamics and promote transcription of the heat shock protein Hsp70. Interestingly, several studies have identified an accumulation of Hsp70 associated with MDD. In a study of post-mortem brain samples from patients with MDD, Hsp70 was significantly elevated in the dorsolateral prefrontal cortex, while antidepressant treatment did not have any modulatory effect[53]. Separately, elevated serum Hsp70 levels were reportedly predictive of premenopausal women who would go on to develop MDD[54], although Hsp70 levels subsequently decreased for women who did not develop MDD. Collectively, this suggests that Hsp70 could be a useful biomarker for MDD risk but it remains to be verified in a younger, or even a healthy, population.

HSF-1 activity is also associated with elevated histone H4 protein levels in somatic tissue during development, leading to reduced transcription of mitochondrial complex IV genes and decreased respiratory capacity[55]. While it has not been linked specifically to histone H4 only, a similar role of neuronal heat shock proteins in moderating the response to oxidative stress is evidenced in D. melanogaster with increased resistance to oxidative stress and extended organismal lifespan, in addition to ameliorating phenotypes associated with Parkinsonism-type genetic models[56]. Collectively, it emphasizes the conserved association between heat shock proteins, oxidative stress and neuronal damage. However, the precise regulatory roles that histone H3 and H4 proteins provide independently to the overall oxidative stress response remain unclear and warrants further investigation. Mitochondrial dysfunction and the accumulation of oxidative stress are crucial factors in the pathophysiology of MDD[57-59], and biomarkers of oxidative stress are elevated in drug-naïve first episode SZ patients[60]. Thus, there is strong interest in targeting oxidative stress deficiencies in MDD, BP and SZ[61] through antioxidant treatments such as N-acetylcysteine[62]. Future studies could use C. elegans to explore the efficacies of various antioxidant compounds in treating heat shock-induced oxidative stress, as well as their underlying modes of action. Studies could also be extended to heat shocking C. elegans pre-treated with antioxidants to better understand the epigenetic regulation of 5-HT neurotransmission.

The dysregulation of transcriptional activity is widely reported in a swathe of psychiatric conditions but the causes have yet to be precisely identified. For example, H3K4me3 has been implicated in the pathophysiology of SZ, BP and MDD, with increased H3K4me3 is associated with three synapsin gene variants in BP and MDD[63] while SZ risk variants are over-represented in association with H3K4me3 in human frontal lobe samples[64]. The latter is a consistent with a separate study examining H3K4me3 association with SZ susceptibility SNPs[65]. While there have been several independent GWAS studies of SZ, there has yet to be an attempt to reconcile the genomic data with epigenomic variation. That would undoubtedly be a tremendous undertaking, but it could further streamline and identify more robust gene candidates in our attempts to pinpoint the primary molecular pathologies underlying SZ. C. elegans could be used to first establish the molecular consequences of such an abnormal epigenetic landscape and resulting transcriptional dysregulation (matched to existing human data), before further behavioural studies are extended to mammalian models. Incidentally, H3K4me3 was identified by Kishimoto et al[10] as being involved with the transgenerational adaptations to other forms of environmental stressors aside from thermal stress, namely heavy metal exposure, hyperosmotic conditions, and transient starvation[10]. Following progenitor exposure to all three stressors, there were consistent increases in progeny fitness up till the F2 generation; however only the epigenetic mechanism mediating adaptation to arsenite exposure was further investigated. Unlike the repressive histone modifications mentioned above, H3K4me3 predominantly marks transcriptional start sites and is part of a regulatory complex that facilitates access and assembly of RNA polymerase 2[66,67]. Kishimoto et al[10] reported that the genetic components (wdr-5.1, ash-2 and set-2) of the H3K4me3 regulatory complex were required to manifest the transgenerational adaptations, implicating histone H3-dependent gene transcription in transgenerational inheritance. Therefore, future work on H3K4me3-regulation transcriptional activity could provide new insight into the molecular pathways affected in SZ, BP and MDD by targeting C. elegans homologs of human risk genes for more specific investigations.

Finally, in a rat model of methamphetamine addiction, there was greater H3K4me3 association with the oxytocin receptor gene that corresponded to increased Oxtr gene expression[68]. As discussed above, strategies to treat addiction-related molecular pathologies by targeting histone modifications will be challenged by having to account for both active and repressive histone marks. The viability of such interventions and their molecular consequences would be ideally be first tested in C. elegans before proceeding to trials in mammalian models.

Malnutrition and starvation at different stages of life have a dramatic impact on mental health. For example, famine exposure in utero was associated with an increased risk for mental illness in females, though surprisingly with no apparent significant effect on males[69]. Developmental malnutrition driven by abnormalities in oxidative stress pathways has been linked to an increased risk for SZ and other psychiatric illness later-in-life[70]. Nutrition ultimately dictates metabolic health and more recent studies reported that fasting insulin levels and body mass index at different ages were predictive of at-risk status for psychosis or depression[71], while fasting blood glucose and serum lipid levels predicted suicide attempters in young patients with MDD[72]. At the opposite end of the age spectrum, geriatric deficiencies in micronutrients such as folic acid, thiamine or cobalamin have been linked to worsened mental health symptoms[73,74]. However, careful regulation of nutrition through caloric restriction or fasting has been proposed to be effective in improving symptoms of MDD[75], indicating that dietary interventions where appropriate would benefit patients. This could be particularly important in conditions whereby medications could have unavoidable metabolic side effects[76]. While epidemiological data flags the importance of nutrition for mental health, we continue to have a very poor understanding of this interactive relationship in the absence of evidence of causality and the underlying molecular mechanisms. Human studies of that nature would be severely limited by inherent genetic and cultural heterogeneities within populations, and there would be strong ethical arguments against the manipulation of subjects’ diets. These issues are circumvented in studies of C. elegans wherein genetic homogeneity is controlled and dietary manipulations are feasible, although as C. elegans feed upon bacteria subtle dietary manipulations may be more easily accomplished using the chemically controlled diets that have been formulated for D. melanogaster. Transgenerational studies of starvation in C. elegans have already been conducted with clear evidence of downstream impacts on progeny fitness. More importantly, these studies have identified epigenetic mechanisms regulating the transgenerational adaptations, and these could potentially be regulating the molecular pathologies driving the malnutrition-related increase in risk for mental illness.

Kishimoto et al[10] reported that progenitor larval starvation triggered increased resistance to oxidative stress of two generations of progeny[10] but did not pursue the underlying epigenetic mechanisms and their associated molecular adaptations. However, previously, it was reported that starvation during the early L4 Larval stage altered the expression of 13 miRNAs in C. elegans[77]. Of the 13, only 2 were downregulated while the miRNAs of the miR-35 family were most highly upregulated. Being a simple organism, there are only 302 known miRNAs in C. elegans compared to over 2000 human miRNAs, so studying their role in transgenerational inheritance and phenotype adaptations is comparatively straightforward. miRNAs are now established to be dysregulated in different human conditions and are the subjects of interest for severe stress-related anxiety disorders such as post-traumatic stress disorder and SZ, as prognostic biomarkers and therapeutic targets. However, their role as epigenetic regulators of pathogenesis are unclear and systematic profiling of individual miRNAs to neuronal circuitry could be one approach to identifying their potential pathogenic roles in psychiatric conditions.

In a cohort study of military combat veterans, 8 differentially expressed blood miRNAs were associated with the diagnosis of post-traumatic stress disorder (PTSD)[78], and their predicted gene targets were implicated in neurotransmission and maintenance of the neural circuitry. Indeed, multiple functional magnetic resonance imaging studies have clearly demonstrated that brain function is compromised in PTSD[79,80]. There is initial evidence to suggest that paternal PTSD may also have the capacity to influence the neural function and behaviour of progeny, and that this is through the inheritance of sperm-borne miRNAs. In the social defeat mouse model of PTSD, both male and female progeny displayed significant anxiety and depression-related behaviours despite themselves not having been subject to stressful interventions[81,82]. It was later independently reported that modelling paternal early life trauma alters sperm miRNAs and exerts significant intergenerational alterations of target genes in the brains of progeny (e.g. ctnnb1, catenin β1 in the hippocampus)[22]. Our own studies have extended that line of evidence by demonstrating the transgenerational effects of paternal stress exposure and altered sperm miRNAs resulting in significant expression differences of the imprinted gene insulin-like growth factor 2, Igf2 in the hippocampus of two generations of progeny[21]. While their downstream target genes may have been discovered to be dysregulated, there is still some controversy regarding the intergenerational inheritance of sperm miRNAs because having altered levels of miRNAs in sperm does not translate to those same miRNAs being dysregulated in offspring brains[23]. Despite the transgenerational implications of paternal PTSD on brain function of their children remaining unknown at this time, a bigger unresolved question is how traumatic stress alters miRNA expression , with one possibility being dysregulation of histone protein modifications and altered chromatin state. Unlike PTSD, which is caused by an external trigger, miRNAs appear to be co-regulated with susceptibility risk genes in SZ. For example, one study has reported an over-representation of miR-9-5p-targeted risk genes while miR-9-2 is located in a genomic region strongly associated with SZ[83]. Given the strong environmental component to both PTSD and SZ, continuing research into stress-induced miRNA changes in C. elegans could be used to further our understanding of the relevant environment x gene interactions underlying the molecular pathogenesis of PTSD and SZ. Other miRNAs have been implicated in stress-related disorders such as members of the miR-34 family, which are differentially expressed in induced pluripotent stem cells derived from SZ patients[41,84]. Among these, and consistent with the neurodevelopmental hypothesis of SZ[85], miR-34a is a key regulator of p73 expression, a p53-family member that is implicated in neuronal differentiation[86]. However, causal evidence is lacking to demonstrate that miR-34a is an epigenetic conduit for environmental stress to impact on brain development resulting in a schizotypy brain phenotype. One feasible experiment to propose would be ablating expression of the C. elegans homolog of miR-34a or the miR-34 family and study the impacts on neuronal differentiation, development and circuit maturation.

Interestingly, Rechavi et al[11] report that progenitor larval starvation was associated with extended longevity in three generations of progeny through the inheritance of small RNAs that regulate genes involved in nutrition, metabolic health and lipid transport[11]. It has been demonstrated in C. elegans that extracellular RNAs (exRNAs) are transported from one generation to the next through intracellular vesicles or even as unpackaged extracellular material[87]. The transgenerational effects of paternal stress exposures[21-23] involve altered small non-coding RNA content of sperm transmitted in microvesicles within the male reproductive organs[88,89], but so far this has only been demonstrated in mouse models[90]. Perhaps not so coincidentally, miRNAs are one of two major exRNA species in human plasma (the other being ribosomal RNAs)[91]. Their presence and relative stability have led to an emerging recognition of their promise as ‘liquid biopsies’ for diseases, but while early adoption has targeted metabolic pathology[92], the correlation of biofluid exRNA levels with psychiatric conditions remain untested. Interestingly, it was reported that chronic injection of serum from a mouse model of trauma into healthy controls was sufficient to recapitulate the intergenerational impact on offspring metabolism[93]. However, miRNA profiling of the serum content was not performed in that study. Very recently, an investigation profiling exRNAs isolated from the plasma of elderly individuals up to 15 years prior to death revealed that the early presence and progressive increase of phosphoglycerate dehydrogenase (PHGDH) exRNA predicted eventual diagnosis of Alzheimer’s disease (confirmed with post mortem pathology testing)[94]. Studies of C. elegans could be used to first determine how stress triggers an elevation of circulating exRNAs. Subsequently, given that biofluid screening of exRNAs is already being used to aid diabetes and AD diagnoses, there appears to be untapped potential for this methodology as a presymptomatic screening tool in psychiatry.

