Minireviews Open Access
Copyright ©The Author(s) 2020. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Psychiatr. May 19, 2020; 10(5): 81-94
Published online May 19, 2020. doi: 10.5498/wjp.v10.i5.81
Neuroendocrine, epigenetic, and intergenerational effects of general anesthetics
Anatoly E Martynyuk, Department of Anesthesiology and the McKnight Brain Institute, University of Florida College of Medicine, Gainesville, FL 32610, United States
Ling-Sha Ju, Timothy E Morey, Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL 32610, United States
Jia-Qiang Zhang, Department of Anesthesiology and Perioperative Medicine, Henan Provincial People’s Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan Province, China
ORCID number: Anatoly E Martynyuk (0000-0002-9915-5584); Ling-Sha Ju (0000-0001-7266-0794); Timothy E Morey (0000-0003-2922-8984); Jia-Qiang Zhang (0000-0002-1367-9644).
Author contributions: The authors conducted literature review and analysis, drafted and critically revised the manuscript, and gave final approval.
Supported by National Institutes of Health, No. R01NS091542; National Natural Science Foundation of China, No. 81771149, No. U1704165.
Conflict-of-interest statement: No potential conflicts of interest.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Corresponding author: Anatoly E Martynyuk, DSc, PhD, Professor, Department of Anesthesiology and the McKnight Brain Institute, University of Florida College of Medicine, 1600 SW Archer Road, PO Box 100254, Gainesville, FL 32610, United States. amartynyuk@anest.ufl.edu
Received: December 24, 2019
Peer-review started: December 24, 2019
First decision: February 20, 2020
Revised: March 18, 2020
Accepted: March 25, 2020
Article in press: March 25, 2020
Published online: May 19, 2020

Abstract

The progress of modern medicine would be impossible without the use of general anesthetics (GAs). Despite advancements in refining anesthesia approaches, the effects of GAs are not fully reversible upon GA withdrawal. Neurocognitive deficiencies attributed to GA exposure may persist in neonates or endure for weeks to years in the elderly. Human studies on the mechanisms of the long-term adverse effects of GAs are needed to improve the safety of general anesthesia but they are hampered not only by ethical limitations specific to human research, but also by a lack of specific biological markers that can be used in human studies to safely and objectively study such effects. The latter can primarily be attributed to an insufficient understanding of the full range of the biological effects induced by GAs and the molecular mechanisms mediating such effects even in rodents, which are far more extensively studied than any other species. Our most recent experimental findings in rodents suggest that GAs may adversely affect many more people than is currently anticipated. Specifically, we have shown that anesthesia with the commonly used GA sevoflurane induces in exposed animals not only neuroendocrine abnormalities (somatic effects), but also epigenetic reprogramming of germ cells (germ cell effects). The latter may pass the neurobehavioral effects of parental sevoflurane exposure to the offspring, who may be affected even at levels of anesthesia that are not harmful to the exposed parents. The large number of patients who require general anesthesia, the even larger number of their future unexposed offspring whose health may be affected, and a growing number of neurodevelopmental disorders of unknown etiology underscore the translational importance of investigating the intergenerational effects of GAs. In this mini review, we discuss emerging experimental findings on neuroendocrine, epigenetic, and intergenerational effects of GAs.

Key Words: Brain, General anesthetic, Sevoflurane, Corticosterone, Cortisol, Histone acetylation, Deoxyribonucleic acid methylation, Intergenerational effects, Gamma aminobutyric acid

Core tip: The GABAergic general anesthetics may act as stressors and endocrine disruptors in neonates and young adults. They may induce two distinct types of long-term adverse effects: Neuroendocrine effects (the somatic effects) and epigenetic reprogramming of germ cells (the germ cell effects). The latter may pass neurobehavioral abnormalities to male offspring. Compared to the somatic cells, the germ cells may be more sensitive to the deleterious effects of general anesthetics, raising the possibility that the offspring may be affected even when levels of anesthesia are not harmful to the exposed parents. Further rigorous experimental testing of all these possibilities is required.



