Abstract
Maternal care during the early postnatal period of altricial mammals is a key factor in the survival and adaptation of offspring to environmental conditions. Natural variations in maternal care and experimental manipulations with maternal-child relationships modeling early-life adversity in laboratory rats and mice have a strong long-term influence on the physiology and behavior of offspring in rats and mice. This literature review is devoted to the latest research on the role of epigenetic mechanisms in these effects of early-life adversity and mother-infant relationship, with a focus on the regulation of hypothalamic-pituitary-adrenal (HPA) axis and brain derived neurotrophic factor (BDNF). An important part of this review is dedicated to pharmacological interventions and epigenetic editing as tools for studying the causal role of epigenetic mechanisms in the development of physiological and behavioral profiles. A special section of the manuscript will discuss the translational potential of the discussed research.
Keywords: early-life adversity, maternal separation, HPA axis, glucocorticoid receptors, epigenetics, HDAC, histone deacetylase inhibitors, BDNF, neuronal plasticity, enrichment
Introduction
In line with the biological embedding theory, early-life experiences have the potential to induce enduring physiological and molecular alterations that become embedded in the biological systems, impacting outcomes like health, well-being, and behavior across the lifespan Hertzman (2012). In studies on humans, the close relationship of adverse early life conditions, mainly associated with the quality of parental care, with cognitive abilities (Kaplan et al., 2001; Nelson III et al., 2007), physical health (Burenkova et al., 2021; Poulton et al., 2002; Wickrama, Conger, & Abraham, 2005), and mental health (Kessler et al., 2010; Mullen, Martin, Anderson, Romans, & Herbison, 1996; Poulton et al., 2002; Wickrama et al., 2005) of adults has been revealed in empirical studies and further confirmed by multiple meta-analyses on cognitive abilities (Goodman, Freeman, & Chalmers, 2019), physical (Jakubowski, Cundiff, & Matthews, 2018) and mental health outcomes (LeMoult et al., 2020; McKay et al., 2022; Trotta, Murray, & Fisher, 2015). In experiments with laboratory animals, effects of early-life adversity (ELA), again related to the quality of parental care, on the future physiological (F. Champagne, Francis, Mar, & Meaney, 2003; Francis, Diorio, Plotsky, & Meaney, 2002) and behavioral (D. Champagne et al., 2008; F. Champagne et al., 2003; Francis, Diorio, Liu, & Meaney, 1999; Francis et al., 2002; Franklin et al., 2010) phenotypes were revealed in multiple rodent models, such as maternal deprivation or separation, variation in the level of maternal care, limited bedding and nesting, and others (Bonapersona et al., 2019; Duque-Quintero et al., 2022; Rocha, Wang, Avila-Quintero, Bloch, & Kaffman, 2021) (Figure 1). One of the underlying mechanisms for the long-term effects of early experience is epigenetic modifications of the genome of nervous cells, common to both humans (Anacker, O’Donnell, & Meaney, 2014; Labonté et al., 2012; Neves, Dinis-Oliveira, & Magalhaes, 2021; Romens, McDonald, Svaren, & Pollak, 2015; Suderman et al., 2014; Suderman et al., 2012; Turecki & Meaney, 2016; Yehuda et al., 2014; T. Zhang, Labonte, Wen, Turecki, & Meaney, 2013) and animals (Anacker et al., 2014; Suderman et al., 2012; Turecki & Meaney, 2016; T. Zhang et al., 2013).
Figure 1.

Simplified scheme of the long-term effects of early-life adversity and mother-infant relationship on physiology and behavior of offspring. Note. GRs = glucocorticoid receptors; DNAm = DNA methylation; HDACi = HDAC inhibitors; HPA axis = hypothalamic-pituitary-adrenal axis; LG-ABN = licking/grooming and arched-back nursing. Activating pathways are indicated by solid lines, inhibitory with dashed ones.
The field of epigenetics explores heritable1, self-sustaining alterations in the transcriptional activity of the genome; these alterations occur without changes in the DNA sequence and persist even in the absence of the initial stimuli that triggered them (Ptashne, 2007; Vanyushin, 2014). Epigenetic modifications are involved at the stage of germ cell formation and the preimplantation stage of embryo development in the process of genomic imprinting (Li, 2002; Reik, Dean, & Walter, 2001). Neurogenesis and gliogenesis at all stages of ontogenesis, starting from the embryonic period, are controlled by epigenetic regulation (Hirabayashi & Gotoh, 2010; J. Hsieh & Gage, 2005). The molecular investigation of epigenetic mechanisms of activity-dependent processes of long-term plasticity in adult neurons at different stages of the ontogenetic development of an organism has provided novel insights into the interplay between the organism’s genotype and the environment in shaping the phenotype (Day & Sweatt, 2011; Kappeler & Meaney, 2010); this perspective aligns with Waddington’s concept of epigenetics (Waddington, 1940). It has become apparent that epigenetic modifications in neurons of the developing brain are a key mechanism in adapting the behavior of developing organism to future anticipated environmental variations, forming the so-called “predictive adaptive response” (Gluckman & Hanson, 2004). For example, high anxiety level can be considered an adaptive stress response in situations where the early environment signals a high-stress environment (Meaney, 2001; Meaney & Szyf, 2005), so an individual with a heightened stress response may be better equipped to cope with and respond to stressors in the future. This could contribute to increased survival in challenging conditions.
The most studied main types of epigenetic modifications that occur in response to environmental signals are DNA methylation and post-translational modifications of histones. Acetylation, as the most common and well-studied histone modification, is a key factor that affects chromatin structure and gene transcription, enabling the interaction of extracellular signals with the genome (Clayton, Hazzalin, & Mahadevan, 2006). DNA methylation and histone modifications can be interrelated processes: The suppression of transcription occurs through an increase in DNA methylation levels, which is accompanied by a decrease in acetylation levels through the activation of histone deacetylases (Z. Chen & Pikaard, 1997; Eden, Hashimshony, Keshet, Cedar, & Thorne, 1998; Jones et al., 1998).
This narrative literature review focuses on the role of epigenetic mechanisms in mediating the enduring impacts of early-life experiences, including those influenced by maternal care levels, on the physiological and behavioral profiles of laboratory rats and mice. The research sheds light on the translational potential of understanding epigenetic processes, paving the way for possible interventions, such as the implementation of histone deacetylase inhibitors and epigenome editing, to modify epigenetic marks and mitigate the long-term consequences of early-life adversity.
Maternal Care and Development of Behavioral and Physiological Phenotype of Laboratory Rodent Offspring
In humans, a close relationship has been found among cognitive abilities (Kaplan et al., 2001; Nelson III et al., 2007), physical health (Burenkova et al., 2021; Poulton et al., 2002; Wickrama et al., 2005), and mental health (Kessler et al., 2010; Mullen et al., 1996; Poulton et al., 2002; Wickrama et al., 2005) on one hand and adverse early life conditions on the other. In particular, dysfunctional family environment in childhood (physical and sexual abuse, psychological pressure, lack of parental care, divorce or death of parents) is often associated with the emergence of psychopathology2 (depression, alcohol, and drug addiction, post-traumatic stress disorder, bipolar disorder, among others) (LeMoult et al., 2020; McKay et al., 2022; Trotta et al., 2015) and antisocial behavior (Barnow, Lucht, & Freyberger, 2001) in adulthood. Conversely, a positive psychological climate in the family and supportive early relationships can act as social buffering and help reduce the negative impact of unfavorable conditions on the child’s emotion regulation skills, positive affect, and self-esteem (Hostinar, Sullivan, & Gunnar, 2014).
During the early postnatal development of all altricial mammals, including humans, maternal control and care are crucial and play a pivotal role in the survival and adaptation of offspring to environmental conditions (Kotenkova, Meshkova, & Shutova, 1989; Kruchenkova, 2009). The quality of the child-maternal relationship determines the physical development of the offspring and their behavior (Bredy, Humpartzoomian, Cain, & Meaney, 2003; Caldji et al., 1998; Francis et al., 1999; D. Liu, Diorio, Day, Francis, & Meaney, 2000). Licking and grooming of altricial rodent pups by a dam in the early postnatal period is the main source of tactile stimulation that regulates their metabolism and is required to facilitate normative developmental processes (Kappeler & Meaney, 2010).
The model of natural variations in maternal care in rodents developed by M. J. Meaney and colleagues has contributed to a comprehensive study of the mechanisms of the long-term influence of maternal care on various aspects of offspring development. Observations of child-maternal relationships in lactating laboratory rats and mice in the first week of life showed that natural variations in maternal care take place (F. Champagne et al., 2003; Pedersen, Vadlamudi, Boccia, & Moy, 2011; Wei, David, Duman, Anisman, & Kaffman, 2010). Specifically, dams with a high level of maternal care, who lick and groom their pups more often and also characterized by longer duration of licking/grooming and arched-back nursing (LG-ABN), and dams with a low LG-ABN level can be identified (F. Champagne et al., 2003; Meaney, 2001; Pedersen et al., 2011; Wei et al., 2010). Despite the fact that differences in the quality of maternal care between females with high and low LG-ABN levels are observed only during the first postnatal week, they have a strong lifelong influence on the physiology and behavior in the offspring (F. Champagne, Diorio, Sharma, & Meaney, 2001). Particularly, offspring of mothers with high LG-ABN levels display high levels of their own maternal care compared to offspring of mothers with low LG-ABN levels (Francis et al., 1999), are less anxious in a new environment (Pedersen et al., 2011), are characterized by lower plasma corticosterone levels after stress exposure (D. Liu et al., 1997), and improved learning in models without significant exposure to stress: either spatial learning/memory in the Morris water maze (Bredy et al., 2003; Bredy, Zhang, Grant, Diorio, & Meaney, 2004; D. Liu et al., 2000) or social learning of food preferences (Lindeyer, Meaney, & Reader, 2013) with no available data for stress-related learning models, such as fear conditioning. In a cross-fostering experiment, females born by low-LG-ABN mothers but reared by high-LG-ABN females demonstrated a high-LG-ABN maternal profile similar to their foster mothers (Weaver et al., 2004). These results indicate an acquired, non-genetic nature of the manifestation of this behavior.
In addition to natural variations of maternal care, experimental manipulations with maternal-child relationships during the early postnatal period could also be critical for the future physiological and behavioral phenotype of the offspring. This has been shown in a number of studies using experimental models of offspring handling (Caldji, Francis, Sharma, Plotsky, & Meaney, 2000; Lesuis, van Hoek, Lucassen, & Krugers, 2017), deprivation (Llorente-Berzal et al., 2012) or separation (Burenkova, Aleksandrova, & Zaraiskaya, 2014a; Burenkova, Aleksandrova, & Zarayskaya, 2012, 2019; Caldji et al., 2000; Franklin et al., 2010) of offspring from the mother, maternal stress (Blaze & Roth, 2017), and others (Davis, Bolton, Hanson, & Guarraci, 2020). Handling (3–15 min separation of pups from the mother) usually has a favorable effect on the future emotional (level of anxiety assessed in the neophagia test, elevated plus maze, open field test, and others) and cognitive characteristics (performance on learning and memory tasks) of the offspring. The effects of this experimental procedure are expressed in a decrease in the level of anxiety (Caldji et al., 2000; Denenberg & Haltmeyer, 1967; S. Levine, 1956; Meerlo, Horvath, Nagy, Bohus, & Koolhaas, 1999) and an improvement in memory in adult offspring (Kosten, Lee, & Kim, 2007; S. Levine, 1956). These effects are probably caused by an increase in the level of maternal care induced by this procedure of short-term separation of offspring from the mother (M. Lee & Williams, 1974; D. Liu et al., 1997; Pryce, Bettschen, & Feldon, 2001; Smotherman, Brown, & Levine, 1977; Wei et al., 2010). On the opposite end of the spectrum are models of early-life adversity (ELA) that typically have negative effects on animals – the models of limited bedding and nesting (LBN), maternal separation (MS) or maternal deprivation (MD) in laboratory rats and mice offspring (Pryce et al., 2005; Tractenberg et al., 2016). The latter two models represent either a single 24 h episode (in the case of MD) or shorter multiple (in the case of MS, most commonly for 3 h, but generally very heterogeneous, from 15 to 480 min) episodes of isolation of offspring from the mother, and sometimes simultaneously from sibs and performed at different time points of the early postnatal period (Tractenberg et al., 2016). Deprivation or separation of the offspring from the mother can have a negative impact on the phenotype of the grown offspring. In particular, they lead to an increase in the level of anxiety tested at the behavioral and physiological levels (Aisa, Tordera, Lasheras, Del Rio, & Ramirez, 2008; Aisa, Tordera, Lasheras, Del Rio, & Ramirez, 2007; R. L. Huot, Plotsky, Lenox, & McNamara, 2002; Kalinichev, Easterling, Plotsky, & Holtzman, 2002), memory impairment (Aisa et al., 2009; Aisa et al., 2008; Aisa et al., 2007; R. L. Huot et al., 2002; Uysal Harzadın et al., 2005), preference for alcohol (Cruz, Quadros, Planeta, & Miczek, 2008; Rebecca L Huot, Thrivikraman, Meaney, & Plotsky, 2001) and cocaine (Moffett et al., 2006), or a decrease in the level of maternal care in adult offspring females (Lovic, Gonzalez, & Fleming, 2001).
The rearing of rat pups during the first week of life by a stressed female is another model of intervention in the mother-child relationship, leading to a change in the phenotype of the offspring. Females exposed to stress by providing limited nesting resources in an unfamiliar environment were characterized by a reduced level of maternal care and an increased proportion of adverse forms of pup-directed behaviors (stepping on, dropping during transport, dragging, actively rejection, rough handling, and neglect); adult females with a history of maltreatment later reproduced abusive behaviors toward their offspring (Roth, Lubin, Funk, & Sweatt, 2009).
The extensive dataset amassed on the impact of ELA on animal behavior facilitated subsequent systematic reviews and meta-analyses. A qualitative systematic review revealed that the deleterious effects of MS appear to be more consistent in certain behavioral phenotypes, such as depressive-like behavior and memory performance, while exhibiting less consistency in anxiety-like behavior (Tractenberg et al., 2016). Subsequent meta-analysis demonstrated that ELA leads to deficits in hippocampal-dependent learning assessed through Morris water maze and novel object recognition tests in adult rodents exposed to two models of ELA: MS and limited bedding and nesting (LBN) (Rocha et al., 2021). In another meta-analysis, Bonapersona et al. (2019) showed that ELA , encompassing maternal separation, maternal deprivation, LBN, and low LG-ABN levels, impairs non-stressful learning (Morris water maze with water temperature >26°C, object and social recognition), while promoting memory formation during stressful learning (Morris water maze with water temperature <24°C, fear conditioning, and shuttle box). Increased defensive behavior was observed in the elevated plus maze and open field test after MS according to the next meta-analysis (D. Wang, Levine, Avila-Quintero, Bloch, & Kaffman, 2020). Consistent with these findings, Bonapersona et al. (2019) demonstrated that ELA increases anxiety-like behavior (assessed in the elevated plus maze, open field test, forced swim test, light/dark box, and some other tests) and decreases social behavior. ELA, including maternal separation, maternal deprivation, LBN, and low LG-ABN levels, also significantly reduced reward responsiveness and social reward directed behaviors without affecting reward learning (Duque-Quintero et al., 2022).