Overall, recent studies have demonstrated the complexity of epigenetic responses implicated in the transgenerational responses to progenitor stress exposure. These include histone modifications, dysregulation of DNA repetitive elements and altered expression of non-coding RNAs. These are also molecular processes shared by humans and have been identified as molecular pathologies of various psychiatric conditions. Thus, studying the epigenetic response of C. elegans to etiologically relevant environmental stressors and the corresponding physiological and behavioural responses will continue to provide further insight into human molecular psychiatry.

EPIGENETIC MODIFICATIONS IDENTIFIED BY TRANSGENERATIONAL STUDIES OF D. MELANOGASTER RELEVANT TO PSYCHIATRY

D. melanogaster has been established as an invertebrate model organism for studying human neurological disorders due to the remarkable evolutionary conservation of multiple human disease-causing genes. D. melanogaster have a higher degree of concordance with humans than C. elegans, with 75% of human diseases estimated to have a D. melanogaster homologue[95]. While also displaying sexual dimorphism in its physiology and behaviour, D. melanogaster have a generational time of only 10-12 d as opposed to approximately 6-9 wk for mice. Thus, in a protracted timeframe and at much lower cost compared to using rodents, multi-generational studies can also be performed to assess transgenerational effects and adaptations of D. melanogaster offspring to various environmental stressors. Additionally, a wide range of established transgenic strains, gene manipulation techniques and tools are readily available[96]. Here, we refer readers to several broad reviews discussing the utility of D. melanogaster research in advancing the understanding of the complex genetic basis for human traits, psychiatric disorders, neurodegeneration, and for drug discovery and screening[97-100]. Of course, the significant limitations of modelling complex neuropsychiatric conditions in D. melanogaster must also be acknowledged. Despite the relative ease in genetic manipulation, neuropsychiatric conditions such as SZ are driven by a combination of multiple genetic and environmental factors, and cannot be simply reduced to and reproduced in single, double or even triple transgenic knockout strains. Furthermore, the myriad of behavioural symptoms requires higher brain function to manifest, for which only mammalian models could be considered as appropriate. However, these reasons should certainly not diminish the utility value of D. melanogaster as a high throughput screening tool for basic neuropathological, molecular or epigenetic markers of disease. Most recently, D. melanogaster have even been used to model insomnia in order to examine the effectiveness of sleep restriction therapy[101]. However, despite these advantages, transgenerational studies in D. melanogaster aimed at examining mechanisms of epigenetic inheritance remain relatively sparse. Yet, the limited research has produced some compelling evidence, nonetheless. In this section, we will summarise key findings by highlighting the transgenerational outcomes of environmental and chemical stress exposures on offspring phenotypes paired with the reported epigenetic processes implicated. We will then flag the neuropsychiatric conditions for which further D. melanogaster research could potentially shed new light on the pathological origins.

D. melanogaster are sensitive to the climate and temperature fluctuations[102,103] and have been instrumental in advancing our understanding of the heat stress response. Heat stress-associated deleterious effects on physiology and behaviour are largely attributed to its denaturing effect on proteins, which undergo abnormal folding, entanglement and unspecific aggregation[104]. In addition to the disruption of singular proteins, heat stress can also disrupt other cellular mechanisms with the culmination of these individual disruptions being cell death[105]. The ubiquitous and highly conserved heat shock response is a complex cascade of different processes, the most central being the transcriptional up-regulation of genes coding for the family of heat shock proteins that were in fact first discovered in D. melanogaster[106,107]. In addition to the metabolic and physiological effects on the exposed organism[108,109], selective thermal variations can dramatically shift D. melanogaster physical phenotypes such as flight ability over generations (impaired by F2 generation and maintained till the F4 generation) in a sex-dependent manner[110,111]. Thus, imposing a suboptimal ambient environment for survival either by changing the housing temperature or through a transient shift of temperature represents the most etiologically relevant approaches to stressing D. melanogaster. These encapsulate studies of both cold tolerance[112] and heat tolerance (discussed in detail below, Table 2), and these allow us to investigate how genetic variation dictates response to the environment or vice versa. Research into the transgenerational effects of heat stress in D. melanogaster have yielded intriguing and robust evidence of altered offspring physiology and heat stress responses. More importantly, those studies have also revealed epigenetic mechanisms that are of particular interest to psychiatry. Perhaps the most compelling demonstrations of environment-directed modifications of D. melanogaster epigenetics resulting in altered gene expression are the transgenerational studies of white gene expression following heat stress. The X chromosome residing white gene encodes for an ATP-binding cassette transporter that facilitates transport of the eye pigment precursors, guanine and tryptophan (red and brown pigment precursors, respectively) into the developing eyes during pupation[113]. Repression of white achieved by inserting the cellular memory module Fab-7 upstream of white to enhance chromatin silencing results in the loss of eye pigmentation[114]. Importantly, the Fab-7-mediated silencing process involves recruitment of Polycomb Group (PcG) proteins, which are essential in the propagation of chromatin structures and regulate gene silencing through S-phase of the cell cycle[115-117]. The mere developmental exposure to a mildly stressful temperature of 29 °C (typical housing temperature is 25 °C) suppressed Fab-7 expression, resulting in the de-repression of white and recovery of red eye pigmentation[118]. Importantly, that de-repression event was heritable down both male and female germ lines up till the F4 generation. That “founder effect” and maintenance of a de-repressed state across multiple generations indicates that inheritance of the temperature-modified chromatin state is maintained by the PcG protein complex. Of relevance to the human epigenome, the PcG protein complexes catalyse the formation and maintenance of the inactive histone mark H3K27me3[118], which as previously mentioned, is widely associated with neuropsychiatric conditions with abnormal histone modification patterns and aberrant gene transcriptional profiles[119]. Yet, the regulation of differentially expressed genes by PcG protein complexes in neuropsychiatric conditions has not been reported. While PcG protein complex function has been of great interest to the oncology field given the tell-tale features of DNA hypermethylation and aberrant transcriptional silencing of tumour suppressor genes[120], a causative role in psychiatric disorders has yet to be established. PcG protein complexes serve as a master regulator of active gene transcription so understanding the intricacies of PcG regulation of chromatin states will be essential if targeting aberrant histone modifications are to be a major therapeutic focus of the future. Aside from changes at the white gene locus, the multi-generational effects of heat shock on other behavioural (social interaction, mating) and physiological (metabolic and endocrine health) parameters in D. melanogaster are yet to be comprehensively studied. It would be very interesting to investigate if PcG protein complexes also have the capacity to affect the social behaviour, cognition and physical attributes of D. melanogaster by manipulating the extent of histone methylation associated with neuropsychiatric risk genes.

Table 2 Studies of transgenerational epigenetic inheritance in Drosophila melanogaster of potential relevance to psychiatric conditions and mammalian preclinical models.
Type of stress (if applicable to study)
Transgenerational shifts in progeny phenotypes
Epigenetic processes implicated in the inheritance process
Ref.
Potentially relevant psychiatric conditions
Ref.
Thermal stress (selection based on intolerance to heat stress)Reduced ability to fly by F2 generation, maintain through to F4 generationEpigenetic mechanism not investigated; aspects of stress physiology that affect flight still unclearKrebs and Thompson[111], 2006Relevance to human health presently unclear.
Mild heat stress(embryos maintained at 29 °C)De-suppression of white gene up to F4 generationDisruption of polycomb group (PcG) protein complex affecting H3K27me3Bantignies et al[114], 2003 Despite multiple reports of altered H3K27me3, the involvement of PcG protein complexes in human psychopathologies has not been established
Heat shock (flies exposed to 37 °C for 1 h)De-suppression of white gene sustained up to F3 generation required repeated exposure to the same paternal stressor. Gradual return to normal upon removal of heat shockDisruption of pATF-2 mediated heterochromatin assemblySeong et al[121], 2011 Rat model of chronic stress reported increased ATF-2 gene expression in the frontal cortex of chronically stressed rats, which is decreased following chronic antidepressant treatmentLaifenfeld et al[122], 2004
pATF-2 levels are increased in post mortem samples of unmedicated vs medicated patients with MDD. No differences detected for bipolar disorder or schizophreniaGourzis et al[177], 2012
Case report of decreased chromosome 1 heterochromatin in FTLD, misdiagnosed as SZ. Altered size distribution of chromosome 1 heterochromatic region in unrelated SZ patients compared to controlsKosower et al[178], 1995
Risperidone inhibition of heterochromatin formation in human liposarcoma cells in vitro, in a process involving PKA signalling; extent of dysregulated heterochromatin in psychosis yet to be exploredFeiner et al[125], 2019
Parental exposure to risperidone led to intergenerational effects on F1 predator avoidance behaviours in zebrafish; potential human effects have not been investigatedKalichak et al[179], 2019
Heat stress (flies raised at 29 °C)Suppression of BX2 transgene cluster over multiple (50) generationsParamutation of BX2 via maternally inherited piRNAs, triggered by heat stress which resulted in active transcription of piRNAs within that gene locusde Vanssay et al[126], 2012 Paramutation is not regarded as an established epigenetic process in mammals
However, readers should be aware of this proof-of-concept study in miceYuan et al[180], 2015
Casier et al[127], 2019 Paternal transmission of “white-tail-tip” phenotype caused by paramutant allele in mice limited to one generation. Maternal miRNAs and piRNAs regulate (inhibit) germline transmission of paramutation
14 piRNAs differentially expressed in AD prefrontal cortex samples vs controlsQiu et al[128], 2017
Sequencing of CSF-derived exosome sncRNA revealed combination of 3 miRNAs and 3 piRNAs detected AD and predicted the conversion of mild–cognitive impaired (MCI) patients to AD dementia. Greater predictive confidence when combining the smallRNA signature with pTau and Aβ 42/40 ratio pathologyJain et al[129], 2019
Forced cohabitation with predator or endoparasitoid wasps Stressed females shift behaviour to laying eggs on food rich in ethanol, and that preference is inherited through five generationsMaternal inheritance of chromosome III and NPF (Drosophila homolog of NPY) gene locus, reduced NPF expression in the fan shaped body of the adult brain drives ethanol preferenceBozler et al[136], 2019 Dysregulation of NPY levels in the brain is a key pathophysiology of drug addiction. Manipulation of NPY neurotransmission has potentially beneficial behavioural outcomes, depending on the drug in questionGonçalves et al[181], 2016
NPY is implicated in human alcohol misuse disordersMayfield et al[137], 2002
NPY is also implicated in rodent models of alcohol misuse disorderMottagui-Tabar et al[138], 2005
Thorsell and Mathe[139], 2017
Badia-Elder et al[140], 2003
Schroeder et al[142], 2005
Robinson et al[141], 2019
Restraint stressPaternal restraint stress affects epigenome, transcriptome and metabolome of F1 progenyStress-induced up-regulation of Upd3 (Drosophila homolog of IL-6) in somatic cells and testes, activating JAK/STAT pathwaySeong et al[145], 2020 Metabolic dysregulation in the F1 offspring derived from male breeders exposed to early postnatal stressvan Steenwyk et al[146], 2018; van Steenwyk et al[93], 2020
Subsequent p38 activation results in dATF-2 deactivation in germ cells leading to decreased H3K9me2 (repressive mark) at target genes. Repressive histone marks inherited by F1 progenyReview of epigenetic mechanisms proposed to underlie intergenerational transmission of paternal traumaYehuda and Lehrner[182], 2018
Childhood adversity associated with altered DNA methylation of HPA axis and immune system genes; potentially inherited by offspringBick et al[154], 2012
Methylphenidate (MPH) treatmentBehavioural response to MPH is genetically variable and intergenerational effects can be observed in F1 offspringMechanism is unknown but MPH resulted in alterations to expression of many histone modifying genesRohde et al[158], 2019 ADHD is highly heritable, but the reasons are unclear despite the identification of candidate genes. Future studies should attempt to identify transgenerationally heritable epigenetic modifications as the basis for genetic vulnerability
Non-human primate studies indicate that MPH treatment affects normal puberty. The transgenerational implications of this finding for humans needs to be followed-upMattison et al[155], 2011
G418 treatment (toxic stress)Exposure of F0 females to G418 resulted in reduction of Polycomb group gene expression in up till F3 generationMaternal Polycomb group expression in early embryogenesis affects expression of the zygotic genome, which can be inheritedStern et al[168], 2014 G418 has been successfully used to rescue PTC deficiencies in a cell culture model for frontotemporal dementia. However, its broader utility for treating neuropsychiatric conditions remains unknownKuang et al[164], 2020
PTC mutations of neuronal UPF3B gene associated with nonspecific mental retardation with or without austismLaumonnier et al[183], 2010

Interestingly, and in contrast to the stable inheritance pattern mediated by PcG protein complexes, heat shock-induced de-repression of white gene expression involving disruption of the heterochromatin assembly was maintained through three generations of embryos but contingent on repeated exposure of the offspring themselves to heat stress[121]. In that study, the transgenerational effects of heat shock were associated with increased phosphorylation of ATF-2, a member of the CREB/ATF family of transcription factors. Interestingly, levels of phosphorylated ATF-2 are reported to be increased in the ventral parieto-occipital region of post-mortem human brains when comparing between medicated and unmedicated patients with depression[122]; it is unknown if pATF-2 Levels could be predictive of a familial history of MDD or other forms of stress-related psychopathology. The D. melanogaster ATF-2 is known to be an essential regulator of heterochromatin assembly through its co-localisation with HP1, a crucial adaptor molecule for DNA methyltransferases that are recruited along the heterochromatin assembly by H3K9me marks. Thus, despite the lack of evidence at this time, it has been speculated that the general disruption of gene expression in psychiatric conditions such as SZ involves a combination of abnormal DNA methylation and histone methyltransferase activity[123,124], and that recurring environmental stress could be key triggers for the familial manifestations of psychosis. It is especially important that this aspect of epigenetic pathology be examined given more recent in vitro evidence that antipsychotics such as risperidone have the capacity to inhibit heterochromatin formation[125].

Studies of heat stress have also uncovered other heat-induced epigenetic responses involving paramutation and the resulting transgenerational inheritance of small non-coding RNAs via the maternal lineage. de Vanssay et al[126] described a paramutation event involving P-transposable-element repression in the germ line (termed trans-silencing effect, TSE) that converted other homologous clusters typically incapable of TSE into strong silencers[126]. The transgenerational effects of this paramutation persisted through 50 generations of progeny and was found to specifically require aubergine gene-mediated piRNA biogenesis but not Dicer-2 mediated siRNA production. Interestingly, this paramutation is triggered by heat stress and the pattern of piRNA up-regulation is transmitted via the maternal lineage[127]. Thus, one of the persistent epigenetic modifications in response to stress in humans could be the emergence of actively transcribed piRNA loci. While piRNAs are not a core focus of molecular psychiatry, piRNAs have started to gain attention in the domain of neurodegenerative diseases after having been found to be differentially expressed in prefrontal cortical tissue of post-mortem AD brains[128]. That has led to questions of their role in disease pathogenesis and the possibility of using them as a reliable biomarker for human disease. In support of the latter notion, miRNA and piRNA profiling of human cerebrospinal fluid-derived exosomes has more recently been proposed to have utility in diagnosing AD, as well as predicting the conversion from mild cognitive impairment to AD dementia[129]. There is sexual dimorphism in the clinical manifestation of AD with more women than men being diagnosed and maternal transmission is more frequently observed than paternal transmission[130]; but the potential involvement of maternally inherited miRNAs or piRNAs to confer AD risk is completely unknown at this time. In D. melanogaster it has been established that piRNAs are maternally inherited and aging is associated with an increased presence of novel heterochromatic-only secondary piRNAs[131-134]. However, evidence of a similar pattern of inheritance role in humans has yet to be discovered. Our understanding of piRNA in the context of psychiatry and behaviour is barely in its infancy, and there remains much to be uncovered regarding the piRNA pathogenesis and its direct consequences across the range of neuropsychiatric diseases. Perhaps further studies in D. melanogaster can uncover novel piRNA-mediated disease mechanisms for psychiatry conditions that are skewed to maternal transmission.

Predator stress is another form of environmental stress that applies to D. melanogaster and studies have revealed that it is sufficiently severe to induce shifts in reproductive behaviours. Females housed in cohabitation conditions with endoparasitoid wasps develop a preference to lay eggs on ethanol-rich food as ethanol protects the larvae from wasp infection[135]. That change in oviposition behaviour was found to be driven by neuropeptide F (the D. melanogaster homolog of Neuropeptide Y, NPY) and persisted despite removal of the endoparasitoid wasps. More impressively, a recent study reported that exposure to predatory wasps is also an environmental stressor that triggers a similar transgenerational modification of egg laying behaviour over five generations[136]. That shift towards ethanol-rich substrates was established to be superficially maternally transmitted and involved inheritance of Chromosome III within which resides the NPF gene that is differentially expressed in the fan shaped body of the adult female brain. Here, it is worth noting that NPY is of major interest to substance misuse disorders and has been implicated in human alcohol use disorder[137-139] as well as in rodent models[140-142]. Since genetic vulnerability remains the core disease-causing factor for humans, and given that unbiased genetic screening, QTL analyses or GWAS studies are easily paired with functional studies in D. melanogaster[143,144], the latter presents as a viable alternative organism to study gene-environment interactions and the triggers that drive alcoholism, with perhaps the next step being a pursuit of the epigenetic mechanisms underlying those pathologies.

Interestingly, by using restraint stress to model strong psychological stress, Seong et al[145] found that paternal stress altered the epigenome, transcriptome, and metabolome in a dATF2 pathway-dependent manner[145]. A host of genes involved in metabolic health (amino acid metabolism, glycolysis, TCA cycle) were differentially expressed in the F1 offspring, which is consistent with the observations of similar paternal stress studies in mice[93,146]. The intergenerational effects in D. melanogaster were proposed to be caused by stress-induced up-regulation of Upd3 gene in the testes [the D. melanogaster homolog of the pro-inflammatory cytokine Interleukin-6 (IL-6)], which was confirmed by overexpression studies of Upd3 in paternal somatic cells with corresponding studies of the offspring outcomes. The overall intergenerational effects were proposed to be mediated by stress-induced increases in Upd3 that causes abnormal phosphorylation of dATF-2 in D. melanogaster germ cells, resulting in decreased H3K9me2 repressive marks that are inherited by the F1 offspring to ultimately disrupt heterochromatin assembly and gene transcription. In humans, it remains to be clarified whether IL-6 (or other pro-inflammatory cytokines) correlates with sperm DNA damage[147,148]. However, it is well-established that inflammation has a significant role in the pathogenesis of various neuropsychiatric conditions including MDD[149-151] and SZ[152,153]. It would be interesting to elucidate the relationship of SNPs and risk gene loci with H3K9me2 repressive marks, and its contribution to the development of those conditions especially in familial cases. Additionally, given initial evidence suggesting that traumatic stress has long-term epigenetic consequences including altering the DNA methylation patterns of genes relevant to HPA axis function and the immune (inflammation) response[154], future D. melanogaster studies should also focus on DNA methylation as a key epigenetic mechanism mediating the transgenerational inheritance of stress-induced pathologies.

Methylphenidate (Ritalin) is a frontline prescription psychostimulant for the treatment of attention deficit hyperactivity disorder (ADHD) and narcolepsy. The increasing frequency of prescription has been the cause for concern regarding over-prescription and overdiagnosis. Methylphenidate treatment has been reported to result in significant developmental delay to puberty with hormonal imbalance in non-human primates[155]. While the impacts on spermatogenesis or sperm health were not investigated in that study, separate work on the major metabolite of methylphenidate, ritalinic acid, has found a significant increase of human sperm motility and viability in vitro[156]. However, any effects of long-term methylphenidate treatment on pubertal growth, sperm development in vivo and the sperm epigenome are unknown presently. D. melanogaster studies have contributed tremendously to advancing our understanding of the genetics of neuropsychiatric conditions. A prime example is they have been used to identify ADHD candidate genes[157] and to determine the transcriptomic response to methylphenidate, which correlate to their locomotor responses to drug treatment[158]. The latter study also identified putative candidate genes through whole genome transcriptomic analysis that accounted for the variability in drug response. Collectively, that body of work establishes D. melanogaster as a valid organism to further probe the transgenerational effects of methylphenidate exposure on male reproductive health and progeny behaviours. The aetiology of ADHD remains poorly understood but epidemiological data indicates approximately 80% heritability for both adults and children[159,160] despite only 22% of the disease liability being linked to common gene variants[161]. Given that knockdown of D. melanogaster homologues of ADHD candidate genes produces abnormal locomotor phenotypes that are also responsive to treatment by ADHD prescription compounds[162,163], D. melanogaster would continue to serve as an ideal organism for future investigations into the epigenetic factors underlying the high degree of heritability of ADHD.

Recently, one study investigating new therapeutic options for treating frontotemporal dementia (FTLD)[164] explored the use of aminoglycosides–a class of gram-negative bacilli antibiotics that have the capacity to induce eukaryotic ribosomal readthrough of premature termination codon (PTC) sequences to yield a full-length protein. Aminoglycosides have successfully been used to treat various diseases involving PTC mutations such as cystic fibrosis[165], Duchenne muscular dystrophy[166] and Rett syndrome[167], but have yet to be employed for neuropsychiatric conditions. In using a cell culture screening assay to conduct proof-of-principle studies with non-sense mutations of progranulin associated with FTLD, Kuang et al[164] identified two aminoglycosides that rescued the expression of the progranulin. It is worth noting that one of those aminoglycosides, G418 (also known as geneticin), has previously been reported to exert transgenerational effects on maternal Polycomb levels in D. melanogaster F2 embryos that persisted into the F3 generation[168]. Importantly, G418 exposure lead to growth retardation and delay in pupation times. While the transgenerational implications of G418 would be minimal since FTLD is associated with advanced aging, we believe it is important that readers be aware of such potential risks to offspring should aminoglycosides continue to be explored as therapeutic options for conditions in a younger fertile population.