Along with the effects of ELA on behavioral phenotypes of offspring, numerous physiological effects related to hypothalamic-pituitary-adrenal (HPA) axis functioning have been demonstrated as well. According to recent meta-analysis, it was displayed in a decrease in hippocampal volume, the number of dendritic branches, and dendritic length and suppresses neurogenesis in adulthood (Joëls, Kraaijenvanger, Sarabdjitsingh, & Bonapersona, 2022). Apparently, the hippocampus is one of the primary targets of exposure to stressors, which could be attributed to a high number of glucocorticoid receptors (GRs) that bind cortisol/corticosterone in this brain structure (Sanchez, Young, Plotsky, & Insel, 2000). In addition to enabling negative feedback in the regulation of HPA axis activity, this brain structure is also critically involved in cognitive functioning (Lisman et al., 2017) and, therefore, may play a role in establishing the associations between ELA and cognitive performance.
However, a question arises concerning the mechanisms that unite stress-induced changes in the functioning of the HPA axis with alterations in the brain and behavior. As comprehensively reviewed by Suri and Vaidya (2013), glucocorticoids alter expression and signaling of brain derived neurotrophic factor (BDNF), either through direct binding to putative glucocorticoid response elements (GREs) on the Bdnf gene promoter regions or via other transcription factors involved in Bdnf transcription. These mechanisms represent how the HPA axis activity influence neurobiological and behavioral stress-induced alterations, considering the pivotal role of BDNF in brain structural and functional plasticity, including hippocampal neurogenesis (H. Schmidt & Duman, 2007), synaptogenesis and spine formation (Yoshii & Constantine-Paton, 2010), and neuronal survival (Lipsky & Marini, 2007). Furthermore, the relationship between the HPA axis and BDNF is bidirectional, since BDNF can directly influence the HPA-axis regulation through alterations of corticotrophin‐releasing hormone (CRH) expression levels (Jeanneteau et al., 2012).
It should be noted that standard experimental batteries for testing the physical, somatosensory, or cognitive development of mice in the early postnatal period require isolation of the offspring from the mother for a period of 30 to 60 minutes. However, the long-term consequences of such procedures are practically not studied, which complicates the interpretation of the results of behavioral phenotyping of adult animals that had experienced testing in longitudinal experiments in the early postnatal period. In this regard, a model of daily intermediate length of 45-minute MS from the 3rd to the 6th postnatal day (PND) has been developed; this procedure was shown to induce stress in pups on both hormonal and behavioral levels (Burenkova, Averkina, Aleksandrova, & Zarayskaya, 2020) and impair olfactory discrimination learning task with imitation of maternal grooming as positive reinforcement at the PND8 (Burenkova et al., 2012) that can be explained by alterations in maternal behavior caused by MS (Burenkova et al., 2020). This early-life experience also negatively affected contextual fear memory formation in adult male offspring subjected to brief MS, though no effect on their emotionality tested in the open field test or elevated plus-maze test was observed (Burenkova et al., 2014a). In female adult offspring subjected to brief MS, this experience altered their maternal behavior (Burenkova et al., 2019).
Long-term modifications of behavior caused by early life events have been also identified in subsequent generations. Experimental evidence has been obtained for the transfer of properties of the behavioral phenotype through epigenetic inheritance in mammals: the offspring of mothers with high-LG-ABN maternal profile in two subsequent generations themselves demonstrate high-LG-ABN maternal profile compared to the offspring of mothers with low-LG-ABN maternal profile (Francis et al., 1999). Another example of an epigenetic exposure that leads to a change in the phenotype of animals over several generations is a model of unpredictable chronic MS combined with unpredictable maternal stress, consisting of either a 20-min restraint or a 5-min forced swim in cold water applied unpredictably and randomly during MS which took place daily for 3 hours from the PND1 to 14; this exposure induced depressive-like behaviors up to the third generation and in a complex gender-dependent manner (Franklin et al., 2010).
Experimental interventions in both natural variations of maternal care and models of ELA can potentially reverse the phenotypes of offspring. In the case of natural variations, this reversal can be achieved through cross-fostering, where the biological offspring of low-LG-ABN mothers reared by high-LG-ABN dams exhibit behavioral profiles similar to the offspring of high-LG-ABN mothers, and vice versa, specifically, in their maternal behavioral profiles and fearfulness in the open-field test of exploration (Francis et al., 1999), spatial learning (Bredy et al., 2003), or object recognition memory (Bredy et al., 2003). Additionally, environmental enrichment has been shown to counteract the negative effects of ELA. For instance, it can mitigate the impact of MS on both HPA activity and behavioral responses to stress (Francis et al., 2002), fearfulness in the open-field test of exploration (Berardo, Fabio, & Pautassi, 2016), anxiety in elevated plus-maze test (Dandi et al., 2018), spatial learning (Cordier et al., 2021; Dandi et al., 2018; Menezes, das Neves, Gonçalves, Benetti, & Mello-Carpes, 2020; Mohammadian, Najafi, & Miladi-Gorji, 2019), and object recognition (Menezes et al., 2020). This suggests that environmental influences can not only shape behavioral profiles but also reverse otherwise potentially deleterious outcomes.
Thus, the phenomenology of the effects of the mother-infant relationship on the subsequent physiological and behavioral profile of the offspring is currently well documented in laboratory rodents. As will be shown below, the HPA axis, in particular, the epigenetic mechanisms of the expression of GRs of the hippocampus, is one of the targets of effects of early-life experience.
Maternal Care Triggers Epigenetic Mechanisms of the Development of the Physiological and Behavioral Phenotype of Laboratory Rodents
The early postnatal period in immature rodents is marked by the presence of the so-called stress hyporesponsive period, SHRP (Schapiro, Geller, & Eiduson, 1962), which is analogous to such a period in children under the age of one year (Gunnar, Brodersen, Krueger, & Rigatuso, 1996; Larson, White, Cochran, Donzella, & Gunnar, 1998). In rats, it lasts from PND3 to 14 (Schapiro et al., 1962; Schoenfeld, Leathem, & Rabii, 1980), in mice, from PND1 to 12 (Cirulli, Santucci, Laviola, Alleva, & Levine, 1994; D’Amato et al., 1992; M. Schmidt, Oitzl, Levine, & de Kloet, 2002). This period is characterized by a reduced level of activity of the HPA system in response to stressors (Schapiro et al., 1962; Schoenfeld et al., 1980; Walker, Perrin, Vale, & Rivier, 1986). Its biological role is to protect the developing central nervous system from the negative effects of high concentrations of glucocorticoids released during stress (Sapolsky & Meaney, 1986). The existence of a period of low sensitivity to stress is suggestively based on the influence of maternal care, which suppresses the activity of the HPA system (De Kloet, Rosenfeld, Van Eekelen, Sutanto, & Levine, 1988; Suchecki, Rosenfeld, & Levine, 1993). This is substantiated by the fact that maternal deprivation is one of the few stressors, the impact of which during this period leads to the activation of the HPA system in pups, namely, to an increase in the blood level of corticosterone (Cirulli et al., 1994; D’Amato et al., 1992; Enthoven et al., 2010; Kuhn, Pauk, & Schanberg, 1990; McCormick, Kehoe, & Kovacs, 1998; Pihoker, Owens, Kuhn, Schanberg, & Nemeroff, 1993; Smith, Kim, van Oers, & Levine, 1997). Consequently, the activation of the HPA system in pups is regarded as one of the key mechanisms underlying the negative impact of maternal deprivation.
An important role in the regulation of the HPA system’s activity is played by GRs in the hippocampus: when corticosterone is released into the bloodstream, this activity is inhibited by the mechanism of negative feedback (Jacobson & Sapolsky, 1991). Adult offspring of high-LG-ABN mothers are characterized by a high expression of GRs, encoded by the Nr3c1 gene, in the hippocampus and, as a result, a more pronounced inhibition of the HPA system, i.e., its less pronounced activity under stress (Francis et al., 2002; Weaver et al., 2005). Manipulations with lactating females that lead to an increase in the level of LG-ABN (for example, handling of offspring) can cause a long-term increase in the expression of GRs in the hippocampus and a decrease in the activity level of the HPA system, in particular, a decrease in the level of corticosterone in blood plasma in response stress in adulthood (Avishai-Eliner, Eghbal-Ahmadi, Tabachnik, Brunson, & Baram, 2001; Francis et al., 1999). On the contrary, manipulations with lactating females that lead to a decrease in the level of LG-ABN, for example, separation of offspring from the mother, cause a decrease in the expression of GRs in the hippocampus (Francis et al., 2002; Ladd, Huot, Thrivikraman, Nemeroff, & Plotsky, 2004) and also an increase in the level of activation of the HPA system manifested in an increase in the adult plasma levels of corticosterone in response to stress (J. Chen et al., 2012; Kalinichev et al., 2002). The impact of maternal care level during the first week of offspring’s life on the enduring modification of HPA system functioning and the expression of hippocampal GRs is grounded in epigenetic mechanisms.
The availability of a DNA strand to various regulatory proteins, including transcription factors, determines the structure of chromatin, thus regulating gene expression (Vanyushin, 2014). The most well-known epigenetic modifications that occur in response to environmental influences are DNA methylation, post-translational modifications of histones, and gene silencing mediated by small RNAs, such as micro RNA (miRNA) and short interfering or silencing RNA (siRNA) (Vanyushin, 2014). In recent years, miRNA as an important regulator of gene expression has gained significant interest for their involvement in neural plasticity and diverse pathological processes (Allen & Dwivedi, 2020; McKibben & Dwivedi, 2021).
DNA methylation is known to cause transcription suppression of certain genes (Boyes & Bird, 1992; C. Hsieh, 1994). According to one of the hypotheses, methyl groups sterically hinder the binding of transcription factors to the DNA strand (Bird, 1986); according to another, methylation promotes the binding with DNA enzymes that suppress transcriptional activity (Bird, 2002). The main factor that determines the structure of chromatin is the state of amino acids at the N-terminus of H3 and H4 histones, i.e., those proteins that are the target of a number of reversible post-translational modifications that affect the structure of the nucleosome and gene transcription (Dorigo, Schalch, Bystricky, & Richmond, 2003; Luger, Mäder, Richmond, Sargent, & Richmond, 1997).
According to the “histone code” hypothesis (Strahl & Allis, 2000), the state of chromatin is dynamic. It is determined by a combination of post-translational modifications of various amino acid residues of histone proteins: acetylation (Sterner & Berger, 2000), phosphorylation (Nowak & Corces, 2004), methylation (Y. Zhang & Reinberg, 2001), ubiquitination (Osley, 2004; Sun & Allis, 2002), sumoylation (Nathan, Sterner, & Berger, 2003), propionylation and butyrylation (Y. Chen et al., 2007), glycosylation (Gugliucci, 1994; Liebich et al., 1993), biotinylation (Rodriguez-Melendez & Zempleni, 2003; Stanley, Griffin, & Zempleni, 2001), carbonylation (Wondrak, Cervantes-Laurean, Jacobson, & Jacobson, 2000), and ADP-ribosylation (Golderer & Gröbner, 1991). Acetylation, maybe the most studied histone modification, is a key factor influencing chromatin structure and gene transcription. It enables the interaction of extracellular signals with the genome (Clayton et al., 2006). In 1964, Allfrey et al. (Allfrey, Faulkner, & Mirsky, 1964) suggested that the level of histone acetylation correlates with the transcriptional activity of genes, with a decreased or increased level of histone acetylation, transcriptional activity is suppressed or activated, respectively. If histones are tightly bound to DNA, this prevents the DNA from interacting with other proteins, including the enzyme RNA polymerase required for transcription. For transcriptional activation, the chromatin structure must be changed. According to one hypothesis, the activation of transcription that occurs during histone acetylation (Z. Wang et al., 2008) is explained by the fact that these modifications decrease the positive charge of histones, due to which histones interact with negatively charged DNA (D. Lee, Hayes, Pruss, & Wolffe, 1993). Acetylation of the amino group of lysine at the N-terminus of the histone molecule neutralizes the positive charge of histones, whereas the affinity between histones and DNA decreases, which results in chromatin decondensation and increases the availability of DNA sites for transcription factors (D. Lee et al., 1993). According to an alternative hypothesis, transcription factors can recognize modified amino acids at the N-terminus of histone molecules and directly interact with them (Vettese-Dadey et al., 1996; Vitolo, Thiriet, & Hayes, 2000). Specific transcription coactivators are also capable of recognizing acetylated lysine residues and binding to acetylated histone “tails” (Dion, Altschuler, Wu, & Rando, 2005; Hong, Schroth, Matthews, Yau, & Bradbury, 1993). The above mechanisms underlie the long-term maintenance of not only stable genomic properties of individual cells but also their ensembles, which provide long-term changes in physiology and behavior (Korzus, Rosenfeld, & Mayford, 2004).
It was shown that in the pups raised by high- and low-LG-ABN mothers, the DNA of the GR promoter (exon 17) in the hippocampus is methylated differently: low and high levels of DNA methylation, correspondingly (Weaver et al., 2004). The levels of histone H3 acetylation in the same GR region of the hippocampus have the opposite trend: offspring of high- and low-LG-ABN mothers were characterized by high and low levels of histone acetylation, correspondingly (Weaver et al., 2004). These differences in the level of DNA methylation and histone acetylation in the brain of offspring appear after the first week of life and persist throughout the lifespan (Weaver et al., 2004; T. Zhang et al., 2013), potentially underlying the above-described emotional (Caldji et al., 2000; Francis et al., 1999; Weaver, Meaney, & Szyf, 2006) and cognitive (Bredy et al., 2003; Bredy et al., 2004; Lindeyer et al., 2013; D. Liu et al., 2000; Pedersen et al., 2011) profiles of adult animals. The long-term influence of the LG-ABN levels on hippocampal GRs expression is enabled through an increase in the expression of the transcription factor NGFI-A (nerve growth factor-inducible factor-A, also known as egr-1, krox-24, zenk, and zif-26) and its binding to exon 17 GR promoter (Weaver et al., 2007). It should be emphasized that although DNA methylation of the GR promoter of animals is transmitted across generations, it can also be reversed with cross-fostering, such as the biological offspring of low-LG-ABN mothers reared by high-LG-ABN dams resemble the offspring of high-LG-ABN mothers, and vice versa (Francis et al., 1999; Weaver et al., 2004). This suggests that environmental influences can not only program epigenetic and behavioral profiles but also reverse otherwise potentially negative outcomes.