CONCLUSION

Looking towards the future, improving the prospects for neuropsychiatric patients requires the field of psychiatry to have a more comprehensive understanding of the causes of various conditions, especially regarding how basic molecular and epigenetic pathologies interact and contribute to the overall disease phenotype. A major step would be the incorporation of epigenome profiling since it is the key molecular intermediary linking genetics (susceptibility) to the environment (stress-related triggers). In highlighting the key findings of studies of C. elegans and D. melanogaster, we hope readers can come to appreciate the value of conducting basic research employing these two key non-mammalian organisms to potentially uncover novel molecular and epigenetic pathologies. Multiple stress-induced epigenetic modifications that affect the individual have significance in a variety of human neurological conditions, but further findings that progeny are also transgenerationally affected will have broader implications for health projections for future generations. At a time when stress (physical and mental) is prevalent and largely unavoidable, there is great urgency to understand the current mental health crisis and work towards new approaches for treatment and prevention. Of course, it is openly acknowledged that complex human behavioural responses and adaptations related to psychopathologies cannot be modelled in simple organisms. However, many fundamental molecular mechanisms that regulate neuronal behaviour have been conserved across phyla, and those molecular and neuronal circuitries can be interrogated in a rapid manner in simple model organisms Therefore, invertebrate research should be regarded as being tremendously beneficial and highly complementary to human and mammalian model research, and further investments should be made in this regard. An expanded combination of clinical studies, rodent models and molecular studies in model organisms provides an extremely powerful multi-tiered approach to understanding the molecular basis of psychiatric disorders. Focusing on the epigenetic pathologies associated with neuropsychiatric conditions will undoubtedly lead to the development of novel approaches for treatment.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Psychiatry

Country/Territory of origin: Australia

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Pethe P S-Editor: Fan JR L-Editor: A P-Editor: Ma YJ

References
1.  Eberle C, Kirchner MF, Herden R, Stichling S. Paternal metabolic and cardiovascular programming of their offspring: A systematic scoping review. PLoS One. 2020;15:e0244826.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 35]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
2.  Pereira SC, Crisóstomo L, Sousa M, Oliveira PF, Alves MG. Metabolic diseases affect male reproduction and induce signatures in gametes that may compromise the offspring health. Environ Epigenet. 2020;6:dvaa019.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
3.  Bowers ME, Yehuda R. Intergenerational Transmission of Stress in Humans. Neuropsychopharmacology. 2016;41:232-244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 264]  [Cited by in F6Publishing: 242]  [Article Influence: 30.3]  [Reference Citation Analysis (0)]
4.  Jawaid A, Roszkowski M, Mansuy IM. Transgenerational Epigenetics of Traumatic Stress. Prog Mol Biol Transl Sci. 2018;158:273-298.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 41]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
5.  Klengel T, Dias BG, Ressler KJ. Models of Intergenerational and Transgenerational Transmission of Risk for Psychopathology in Mice. Neuropsychopharmacology. 2016;41:219-231.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 75]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
6.  Rompala GR, Homanics GE. Intergenerational Effects of Alcohol: A Review of Paternal Preconception Ethanol Exposure Studies and Epigenetic Mechanisms in the Male Germline. Alcohol Clin Exp Res. 2019;43:1032-1045.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 34]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
7.  Cook DE, Zdraljevic S, Tanny RE, Seo B, Riccardi DD, Noble LM, Rockman MV, Alkema MJ, Braendle C, Kammenga JE, Wang J, Kruglyak L, Félix MA, Lee J, Andersen EC. The Genetic Basis of Natural Variation in Caenorhabditis elegans Telomere Length. Genetics. 2016;204:371-383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 72]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
8.  Maklakov AA, Carlsson H, Denbaum P, Lind MI, Mautz B, Hinas A, Immler S. Antagonistically pleiotropic allele increases lifespan and late-life reproduction at the cost of early-life reproduction and individual fitness. Proc Biol Sci. 2017;284.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
9.  Das S, Ooi FK, Cruz Corchado J, Fuller LC, Weiner JA, Prahlad V. Serotonin signaling by maternal neurons upon stress ensures progeny survival. Elife. 2020;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 26]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
10.  Kishimoto S, Uno M, Okabe E, Nono M, Nishida E. Environmental stresses induce transgenerationally inheritable survival advantages via germline-to-soma communication in Caenorhabditis elegans. Nat Commun. 2017;8:14031.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 100]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
11.  Rechavi O, Houri-Ze'evi L, Anava S, Goh WSS, Kerk SY, Hannon GJ, Hobert O. Starvation-induced transgenerational inheritance of small RNAs in C. elegans. Cell. 2014;158:277-287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 425]  [Cited by in F6Publishing: 317]  [Article Influence: 31.7]  [Reference Citation Analysis (0)]
12.  Bale TL. Epigenetic and transgenerational reprogramming of brain development. Nat Rev Neurosci. 2015;16:332-344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 316]  [Cited by in F6Publishing: 322]  [Article Influence: 35.8]  [Reference Citation Analysis (0)]
13.  Müller D, Grevet EH, da Silva BS, Charão MF, Rovaris DL, Bau CHD. The neuroendocrine modulation of global DNA methylation in neuropsychiatric disorders. Mol Psychiatry. 2021;26:66-69.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 10]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
14.  Saxena R, Babadi M, Namvarhaghighi H, Roullet FI. Role of environmental factors and epigenetics in autism spectrum disorders. Prog Mol Biol Transl Sci. 2020;173:35-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 12]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
15.  Tsankova N, Renthal W, Kumar A, Nestler EJ. Epigenetic regulation in psychiatric disorders. Nat Rev Neurosci. 2007;8:355-367.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 995]  [Cited by in F6Publishing: 898]  [Article Influence: 52.8]  [Reference Citation Analysis (0)]
16.  Yao B, Christian KM, He C, Jin P, Ming GL, Song H. Epigenetic mechanisms in neurogenesis. Nat Rev Neurosci. 2016;17:537-549.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 212]  [Cited by in F6Publishing: 242]  [Article Influence: 30.3]  [Reference Citation Analysis (0)]
17.  Nagy C, Turecki G. Transgenerational epigenetic inheritance: an open discussion. Epigenomics. 2015;7:781-790.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 50]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
18.  Brass KE, Herndon N, Gardner SA, Grindstaff JL, Campbell P. Intergenerational effects of paternal predator cue exposure on behavior, stress reactivity, and neural gene expression. Horm Behav. 2020;124:104806.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
19.  Cuarenta A, Kigar SL, Henion IC, Chang L, Bakshi VP, Auger AP. Early life stress during the neonatal period alters social play and Line1 during the juvenile stage of development. Sci Rep. 2021;11:3549.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 11]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
20.  Fennell KA, Busby RGG, Li S, Bodden C, Stanger SJ, Nixon B, Short AK, Hannan AJ, Pang TY. Limitations to intergenerational inheritance: subchronic paternal stress preconception does not influence offspring anxiety. Sci Rep. 2020;10:16050.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
21.  Short AK, Fennell KA, Perreau VM, Fox A, O'Bryan MK, Kim JH, Bredy TW, Pang TY, Hannan AJ. Elevated paternal glucocorticoid exposure alters the small noncoding RNA profile in sperm and modifies anxiety and depressive phenotypes in the offspring. Transl Psychiatry. 2016;6:e837.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 136]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
22.  Gapp K, Jawaid A, Sarkies P, Bohacek J, Pelczar P, Prados J, Farinelli L, Miska E, Mansuy IM. Implication of sperm RNAs in transgenerational inheritance of the effects of early trauma in mice. Nat Neurosci. 2014;17:667-669.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 806]  [Cited by in F6Publishing: 754]  [Article Influence: 75.4]  [Reference Citation Analysis (0)]
23.  Rodgers AB, Morgan CP, Bronson SL, Revello S, Bale TL. Paternal stress exposure alters sperm microRNA content and reprograms offspring HPA stress axis regulation. J Neurosci. 2013;33:9003-9012.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 527]  [Cited by in F6Publishing: 498]  [Article Influence: 45.3]  [Reference Citation Analysis (0)]
24.  Rodgers AB, Morgan CP, Leu NA, Bale TL. Transgenerational epigenetic programming via sperm microRNA recapitulates effects of paternal stress. Proc Natl Acad Sci U S A. 2015;112:13699-13704.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 448]  [Cited by in F6Publishing: 455]  [Article Influence: 50.6]  [Reference Citation Analysis (0)]
25.  Sullivan PF, Kendler KS, Neale MC. Schizophrenia as a complex trait: evidence from a meta-analysis of twin studies. Arch Gen Psychiatry. 2003;60:1187-1192.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1536]  [Cited by in F6Publishing: 1494]  [Article Influence: 74.7]  [Reference Citation Analysis (0)]
26.  Lichtenstein P, Yip BH, Björk C, Pawitan Y, Cannon TD, Sullivan PF, Hultman CM. Common genetic determinants of schizophrenia and bipolar disorder in Swedish families: a population-based study. Lancet. 2009;373:234-239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1472]  [Cited by in F6Publishing: 1404]  [Article Influence: 93.6]  [Reference Citation Analysis (0)]
27.  Wray NR, Gottesman II. Using summary data from the danish national registers to estimate heritabilities for schizophrenia, bipolar disorder, and major depressive disorder. Front Genet. 2012;3:118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 137]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
28.  Pettersson E, Lichtenstein P, Larsson H, Song J; Attention Deficit/Hyperactivity Disorder Working Group of the iPSYCH-Broad-PGC Consortium, Autism Spectrum Disorder Working Group of the iPSYCH-Broad-PGC Consortium, Bipolar Disorder Working Group of the PGC, Eating Disorder Working Group of the PGC, Major Depressive Disorder Working Group of the PGC, Obsessive Compulsive Disorders and Tourette Syndrome Working Group of the PGC, Schizophrenia CLOZUK, Substance Use Disorder Working Group of the PGC, Agrawal A, Børglum AD, Bulik CM, Daly MJ, Davis LK, Demontis D, Edenberg HJ, Grove J, Gelernter J, Neale BM, Pardiñas AF, Stahl E, Walters JTR, Walters R, Sullivan PF, Posthuma D, Polderman TJC. Genetic influences on eight psychiatric disorders based on family data of 4 408 646 full and half-siblings, and genetic data of 333 748 cases and controls. Psychol Med. 2019;49:1166-1173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 79]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
29.  Gupta A, Chauhan NR, Chowdhury D, Singh A, Meena RC, Chakrabarti A, Singh SB. Heat stress modulated gastrointestinal barrier dysfunction: role of tight junctions and heat shock proteins. Scand J Gastroenterol. 2017;52:1315-1319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
30.  Das R, Sailo L, Verma N, Bharti P, Saikia J, Imtiwati, Kumar R. Impact of heat stress on health and performance of dairy animals: A review. Vet World. 2016;9:260-268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 227]  [Article Influence: 28.4]  [Reference Citation Analysis (0)]
31.  Iossa G, Maury C, Fletcher RM, Eady PE. Temperature-induced developmental plasticity in Plodia interpunctella: Reproductive behaviour and sperm length. J Evol Biol. 2019;32:675-682.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 5]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
32.  Miwa Y, Koganezawa M, Yamamoto D. Antennae sense heat stress to inhibit mating and promote escaping in Drosophila females. J Neurogenet. 2018;32:353-363.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 4]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
33.  Johnson JS, Abuajamieh M, Victoria Sanz Fernandez M, Seibert JT, Stoakes SK, Keating AF, Ross JW, Selsby JT, Rhoads RP, Baumgard LH. The impact of in utero heat stress and nutrient restriction on progeny body composition. J Therm Biol. 2015;53:143-150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
34.  Wang J, Liu X, Dong M, Sun X, Xiao J, Zeng W, Hu J, Li X, Guo L, Rong Z, He G, Sun J, Ning D, Chen D, Zhang Y, Zhang B, Ma W, Liu T. Associations of maternal ambient temperature exposures during pregnancy with the placental weight, volume and PFR: A birth cohort study in Guangzhou, China. Environ Int. 2020;139:105682.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
35.  Cianconi P, Betrò S, Janiri L. The Impact of Climate Change on Mental Health: A Systematic Descriptive Review. Front Psychiatry. 2020;11:74.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 421]  [Cited by in F6Publishing: 233]  [Article Influence: 58.3]  [Reference Citation Analysis (0)]
36.  Hein NT, Ciampitti IA, Jagadish SVK. Bottlenecks and opportunities in field-based high-throughput phenotyping for heat and drought stress. J Exp Bot. 2021;72:5102-5116.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
37.  Waqas MA, Wang X, Zafar SA, Noor MA, Hussain HA, Azher Nawaz M, Farooq M. Thermal Stresses in Maize: Effects and Management Strategies. Plants (Basel). 2021;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
38.  Morrell JM. Heat stress and bull fertility. Theriogenology. 2020;153:62-67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 45]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
39.  van Wettere WHEJ, Kind KL, Gatford KL, Swinbourne AM, Leu ST, Hayman PT, Kelly JM, Weaver AC, Kleemann DO, Walker SK. Review of the impact of heat stress on reproductive performance of sheep. J Anim Sci Biotechnol. 2021;12:26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 44]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
40.  Pembrey M, Saffery R, Bygren LO; Network in Epigenetic Epidemiology;  Network in Epigenetic Epidemiology. Human transgenerational responses to early-life experience: potential impact on development, health and biomedical research. J Med Genet. 2014;51:563-572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 204]  [Cited by in F6Publishing: 190]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
41.  Zhang S, Li F, Zhou T, Wang G, Li Z. Caenorhabditis elegans as a Useful Model for Studying Aging Mutations. Front Endocrinol (Lausanne). 2020;11:554994.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 67]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
42.  Briese M, Esmaeili B, Fraboulet S, Burt EC, Christodoulou S, Towers PR, Davies KE, Sattelle DB. Deletion of smn-1, the Caenorhabditis elegans ortholog of the spinal muscular atrophy gene, results in locomotor dysfunction and reduced lifespan. Hum Mol Genet. 2009;18:97-104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 73]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
43.  Klosin A, Casas E, Hidalgo-Carcedo C, Vavouri T, Lehner B. Transgenerational transmission of environmental information in C. elegans. Science. 2017;356:320-323.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 255]  [Cited by in F6Publishing: 254]  [Article Influence: 36.3]  [Reference Citation Analysis (0)]
44.  Darby MM, Sabunciyan S. Repetitive elements and epigenetic marks in behavior and psychiatric disease. Adv Genet. 2014;86:185-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
45.  Deb-Rinker P, Klempan TA, O'Reilly RL, Torrey EF, Singh SM. Molecular characterization of a MSRV-like sequence identified by RDA from monozygotic twin pairs discordant for schizophrenia. Genomics. 1999;61:133-144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 50]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
46.  Karlsson H, Bachmann S, Schröder J, McArthur J, Torrey EF, Yolken RH. Retroviral RNA identified in the cerebrospinal fluids and brains of individuals with schizophrenia. Proc Natl Acad Sci U S A. 2001;98:4634-4639.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 263]  [Cited by in F6Publishing: 274]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