In addition to the model of natural variations in maternal care, similar epigenetic effects have been shown in models of MS. MS increased DNA methylation levels within the exon 17 GR promoter region in the hippocampus of adult rats (Zhu et al., 2017) and mice (Kember et al., 2012; Kundakovic, Lim, Gudsnuk, & Champagne, 2013). The systematic review revealed a trend for a downregulation of GR mRNA expression and an increase in GR DNA methylation in response to MS (Tractenberg et al., 2016). Conversely, MS decreased the levels of histone acetylation in several studies. MS of rat pups from PND1 to 14 led to a decrease in the level of total (not associated with specific genes’ promoters) H3 and H4 histone acetylation in the mesolimbic structures, dorsal caudate putamen and nucleus accumbens core in adulthood (Tesone‐Coelho et al., 2015). Separation from the mother and siblings in the first two weeks of life reduced the level of total histone H4 acetylation at the PND21 in the forebrain neocortex of mice (A. Levine, Worrell, Zimnisky, & Schmauss, 2012). MD on PND9 in rats led to a decrease in histone H3 acetylation at lysine 9 in the ventral tegmental area (VTA) on PND14–21 (Shepard et al., 2018).
In the hippocampus, the levels of acetylated histones H3 and H4 were significantly reduced at the Bdnf gene promoter IV in the hippocampus of adult rats after MS (Seo et al., 2016). This and other studies thus show that the rearing environment in the first week of life affects the expression of the Bdnf gene, one of the key genes involved in neuronal plasticity (Yamada, Mizuno, & Nabeshima, 2002), through the epigenetic mechanisms. MS increased DNA methylation levels within the Bdnf IX promoter in the hippocampus of adult mice (Kundakovic et al., 2013). Nursing by a stressed female during the first week of life reduced Bdnf gene mRNA expression and increased the methylation level of the Bdnf gene promoter in the prefrontal cortex, both in the first week of life and in adult animals (Roth et al., 2009). Conversely, communal nursing, characterized by an increased level of maternal care received by offspring, led to an increase in the level of histone acetylation at the I, IV, VI, and VII promoters of the Bdnf gene (Branchi, Karpova, D’Andrea, Castren, & Alleva, 2011). The systematic review revealed a trend for an increase in Bdnf DNA methylation in response to MS (Tractenberg et al., 2016). In light of the previously mentioned interplay between the HPA axis and BDNF (Jeanneteau et al., 2012; Suri & Vaidya, 2013), these studies, along with research on GRs, provide evidence for the mutual reciprocal modulation of epigenetic molecular mechanisms that regulate stress and neuronal plasticity systems in the brain in response to alterations in mother-infant relationships in laboratory rodents.
It is important to note that the long-term impact of ELA in rats and humans is rooted in the same epigenetic mechanisms (Labonté et al., 2012; Romens et al., 2015; Suderman et al., 2014; Suderman et al., 2012; Turecki & Meaney, 2016; Yehuda et al., 2014). According to a systematic review published in 2016 by Turecki and Meaney, since the publication of the first data on the different levels of DNA methylation of the GR promoter (exon 17 GR promoter) in the hippocampus of the offspring of rats with different levels of maternal care, the number of studies on the effects of early-life experience, parental stress, and psychopathology on the methylation status of the GR had reached forty, of which 13 were carried out in animals and 27 in humans (Turecki & Meaney, 2016). Most of these studies determined the methylation status of exon 17 in rats and exon 1F in humans. Among the studies on the impact of ELA, an increase in the methylation level of these exons was shown in 89% of human studies and 70% of animal studies (Turecki & Meaney, 2016).
Thus, the rearing environment of altricial mammals, mediated by maternal care, plays a key role in the development of their phenotype, and epigenetic mechanisms might be essential “injectors” of the early environment under the “skin” of the pups. However, to establish a causal rather than merely correlational relationship between these two outcomes of ELA exposure – alteration of behavioral and epigenetic phenotype – experimental manipulation of the epigenetic profile is required, and the use of pharmacological agents that modulate the epigenetic mechanisms underlying these long-term negative consequences can serve to this purpose. Furthermore, it is of applied importance to correct the negative consequences caused by disturbances in mother-child relationships in the early postnatal period using pharmacological agents. Inhibitors of histone deacetylases (HDACs) and DNA methyltransferase (DNMT) inhibitors may serve as such agents.
Pharmacological Interventions and Epigenetic Editing as Tools for Studying the Role of Epigenetic Mechanisms in Nervous System Plasticity
HDACs are a family of enzymes consisting in humans of 18 HDAC enzymes (Seto & Yoshida, 2014) that catalyze acetyl groups removal from the lysine residues on both histone and nonhistone proteins (Seto & Yoshida, 2014). Some of these HDACs were proven to play a role in both physiological and pathological neuronal development and function (Guan et al., 2009; Kelly & Cowley, 2013; Parra, 2015; Reichert, Choukrallah, & Matthias, 2012). One of the methods of intervention in epigenetic mechanisms is HDAC inhibition, which leads to an increase in the level of histone acetylation in the brain and is accompanied by an increase in transcriptional activity (Sinn et al., 2007). The most widely used HDAC inhibitors block class I HDACs (trichostatin A, sodium butyrate, valproic acid, suberoylanilide hydroxamic acid (SAHA)), class II HDACs (TSA, sodium butyrate, and SAHA), or class IV HDACs (SAHA); each of them has a different degree of selectivity for HDACs isoforms (Grinkevich, 2014; Roth & Sweatt, 2009). The development of new, more specific inhibitors for each of the 18 existing types of HDAC is actively pursued in cancer research. However, these developments have not yet extended to the field covered in this review. Moreover, the differences in the effects of existing inhibitors in this context have yet to undergo careful examination, although some research has already demonstrated the varying effects of different HDAC inhibitors on learning and memory performance (Guan et al., 2009).
A large proportion of studies using HDAC inhibitors are devoted to the role of epigenetic mechanisms in the plasticity of the mature nervous system. For example, an increase in the level of histone acetylation in the brain caused by prolonged intraperitoneal administration of HDAC inhibitors (TSA, sodium valproate, sodium butyrate) triggers the processes of eye-dominant plasticity, which is usually observed only during the critical period of development of the visual system (Putignano et al., 2007; Vetencourt, Tiraboschi, Spolidoro, Castrén, & Maffei, 2011). In addition, restoration of visual functions is observed in adult animals with such a form of visual acuity impairment as amblyopia caused by prior monocular deprivation during the critical period of development of the visual system (Silingardi, Scali, Belluomini, & Pizzorusso, 2010). Another example of the plasticity of the mature nervous system is learning. The administration of HDAC inhibitors shortly before (from 30 min to 2 h) or immediately after training improves long-term memory and learning performance in a number of adult animal models: enhances memory for conditioned fear in mice and rats (Bredy & Barad, 2008; Guan et al., 2009; Levenson et al., 2004; Vecsey et al., 2007), facilitates memory in object recognition training in mice (Stefanko, Barrett, Ly, Reolon, & Wood, 2009), and potentiates the formation of long-term memory in passive avoidance task in chickens (Toropova, Anokhin, & Tiunova, 2014).
In the model of variation in the level of maternal care in rats, it should be noted that the reversibility of the epigenetic status acquired in the early postnatal period was experimentally shown in the adult offspring of low-LG-ABN mothers. Specifically, intracerebral (intraventricular) administration of TSA into the brains of adult offspring of low-LG-ABN mothers significantly reduced the level of DNA methylation of the GR promoter and increased the level of histone H3 acetylation in the region of the same promoter in the hippocampus to levels observed in the offspring of high-LG-ABN mothers (Weaver et al., 2004). In line with it, the level of corticosterone in the blood, normally elevated in the low-LG-ABN mothers in response to acute stress (immobilization for 20 min), decreased to the levels of the offspring of high-LG-ABN mothers (Weaver et al., 2004). In addition, the administration of TSA to adult offspring of low-LG-ABN mothers reduced anxiety levels in the open field test to the level observed in the offspring of high-LG-ABN mothers (Weaver et al., 2006).
In the model of MS, the administration of sodium butyrate to adult animals that underwent maternal separation (3 hours daily, PND1–14) increased total histone H3 acetylation levels in the hippocampus compared to saline administration (Albuquerque Filho et al., 2017). Furthermore, sodium butyrate restored object recognition memory in MS animals (Albuquerque Filho et al., 2017). This study also demonstrated an increase in BDNF levels in the hippocampus after the administration of sodium butyrate in the MS group but not in the non-MS group with a positive correlation was observed between BDNF levels and memory performance across all groups combined (Albuquerque Filho et al., 2017).
Studies of the effect of blockade of HDAC in the first week of life, which is a critical period for the development of the future epigenetic status of animals, are practically absent. It is important to note that the solvent commonly used to dissolve TSA, dimethyl sulfoxide (DMSO), is known to be toxic to the developing nervous system (Lobanov, Khokhlova, Suvorova, Zaraiskaya, & Murashev, 2008). In contrast, HDAC inhibitor sodium valproate does not pose this drawback, as it is readily soluble in saline. Studies have shown that its subcutaneous administration at a dose of 50 mg/kg on the mice PND1 significantly increases histone H3 acetylation levels in the brain and, at the same time, is well tolerated by them (Murray, Hien, de Vries, & Forger, 2009). This substance is also known as an anticonvulsant drug, the mechanism of action of which is attributed to the activation of the inhibitory GABAergic system, the suppression of the excitatory glutamatergic system, and the general decrease in brain metabolism when used in therapeutic doses. However, with the administration of smaller doses (such as 50 mg/kg), these effects are not observed (Johannessen & Johannessen, 2003). In our experiments, we showed that the blockade of histone deacetylases by sodium valproate, administered subcutaneously at a dose of 50 mg/kg on PND3–6 led to an improvement in olfactory discrimination learning task with imitation of maternal grooming as positive reinforcement impaired by MS procedure in a gender-dependent manner, i.e., in males, but not in females (Burenkova, Aleksandrova, & Zaraiskaya, 2014b). At the same time, the use of this scheme of administration of valproate did not lead to a violation of the physical, somatosensory development and social behavior of 129Sv mice in the nesting period (Burenkova, Aleksandrova, & Zarayskaya, 2015).
The results obtained indicate the possibility of further use of sodium valproate at the indicated dosage for experimental modulation of the level of histone acetylation in the developing brain (Burenkova et al., 2019). The long-term influence of the administration of histone deacetylase inhibitors on the physiological and behavioral phenotype of adult animals to correct those negative consequences of environmental influences in the early postnatal period that affect epigenetic molecular and cellular mechanisms remains poorly explored. The results obtained to date in adult males are showing that sodium valproate selectively increased the anxiety of adult male mice in open-field and elevated plus-maze tests (Burenkova et al., 2014a). At the same time, we revealed that 50 mg/kg sodium valproate injections prevented the reduction of the level of maternal nursing associated with licking-grooming and pup investigation induced by MS and pain from injections in offspring females later in adulthood (Burenkova et al., 2019). However, it appears that the positive effects of valproate might be constrained by a specific range of concentrations. To begin with, a single prenatal (on E10-E12, which approximately corresponds to the first trimester of human embryonic life) or multiple postnatal (in the range of PND0–14, which corresponds to the third trimester of human embryonic life) exposure of high valproate dosages (300–600 mg/kg) is widely utilized for modeling autism spectrum disorder since it recapitulates autism-related phenotypes, including deficits in social interactions (Elnahas et al., 2021; Kim et al., 2011; Moldrich et al., 2013). The mechanisms underlying these behavioral effects may involve elevated brain inflammatory cytokines, oxidative stress, histological neurodegeneration, and apoptosis (Elnahas et al., 2021). The effects of lower but still high postnatal valproate dosage are generally in line with the prenatal effects of valproate. Valproate injections on PND6–12 (150 mg/kg) decreased the interest in new social contacts on PND36 in rats (Gedzun et al., 2021). The injection of a dosage of 400 mg/kg to rat pups during the first week of life resulted in later maternal adverse behaviors toward their own pups in adulthood (Collins et al., 2022). Furthermore, valproate (200 mg/kg) administered on PND4–11 led to significant delays in weight gain and motor impairments in the nesting and adolescent period (Bath & Pimentel, 2017). Similarly, valproate injections on PND6–12 (150 mg/kg) led to a delay in early motor development and hyperactivity in the open field test on PND21 in rats (Gedzun et al., 2021).
A 400 mg/kg dose of another HDAC inhibitor, sodium butyrate, dissolved in a 5% sucrose solution and delivered orally before each maltreatment episode (exposure to stressed dams) on PND1–7 prevented the maltreatment-induced increase in methylation at Bdnf exon IX in the prefrontal cortex of male pups on PND8, though not in female pups (Doherty, Chajes, Reich, Duffy, & Roth, 2019). This outcome could be indicative of crosstalk between histone acetylation and DNA methylation levels, as demonstrated in earlier cell culture studies (Cervoni & Szyf, 2001). Decreased after MD on PND9 Bdnf mRNA levels were reversed 24 hours after a single injection of the selective class I HDAC inhibitor CI-994 (also known as N-acetyldinaline or tacedinaline; dissolved in 1% Tween80, which is considered not toxic) on PND14–21 (Shepard et al., 2018).
The second of the two most frequently utilized pharmacological tools in the study of epigenetic mechanisms related to ELA is the inhibition of DNA methyltransferase (DNMT). DNMTs constitute a family of enzymes, consisting in humans of five members, with DNMT1, DNMT3A, and DNMT3B being the primary ones; DNMT1 maintains existing DNA methylation patterns during DNA replication, whereas DNMT3A and DNMT3B are responsible for de novo DNA methylation (Lyko, 2018). Inhibition of DNMTs results in a decrease in DNA methylation, and commonly used inhibitors include zebularine, azacitidine, and decitabine (Weaver et al., 2017). Pertaining to the focus of this review, treatment with zebularine in adult animals reversed the effects of nursing by a stressed female during the first week of life in rats, leading to reduced Bdnf gene mRNA expression and increased DNA methylation of the Bdnf gene promoter in the prefrontal cortex (Roth et al., 2009). Similarly, zebularine rescued aberrations in maternal behavior caused by maltreatment exposure (LBN model), but its treatment resulted in an increase in DNA methylation levels of Bdnf exon IV and a decrease in its expression in the medial preoptic area (MPOA) of maltreated animals (Keller, Doherty, & Roth, 2019). Additionally, zebularine disturbed behavior in dams without a history of maltreatment, leading to higher levels of adverse behavior toward offspring compared to vehicle-treated controls (Keller et al., 2019). The negative behavioral effects of zebularine were also demonstrated in other studies, where it prevented memory consolidation (Lubin, Roth, & Sweatt, 2008; Miller & Sweatt, 2007). More precise mechanism of zebularine’s action still remains poorly understood, as it inhibits DNMT1 activity through intercalation into DNA during replication, potentially limiting its applicability in postmitotic neuronal cells. In contrast, the use of small molecule inhibitors that do not incorporate into DNA and act more as enzymatic DNMT inhibitors, such as RG108 (N-phthalyl-1-tryptophan) (Zheng, Wu, Chen, & Goodman, 2008), represent a more promising epigenetic tool, although it has not been tested for alleviating the effects of ELA yet.
Therefore, studies on the impact of HDACs and DNMTs inhibition during critical developmental periods and adulthood highlight their potential to reverse epigenetic changes associated with ELA. These inhibitors can be an important tool in understanding and manipulating epigenetic mechanisms in the context of brain development and function and in mitigating the consequences of early environmental influences on neural development. However, further study of the mechanisms of their action, as well as the examination of appropriate dosages and potential adverse effects, is warranted. Furthermore, an important caveat of using these inhibitors is their non-specific effects on particular genes or sequences. In other words, it is not possible to use HDAC inhibitors to target specific genes or loci, and this limitation could potentially restrict their therapeutic efficacy. This drawback can be addressed through the utilization of epigenome editing techniques.