47.  Karlsson H, Schröder J, Bachmann S, Bottmer C, Yolken RH. HERV-W-related RNA detected in plasma from individuals with recent-onset schizophrenia or schizoaffective disorder. Mol Psychiatry. 2004;9:12-13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 74]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
48.  Yao Y, Schröder J, Nellåker C, Bottmer C, Bachmann S, Yolken RH, Karlsson H. Elevated levels of human endogenous retrovirus-W transcripts in blood cells from patients with first episode schizophrenia. Genes Brain Behav. 2008;7:103-112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 38]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
49.  Lee TW, David HS, Engstrom AK, Carpenter BS, Katz DJ. Repressive H3K9me2 protects lifespan against the transgenerational burden of COMPASS activity in C. elegans. Elife. 2019;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 22]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
50.  Chase KA, Feiner B, Ramaker MJ, Hu E, Rosen C, Sharma RP. Examining the effects of the histone methyltransferase inhibitor BIX-01294 on histone modifications and gene expression in both a clinical population and mouse models. PLoS One. 2019;14:e0216463.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
51.  Kim TK, Lee JE, Kim JE, Park JY, Choi J, Kim H, Lee EH, Han PL. G9a-Mediated Regulation of OXT and AVP Expression in the Basolateral Amygdala Mediates Stress-Induced Lasting Behavioral Depression and Its Reversal by Exercise. Mol Neurobiol. 2016;53:2843-2856.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 31]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
52.  Heller EA, Hamilton PJ, Burek DD, Lombroso SI, Peña CJ, Neve RL, Nestler EJ. Targeted Epigenetic Remodeling of the Cdk5 Gene in Nucleus Accumbens Regulates Cocaine- and Stress-Evoked Behavior. J Neurosci. 2016;36:4690-4697.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 72]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
53.  Martín-Hernández D, Caso JR, Javier Meana J, Callado LF, Madrigal JLM, García-Bueno B, Leza JC. Intracellular inflammatory and antioxidant pathways in postmortem frontal cortex of subjects with major depression: effect of antidepressants. J Neuroinflammation. 2018;15:251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 52]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
54.  Pasquali MA, Harlow BL, Soares CN, Otto MW, Cohen LS, Minuzzi L, Gelain DP, Moreira JCF, Frey BN. A longitudinal study of neurotrophic, oxidative, and inflammatory markers in first-onset depression in midlife women. Eur Arch Psychiatry Clin Neurosci. 2018;268:771-781.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
55.  Sural S, Liang CY, Wang FY, Ching TT, Hsu AL. HSB-1/HSF-1 pathway modulates histone H4 in mitochondria to control mtDNA transcription and longevity. Sci Adv. 2020;6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
56.  Liao PC, Lin HY, Yuh CH, Yu LK, Wang HD. The effect of neuronal expression of heat shock proteins 26 and 27 on lifespan, neurodegeneration, and apoptosis in Drosophila. Biochem Biophys Res Commun. 2008;376:637-641.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 35]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
57.  Allen J, Caruncho HJ, Kalynchuk LE. Severe life stress, mitochondrial dysfunction, and depressive behavior: A pathophysiological and therapeutic perspective. Mitochondrion. 2021;56:111-117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 23]  [Article Influence: 5.8]  [Reference Citation Analysis (1)]
58.  Shao A, Lin D, Wang L, Tu S, Lenahan C, Zhang J. Oxidative Stress at the Crossroads of Aging, Stroke and Depression. Aging Dis. 2020;11:1537-1566.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 56]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
59.  Weger M, Alpern D, Cherix A, Ghosal S, Grosse J, Russeil J, Gruetter R, de Kloet ER, Deplancke B, Sandi C. Mitochondrial gene signature in the prefrontal cortex for differential susceptibility to chronic stress. Sci Rep. 2020;10:18308.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 32]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
60.  Tao Q, Miao Y, Li H, Yuan X, Huang X, Wang Y, Andreassen OA, Fan X, Yang Y, Song X. Insulin Resistance and Oxidative Stress: In Relation to Cognitive Function and Psychopathology in Drug-Naïve, First-Episode Drug-Free Schizophrenia. Front Psychiatry. 2020;11:537280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
61.  Morris G, Walker AJ, Walder K, Berk M, Marx W, Carvalho AF, Maes M, Puri BK. Increasing Nrf2 Activity as a Treatment Approach in Neuropsychiatry. Mol Neurobiol. 2021;58:2158-2182.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 21]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
62.  Fan C, Long Y, Wang L, Liu X, Liu Z, Lan T, Li Y, Yu SY. N-Acetylcysteine Rescues Hippocampal Oxidative Stress-Induced Neuronal Injury via Suppression of p38/JNK Signaling in Depressed Rats. Front Cell Neurosci. 2020;14:554613.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
63.  Cruceanu C, Alda M, Nagy C, Freemantle E, Rouleau GA, Turecki G. H3K4 tri-methylation in synapsin genes leads to different expression patterns in bipolar disorder and major depression. Int J Neuropsychopharmacol. 2013;16:289-299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 60]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
64.  Girdhar K, Hoffman GE, Jiang Y, Brown L, Kundakovic M, Hauberg ME, Francoeur NJ, Wang YC, Shah H, Kavanagh DH, Zharovsky E, Jacobov R, Wiseman JR, Park R, Johnson JS, Kassim BS, Sloofman L, Mattei E, Weng Z, Sieberts SK, Peters MA, Harris BT, Lipska BK, Sklar P, Roussos P, Akbarian S. Cell-specific histone modification maps in the human frontal lobe link schizophrenia risk to the neuronal epigenome. Nat Neurosci. 2018;21:1126-1136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 73]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
65.  Niu HM, Yang P, Chen HH, Hao RH, Dong SS, Yao S, Chen XF, Yan H, Zhang YJ, Chen YX, Jiang F, Yang TL, Guo Y. Comprehensive functional annotation of susceptibility SNPs prioritized 10 genes for schizophrenia. Transl Psychiatry. 2019;9:56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
66.  Benayoun BA, Pollina EA, Ucar D, Mahmoudi S, Karra K, Wong ED, Devarajan K, Daugherty AC, Kundaje AB, Mancini E, Hitz BC, Gupta R, Rando TA, Baker JC, Snyder MP, Cherry JM, Brunet A. H3K4me3 breadth is linked to cell identity and transcriptional consistency. Cell. 2014;158:673-688.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 368]  [Cited by in F6Publishing: 326]  [Article Influence: 32.6]  [Reference Citation Analysis (0)]
67.  Liu X, Wang C, Liu W, Li J, Li C, Kou X, Chen J, Zhao Y, Gao H, Wang H, Zhang Y, Gao Y, Gao S. Distinct features of H3K4me3 and H3K27me3 chromatin domains in pre-implantation embryos. Nature. 2016;537:558-562.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 404]  [Cited by in F6Publishing: 410]  [Article Influence: 51.3]  [Reference Citation Analysis (0)]
68.  Aguilar-Valles A, Vaissière T, Griggs EM, Mikaelsson MA, Takács IF, Young EJ, Rumbaugh G, Miller CA. Methamphetamine-associated memory is regulated by a writer and an eraser of permissive histone methylation. Biol Psychiatry. 2014;76:57-65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 65]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
69.  Huang C, Phillips MR, Zhang Y, Zhang J, Shi Q, Song Z, Ding Z, Pang S, Martorell R. Malnutrition in early life and adult mental health: evidence from a natural experiment. Soc Sci Med. 2013;97:259-266.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 55]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
70.  Mhillaj E, Morgese MG, Trabace L. Early life and oxidative stress in psychiatric disorders: what can we learn from animal models? Curr Pharm Des. 2015;21:1396-1403.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
71.  Perry BI, Stochl J, Upthegrove R, Zammit S, Wareham N, Langenberg C, Winpenny E, Dunger D, Jones PB, Khandaker GM. Longitudinal Trends in Childhood Insulin Levels and Body Mass Index and Associations With Risks of Psychosis and Depression in Young Adults. JAMA Psychiatry. 2021;78:416-425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 33]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
72.  Zhao K, Zhou S, Shi X, Chen J, Zhang Y, Fan K, Zhang X, Wang W, Tang W. Potential metabolic monitoring indicators of suicide attempts in first episode and drug naive young patients with major depressive disorder: a cross-sectional study. BMC Psychiatry. 2020;20:387.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 12]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
73.  Harris D, Haboubi N. Malnutrition screening in the elderly population. J R Soc Med. 2005;98:411-414.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 55]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
74.  Koster A, van Gool CH, Kempen GI, Penninx BW, Lee JS, Rubin SM, Tylavsky FA, Yaffe K, Newman AB, Harris TB, Pahor M, Ayonayon HN, van Eijk JT, Kritchevsky SB; Health ABC Study. Late-life depressed mood and weight change contribute to the risk of each other. Am J Geriatr Psychiatry. 2010;18:236-244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
75.  Igwe O, Sone M, Matveychuk D, Baker GB, Dursun SM. A review of effects of calorie restriction and fasting with potential relevance to depression. Prog Neuropsychopharmacol Biol Psychiatry. 2020;110206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 16]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
76.  Cao H, Meng Y, Li X, Ma X, Deng W, Guo W, Li T. The metabolic effects of antipsychotics in the early stage of treatment in first-episode patients with schizophrenia: A real-world study in a naturalistic setting. J Psychiatr Res. 2020;129:265-271.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
77.  Garcia-Segura L, Abreu-Goodger C, Hernandez-Mendoza A, Dimitrova Dinkova TD, Padilla-Noriega L, Perez-Andrade ME, Miranda-Rios J. High-Throughput Profiling of Caenorhabditis elegans Starvation-Responsive microRNAs. PLoS One. 2015;10:e0142262.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
78.  Martin CG, Kim H, Yun S, Livingston W, Fetta J, Mysliwiec V, Baxter T, Gill JM. Circulating miRNA associated with posttraumatic stress disorder in a cohort of military combat veterans. Psychiatry Res. 2017;251:261-265.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 29]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
79.  Francati V, Vermetten E, Bremner JD. Functional neuroimaging studies in posttraumatic stress disorder: review of current methods and findings. Depress Anxiety. 2007;24:202-218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 207]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
80.  Hughes KC, Shin LM. Functional neuroimaging studies of post-traumatic stress disorder. Expert Rev Neurother. 2011;11:275-285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 183]  [Cited by in F6Publishing: 203]  [Article Influence: 15.6]  [Reference Citation Analysis (0)]
81.  Cunningham AM, Walker DM, Ramakrishnan A, Doyle MA, Bagot RC, Cates HM, Peña CJ, Issler O, Lardner C, Browne C, Russo SJ, Shen L, Nestler EJ. Sperm transcriptional state associated with paternal transmission of stress phenotypes. J Neurosci. 2021;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 11]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
82.  Dietz DM, Laplant Q, Watts EL, Hodes GE, Russo SJ, Feng J, Oosting RS, Vialou V, Nestler EJ. Paternal transmission of stress-induced pathologies. Biol Psychiatry. 2011;70:408-414.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 252]  [Cited by in F6Publishing: 225]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
83.  Hauberg ME, Roussos P, Grove J, Børglum AD, Mattheisen M; Schizophrenia Working Group of the Psychiatric Genomics Consortium. Analyzing the Role of MicroRNAs in Schizophrenia in the Context of Common Genetic Risk Variants. JAMA Psychiatry. 2016;73:369-377.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 63]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
84.  Andolina D, Di Segni M, Bisicchia E, D'Alessandro F, Cestari V, Ventura A, Concepcion C, Puglisi-Allegra S, Ventura R. Effects of lack of microRNA-34 on the neural circuitry underlying the stress response and anxiety. Neuropharmacology. 2016;107:305-316.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 48]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
85.  Owen MJ, O'Donovan MC, Thapar A, Craddock N. Neurodevelopmental hypothesis of schizophrenia. Br J Psychiatry. 2011;198:173-175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 351]  [Cited by in F6Publishing: 315]  [Article Influence: 24.2]  [Reference Citation Analysis (0)]
86.  Agostini M, Tucci P, Killick R, Candi E, Sayan BS, Rivetti di Val Cervo P, Nicotera P, McKeon F, Knight RA, Mak TW, Melino G. Neuronal differentiation by TAp73 is mediated by microRNA-34a regulation of synaptic protein targets. Proc Natl Acad Sci U S A. 2011;108:21093-21098.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 156]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
87.  Marré J, Traver EC, Jose AM. Extracellular RNA is transported from one generation to the next in Caenorhabditis elegans. Proc Natl Acad Sci U S A. 2016;113:12496-12501.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 65]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
88.  Pang TYC, Short AK, Bredy TW, Hannan AJ. Transgenerational paternal transmission of acquired traits: Stress-induced modification of the sperm regulatory transcriptome and offspring phenotypes. Curr Opin Behav Sci. 2017;14:140-147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 28]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
89.  Trigg NA, Eamens AL, Nixon B. The contribution of epididymosomes to the sperm small RNA profile. Reproduction. 2019;157:R209-R223.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 53]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
90.  Chan JC, Morgan CP, Adrian Leu N, Shetty A, Cisse YM, Nugent BM, Morrison KE, Jašarević E, Huang W, Kanyuch N, Rodgers AB, Bhanu NV, Berger DS, Garcia BA, Ament S, Kane M, Neill Epperson C, Bale TL. Reproductive tract extracellular vesicles are sufficient to transmit intergenerational stress and program neurodevelopment. Nat Commun. 2020;11:1499.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 104]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