Epigenome editing is a promising approach that involves the targeted modification of an epigenome at specific sites by employing molecules that bind to specific DNA sequences and modify them or adjacent histones. This approach is based on three different systems that can recognize nucleotide sequence: zinc-finger nucleases (ZFNs), transcription activator-like effectors (TALEs), and clustered regularly interspaced short palindromic repeats (CRISPR) interacting with dCas9 nucleases (Day, 2014). In this review, we will focus on CRISPR/dCas9-based approach as the most recent and effective one.
The traditional CRISPR/Cas9 system uses a guide RNA (gRNA) to target the Cas9 protein to specific DNA sequences, where the Cas9 induces double-strand breaks in the DNA. In contrast, CRISPR/dCas9 enables the targeting of specific genomic locations without inducing DNA breaks, as the Cas9 protein is altered to lack its endonuclease (cutting) activity, rendering it deactivated or ‘dead,’ for which the ‘d’ stands in its name (Whinn et al., 2019). Thus, CRISPR/dCas9 can be employed for the targeted modification of epigenetic marks, such as DNA methylation or histone modifications, without causing changes in the DNA sequence. For this purpose, CRISPR/dCas9 complex is fused with epigenetic modifying enzymes such as Tet13 or DNMTs for the modulation of DNA methylation (X. Liu et al., 2016), or histone acetyltransferase (HAT) p300 or HDACs for the modulation of histone acetylation (Hilton et al., 2015).
In vivo, epigenome editing with CRISPR-dCas9 has already been validated on both molecular (X. Liu et al., 2016) and behavioral (Bohnsack et al., 2022) levels. X. Liu et al. (2016) demonstrated that targeted demethylation of the Bdnf promoter IV by dCas9-Tet1 injection into different regions of the same brain induced Bdnf gene expression in post-mitotic neurons. Bohnsack et al. (2022) used dCas9-p300 injection to the amygdala, which led to the attenuation of adult anxiety and excessive alcohol drinking in a rat model of adolescent alcohol exposure. No data on correction of the effects of ELA on the behavioral, physiological, or epigenetic profiles seem to be available currently.
Despite the obvious advantages of the method of epigenome editing, it also has some disadvantages that limit its widespread use. First, the necessity of precise viral injections to the brain can be challenging to carry out. Second, it can involve off-target effects – unintended modifications that occur at genomic loci other than the intended target site (X. H. Zhang, Tee, Wang, Huang, & Yang, 2015). Third, to be efficient, methylation requires prolonged high expression of dCas9-DNMT3A that declines rapidly after the termination of dCas9-DNMT3A expression (Sapozhnikov & Szyf, 2023). Four, DNMTs exhibit a non-specific activity, independent of targeting, that makes them unsuitable for precise epigenetic editing (Sapozhnikov & Szyf, 2023). Five, this technique is selective to the gene but not the position of acetylated histones. In conclusion, the use of epigenetic editing holds the potential to advance our comprehension of the causal connections between ELA and behavioral, physiological, as well as epigenetic profiles. However, significant improvements in this technique are necessary.
Translational Potential of Mother-Infant Relationship Models
An important result of the research discussed in this review is the fact that the target genes of the detected epigenetic modifications in human and animal brain cells can be similar and be controlled by the same epigenetic mechanisms (Turecki & Meaney, 2016). This indicates both the need for a wider range of studies of epigenetic modifications in developing brains and the possibility of using biological models of adverse early experiences as translational ones.
Translational research is a widely used concept whose meaning has been evolving since its introduction in the literature at the turn of the 21st century as the process signifying the cycle of discoveries “from the bench to the bedside and back again” (Fort, Herr, Shaw, Gutzman, & Starren, 2017). In short, both the definition and the structure of translational research have evolved, reflecting the growing complexity and intensity of the field. Yet, the common notion of numerous various definitions is in attempting to identify and bridge a supposed gap between knowledge produced at the lab bench and its use at the clinical bedside, which needs to be closed to help society harvest the dividends of its support of and investments in scientific research (van der Laan & Boenink, 2015). The current consensus definition of translational research points to a hierarchical structure of interdisciplinary work, dissecting the continuous flow of translational research into five connected phases, T1-T0. Thus, the translational cycle includes (1) T1, where observations and ideas from basic research are verified through early testing in humans; (2) T2 involves the establishment of effectiveness of the basic finding in humans and formulating clinical guidelines; (3) T3 primarily focuses on implementation and dissemination research; (4) T4 focuses on outcomes and effectiveness in human subpopulations and populations; and (5) T0 covers research such as genome-wide association studies which wrap back around to basic research.
The research presented in this narrative review is mostly localized at the T1 phase of translation. Importantly, although most human studies are limited to observation and registration of association, model organism studies can create temporal sequences of events that are characteristic of developmental milestones across species. It is particularly relevant to studies of the impact of early life events on lifespan outcomes. Thus, with an eye to translational applications for human research and practice derivable from rodent studies, Bonapersona and colleagues (2019) conducted a multi-level (animals, outcomes, and experiments) large-scale (400+ independent experiments involving ~8600 animals) meta-analyses of the impact of aberrant maternal care on specific behavioral domains. Among numerous associations between aberrant maternal behavior and subsequent developmental outcomes, the authors stress the following as most interesting. First, they observed that aberrant maternal behavior has differential effects on memory by strengthening memory in stressogenic but hampering it in non-stressful situations. Second, faulty maternal care increases anxiety and decreases social behavior, especially in males. Importantly, in line with human work on cumulative stress (Suor, Sturge-Apple, Davies, Cicchetti, & Manning, 2015), the effects are amplified when rodents experience additional stressful life events (Daskalakis, Bagot, Parker, Vinkers, & de Kloet, 2013).
Moreover, in addition to literature summaries on the general effects of maternal care on subsequent development, there are targeted reviews aimed at evaluating the benefits of rodent models for advancing research on various aspects of human development and disease. Thus, Ham and colleagues (2023) conducted a cross-species review of rodents and humans on synchrony in parent-offspring social interactions across development. Concluding their review, the authors commented on both similar and distinct qualities of the developmental synchrony across the species, stressing that such comparative analyses can generate a deeper understanding of the mechanics of parent-offspring interactions and their typicality and atypicality and related lifespan consequences. The obvious targets for such translational investigations are offspring cries, physical touching, feeding behavior, and social interactions, among other complex behaviors.
To illustrate, altered methylation patterns of the Bdnf gene have been observed in human cord blood following high maternal bisphenol A, BPA, levels during pregnancy (Kundakovic et al., 2015). This result is aligned with the results of model studies in which developmental exposure to endocrine disruptors, such as BPA, results in drastic organismal changes driven by epigenetic mechanisms (Kundakovic & Champagne, 2011). Similarly, diet has been shown to be highly impactful on patterns of methylation in model organisms both during gestation (Reuter, Gupta, Park, Goel, & Aggarwal, 2011; Waterland & Jirtle, 2003) and outside (Milagro et al., 2009) of developmental periods. These findings have been paralleled by studies of severe dietary limitations as those during famine (Heijmans et al., 2008) in humans.
In general, model organisms provide a unique platform for understanding the epigenetic mechanics of fundamental processes unfolding in typical and atypical human development. Given the enormous corpora of the literature on the role of early life effects in the development of human and model organisms, careful comparative analyses similar to those presented by Ham and colleagues (2023) are needed. Findings from such analyses might permit the development of the interventions that might be evaluated at T2 of the translational cycle.
Conclusion
Research efforts in recent decades have focused on uncovering the epigenetic mechanisms underlying the development of physiological and behavioral phenotypes through the interaction of the organism with the environment in the early stages of the postnatal period. Maternal (parental) care is unique among other environmental factors as it determines the survival and adaptation of offspring to environmental conditions. Epigenetic modifications that occur in the neurons of the maturing brain in response to specific levels of parental care are apparently one of the key mechanisms on which this adaptation is based on.
An extremely important property of the detected epigenetic modifications is their reversibility (Weaver et al., 2004). The possibility of this reversibility underlies the importance of developing approaches that elucidate the mechanisms that trigger epigenetic modifications in the brain and mediate the influence of early developmental conditions. Given that a significant implication of “reversibility” is the potential for correction and “reprogramming” of the negative consequences of early-life experiences (Vaiserman, Voitenko, & Mekhova, 2011), further studies on precise techniques that modulate gene- and cell-specific levels of modified histones and DNA methylation are of utmost importance.
Acknowledgment
This work was supported by the awards from the Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD (Grants P50HD052117, PI Jack Fletcher and P20HD091005, PI Elena L. Grigorenko) to the University of Houston, by the award from the Russian Foundation for Basic Research (Grant 16-34-00253, PI Olga V. Burenkova), and by the Ministry of Science and Higher Education of the Russian Federation (Agreement 075-10-2021-093, Project COG-RND-2105, PI Elena L. Grigorenko). We are grateful to Lauren Elderton for her editorial support.
Footnotes
Conflict of Interest Statement
The authors declare no conflict of interest.
Either non-gamete or gamete-mediated. In the first case, for multigenerational transmission of the phenotype, the presence of an environmental factor is required, typically during a critical period in development. For instance, the transmission of a specific maternal profile (low- or high-LG-ABN) from generation to generation is impossible without the impact on the offspring of each subsequent generation by the corresponding maternal care level (Szyf, McGowan, & Meaney, 2008). In the second case, the inheritance of the phenotype is independent of the recurring exposure of an environmental factor and is determined by gametic epigenetic inheritance (Franklin et al., 2010; Skinner & Guerrero-Bosagna, 2009).
However, a nonlinear U-shaped relationship was also demonstrated between exposure to early-life adversity and subsequent resilience and improved mental health, indicating that individuals with some lifetime adversity reported better outcomes than those with a high level of adversity or no history of adversity (Collins et al., 2023; Seery, Holman, & Silver, 2010).
Tet1is enzyme involved in the process of DNA demethylation
References
- Aisa B, Gil-Bea FJ, Marcos B, Tordera R, Lasheras B, Del Rio J, & Ramirez MJ (2009). Neonatal stress affects vulnerability of cholinergic neurons and cognition in the rat: involvement of the HPA axis. Psychoneuroendocrinology, 34(10), 1495–1505. doi: 10.1016/j.psyneuen.2009.05.003 [DOI] [PubMed] [Google Scholar]
- Aisa B, Tordera R, Lasheras B, Del Rio J, & Ramirez M (2008). Effects of maternal separation on hypothalamic–pituitary–adrenal responses, cognition and vulnerability to stress in adult female rats. Neuroscience, 154(4), 1218–1226. doi: 10.1016/j.neuroscience.2008.05.011 [DOI] [PubMed] [Google Scholar]
- Aisa B, Tordera R, Lasheras B, Del Rio J, & Ramirez MJ (2007). Cognitive impairment associated to HPA axis hyperactivity after maternal separation in rats. Psychoneuroendocrinology, 32(3), 256–266. doi: 10.1016/j.psyneuen.2006.12.013 [DOI] [PubMed] [Google Scholar]
- Albuquerque Filho MO, de Freitas BS, Garcia RC, Crivelaro PC, Schroder N, & de Lima MN (2017). Dual influences of early-life maternal deprivation on histone deacetylase activity and recognition memory in rats. Neuroscience, 344, 360–370. doi: 10.1016/j.neuroscience.2016.12.054 [DOI] [PubMed] [Google Scholar]
- Allen L, & Dwivedi Y (2020). MicroRNA mediators of early life stress vulnerability to depression and suicidal behavior. Molecular psychiatry, 25(2), 308–320. doi: 10.1038/s41380-019-0597-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Allfrey VG, Faulkner R, & Mirsky A (1964). Acetylation and methylation of histones and their possible role in the regulation of RNA synthesis. Proceedings of the National Academy of Sciences, 51(5), 786–794. doi: 10.1073/pnas.51.5.786 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Anacker C, O’Donnell K, & Meaney M (2014). Early life adversity and the epigenetic programming of hypothalamic-pituitary-adrenal function. Dialogues in clinical neuroscience, 16(3), 321–333. doi: 10.31887/DCNS.2014.16.3/canacker [DOI] [PMC free article] [PubMed] [Google Scholar]