91.  Danielson KM, Rubio R, Abderazzaq F, Das S, Wang YE. High Throughput Sequencing of Extracellular RNA from Human Plasma. PLoS One. 2017;12:e0164644.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 53]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
92.  Shah R, Murthy V, Pacold M, Danielson K, Tanriverdi K, Larson MG, Hanspers K, Pico A, Mick E, Reis J, de Ferranti S, Freinkman E, Levy D, Hoffmann U, Osganian S, Das S, Freedman JE. Extracellular RNAs Are Associated With Insulin Resistance and Metabolic Phenotypes. Diabetes Care. 2017;40:546-553.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 67]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
93.  van Steenwyk G, Gapp K, Jawaid A, Germain PL, Manuella F, Tanwar DK, Zamboni N, Gaur N, Efimova A, Thumfart KM, Miska EA, Mansuy IM. Involvement of circulating factors in the transmission of paternal experiences through the germline. EMBO J. 2020;39:e104579.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
94.  Yan Z, Zhou Z, Wu Q, Chen ZB, Koo EH, Zhong S. Presymptomatic Increase of an Extracellular RNA in Blood Plasma Associates with the Development of Alzheimer's Disease. Curr Biol. 2020;30:1771-1782.e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 22]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
95.  Lloyd TE, Taylor JP. Flightless flies: Drosophila models of neuromuscular disease. Ann N Y Acad Sci. 2010;1184:e1-20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 108]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
96.  Mackay TF, Richards S, Stone EA, Barbadilla A, Ayroles JF, Zhu D, Casillas S, Han Y, Magwire MM, Cridland JM, Richardson MF, Anholt RR, Barrón M, Bess C, Blankenburg KP, Carbone MA, Castellano D, Chaboub L, Duncan L, Harris Z, Javaid M, Jayaseelan JC, Jhangiani SN, Jordan KW, Lara F, Lawrence F, Lee SL, Librado P, Linheiro RS, Lyman RF, Mackey AJ, Munidasa M, Muzny DM, Nazareth L, Newsham I, Perales L, Pu LL, Qu C, Ràmia M, Reid JG, Rollmann SM, Rozas J, Saada N, Turlapati L, Worley KC, Wu YQ, Yamamoto A, Zhu Y, Bergman CM, Thornton KR, Mittelman D, Gibbs RA. The Drosophila melanogaster Genetic Reference Panel. Nature. 2012;482:173-178.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1231]  [Cited by in F6Publishing: 1102]  [Article Influence: 91.8]  [Reference Citation Analysis (0)]
97.  Engel GL, Taber K, Vinton E, Crocker AJ. Studying alcohol use disorder using Drosophila melanogaster in the era of 'Big Data'. Behav Brain Funct. 2019;15:7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 17]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
98.  Narayanan AS, Rothenfluh A. I Believe I Can Fly! Neuropsychopharmacology. 2016;41:1439-1446.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
99.  van Alphen B, van Swinderen B. Drosophila strategies to study psychiatric disorders. Brain Res Bull. 2013;92:1-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 53]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
100.  Wangler MF, Hu Y, Shulman JM. Drosophila and genome-wide association studies: a review and resource for the functional dissection of human complex traits. Dis Model Mech. 2017;10:77-88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 33]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
101.  Belfer SJ, Bashaw AG, Perlis ML, Kayser MS. A Drosophila model of sleep restriction therapy for insomnia. Mol Psychiatry. 2021;26:492-507.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
102.  Klepsatel P, Gáliková M, Xu Y, Kühnlein RP. Thermal stress depletes energy reserves in Drosophila. Sci Rep. 2016;6:33667.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 63]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
103.  Klepsatel P, Wildridge D, Gáliková M. Temperature induces changes in Drosophila energy stores. Sci Rep. 2019;9:5239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 29]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
104.  Wallace EW, Kear-Scott JL, Pilipenko EV, Schwartz MH, Laskowski PR, Rojek AE, Katanski CD, Riback JA, Dion MF, Franks AM, Airoldi EM, Pan T, Budnik BA, Drummond DA. Reversible, Specific, Active Aggregates of Endogenous Proteins Assemble upon Heat Stress. Cell. 2015;162:1286-1298.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 311]  [Cited by in F6Publishing: 318]  [Article Influence: 35.3]  [Reference Citation Analysis (0)]
105.  Richter K, Haslbeck M, Buchner J. The heat shock response: life on the verge of death. Mol Cell. 2010;40:253-266.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1205]  [Cited by in F6Publishing: 1231]  [Article Influence: 87.9]  [Reference Citation Analysis (0)]
106.  Guertin MJ, Petesch SJ, Zobeck KL, Min IM, Lis JT. Drosophila heat shock system as a general model to investigate transcriptional regulation. Cold Spring Harb Symp Quant Biol. 2010;75:1-9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 60]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
107.  Teves SS, Henikoff S. The heat shock response: A case study of chromatin dynamics in gene regulation. Biochem Cell Biol. 2013;91:42-48.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
108.  Kristensen TN, Sørensen JG, Loeschcke V. Mild heat stress at a young age in Drosophila melanogaster leads to increased Hsp70 synthesis after stress exposure later in life. J Genet. 2003;82:89-94.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 29]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
109.  Sejerkilde M, Sørensen JG, Loeschcke V. Effects of cold- and heat hardening on thermal resistance in Drosophila melanogaster. J Insect Physiol. 2003;49:719-726.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 67]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
110.  Folk DG, Zwollo P, Rand DM, Gilchrist GW. Selection on knockdown performance in Drosophila melanogaster impacts thermotolerance and heat-shock response differently in females and males. J Exp Biol. 2006;209:3964-3973.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 49]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
111.  Krebs RA, Thompson KA. Direct and correlated effects of selection on flight after exposure to thermal stress in Drosophila melanogaster. Genetica. 2006;128:217-225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
112.  Ørsted M, Hoffmann AA, Rohde PD, Sørensen P, Kristensen TN. Strong impact of thermal environment on the quantitative genetic basis of a key stress tolerance trait. Heredity (Edinb). 2019;122:315-325.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
113.  Mackenzie SM, Brooker MR, Gill TR, Cox GB, Howells AJ, Ewart GD. Mutations in the white gene of Drosophila melanogaster affecting ABC transporters that determine eye colouration. Biochim Biophys Acta. 1999;1419:173-185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in F6Publishing: 125]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
114.  Bantignies F, Grimaud C, Lavrov S, Gabut M, Cavalli G. Inheritance of Polycomb-dependent chromosomal interactions in Drosophila. Genes Dev. 2003;17:2406-2420.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 199]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
115.  Campos EI, Stafford JM, Reinberg D. Epigenetic inheritance: histone bookmarks across generations. Trends Cell Biol. 2014;24:664-674.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 100]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
116.  Francis NJ. Mechanisms of epigenetic inheritance: copying of polycomb repressed chromatin. Cell Cycle. 2009;8:3521-3526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
117.  Lanzuolo C, Lo Sardo F, Orlando V. Concerted epigenetic signatures inheritance at PcG targets through replication. Cell Cycle. 2012;11:1296-1300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
118.  Brown JL, Sun MA, Kassis JA. Global changes of H3K27me3 domains and Polycomb group protein distribution in the absence of recruiters Spps or Pho. Proc Natl Acad Sci U S A. 2018;115:E1839-E1848.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 27]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
119.  Bredy TW, Sun YE, Kobor MS. How the epigenome contributes to the development of psychiatric disorders. Dev Psychobiol. 2010;52:331-342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
120.  Ohm JE, McGarvey KM, Yu X, Cheng L, Schuebel KE, Cope L, Mohammad HP, Chen W, Daniel VC, Yu W, Berman DM, Jenuwein T, Pruitt K, Sharkis SJ, Watkins DN, Herman JG, Baylin SB. A stem cell-like chromatin pattern may predispose tumor suppressor genes to DNA hypermethylation and heritable silencing. Nat Genet. 2007;39:237-242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 837]  [Cited by in F6Publishing: 809]  [Article Influence: 47.6]  [Reference Citation Analysis (0)]
121.  Seong KH, Li D, Shimizu H, Nakamura R, Ishii S. Inheritance of stress-induced, ATF-2-dependent epigenetic change. Cell. 2011;145:1049-1061.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 230]  [Cited by in F6Publishing: 220]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]
122.  Laifenfeld D, Karry R, Grauer E, Klein E, Ben-Shachar D. ATF2, a member of the CREB/ATF family of transcription factors, in chronic stress and consequent to antidepressant treatment: animal models and human post-mortem brains. Neuropsychopharmacology. 2004;29:589-597.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
123.  Sharma RP, Gavin DP, Chase KA. Heterochromatin as an incubator for pathology and treatment non-response: implication for neuropsychiatric illness. Pharmacogenomics J. 2012;12:361-367.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 16]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
124.  Zhu Y, Sun D, Jakovcevski M, Jiang Y. Epigenetic mechanism of SETDB1 in brain: implications for neuropsychiatric disorders. Transl Psychiatry. 2020;10:115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 16]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
125.  Feiner B, Chase KA, Melbourne JK, Rosen C, Sharma RP. Risperidone effects on heterochromatin: the role of kinase signaling. Clin Exp Immunol. 2019;196:67-75.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
126.  de Vanssay A, Bougé AL, Boivin A, Hermant C, Teysset L, Delmarre V, Antoniewski C, Ronsseray S. Paramutation in Drosophila linked to emergence of a piRNA-producing locus. Nature. 2012;490:112-115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 157]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
127.  Casier K, Delmarre V, Gueguen N, Hermant C, Viodé E, Vaury C, Ronsseray S, Brasset E, Teysset L, Boivin A. Environmentally-induced epigenetic conversion of a piRNA cluster. Elife. 2019;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
128.  Qiu W, Guo X, Lin X, Yang Q, Zhang W, Zhang Y, Zuo L, Zhu Y, Li CR, Ma C, Luo X. Transcriptome-wide piRNA profiling in human brains of Alzheimer's disease. Neurobiol Aging. 2017;57:170-177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 56]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
129.  Jain G, Stuendl A, Rao P, Berulava T, Pena Centeno T, Kaurani L, Burkhardt S, Delalle I, Kornhuber J, Hüll M, Maier W, Peters O, Esselmann H, Schulte C, Deuschle C, Synofzik M, Wiltfang J, Mollenhauer B, Maetzler W, Schneider A, Fischer A. A combined miRNA-piRNA signature to detect Alzheimer's disease. Transl Psychiatry. 2019;9:250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 63]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
130.  Heggeli KA, Crook J, Thomas C, Graff-Radford N. Maternal transmission of Alzheimer disease. Alzheimer Dis Assoc Disord. 2012;26:364-366.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 10]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
131.  Blumenstiel JP, Hartl DL. Evidence for maternally transmitted small interfering RNA in the repression of transposition in Drosophila virilis. Proc Natl Acad Sci U S A. 2005;102:15965-15970.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 100]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
132.  Brennecke J, Malone CD, Aravin AA, Sachidanandam R, Stark A, Hannon GJ. An epigenetic role for maternally inherited piRNAs in transposon silencing. Science. 2008;322:1387-1392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 592]  [Cited by in F6Publishing: 532]  [Article Influence: 33.3]  [Reference Citation Analysis (0)]
133.  Grentzinger T, Armenise C, Brun C, Mugat B, Serrano V, Pelisson A, Chambeyron S. piRNA-mediated transgenerational inheritance of an acquired trait. Genome Res. 2012;22:1877-1888.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 127]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
134.  Khurana JS, Wang J, Xu J, Koppetsch BS, Thomson TC, Nowosielska A, Li C, Zamore PD, Weng Z, Theurkauf WE. Adaptation to P element transposon invasion in Drosophila melanogaster. Cell. 2011;147:1551-1563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 190]  [Cited by in F6Publishing: 163]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
135.  Kacsoh BZ, Lynch ZR, Mortimer NT, Schlenke TA. Fruit flies medicate offspring after seeing parasites. Science. 2013;339:947-950.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 116]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
136.  Bozler J, Kacsoh BZ, Bosco G. Transgeneratonal inheritance of ethanol preference is caused by maternal NPF repression. Elife. 2019;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
137.  Mayfield RD, Lewohl JM, Dodd PR, Herlihy A, Liu J, Harris RA. Patterns of gene expression are altered in the frontal and motor cortices of human alcoholics. J Neurochem. 2002;81:802-813.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 262]  [Cited by in F6Publishing: 275]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
138.  Mottagui-Tabar S, Prince JA, Wahlestedt C, Zhu G, Goldman D, Heilig M. A novel single nucleotide polymorphism of the neuropeptide Y (NPY) gene associated with alcohol dependence. Alcohol Clin Exp Res. 2005;29:702-707.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 52]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
139.  Thorsell A, Mathé AA. Neuropeptide Y in Alcohol Addiction and Affective Disorders. Front Endocrinol (Lausanne). 2017;8:178.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 45]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
140.  Badia-Elder NE, Stewart RB, Powrozek TA, Murphy JM, Li TK. Effects of neuropeptide Y on sucrose and ethanol intake and on anxiety-like behavior in high alcohol drinking (HAD) and low alcohol drinking (LAD) rats. Alcohol Clin Exp Res. 2003;27:894-899.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 38]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
141.  Robinson SL, Marrero IM, Perez-Heydrich CA, Sepulveda-Orengo MT, Reissner KJ, Thiele TE. Medial prefrontal cortex neuropeptide Y modulates binge-like ethanol consumption in C57BL/6J mice. Neuropsychopharmacology. 2019;44:1132-1140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 19]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
142.  Schroeder JP, Overstreet DH, Hodge CW. The neuropeptide-Y Y5 receptor antagonist L-152,804 decreases alcohol self-administration in inbred alcohol-preferring (iP) rats. Alcohol. 2005;36:179-186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 30]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
143.  Chvilicek MM, Titos I, Rothenfluh A. The Neurotransmitters Involved in Drosophila Alcohol-Induced Behaviors. Front Behav Neurosci. 2020;14:607700.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
144.  Lathen DR, Merrill CB, Rothenfluh A. Flying Together: Drosophila as a Tool to Understand the Genetics of Human Alcoholism. Int J Mol Sci. 2020;21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
145.  Seong KH, Ly NH, Katou Y, Yokota N, Nakato R, Murakami S, Hirayama A, Fukuda S, Kang S, Soga T, Shirahige K, Ishii S. Paternal restraint stress affects offspring metabolism via ATF-2 dependent mechanisms in Drosophila melanogaster germ cells. Commun Biol. 2020;3:208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
146.  van Steenwyk G, Roszkowski M, Manuella F, Franklin TB, Mansuy IM. Transgenerational inheritance of behavioral and metabolic effects of paternal exposure to traumatic stress in early postnatal life: evidence in the 4th generation. Environ Epigenet. 2018;4:dvy023.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 47]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
147.  Aghazarian A, Huf W, Pflüger H, Klatte T. The association of seminal leucocytes, interleukin-6 and interleukin-8 with sperm DNA fragmentation: A prospective study. Andrologia. 2019;51:e13428.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
148.  Kopa Z, Wenzel J, Papp GK, Haidl G. Role of granulocyte elastase and interleukin-6 in the diagnosis of male genital tract inflammation. Andrologia. 2005;37:188-194.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 76]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
149.  Hodes GE, Ménard C, Russo SJ. Integrating Interleukin-6 into depression diagnosis and treatment. Neurobiol Stress. 2016;4:15-22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 164]  [Article Influence: 20.5]  [Reference Citation Analysis (0)]
150.  Kakeda S, Watanabe K, Katsuki A, Sugimoto K, Igata N, Ueda I, Igata R, Abe O, Yoshimura R, Korogi Y. Relationship between interleukin (IL)-6 and brain morphology in drug-naïve, first-episode major depressive disorder using surface-based morphometry. Sci Rep. 2018;8:10054.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 50]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
151.  Ting EY, Yang AC, Tsai SJ. Role of Interleukin-6 in Depressive Disorder. Int J Mol Sci. 2020;21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 152]  [Article Influence: 38.0]  [Reference Citation Analysis (0)]
152.  Borovcanin MM, Jovanovic I, Radosavljevic G, Pantic J, Minic Janicijevic S, Arsenijevic N, Lukic ML. Interleukin-6 in Schizophrenia-Is There a Therapeutic Relevance? Front Psychiatry. 2017;8:221.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 41]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
153.  Chase KA, Cone JJ, Rosen C, Sharma RP. The value of interleukin 6 as a peripheral diagnostic marker in schizophrenia. BMC Psychiatry. 2016;16:152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 43]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
154.  Bick J, Naumova O, Hunter S, Barbot B, Lee M, Luthar SS, Raefski A, Grigorenko EL. Childhood adversity and DNA methylation of genes involved in the hypothalamus-pituitary-adrenal axis and immune system: whole-genome and candidate-gene associations. Dev Psychopathol. 2012;24:1417-1425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 95]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
155.  Mattison DR, Plant TM, Lin HM, Chen HC, Chen JJ, Twaddle NC, Doerge D, Slikker W Jr, Patton RE, Hotchkiss CE, Callicott RJ, Schrader SM, Turner TW, Kesner JS, Vitiello B, Petibone DM, Morris SM. Pubertal delay in male nonhuman primates (Macaca mulatta) treated with methylphenidate. Proc Natl Acad Sci U S A. 2011;108:16301-16306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 29]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
156.  Harlev A, Henkel R, Samanta L, Agarwal A. Ritalinic Acid Stimulates Human Sperm Motility and Maintains Vitality In Vitro. World J Mens Health. 2020;38:61-67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
157.  Rohde PD, Madsen LS, Neumann Arvidson SM, Loeschcke V, Demontis D, Kristensen TN. Testing candidate genes for attention-deficit/hyperactivity disorder in fruit flies using a high throughput assay for complex behavior. Fly (Austin). 2016;10:25-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 7]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
158.  Rohde PD, Jensen IR, Sarup PM, Ørsted M, Demontis D, Sørensen P, Kristensen TN. Genetic Signatures of Drug Response Variability in Drosophila melanogaster. Genetics. 2019;213:633-650.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
159.  Banaschewski T, Becker K, Scherag S, Franke B, Coghill D. Molecular genetics of attention-deficit/hyperactivity disorder: an overview. Eur Child Adolesc Psychiatry. 2010;19:237-257.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 176]  [Cited by in F6Publishing: 181]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
160.  Faraone SV, Asherson P, Banaschewski T, Biederman J, Buitelaar JK, Ramos-Quiroga JA, Rohde LA, Sonuga-Barke EJ, Tannock R, Franke B. Attention-deficit/hyperactivity disorder. Nat Rev Dis Primers. 2015;1:15020.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 745]  [Cited by in F6Publishing: 774]  [Article Influence: 86.0]  [Reference Citation Analysis (0)]
161.  Demontis D, Walters RK, Martin J, Mattheisen M, Als TD, Agerbo E, Baldursson G, Belliveau R, Bybjerg-Grauholm J, Bækvad-Hansen M, Cerrato F, Chambert K, Churchhouse C, Dumont A, Eriksson N, Gandal M, Goldstein JI, Grasby KL, Grove J, Gudmundsson OO, Hansen CS, Hauberg ME, Hollegaard MV, Howrigan DP, Huang H, Maller JB, Martin AR, Martin NG, Moran J, Pallesen J, Palmer DS, Pedersen CB, Pedersen MG, Poterba T, Poulsen JB, Ripke S, Robinson EB, Satterstrom FK, Stefansson H, Stevens C, Turley P, Walters GB, Won H, Wright MJ; ADHD Working Group of the Psychiatric Genomics Consortium (PGC);  Early Lifecourse & Genetic Epidemiology (EAGLE) Consortium;  23andMe Research Team, Andreassen OA, Asherson P, Burton CL, Boomsma DI, Cormand B, Dalsgaard S, Franke B, Gelernter J, Geschwind D, Hakonarson H, Haavik J, Kranzler HR, Kuntsi J, Langley K, Lesch KP, Middeldorp C, Reif A, Rohde LA, Roussos P, Schachar R, Sklar P, Sonuga-Barke EJS, Sullivan PF, Thapar A, Tung JY, Waldman ID, Medland SE, Stefansson K, Nordentoft M, Hougaard DM, Werge T, Mors O, Mortensen PB, Daly MJ, Faraone SV, Børglum AD, Neale BM. Discovery of the first genome-wide significant risk loci for attention deficit/hyperactivity disorder. Nat Genet. 2019;51:63-75.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1204]  [Cited by in F6Publishing: 1135]  [Article Influence: 227.0]  [Reference Citation Analysis (0)]
162.  van der Voet M, Harich B, Franke B, Schenck A. ADHD-associated dopamine transporter, latrophilin and neurofibromin share a dopamine-related locomotor signature in Drosophila. Mol Psychiatry. 2016;21:565-573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 73]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
163.  van Swinderen B, Brembs B. Attention-like deficit and hyperactivity in a Drosophila memory mutant. J Neurosci. 2010;30:1003-1014.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 45]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
164.  Kuang L, Hashimoto K, Huang EJ, Gentry MS, Zhu H. Frontotemporal dementia non-sense mutation of progranulin rescued by aminoglycosides. Hum Mol Genet. 2020;29:624-634.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
165.  Howard M, Frizzell RA, Bedwell DM. Aminoglycoside antibiotics restore CFTR function by overcoming premature stop mutations. Nat Med. 1996;2:467-469.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 349]  [Cited by in F6Publishing: 329]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
166.  Howard MT, Anderson CB, Fass U, Khatri S, Gesteland RF, Atkins JF, Flanigan KM. Readthrough of dystrophin stop codon mutations induced by aminoglycosides. Ann Neurol. 2004;55:422-426.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 90]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
167.  Vecsler M, Ben Zeev B, Nudelman I, Anikster Y, Simon AJ, Amariglio N, Rechavi G, Baasov T, Gak E. Ex vivo treatment with a novel synthetic aminoglycoside NB54 in primary fibroblasts from Rett syndrome patients suppresses MECP2 nonsense mutations. PLoS One. 2011;6:e20733.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 40]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
168.  Stern S, Snir O, Mizrachi E, Galili M, Zaltsman I, Soen Y. Reduction in maternal Polycomb levels contributes to transgenerational inheritance of a response to toxic stress in flies. J Physiol. 2014;592:2343-2355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
169.  Misiak B, Ricceri L, Sąsiadek MM. Transposable Elements and Their Epigenetic Regulation in Mental Disorders: Current Evidence in the Field. Front Genet. 2019;10:580.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 50]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
170.  Billingsley KJ, Lättekivi F, Planken A, Reimann E, Kurvits L, Kadastik-Eerme L, Kasterpalu KM, Bubb VJ, Quinn JP, Kõks S, Taba P. Analysis of repetitive element expression in the blood and skin of patients with Parkinson's disease identifies differential expression of satellite elements. Sci Rep. 2019;9:4369.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
171.  Kandratavicius L, Hallak JE, Carlotti CG Jr, Assirati JA Jr, Leite JP. Hippocampal expression of heat shock proteins in mesial temporal lobe epilepsy with psychiatric comorbidities and their relation to seizure outcome. Epilepsia. 2014;55:1834-1843.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 31]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
172.  Kreher J, Takasaki T, Cockrum C, Sidoli S, Garcia BA, Jensen ON, Strome S. Distinct Roles of Two Histone Methyltransferases in Transmitting H3K36me3-Based Epigenetic Memory Across Generations in Caenorhabditis elegans. Genetics. 2018;210:969-982.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 26]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
173.  Greer EL, Beese-Sims SE, Brookes E, Spadafora R, Zhu Y, Rothbart SB, Aristizábal-Corrales D, Chen S, Badeaux AI, Jin Q, Wang W, Strahl BD, Colaiácovo MP, Shi Y. A histone methylation network regulates transgenerational epigenetic memory in C. elegans. Cell Rep. 2014;7:113-126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 118]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
174.  Su Y, Liu X, Lian J, Deng C. Epigenetic histone modulations of PPARγ and related pathways contribute to olanzapine-induced metabolic disorders. Pharmacol Res. 2020;155:104703.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
175.  Vallianatos CN, Farrehi C, Friez MJ, Burmeister M, Keegan CE, Iwase S. Altered Gene-Regulatory Function of KDM5C by a Novel Mutation Associated With Autism and Intellectual Disability. Front Mol Neurosci. 2018;11:104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 38]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
176.  Zhao D, Lin M, Chen J, Pedrosa E, Hrabovsky A, Fourcade HM, Zheng D, Lachman HM. MicroRNA Profiling of Neurons Generated Using Induced Pluripotent Stem Cells Derived from Patients with Schizophrenia and Schizoaffective Disorder, and 22q11.2 Del. PLoS One. 2015;10:e0132387.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 74]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
177.  Gourzis P, Skokou M, Polychronopoulos P, Soubasi E, Triantaphyllidou IE, Aravidis C, Sarela AI, Kosmaidou Z. Frontotemporal dementia, manifested as schizophrenia, with decreased heterochromatin on chromosome 1. Case Rep Psychiatry. 2012;2012:937518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 4]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
178.  Kosower NS, Gerad L, Goldstein M, Parasol N, Zipser Y, Ragolsky M, Rozencwaig S, Elkabetz E, Abramovitch Y, Lerer B. Constitutive heterochromatin of chromosome 1 and Duffy blood group alleles in schizophrenia. Am J Med Genet. 1995;60:133-138.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 27]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
179.  Kalichak F, de Alcantara Barcellos HH, Idalencio R, Koakoski G, Soares SM, Pompermaier A, Rossini M, Barcellos LJG. Persistent and transgenerational effects of risperidone in zebrafish. Environ Sci Pollut Res Int. 2019;26:26293-26303.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
180.  Yuan S, Oliver D, Schuster A, Zheng H, Yan W. Breeding scheme and maternal small RNAs affect the efficiency of transgenerational inheritance of a paramutation in mice. Sci Rep. 2015;5:9266.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 40]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
181.  Gonçalves J, Martins J, Baptista S, Ambrósio AF, Silva AP. Effects of drugs of abuse on the central neuropeptide Y system. Addict Biol. 2016;21:755-765.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 26]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
182.  Yehuda R, Lehrner A. Intergenerational transmission of trauma effects: putative role of epigenetic mechanisms. World Psychiatry. 2018;17:243-257.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 238]  [Cited by in F6Publishing: 177]  [Article Influence: 29.5]  [Reference Citation Analysis (0)]
183.  Laumonnier F, Shoubridge C, Antar C, Nguyen LS, Van Esch H, Kleefstra T, Briault S, Fryns JP, Hamel B, Chelly J, Ropers HH, Ronce N, Blesson S, Moraine C, Gécz J, Raynaud M. Mutations of the UPF3B gene, which encodes a protein widely expressed in neurons, are associated with nonspecific mental retardation with or without autism. Mol Psychiatry. 2010;15:767-776.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 81]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]