- Avishai-Eliner S, Eghbal-Ahmadi M, Tabachnik E, Brunson KL, & Baram TZ (2001). Down-regulation of hypothalamic corticotropin-releasing hormone messenger ribonucleic acid (mRNA) precedes early-life experience-induced changes in hippocampal glucocorticoid receptor mRNA. Endocrinology, 142(1), 89–97. doi: 10.1210/endo.142.1.7917 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barnow S, Lucht M, & Freyberger HJ (2001). Influence of punishment, emotional rejection, child abuse, and broken home on aggression in adolescence: an examination of aggressive adolescents in Germany. Psychopathology, 34(4), 167–173. doi: 10.1159/000049302 [DOI] [PubMed] [Google Scholar]
- Bath KG, & Pimentel T (2017). Effect of early postnatal exposure to valproate on neurobehavioral development and regional BDNF expression in two strains of mice. Epilepsy & Behavior, 70(Pt A), 110–117. doi: 10.1016/j.yebeh.2017.02.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berardo LR, Fabio MC, & Pautassi RM (2016). Post-weaning Environmental Enrichment, But Not Chronic Maternal Isolation, Enhanced Ethanol Intake during Periadolescence and Early Adulthood. Frontiers in behavioral neuroscience, 10, 195. doi: 10.3389/fnbeh.2016.00195 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bird A (1986). CpG-rich islands and the function of DNA methylation. Nature, 321(6067), 209–213. doi: 10.1038/321209a0 [DOI] [PubMed] [Google Scholar]
- Bird A (2002). DNA methylation patterns and epigenetic memory. Genes & Development, 16(1), 6–21. doi: 10.1101/gad.947102 [DOI] [PubMed] [Google Scholar]
- Blaze J, & Roth TL (2017). Caregiver maltreatment causes altered neuronal DNA methylation in female rodents. Development and psychopathology, 29(2), 477–489. doi: 10.1017/S0954579417000128 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bohnsack JP, Zhang H, Wandling GM, He D, Kyzar EJ, Lasek AW, & Pandey SC (2022). Targeted epigenomic editing ameliorates adult anxiety and excessive drinking after adolescent alcohol exposure. Science Advances, 8(18), eabn2748. doi: 10.1126/sciadv.abn2748 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonapersona V, Kentrop J, Van Lissa CJ, van der Veen R, Joels M, & Sarabdjitsingh RA (2019). The behavioral phenotype of early life adversity: A 3-level meta-analysis of rodent studies. Neuroscience & Biobehavioral Reviews, 102, 299–307. doi: 10.1016/j.neubiorev.2019.04.021 [DOI] [PubMed] [Google Scholar]
- Boyes J, & Bird A (1992). Repression of genes by DNA methylation depends on CpG density and promoter strength: evidence for involvement of a methyl-CpG binding protein. The EMBO journal, 11(1), 327–333. doi: 10.1002/j.1460-2075.1992.tb05055.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Branchi I, Karpova NN, D’Andrea I, Castren E, & Alleva E (2011). Epigenetic modifications induced by early enrichment are associated with changes in timing of induction of BDNF expression. Neuroscience Letters, 495(3), 168–172. doi: 10.1016/j.neulet.2011.03.038 [DOI] [PubMed] [Google Scholar]
- Bredy T, & Barad M (2008). The histone deacetylase inhibitor valproic acid enhances acquisition, extinction, and reconsolidation of conditioned fear. Learning & Memory, 15(1), 39–45. doi: 10.1101/lm.801108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bredy T, Humpartzoomian R, Cain D, & Meaney M (2003). Partial reversal of the effect of maternal care on cognitive function through environmental enrichment. Neuroscience, 118(2), 571–576. doi: 10.1016/S0306-4522(02)00918-1 [DOI] [PubMed] [Google Scholar]
- Bredy T, Zhang TY, Grant RJ, Diorio J, & Meaney MJ (2004). Peripubertal environmental enrichment reverses the effects of maternal care on hippocampal development and glutamate receptor subunit expression. European Journal of Neuroscience, 20(5), 1355–1362. doi: 10.1111/j.1460-9568.2004.03599.x [DOI] [PubMed] [Google Scholar]
- Burenkova O, Aleksandrova E, & Zaraiskaya I (2014a). Administration of histone deacetylase inhibitor during neonatal period changes emotionality of adult male 129Sv mice. Bulletin of Experimental Biology and Medicine, 156(5), 620–623. doi: 10.1007/s10517-014-2409-0 [DOI] [PubMed] [Google Scholar]
- Burenkova O, Aleksandrova E, & Zaraiskaya I (2014b). Gender-Dependent Actions of the Histone Deacetylase Blocker Sodium Valproate on Olfactory Learning in 129Sv Mice during the Early Postnatal Period. Neuroscience and Behavioral Physiology, 44, 1008–1013. doi: 10.1007/s11055-014-0017-0 [DOI] [Google Scholar]
- Burenkova O, Aleksandrova E, & Zarayskaya I (2012). Maternal Deprivation in Early Ontogeny Impairs Olfactory Learning with Mother’s Grooming Imitation in 129Sv Mice. Bulletin of Experimental Biology and Medicine, 153, 761–763. doi: 10.1007/s10517-012-1820-7 [DOI] [PubMed] [Google Scholar]
- Burenkova O, Aleksandrova E, & Zarayskaya I (2015). Effects of histone deacetylase inhibitor sodium valproate on the physical and behavioral development of 129SV mice. Patologicheskaia Fiziologiia i Eksperimental’naia Terapiia, 59(2), 40–45. [PubMed] [Google Scholar]
- Burenkova O, Aleksandrova E, & Zarayskaya I (2019). Effects of early-life stress and HDAC inhibition on maternal behavior in mice. Behavioral Neuroscience, 133(1), 39–49. doi: 10.1037/bne0000284 [DOI] [PubMed] [Google Scholar]
- Burenkova O, Averkina A, Aleksandrova E, & Zarayskaya IY (2020). Brief but enough: 45-min maternal separation elicits behavioral and physiological responses in neonatal mice and changes in dam maternal behavior. Physiology & behavior, 222, 112877. doi: 10.1016/j.physbeh.2020.112877 [DOI] [PubMed] [Google Scholar]
- Burenkova O, Podturkin A, Naumova O, Hein S, Li N, Cicchetti D, … Grigorenko E (2021). Neuroendocrine and autonomic stress systems activity in young adults raised by mothers with mental health and substance abuse problems: A prospective cohort study. Developmental psychobiology, 63(8), e22213. doi: 10.1002/dev.22213 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Caldji C, Francis D, Sharma S, Plotsky PM, & Meaney MJ (2000). The effects of early rearing environment on the development of GABAA and central benzodiazepine receptor levels and novelty-induced fearfulness in the rat. Neuropsychopharmacology, 22(3), 219–229. doi: 10.1016/S0893-133X(99)00110-4 [DOI] [PubMed] [Google Scholar]
- Caldji C, Tannenbaum B, Sharma S, Francis D, Plotsky PM, & Meaney MJ (1998). Maternal care during infancy regulates the development of neural systems mediating the expression of fearfulness in the rat. Proceedings of the National Academy of Sciences, 95(9), 5335–5340. doi: 10.1073/pnas.95.9.5335 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cervoni N, & Szyf M (2001). Demethylase activity is directed by histone acetylation. Journal of Biological Chemistry, 276(44), 40778–40787. doi: 10.1074/jbc.M103921200 [DOI] [PubMed] [Google Scholar]
- Champagne D, Bagot R, van Hasselt F, Ramakers G, Meaney M, de Kloet E, … Krugers H (2008). Maternal care and hippocampal plasticity: evidence for experience-dependent structural plasticity, altered synaptic functioning, and differential responsiveness to glucocorticoids and stress. Journal of Neuroscience, 28(23), 6037–6045. doi: 10.1523/JNEUROSCI.0526-08.2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Champagne F, Diorio J, Sharma S, & Meaney MJ (2001). Naturally occurring variations in maternal behavior in the rat are associated with differences in estrogen-inducible central oxytocin receptors. Proceedings of the National Academy of Sciences, 98(22), 12736–12741. doi: 10.1073/pnas.221224598 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Champagne F, Francis D, Mar A, & Meaney M (2003). Variations in maternal care in the rat as a mediating influence for the effects of environment on development. Physiology & behavior, 79(3), 359–371. doi: 10.1016/s0031-9384(03)00149-5 [DOI] [PubMed] [Google Scholar]
- Chen J, Evans AN, Liu Y, Honda M, Saavedra JM, & Aguilera G (2012). Maternal deprivation in rats is associated with corticotrophin‐releasing hormone (CRH) promoter hypomethylation and enhances CRH transcriptional responses to stress in adulthood. Journal of neuroendocrinology, 24(7), 1055–1064. doi: 10.1111/j.1365-2826.2012.02306.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Y, Sprung R, Tang Y, Ball H, Sangras B, Kim SC, … Zhao Y (2007). Lysine propionylation and butyrylation are novel post-translational modifications in histones. Molecular & Cellular Proteomics, 6(5), 812–819. doi: 10.1074/mcp.M700021-MCP200 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Z, & Pikaard C (1997). Epigenetic silencing of RNA polymerase I transcription: a role for DNA methylation and histone modification in nucleolar dominance. Genes & Development, 11(16), 2124–2136. doi: 10.1101/gad.11.16.2124 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cirulli F, Santucci D, Laviola G, Alleva E, & Levine S (1994). Behavioral and hormonal responses to stress in the newborn mouse: effects of maternal deprivation and chlordiazepoxide. Developmental psychobiology, 27(5), 301–316. doi: 10.1002/dev.420270505 [DOI] [PubMed] [Google Scholar]
- Clayton AL, Hazzalin CA, & Mahadevan LC (2006). Enhanced histone acetylation and transcription: a dynamic perspective. Molecular Cell, 23(3), 289–296. doi: 10.1016/j.molcel.2006.06.017 [DOI] [PubMed] [Google Scholar]
- Collins N, Campbell T, Donoghue K, Ghosh U, Smith J, O’Shea M, … Roth T (2023). Early life stress and the role of environmental and molecular moderators in the ontology of pathological and resilient behavioral phenotypes. Molecular Psychology, 1, 3. doi: 10.12688/molpsychol.17482.2 [DOI] [Google Scholar]
- Collins N, Zimmerman CW, Phillips NLH, Fern S, Doherty TS, & Roth TL (2022). Developmental administration of valproic acid alters DNA methylation and maternal behavior. Developmental psychobiology, 64(3), e22231. doi: 10.1002/dev.22231 [DOI] [PubMed] [Google Scholar]
- Cordier JM, Aguggia JP, Danelon V, Mir FR, Rivarola MA, & Masco D (2021). Postweaning Enriched Environment Enhances Cognitive Function and Brain-Derived Neurotrophic Factor Signaling in the Hippocampus in Maternally Separated Rats. Neuroscience, 453, 138–147. doi: 10.1016/j.neuroscience.2020.09.058 [DOI] [PubMed] [Google Scholar]
- Cruz FC, Quadros IM, Planeta C, & Miczek KA (2008). Maternal separation stress in male mice: long-term increases in alcohol intake. Psychopharmacology, 201(3), 459–468. doi: 10.1007/s00213-008-1307-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- D’Amato FR, Cabib S, Puglisi-Allegra S, Patacchioli FR, Cigliana G, Maccari S, & Angelucci L (1992). Effects of acute and repeated exposure to stress on the hypothalamo-pituitary-adrenocortical activity in mice during postnatal development. Hormones and Behavior, 26(4), 474–485. doi: 10.1016/0018-506x(92)90015-n [DOI] [PubMed] [Google Scholar]
- Dandi E, Kalamari A, Touloumi O, Lagoudaki R, Nousiopoulou E, Simeonidou C, … Tata DA (2018). Beneficial effects of environmental enrichment on behavior, stress reactivity and synaptophysin/BDNF expression in hippocampus following early life stress. International Journal of Developmental Neuroscience, 67, 19–32. doi: 10.1016/j.ijdevneu.2018.03.003 [DOI] [PubMed] [Google Scholar]
- Daskalakis NP, Bagot RC, Parker KJ, Vinkers CH, & de Kloet ER (2013). The three-hit concept of vulnerability and resilience: toward understanding adaptation to early-life adversity outcome. Psychoneuroendocrinology, 38, 1858–1873. doi: 10.1016/j.psyneuen.2013.06.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davis LK, Bolton JL, Hanson H, & Guarraci FA (2020). Modified limited bedding and nesting is a model of early-life stress that affects reproductive physiology and behavior in female and male Long-Evans rats. Physiology & behavior, 224, 113037. doi: 10.1016/j.physbeh.2020.113037 [DOI] [PubMed] [Google Scholar]
- Day JJ (2014). New approaches to manipulating the epigenome. Dialogues in clinical neuroscience, 16(3), 345–357. doi: 10.31887/DCNS.2014.16.3/jday [DOI] [PMC free article] [PubMed] [Google Scholar]
- Day JJ, & Sweatt JD (2011). Epigenetic mechanisms in cognition. Neuron, 70(5), 813–829. doi: 10.1016/j.neuron.2011.05.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Kloet ER, Rosenfeld P, Van Eekelen JA, Sutanto W, & Levine S (1988). Stress, glucocorticoids and development. Progress in Brain Research, 73, 101–120. doi: 10.1016/S0079-6123(08)60500-2 [DOI] [PubMed] [Google Scholar]
- Denenberg VH, & Haltmeyer GC (1967). Test of the monotonicity hypothesis concerning infantile stimulation and emotional reactivity. Journal of Comparative and Physiological Psychology, 63(3), 394–396. doi: 10.1037/h0024616 [DOI] [PubMed] [Google Scholar]
- Dion MF, Altschuler SJ, Wu LF, & Rando OJ (2005). Genomic characterization reveals a simple histone H4 acetylation code. Proceedings of the National Academy of Sciences, 102(15), 5501–5506. doi: 10.1073/pnas.0500136102 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doherty TS, Chajes JR, Reich L, Duffy HBD, & Roth TL (2019). Preventing epigenetic traces of caregiver maltreatment: A role for HDAC inhibition. International Journal of Developmental Neuroscience, 78, 178–184. doi: 10.1016/j.ijdevneu.2019.05.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dorigo B, Schalch T, Bystricky K, & Richmond TJ (2003). Chromatin fiber folding: requirement for the histone H4 N-terminal tail. Journal of Molecular Biology, 327(1), 85–96. doi: 10.1016/s0022-2836(03)00025-1 [DOI] [PubMed] [Google Scholar]
- Duque-Quintero M, Hooijmans CR, Hurowitz A, Ahmed A, Barris B, Homberg JR, … Atsak P (2022). Enduring effects of early-life adversity on reward processes: A systematic review and meta-analysis of animal studies. Neuroscience & Biobehavioral Reviews, 142, 104849. doi: 10.1016/j.neubiorev.2022.104849 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eden S, Hashimshony T, Keshet I, Cedar H, & Thorne A (1998). DNA methylation models histone acetylation. Nature, 394(6696), 842–842. doi: 10.1038/29680 [DOI] [PubMed] [Google Scholar]
- Elnahas EM, Abuelezz SA, Mohamad MI, Nabil MM, Abdelraouf SM, Bahaa N, … Aboul-Fotouh S (2021). Validation of prenatal versus postnatal valproic acid rat models of autism: A behavioral and neurobiological study. Progress in Neuro-Psychopharmacology and Biological Psychiatry, 108, 110185. doi: 10.1016/j.pnpbp.2020.110185 [DOI] [PubMed] [Google Scholar]
- Enthoven L, Schmidt M, Cheung Y, van der Mark M, De Kloet E, & Oitzl M (2010). Ontogeny of the HPA axis of the CD1 mouse following 24 h maternal deprivation at pnd 3. International Journal of Developmental Neuroscience, 28(2), 217–224. doi: 10.1016/j.ijdevneu.2009.10.006 [DOI] [PubMed] [Google Scholar]
- Fort DG, Herr TM, Shaw PL, Gutzman KE, & Starren JB (2017). Mapping the evolving definitions of translational research. Journal of Clinical and Translational Science, 1, 60–66. doi: 10.1017/cts.2016.10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Francis D, Diorio J, Liu D, & Meaney M (1999). Nongenomic transmission across generations of maternal behavior and stress responses in the rat. Science, 286(5442), 1155–1158. doi: 10.1126/science.286.5442.1155 [DOI] [PubMed] [Google Scholar]
- Francis D, Diorio J, Plotsky P, & Meaney M (2002). Environmental enrichment reverses the effects of maternal separation on stress reactivity. Journal of Neuroscience, 22(18), 7840–7843. doi: 10.1523/JNEUROSCI.22-18-07840.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Franklin TB, Russig H, Weiss IC, Graff J, Linder N, Michalon A, … Mansuy IM (2010). Epigenetic transmission of the impact of early stress across generations. Biological Psychiatry, 68(5), 408–415. doi: 10.1016/j.biopsych.2010.05.036 [DOI] [PubMed] [Google Scholar]
- Gedzun V, Svinov M, Sarycheva NY, Shlapakova P, Dovbnyuk K, & Dubynin V (2021). Effects of Prenatal and Early Postnatal Administration of Valproate on Behavior and Cyhtological Characteristics in Wistar Rats. Neuroscience and Behavioral Physiology, 51, 639–647. doi: 10.1007/s11055-021-01117-y [DOI] [Google Scholar]
- Gluckman PD, & Hanson MA (2004). Living with the past: evolution, development, and patterns of disease. Science, 305(5691), 1733–1736. doi: 10.1126/science.1095292 [DOI] [PubMed] [Google Scholar]
- Golderer G, & Gröbner P (1991). ADP-ribosylation of core histones and their acetylated subspecies. Biochemical Journal, 277(3), 607–610. doi: 10.1042/bj2770607 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goodman JB, Freeman EE, & Chalmers KA (2019). The relationship between early life stress and working memory in adulthood: A systematic review and meta-analysis. Memory, 27(6), 868–880. doi: 10.1080/09658211.2018.1561897 [DOI] [PubMed] [Google Scholar]
- Grinkevich L (2014). Epigenetics and the formation of long-term memory. Neuroscience and Behavioral Physiology, 44, 200–213. doi:0097–0549/14/4402–0200 [Google Scholar]
- Guan JS, Haggarty SJ, Giacometti E, Dannenberg JH, Joseph N, Gao J, … Tsai LH (2009). HDAC2 negatively regulates memory formation and synaptic plasticity. Nature, 459(7243), 55–60. doi: 10.1038/nature07925 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gugliucci A (1994). Advanced glycation of rat liver histone octamers: an in vitro study. Biochemical and Biophysical Research Communications, 203(1), 588–593. doi: 10.1006/bbrc.1994.2223 [DOI] [PubMed] [Google Scholar]
- Gunnar MR, Brodersen L, Krueger K, & Rigatuso J (1996). Dampening of adrenocortical responses during infancy: normative changes and individual differences. Child Development, 67(3), 877–889. doi: 10.1111/j.1467-8624.1996.tb01770.x [DOI] [PubMed] [Google Scholar]
- Ham GX, Lim KE, Augustine GJ, & Leong V (2023). Synchrony in parent-offspring social interactions across development: A cross-species review of rodents and humans. Journal of Neuroendocrinology, 35, e13241. doi: 10.1111/jne.13241 [DOI] [PubMed] [Google Scholar]
- Heijmans BT, Tobi EW, Stein AD, Putter H, Blauw GJ, Susser ES, … Lumey LH (2008). Persistent epigenetic differences associated with prenatal exposure to famine in humans. Proceedings of the National Academy of Sciences, 105, 17046–17049. doi: 10.1073/pnas.0806560105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hertzman C (2012). Putting the concept of biological embedding in historical perspective. Proceedings of the National Academy of Sciences, 109 Suppl 2(Suppl 2), 17160–17167. doi: 10.1073/pnas.1202203109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hilton IB, D’Ippolito AM, Vockley CM, Thakore PI, Crawford GE, Reddy TE, & Gersbach CA (2015). Epigenome editing by a CRISPR-Cas9-based acetyltransferase activates genes from promoters and enhancers. Nature biotechnology, 33(5), 510–517. doi: 10.1038/nbt.3199 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hirabayashi Y, & Gotoh Y (2010). Epigenetic control of neural precursor cell fate during development. Nature Reviews Neuroscience, 11(6), 377–388. doi: 10.1038/nrn2810 [DOI] [PubMed] [Google Scholar]
- Hong L, Schroth G, Matthews H, Yau P, & Bradbury E (1993). Studies of the DNA binding properties of histone H4 amino terminus. Thermal denaturation studies reveal that acetylation markedly reduces the binding constant of the H4 “tail” to DNA. Journal of Biological Chemistry, 268(1), 305–314. doi: 10.1016/S0021-9258(18)54150-8 [DOI] [PubMed] [Google Scholar]
- Hostinar CE, Sullivan RM, & Gunnar MR (2014). Psychobiological mechanisms underlying the social buffering of the hypothalamic–pituitary–adrenocortical axis: A review of animal models and human studies across development. Psychological Bulletin, 140(1), 256. doi: 10.1037/a0032671 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hsieh C (1994). Dependence of transcriptional repression on CpG methylation density. Molecular and Cellular Biology, 14(8), 5487–5494. doi: 10.1128/mcb.14.8.5487-5494.1994 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hsieh J, & Gage FH (2005). Chromatin remodeling in neural development and plasticity. Current Opinion in Cell Biology, 17(6), 664–671. doi: 10.1016/j.ceb.2005.09.002 [DOI] [PubMed] [Google Scholar]
- Huot RL, Plotsky PM, Lenox RH, & McNamara RK (2002). Neonatal maternal separation reduces hippocampal mossy fiber density in adult Long Evans rats. Brain Research, 950(1–2), 52–63. doi: 10.1016/s0006-8993(02)02985-2 [DOI] [PubMed] [Google Scholar]
- Huot RL, Thrivikraman K, Meaney MJ, & Plotsky PM (2001). Development of adult ethanol preference and anxiety as a consequence of neonatal maternal separation in Long Evans rats and reversal with antidepressant treatment. Psychopharmacology, 158, 366–373. doi: 10.1007/s002130100701 [DOI] [PubMed] [Google Scholar]
- Jacobson L, & Sapolsky R (1991). The role of the hippocampus in feedback regulation of the hypothalamic-pituitary-adrenocortical axis. Endocrine Reviews, 12(2), 118–134. doi: 10.1210/edrv-12-2-118 [DOI] [PubMed] [Google Scholar]
- Jakubowski KP, Cundiff JM, & Matthews KA (2018). Cumulative childhood adversity and adult cardiometabolic disease: A meta-analysis. Health Psychology, 37(8), 701–715. doi: 10.1037/hea0000637 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jeanneteau FD, Lambert WM, Ismaili N, Bath KG, Lee FS, Garabedian MJ, & Chao MV (2012). BDNF and glucocorticoids regulate corticotrophin-releasing hormone (CRH) homeostasis in the hypothalamus. Proceedings of the National Academy of Sciences, 109(4), 1305–1310. doi: 10.1073/pnas.1114122109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Joëls M, Kraaijenvanger E, Sarabdjitsingh RA, & Bonapersona V (2022). Structural changes after early life adversity in rodents: a systematic review with meta-analysis. bioRxiv. doi: 10.1101/2022.01.17.476580v1 [DOI] [Google Scholar]
- Johannessen CU, & Johannessen SI (2003). Valproate: past, present, and future. CNS Drug Reviews, 9(2), 199–216. doi: 10.1111/j.1527-3458.2003.tb00249.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jones PL, Veenstra GJ, Wade PA, Vermaak D, Kass SU, Landsberger N, … Wolffe AP (1998). Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription. Nature Genetics, 19(2), 187–191. doi: 10.1038/561 [DOI] [PubMed] [Google Scholar]
- Kalinichev M, Easterling KW, Plotsky PM, & Holtzman SG (2002). Long-lasting changes in stress-induced corticosterone response and anxiety-like behaviors as a consequence of neonatal maternal separation in Long–Evans rats. Pharmacology Biochemistry and Behavior, 73(1), 131–140. doi: 10.1016/S0091-3057(02)00781-5 [DOI] [PubMed] [Google Scholar]
- Kaplan GA, Turrell G, Lynch JW, Everson SA, Helkala EL, & Salonen JT (2001). Childhood socioeconomic position and cognitive function in adulthood. International Journal of Epidemiology, 30(2), 256–263. doi: 10.1093/ije/30.2.256 [DOI] [PubMed] [Google Scholar]
- Kappeler L, & Meaney MJ (2010). Epigenetics and parental effects. Bioessays, 32(9), 818–827. doi: 10.1002/bies.201000015 [DOI] [PubMed] [Google Scholar]
- Keller SM, Doherty TS, & Roth TL (2019). Pharmacological manipulation of DNA methylation normalizes maternal behavior, DNA methylation, and gene expression in dams with a history of maltreatment. Scientific Reports, 9(1), 10253. doi: 10.1038/s41598-019-46539-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kelly RD, & Cowley SM (2013). The physiological roles of histone deacetylase (HDAC) 1 and 2: complex co-stars with multiple leading parts. Biochemical Society Transactions, 41(3), 741–749. doi: 10.1042/BST20130010 [DOI] [PubMed] [Google Scholar]
- Kember R, Dempster E, Lee T, Schalkwyk L, Mill J, & Fernandes C (2012). Maternal separation is associated with strain‐specific responses to stress and epigenetic alterations to Nr3c1, Avp, and Nr4a1 in mouse. Brain and behavior, 2(4), 455–467. doi: 10.1002/brb3.69 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kessler RC, McLaughlin KA, Green JG, Gruber MJ, Sampson NA, Zaslavsky AM, … Williams DR (2010). Childhood adversities and adult psychopathology in the WHO World Mental Health Surveys. The British Journal of Psychiatry, 197(5), 378–385. doi: 10.1192/bjp.bp.110.080499 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim KC, Kim P, Go HS, Choi CS, Yang S-I, Cheong JH, … Ko KH (2011). The critical period of valproate exposure to induce autistic symptoms in Sprague–Dawley rats. Toxicology Letters, 201(2), 137–142. doi: 10.1016/j.toxlet.2010.12.018 [DOI] [PubMed] [Google Scholar]
- Korzus E, Rosenfeld MG, & Mayford M (2004). CBP histone acetyltransferase activity is a critical component of memory consolidation. Neuron, 42(6), 961–972. doi: 10.1016/j.neuron.2004.06.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kosten TA, Lee HJ, & Kim JJ (2007). Neonatal handling alters learning in adult male and female rats in a task-specific manner. Brain Research, 1154, 144–153. doi: 10.1016/j.brainres.2007.03.081 [DOI] [PubMed] [Google Scholar]
- Kotenkova E, Meshkova N, & Shutova M (1989). On rats and mice: Nauka. [Google Scholar]
- Kruchenkova E (2009). Maternal behavior in mammals: Krasand. [Google Scholar]
- Kuhn CM, Pauk J, & Schanberg SM (1990). Endocrine responses to mother-infant separation in developing rats. Developmental psychobiology, 23(5), 395–410. doi: 10.1002/dev.420230503 [DOI] [PubMed] [Google Scholar]
- Kundakovic M, & Champagne FA (2011). Epigenetic perspective on the developmental effects of bisphenol A. Brain, Behavior, and Immunity, 25, 1084–1093. doi: 10.1016/j.bbi.2011.02.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kundakovic M, Gudsnuk K, Herbstman JB, Tang D, Perera FP, & Champagne FA (2015). DNA methylation of BDNF as a biomarker of early-life adversity. Proceedings of the National Academy of Sciences, 112, 6807–6813. doi: 10.1073/pnas.1408355111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kundakovic M, Lim S, Gudsnuk K, & Champagne FA (2013). Sex-specific and strain-dependent effects of early life adversity on behavioral and epigenetic outcomes. Frontiers in Psychiatry, 4, 78. doi: 10.3389/fpsyt.2013.00078 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Labonté B, Suderman M, Maussion G, Navaro L, Yerko V, Mahar I, … Meaney MJ (2012). Genome-wide epigenetic regulation by early-life trauma. Archives of general psychiatry, 69(7), 722–731. doi: 10.1001/archgenpsychiatry.2011.2287 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ladd CO, Huot RL, Thrivikraman KV, Nemeroff CB, & Plotsky PM (2004). Long-term adaptations in glucocorticoid receptor and mineralocorticoid receptor mRNA and negative feedback on the hypothalamo-pituitary-adrenal axis following neonatal maternal separation. Biological Psychiatry, 55(4), 367–375. doi: 10.1016/j.biopsych.2003.10.007 [DOI] [PubMed] [Google Scholar]
- Larson MC, White BP, Cochran A, Donzella B, & Gunnar M (1998). Dampening of the cortisol response to handling at 3 months in human infants and its relation to sleep, circadian cortisol activity, and behavioral distress. Developmental Psychobiology: The Journal of the International Society for Developmental Psychobiology, 33(4), 327–337. doi: [DOI] [PubMed] [Google Scholar]
- Lee D, Hayes J, Pruss D, & Wolffe A (1993). A positive role for histone acetylation in transcription factor access to nucleosomal DNA. Cell, 72(1), 73–84. doi: 10.1016/0092-8674(93)90051-q [DOI] [PubMed] [Google Scholar]
- Lee M, & Williams D (1974). Changes in licking behaviour of rat mother following handling of young. Animal Behaviour, 22(3), 679–681. doi: 10.1016/S0003-3472(74)80016-3 [DOI] [Google Scholar]
- LeMoult J, Humphreys KL, Tracy A, Hoffmeister JA, Ip E, & Gotlib IH (2020). Meta-analysis: Exposure to Early Life Stress and Risk for Depression in Childhood and Adolescence. Journal of the American Academy of Child & Adolescent Psychiatry, 59(7), 842–855. doi: 10.1016/j.jaac.2019.10.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lesuis SL, van Hoek BA, Lucassen PJ, & Krugers HJ (2017). Early postnatal handling reduces hippocampal amyloid plaque formation and enhances cognitive performance in APPswe/PS1dE9 mice at middle age. Neurobiology of Learning and Memory, 144, 27–35. doi: 10.1016/j.nlm.2017.05.016 [DOI] [PubMed] [Google Scholar]
- Levenson JM, O’Riordan KJ, Brown KD, Trinh MA, Molfese DL, & Sweatt JD (2004). Regulation of histone acetylation during memory formation in the hippocampus. Journal of Biological Chemistry, 279(39), 40545–40559. doi: 10.1074/jbc.M402229200 [DOI] [PubMed] [Google Scholar]
- Levine A, Worrell TR, Zimnisky R, & Schmauss C (2012). Early life stress triggers sustained changes in histone deacetylase expression and histone H4 modifications that alter responsiveness to adolescent antidepressant treatment. Neurobiology of disease, 45(1), 488–498. doi: 10.1016/j.nbd.2011.09.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levine S (1956). A further study of infantile handling and adult avoidance learning. Journal of Personality, 25, 70–80. doi: 10.1111/j.1467-6494.1956.tb01289.x [DOI] [PubMed] [Google Scholar]
- Li E (2002). Chromatin modification and epigenetic reprogramming in mammalian development. Nature Reviews Genetics, 3(9), 662–673. doi: 10.1038/nrg887 [DOI] [PubMed] [Google Scholar]
- Liebich HM, Gesele E, Wirth C, Woll J, Jobst K, & Lakatos A (1993). Non-enzymatic glycation of histones. Biological Mass Spectrometry, 22(2), 121–123. doi: 10.1002/bms.1200220204 [DOI] [PubMed] [Google Scholar]
- Lindeyer CM, Meaney MJ, & Reader SM (2013). Early maternal care predicts reliance on social learning about food in adult rats. Developmental psychobiology, 55(2), 168–175. doi: 10.1002/dev.21009 [DOI] [PubMed] [Google Scholar]
- Lipsky RH, & Marini AM (2007). Brain-derived neurotrophic factor in neuronal survival and behavior-related plasticity. Annals of the New York Academy of Sciences, 1122(1), 130–143. doi: 10.1196/annals.1403.009 [DOI] [PubMed] [Google Scholar]
- Lisman J, Buzsaki G, Eichenbaum H, Nadel L, Ranganath C, & Redish AD (2017). Viewpoints: how the hippocampus contributes to memory, navigation and cognition. Nature Neuroscience, 20(11), 1434–1447. doi: 10.1038/nn.4661 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu D, Diorio J, Day JC, Francis DD, & Meaney MJ (2000). Maternal care, hippocampal synaptogenesis and cognitive development in rats. Nature Neuroscience, 3(8), 799–806. doi: 10.1038/77702 [DOI] [PubMed] [Google Scholar]
- Liu D, Diorio J, Tannenbaum B, Caldji C, Francis D, Freedman A, … Meaney MJ (1997). Maternal care, hippocampal glucocorticoid receptors, and hypothalamic-pituitary-adrenal responses to stress. Science, 277(5332), 1659–1662. doi: 10.1126/science.277.5332.1659 [DOI] [PubMed] [Google Scholar]
- Liu X, Wu H, Ji X, Stelzer Y, Wu X, Czauderna S, … Jaenisch R (2016). Editing DNA Methylation in the Mammalian Genome. Cell, 167(1), 233–247 e217. doi: 10.1016/j.cell.2016.08.056 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Llorente-Berzal A, Mela V, Borcel E, Valero M, Lopez-Gallardo M, Viveros MP, & Marco EM (2012). Neurobehavioral and metabolic long-term consequences of neonatal maternal deprivation stress and adolescent olanzapine treatment in male and female rats. Neuropharmacology, 62(3), 1332–1341. doi: 10.1016/j.neuropharm.2011.07.031 [DOI] [PubMed] [Google Scholar]
- Lobanov A, Khokhlova O, Suvorova M, Zaraiskaya I, & Murashev A (2008). Features of somatic maturation and sensorimotor development in C57BL/6 mice in early ontogenesis under prenatal exposure to cytosine arabinose Zhurnal Vysshei Nervnoi Deyatelnosti Imeni I.P. Pavlova, 58(1), 98–110. [PubMed] [Google Scholar]
- Lovic V, Gonzalez A, & Fleming AS (2001). Maternally separated rats show deficits in maternal care in adulthood. Developmental Psychobiology: The Journal of the International Society for Developmental Psychobiology, 39(1), 19–33. doi: 10.1002/dev.1024 [DOI] [PubMed] [Google Scholar]
- Lubin FD, Roth TL, & Sweatt JD (2008). Epigenetic regulation of BDNF gene transcription in the consolidation of fear memory. Journal of Neuroscience, 28(42), 10576–10586. doi: 10.1523/JNEUROSCI.1786-08.2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luger K, Mäder AW, Richmond RK, Sargent DF, & Richmond TJ (1997). Crystal structure of the nucleosome core particle at 2.8 Å resolution. Nature, 389(6648), 251–260. doi: 10.1038/38444 [DOI] [PubMed] [Google Scholar]
- Lyko F (2018). The DNA methyltransferase family: a versatile toolkit for epigenetic regulation. Nature Reviews Genetics, 19(2), 81–92. doi: 10.1038/nrg.2017.80 [DOI] [PubMed] [Google Scholar]
- McCormick CM, Kehoe P, & Kovacs S (1998). Corticosterone release in response to repeated, short episodes of neonatal isolation: evidence of sensitization. International Journal of Developmental Neuroscience, 16(3–4), 175–185. doi: 10.1016/s0736-5748(98)00026-4 [DOI] [PubMed] [Google Scholar]
- McKay MT, Kilmartin L, Meagher A, Cannon M, Healy C, & Clarke MC (2022). A revised and extended systematic review and meta-analysis of the relationship between childhood adversity and adult psychiatric disorder. Journal of Psychiatric Research. doi: 10.1016/j.jpsychires.2022.10.015 [DOI] [PubMed] [Google Scholar]
- McKibben LA, & Dwivedi Y (2021). Early-life stress induces genome-wide sex-dependent miRNA expression and correlation across limbic brain areas in rats. Epigenomics, 13(13), 1031–1056. doi: 10.2217/epi-2021-0037 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meaney M (2001). Maternal care, gene expression, and the transmission of individual differences in stress reactivity across generations. Annual Review of Neuroscience, 24(1), 1161–1192. doi: 10.1146/annurev.neuro.24.1.1161 [DOI] [PubMed] [Google Scholar]
- Meaney M, & Szyf M (2005). Maternal care as a model for experience-dependent chromatin plasticity? Trends in neurosciences, 28(9), 456–463. doi: 10.1016/j.tins.2005.07.006 [DOI] [PubMed] [Google Scholar]
- Meerlo P, Horvath KM, Nagy GM, Bohus B, & Koolhaas JM (1999). The influence of postnatal handling on adult neuroendocrine and behavioural stress reactivity. Journal of Neuroendocrinology, 11(12), 925–933. doi: 10.1046/j.1365-2826.1999.00409.x [DOI] [PubMed] [Google Scholar]
- Menezes J, das Neves B-HS, Gonçalves R, Benetti F, & Mello-Carpes PB (2020). Maternal deprivation impairs memory and cognitive flexibility, effect that is avoided by environmental enrichment. Behavioural Brain Research, 381, 112468. doi: 10.1016/j.bbr.2020.112468 [DOI] [PubMed] [Google Scholar]
- Milagro FI, Campión J, García-Díaz DF, Goyenechea E, Paternain L, & Martínez JA (2009). High fat diet-induced obesity modifies the methylation pattern of leptin promoter in rats. Journal of Physiology and Biochemistry, 65, 1–9. doi: 10.1007/bf03165964 [DOI] [PubMed] [Google Scholar]
- Miller CA, & Sweatt JD (2007). Covalent modification of DNA regulates memory formation. Neuron, 53(6), 857–869. doi: 10.1016/j.neuron.2007.02.022 [DOI] [PubMed] [Google Scholar]
- Moffett MC, Harley J, Francis D, Sanghani SP, Davis WI, & Kuhar MJ (2006). Maternal separation and handling affects cocaine self-administration in both the treated pups as adults and the dams. Journal of Pharmacology and Experimental Therapeutics, 317(3), 1210–1218. doi: 10.1124/jpet.106.101139 [DOI] [PubMed] [Google Scholar]
- Mohammadian J, Najafi M, & Miladi-Gorji H (2019). Effect of enriched environment during adolescence on spatial learning and memory, and voluntary consumption of morphine in maternally separated rats in adulthood. Developmental psychobiology, 61(4), 615–625. doi: 10.1002/dev.21808 [DOI] [PubMed] [Google Scholar]
- Moldrich RX, Leanage G, She D, Dolan-Evans E, Nelson M, Reza N, & Reutens DC (2013). Inhibition of histone deacetylase in utero causes sociability deficits in postnatal mice. Behavioural Brain Research, 257, 253–264. doi: 10.1016/j.bbr.2013.09.049 [DOI] [PubMed] [Google Scholar]
- Mullen PE, Martin JL, Anderson JC, Romans SE, & Herbison GP (1996). The long-term impact of the physical, emotional, and sexual abuse of children: a community study. Child Abuse & Neglect, 20(1), 7–21. doi: 10.1016/0145-2134(95)00112-3 [DOI] [PubMed] [Google Scholar]
- Murray EK, Hien A, de Vries GJ, & Forger NG (2009). Epigenetic control of sexual differentiation of the bed nucleus of the stria terminalis. Endocrinology, 150(9), 4241–4247. doi: 10.1210/en.2009-0458 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nathan D, Sterner DE, & Berger SL (2003). Histone modifications: Now summoning sumoylation. Proceedings of the National Academy of Sciences, 100(23), 13118–13120. doi: 10.1073/pnas.2436173100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nelson CA III, Zeanah CH, Fox NA, Marshall PJ, Smyke AT, & Guthrie D (2007). Cognitive recovery in socially deprived young children: The Bucharest Early Intervention Project. Science, 318(5858), 1937–1940. doi: 10.1126/science.114392 [DOI] [PubMed] [Google Scholar]
- Neves I, Dinis-Oliveira RJ, & Magalhaes T (2021). Epigenomic mediation after adverse childhood experiences: a systematic review and meta-analysis. Forensic Sciences Research, 6(2), 103–114. doi: 10.1080/20961790.2019.1641954 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nowak SJ, & Corces VG (2004). Phosphorylation of histone H3: a balancing act between chromosome condensation and transcriptional activation. TRENDS in Genetics, 20(4), 214–220. doi: 10.1016/j.tig.2004.02.007 [DOI] [PubMed] [Google Scholar]
- Osley MA (2004). H2B ubiquitylation: the end is in sight. Biochimica et Biophysica Acta (BBA)-Gene Structure and Expression, 1677(1–3), 74–78. doi: 10.1016/j.bbaexp.2003.10.013 [DOI] [PubMed] [Google Scholar]
- Parra M (2015). Class II a HDAC s–new insights into their functions in physiology and pathology. The FEBS journal, 282(9), 1736–1744. doi: 10.1111/febs.13061 [DOI] [PubMed] [Google Scholar]
- Pedersen CA, Vadlamudi S, Boccia ML, & Moy SS (2011). Variations in maternal behavior in C57BL/6J mice: behavioral comparisons between adult offspring of high and low pup-licking mothers. Frontiers in psychiatry, 2(42). doi: 10.3389/fpsyt.2011.00042 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pihoker C, Owens MJ, Kuhn CM, Schanberg SM, & Nemeroff CB (1993). Maternal separation in neonatal rats elicits activation of the hypothalamic-pituitary-adrenocortical axis: a putative role for corticotropin-releasing factor. Psychoneuroendocrinology, 18(7), 485–493. doi: 10.1016/0306-4530(93)90042-j [DOI] [PubMed] [Google Scholar]
- Poulton R, Caspi A, Milne BJ, Thomson WM, Taylor A, Sears MR, & Moffitt TE (2002). Association between children’s experience of socioeconomic disadvantage and adult health: a life-course study. The lancet, 360(9346), 1640–1645. doi: 10.1016/S0140-6736(02)11602-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pryce CR, Bettschen D, & Feldon J (2001). Comparison of the effects of early handling and early deprivation on maternal care in the rat. Developmental psychobiology, 38(4), 239–251. doi: 10.1002/dev.1018 [DOI] [PubMed] [Google Scholar]
- Pryce CR, Ruedi-Bettschen D, Dettling AC, Weston A, Russig H, Ferger B, & Feldon J (2005). Long-term effects of early-life environmental manipulations in rodents and primates: Potential animal models in depression research. Neuroscience & Biobehavioral Reviews, 29(4–5), 649–674. doi: 10.1016/j.neubiorev.2005.03.011 [DOI] [PubMed] [Google Scholar]
- Ptashne M (2007). On the use of the word ‘epigenetic’. Current Biology, 17(7), R233–R236. doi: 10.1016/j.cub.2007.02.030. [DOI] [PubMed] [Google Scholar]
- Putignano E, Lonetti G, Cancedda L, Ratto G, Costa M, Maffei L, & Pizzorusso T (2007). Developmental downregulation of histone posttranslational modifications regulates visual cortical plasticity. Neuron, 53(5), 747–759. doi: 10.1016/j.neuron.2007.02.007 [DOI] [PubMed] [Google Scholar]
- Reichert N, Choukrallah MA, & Matthias P (2012). Multiple roles of class I HDACs in proliferation, differentiation, and development. Cellular and Molecular Life Sciences, 69(13), 2173–2187. doi: 10.1007/s00018-012-0921-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reik W, Dean W, & Walter J (2001). Epigenetic reprogramming in mammalian development. Science, 293(5532), 1089–1093. doi: 10.1126/science.1063443 [DOI] [PubMed] [Google Scholar]
- Reuter S, Gupta S, Park B, Goel A, & Aggarwal B (2011). Epigenetic changes induced by curcumin and other natural compounds. Genes and Nutrition, 6, 93–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rocha M, Wang D, Avila-Quintero V, Bloch MH, & Kaffman A (2021). Deficits in hippocampal-dependent memory across different rodent models of early life stress: systematic review and meta-analysis. Translational Psychiatry, 11(1), 231. doi: 10.1038/s41398-021-01352-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rodriguez-Melendez R, & Zempleni J (2003). Regulation of gene expression by biotin (review). The Journal of Nutritional Biochemistry, 14(12), 680–690. doi: 10.1016/j.jnutbio.2003.07.001 [DOI] [PubMed] [Google Scholar]
- Romens SE, McDonald J, Svaren J, & Pollak SD (2015). Associations between early life stress and gene methylation in children. Child development, 86(1), 303–309. doi: 10.1111/cdev.12270 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roth TL, Lubin FD, Funk AJ, & Sweatt JD (2009). Lasting epigenetic influence of early-life adversity on the BDNF gene. Biological Psychiatry, 65(9), 760–769. doi: 10.1016/j.biopsych.2008.11.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roth TL, & Sweatt JD (2009). Regulation of chromatin structure in memory formation. Current Opinion in Neurobiology, 19(3), 336–342. doi: 10.1016/j.conb.2009.05.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sanchez MM, Young LJ, Plotsky PM, & Insel TR (2000). Distribution of corticosteroid receptors in the rhesus brain: relative absence of glucocorticoid receptors in the hippocampal formation. Journal of Neuroscience, 20(12), 4657–4668. doi: 10.1523/JNEUROSCI.20-12-04657.2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sapolsky RM, & Meaney MJ (1986). Maturation of the adrenocortical stress response: neuroendocrine control mechanisms and the stress hyporesponsive period. Brain Research Reviews, 396(1), 64–76. doi: 10.1016/s0006-8993(86)80190-1 [DOI] [PubMed] [Google Scholar]
- Sapozhnikov DM, & Szyf M (2023). Increasing Specificity of Targeted DNA Methylation Editing by Non-Enzymatic CRISPR/dCas9-Based Steric Hindrance. Biomedicines, 11(5), 1238. doi: 10.3390/biomedicines11051238 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schapiro S, Geller E, & Eiduson S (1962). Neonatal adrenal cortical response to stress and vasopressin. Proceedings of the Society for Experimental Biology and Medicine, 109(4), 937–941. doi: 10.3181/00379727-109-27384 [DOI] [PubMed] [Google Scholar]
- Schmidt H, & Duman R (2007). The role of neurotrophic factors in adult hippocampal neurogenesis, antidepressant treatments and animal models of depressive-like behavior. Behavioural pharmacology, 18(5–6), 391–418. doi: 10.1097/FBP.0b013e3282ee2aa8 [DOI] [PubMed] [Google Scholar]
- Schmidt M, Oitzl M, Levine S, & de Kloet E (2002). The HPA system during the postnatal development of CD1 mice and the effects of maternal deprivation. Developmental Brain Research, 139(1), 39–49. doi: 10.1016/s0165-3806(02)00519-9 [DOI] [PubMed] [Google Scholar]
- Schoenfeld NM, Leathem JH, & Rabii J (1980). Maturation of adrenal stress responsiveness in the rat. Neuroendocrinology, 31(2), 101–105. doi: 10.1159/000123058 [DOI] [PubMed] [Google Scholar]
- Seery MD, Holman EA, & Silver RC (2010). Whatever does not kill us: cumulative lifetime adversity, vulnerability, and resilience. Journal of personality and social psychology, 99(6), 1025. doi: 10.1037/a0021344 [DOI] [PubMed] [Google Scholar]
- Seo MK, Ly NN, Lee CH, Cho HY, Choi CM, Lee JG, … Park SW (2016). Early life stress increases stress vulnerability through BDNF gene epigenetic changes in the rat hippocampus. Neuropharmacology, 105, 388–397. doi: 10.1016/j.neuropharm.2016.02.009 [DOI] [PubMed] [Google Scholar]
- Seto E, & Yoshida M (2014). Erasers of histone acetylation: the histone deacetylase enzymes. Cold Spring Harbor perspectives in biology, 6(4), a018713. doi: 10.1101/cshperspect.a018713 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shepard RD, Gouty S, Kassis H, Berenji A, Zhu W, Cox BM, & Nugent FS (2018). Targeting histone deacetylation for recovery of maternal deprivation-induced changes in BDNF and AKAP150 expression in the VTA. Experimental neurology, 309, 160–168. doi: 10.1016/j.expneurol.2018.08.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Silingardi D, Scali M, Belluomini G, & Pizzorusso T (2010). Epigenetic treatments of adult rats promote recovery from visual acuity deficits induced by long-term monocular deprivation. European Journal of Neuroscience, 31(12), 2185–2192. doi: 10.1111/j.1460-9568.2010.07261.x [DOI] [PubMed] [Google Scholar]
- Sinn DI, Kim SJ, Chu K, Jung KH, Lee ST, Song EC, … Roh JK (2007). Valproic acid-mediated neuroprotection in intracerebral hemorrhage via histone deacetylase inhibition and transcriptional activation. Neurobiology of Disease, 26(2), 464–472. doi: 10.1016/j.nbd.2007.02.006 [DOI] [PubMed] [Google Scholar]
- Skinner MK, & Guerrero-Bosagna C (2009). Environmental signals and transgenerational epigenetics. Epigenomics, 1(1), 111–117. doi: 10.2217/epi.09.11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith MA, Kim SY, van Oers HJ, & Levine S (1997). Maternal deprivation and stress induce immediate early genes in the infant rat brain. Endocrinology, 138(11), 4622–4628. doi:10.1210/endo.138.11.5529 10.1016/0018-506X(77)90041-1 [DOI] [PubMed] [Google Scholar]
- Smotherman WP, Brown CP, & Levine S (1977). Maternal responsiveness following differential pup treatment and mother-pup interactions. Hormones and Behavior, 8(2), 242–253. doi: 10.1016/0018-506x(77)90041-1 [DOI] [PubMed] [Google Scholar]
- Stanley JS, Griffin JB, & Zempleni J (2001). Biotinylation of histones in human cells. Effects of cell proliferation. European Journal of Biochemistry, 268(20), 5424–5429. doi: 10.1046/j.0014-2956.2001.02481.x [DOI] [PubMed] [Google Scholar]
- Stefanko DP, Barrett RM, Ly AR, Reolon GK, & Wood MA (2009). Modulation of long-term memory for object recognition via HDAC inhibition. Proceedings of the National Academy of Sciences, 106(23), 9447–9452. doi: 10.1073/pnas.0903964106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sterner DE, & Berger SL (2000). Acetylation of histones and transcription-related factors. Microbiology and Molecular Biology Reviews, 64(2), 435–459. doi: 10.1128/MMBR.64.2.435-459.2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Strahl BD, & Allis CD (2000). The language of covalent histone modifications. Nature, 403(6765), 41–45. doi: 10.1038/47412 [DOI] [PubMed] [Google Scholar]
- Suchecki D, Rosenfeld P, & Levine S (1993). Maternal regulation of the hypothalamic-pituitary-adrenal axis in the infant rat: the roles of feeding and stroking. Developmental brain research, 75(2), 185–192. doi: 10.1016/0165-3806(93)90022-3 [DOI] [PubMed] [Google Scholar]
- Suderman M, Borghol N, Pappas J, Pinto Pereira S, Pembrey M, Hertzman C, … Szyf M (2014). Childhood abuse is associated with methylation of multiple loci in adult DNA. BMC medical genomics, 7, 1–12. doi: 10.1186/1755-8794-7-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suderman M, McGowan PO, Sasaki A, Huang TC, Hallett MT, Meaney MJ, … Szyf M (2012). Conserved epigenetic sensitivity to early life experience in the rat and human hippocampus. Proceedings of the National Academy of Sciences, 109 Suppl 2(Suppl 2), 17266–17272. doi: 10.1073/pnas.1121260109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun ZW, & Allis CD (2002). Ubiquitination of histone H2B regulates H3 methylation and gene silencing in yeast. Nature, 418(6893), 104–108. doi: 10.1038/nature00883 [DOI] [PubMed] [Google Scholar]
- Suor JH, Sturge-Apple ML, Davies PT, Cicchetti D, & Manning LG (2015). Tracing differential pathways of risk: Associations among family adversity, cortisol, and cognitive functioning in childhood. Child Development, 86, 1142–1158. doi: 10.1111/cdev.12376 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suri D, & Vaidya VA (2013). Glucocorticoid regulation of brain-derived neurotrophic factor: relevance to hippocampal structural and functional plasticity. Neuroscience, 239, 196–213. doi: 10.1016/j.neuroscience.2012.08.065 [DOI] [PubMed] [Google Scholar]
- Szyf M, McGowan P, & Meaney MJ (2008). The social environment and the epigenome. Environmental and molecular mutagenesis, 49(1), 46–60. doi: 10.1002/em.20357 [DOI] [PubMed] [Google Scholar]
- Tesone‐Coelho C, Morel LJ, Bhatt J, Estevez L, Naudon L, Giros B, … Daugé V (2015). Vulnerability to opiate intake in maternally deprived rats: implication of MeCP 2 and of histone acetylation. Addiction Biology, 20(1), 120–131. doi: 10.1111/adb.12084 [DOI] [PubMed] [Google Scholar]
- Toropova K, Anokhin K, & Tiunova A (2014). Inhibition of histone deacetylation in the brain modulates the expression of transcription factors c-fos and ZENK and potentiates the formation of long-term memory in newborn chickens. Zhurnal Vysshei Nervnoi Deyatelnosti Imeni I.P. Pavlova, 54(5), 551–561. [PubMed] [Google Scholar]
- Tractenberg SG, Levandowski ML, de Azeredo LA, Orso R, Roithmann LG, Hoffmann ES, … Grassi-Oliveira R (2016). An overview of maternal separation effects on behavioural outcomes in mice: Evidence from a four-stage methodological systematic review. Neuroscience & Biobehavioral Reviews, 68, 489–503. doi: 10.1016/j.neubiorev.2016.06.021 [DOI] [PubMed] [Google Scholar]
- Trotta A, Murray RM, & Fisher HL (2015). The impact of childhood adversity on the persistence of psychotic symptoms: a systematic review and meta-analysis. Psychological medicine, 45(12), 2481–2498. doi: 10.1017/S0033291715000574 [DOI] [PubMed] [Google Scholar]
- Turecki G, & Meaney MJ (2016). Effects of the Social Environment and Stress on Glucocorticoid Receptor Gene Methylation: A Systematic Review. Biological Psychiatry, 79(2), 87–96. doi: 10.1016/j.biopsych.2014.11.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uysal Harzadın N, Ozdemir D, Dayı A, Yalaz G, Baltaci A, & Bediz C (2005). Effects of maternal deprivation on melatonin production and cognition in adolescent male and female rats. Neuroendocrinology Letters, 26(5), 555–560. [PubMed] [Google Scholar]
- Vaiserman A, Voitenko V, & Mekhova L (2011). Epigenetic epidemiology of age-related diseases. Russian Journal of Developmental Biology, 42, 25–42. doi: 10.1134/S1062360411010127 [DOI] [PubMed] [Google Scholar]
- van der Laan AL, & Boenink M (2015). Beyond Bench and Bedside: Disentangling the Concept of Translational Research. Health Care Analysis, 23(1), 32–49. doi: 10.1007/s10728-012-0236-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vanyushin B (2014). Epigenetics today and tomorrow. Russian Journal of Genetics: Applied Research, 4, 168–188. doi: 10.1134/S2079059714030083 [DOI] [Google Scholar]
- Vecsey CG, Hawk JD, Lattal KM, Stein JM, Fabian SA, Attner MA, … Wood MA (2007). Histone deacetylase inhibitors enhance memory and synaptic plasticity via CREB:CBP-dependent transcriptional activation. Journal of Neuroscience, 27(23), 6128–6140. doi: 10.1523/JNEUROSCI.0296-07.2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vetencourt JFM, Tiraboschi E, Spolidoro M, Castrén E, & Maffei L (2011). Serotonin triggers a transient epigenetic mechanism that reinstates adult visual cortex plasticity in rats. European Journal of Neuroscience, 33(1), 49–57. doi: 10.1111/j.1460-9568.2010.07488.x [DOI] [PubMed] [Google Scholar]
- Vettese-Dadey M, Grant PA, Hebbes TR, Crane- Robinson C, Allis CD, & Workman JL (1996). Acetylation of histone H4 plays a primary role in enhancing transcription factor binding to nucleosomal DNA in vitro. EMBO J, 15(10), 2508–2518. doi: 10.1002/j.1460-2075.1996.tb00608.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vitolo JM, Thiriet C, & Hayes JJ (2000). The H3-H4 N-terminal tail domains are the primary mediators of transcription factor IIIA access to 5S DNA within a nucleosome. Molecular and Cellular Biology, 20(6), 2167–2175. doi: 10.1128/MCB.20.6.2167-2175.2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Waddington CH (1940). Organisers and genes. Organisers and genes. [Google Scholar]
- Walker CD, Perrin M, Vale W, & Rivier C (1986). Ontogeny of the stress response in the rat: role of the pituitary and the hypothalamus. Endocrinology, 118(4), 1445–1451. doi: 10.1210/endo-118-4-1445 [DOI] [PubMed] [Google Scholar]
- Wang D, Levine JLS, Avila-Quintero V, Bloch M, & Kaffman A (2020). Systematic review and meta-analysis: effects of maternal separation on anxiety-like behavior in rodents. Translational Psychiatry, 10(1), 174. doi: 10.1038/s41398-020-0856-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Z, Zang C, Rosenfeld JA, Schones DE, Barski A, Cuddapah S, … Zhao K (2008). Combinatorial patterns of histone acetylations and methylations in the human genome. Nature Genetics, 40(7), 897–903. doi: 10.1038/ng.154 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Waterland RA, & Jirtle RL (2003). Transposable elements: targets for early nutritional effects on epigenetic gene regulation. Molecular and Cellular Biology, 23, 5293–5300. doi: 10.1128/mcb.23.15.5293-5300.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weaver IC, Cervoni N, Champagne FA, D’Alessio AC, Sharma S, Seckl JR, … Meaney MJ (2004). Epigenetic programming by maternal behavior. Nature Neuroscience, 7(8), 847–854. doi: 10.1038/nn1276 [DOI] [PubMed] [Google Scholar]
- Weaver IC, Champagne FA, Brown SE, Dymov S, Sharma S, Meaney MJ, & Szyf M (2005). Reversal of maternal programming of stress responses in adult offspring through methyl supplementation: altering epigenetic marking later in life. Journal of Neuroscience, 25(47), 11045–11054. doi: 10.1523/JNEUROSCI.3652-05.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weaver IC, D’Alessio AC, Brown SE, Hellstrom IC, Dymov S, Sharma S, … Meaney MJ (2007). The transcription factor nerve growth factor-inducible protein a mediates epigenetic programming: altering epigenetic marks by immediate-early genes. Journal of Neuroscience, 27(7), 1756–1768. doi: 10.1523/JNEUROSCI.4164-06.2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weaver IC, Korgan AC, Lee K, Wheeler RV, Hundert AS, & Goguen D (2017). Stress and the Emerging Roles of Chromatin Remodeling in Signal Integration and Stable Transmission of Reversible Phenotypes. Front Behav Neurosci, 11, 41. doi: 10.3389/fnbeh.2017.00041 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weaver IC, Meaney MJ, & Szyf M (2006). Maternal care effects on the hippocampal transcriptome and anxiety-mediated behaviors in the offspring that are reversible in adulthood. Proceedings of the National Academy of Sciences, 103(9), 3480–3485. doi: 10.1073/pnas.050752610 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wei L, David A, Duman RS, Anisman H, & Kaffman A (2010). Early life stress increases anxiety-like behavior in Balbc mice despite a compensatory increase in levels of postnatal maternal care. Hormones and Behavior, 57(4–5), 396–404. doi: 10.1016/j.yhbeh.2010.01.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Whinn KS, Kaur G, Lewis JS, Schauer GD, Mueller SH, Jergic S, … Bruchez MP (2019). Nuclease dead Cas9 is a programmable roadblock for DNA replication. Scientific Reports, 9(1), 13292. doi: 10.1038/s41598-019-49837-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wickrama K, Conger RD, & Abraham WT (2005). Early adversity and later health: The intergenerational transmission of adversity through mental disorder and physical illness. The Journals of Gerontology Series B: Psychological Sciences and Social Sciences, 60(Special_Issue_2), S125–S129. doi: 10.1093/geronb/60.Special_Issue_2.S125 [DOI] [PubMed] [Google Scholar]
- Wondrak GT, Cervantes-Laurean D, Jacobson EL, & Jacobson MK (2000). Histone carbonylation in vivo and in vitro. Biochemical Journal, 351 Pt 3(Pt 3), 769–777. doi: 10.1042/bj3510769 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yamada K, Mizuno M, & Nabeshima T (2002). Role for brain-derived neurotrophic factor in learning and memory. Life Sciences, 70(7), 735–744. doi: 10.1016/s0024-3205(01)01461-8 [DOI] [PubMed] [Google Scholar]
- Yehuda R, Daskalakis NP, Lehrner A, Desarnaud F, Bader HN, Makotkine I, … Meaney MJ (2014). Influences of maternal and paternal PTSD on epigenetic regulation of the glucocorticoid receptor gene in Holocaust survivor offspring. American Journal of Psychiatry, 171(8), 872–880. doi: 10.1176/appi.ajp.2014.13121571 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yoshii A, & Constantine-Paton M (2010). Postsynaptic BDNF-TrkB signaling in synapse maturation, plasticity, and disease. Dev Neurobiol, 70(5), 304–322. doi: 10.1002/dneu.20765 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang T, Labonte B, Wen XL, Turecki G, & Meaney MJ (2013). Epigenetic mechanisms for the early environmental regulation of hippocampal glucocorticoid receptor gene expression in rodents and humans. Neuropsychopharmacology, 38(1), 111–123. doi: 10.1038/npp.2012.149 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang XH, Tee LY, Wang XG, Huang QS, & Yang SH (2015). Off-target Effects in CRISPR/Cas9-mediated Genome Engineering. Molecular Therapy-Nucleic Acids, 4(11), e264. doi: 10.1038/mtna.2015.37 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Y, & Reinberg D (2001). Transcription regulation by histone methylation: interplay between different covalent modifications of the core histone tails. Genes & development, 15(18), 2343–2360. doi: 10.1101/gad.927301 [DOI] [PubMed] [Google Scholar]
- Zheng YG, Wu J, Chen Z, & Goodman M (2008). Chemical regulation of epigenetic modifications: opportunities for new cancer therapy. Medicinal research reviews, 28(5), 645–687. doi: 10.1002/med.20120 [DOI] [PubMed] [Google Scholar]
- Zhu Y, Wang Y, Yao R, Hao T, Cao J, Huang H, … Wu Y (2017). Enhanced neuroinflammation mediated by DNA methylation of the glucocorticoid receptor triggers cognitive dysfunction after sevoflurane anesthesia in adult rats subjected to maternal separation during the neonatal period. Journal of Neuroinflammation, 14(1), 6. doi: 10.1186/s12974-016-0782-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