INTRODUCTION

The number of surgeries performed globally has rapidly increased from 226.4 million in 2004 to 312.9 million in 2012, according to 2016 World Health Organization estimates[1]. Most of these surgeries and many non-surgical procedures require general anesthesia, which can be viewed as a state of pharmacologically induced “reversible brain coma”[2]. Despite complete reversibility of the primary effect of general anesthetics (GAs), i.e., induction of the general anesthesia state, many studies in humans and almost all studies in laboratory animals provide evidence that GAs may leave persistent footprints of their brief presence in the body, i.e., anesthetic exposure may lead to long-lasting functional abnormalities[3-7]. Investigations of anesthesia-induced abnormalities are currently restricted primarily to evaluating neurocognitive function in the exposed subjects[8-12], although it is biologically plausible that GAs may affect other functions/systems because their actions are not limited to neuronal effects involved in the mediation of a general anesthesia state. Still, as a result of these studies, the long-term adverse effects of GAs are an increasingly recognized health concern, especially in the very young and elderly[3-7]. The adverse effects of GAs during the perinatal period are of special concern[13-17] because mounting evidence indicates that at this stage of life the central nervous system and other body systems are highly susceptible to reprogramming by environmental factors/stressors[18-21]. Such environmental factors may include GAs, given the multiple molecular targets known to mediate their actions in the brain and throughout the body[22-25]. In support of this contention are reports of learning disabilities, long-term memory impairment, and attention-deficit hyperactivity disorders in patients who had anesthesia early in life[3,5,11,26,27]. Although several recent studies have not found negative neurocognitive consequences of relatively short (≤ 1 h) anesthesia exposures in children[6,7,28], both clinical and laboratory studies, including the most recent clinical assessments[29], agree that prolonged or repeated exposures to GAs that are frequently required for very sick children may result in significant neurocognitive abnormalities later in life[30]. Furthermore, a recent report of the effects on brain development of relatively short anesthesia exposure for cesarean delivery[31] further confirms that the current understanding of this phenomenon remains in an early stage.

Because of a widely accepted dogma that the brain is most susceptible to the deleterious effects of environmental stressors at the extreme of ages, investigations of the long-term adverse effects of GAs in young adults are scarce even in animal models. Several studies have assessed the effects of GAs in young adult rats, primarily using rats of this age as comparisons to other age groups[4,32-34]. Aside from the fact that these studies found long-term effects of isoflurane in young adult rats, different isoflurane concentrations and exposure regimens make it difficult to compare the effects across these studies. Clearly, further research is needed to elucidate the full range of long-term effects of GAs in young adults. Importantly, the germ cells, which pass the genetic and epigenetic information from parents to offspring, can be susceptible to epigenetic reprogramming by environmental factors throughout the lifespan[35-39].

Laboratory and clinical studies provide evidence that alcohol, stress, endocrine disruptors, obesity, and even physical exercise may affect embryonic development and the phenotype of the offspring[40-44]. GAs share many molecular mechanisms of action with alcohol[45-49] and may act as endocrine disruptors and environmental stressors in animal models and humans. The spectrum of molecular actions of GAs and susceptibility of germ cells to epigenetic reprogramming by environmental factors across the lifespan support the possibility that the offspring may be affected by parental exposure to GAs regardless of the parental age at the time of exposure to GAs. In this mini review, we discuss emerging experimental findings on the neuroendocrine, epigenetic, and intergenerational effects of GAs.

NEUROENDOCRINE, EPIGENETIC, AND SECOND-GENERATION EFFECTS OF GENERAL ANESTHETICS

Research studies on the epigenetic multigenerational effects of environmental factors, such as alcohol, stress, endocrine disruptors, and others, have changed our thinking about the susceptibility of somatic and germ cells to alterations by environmental factors and the persistence of such alterations not only across the lifespan, but also through generations[36-38,40,42]. The potential of GAs to induce similar epigenetic effects, including reprogramming of the germ cell epigenome, and by extension intergenerational effects is supported by the notion that GAs share many molecular mechanisms of action with alcohol and may act as endocrine disruptors and environmental stressors in animal models and humans[45-49].

Endocrine disruptors can broadly be defined as agents that interfere with the functioning of the endocrine system. In support of the neuroendocrine effects of GAs are reports of significant rises in cortisol levels in pediatric patients after surgery or after anesthesia without surgery in healthy children[50,51]. Also, measurements of salivary cortisol levels in response to different levels of sedation in healthy children found a more than threefold increase in cortisol values, with the highest cortisol levels during the recovery phase[52]. Adult patients who received isoflurane-based tracheal general anesthesia compared to those who received bupivacaine-based epidural anesthesia had more than two times higher plasma levels of cortisol at the end of surgery, which was also more than four times higher compared to baseline levels in the same patients[53]. For a recent comprehensive review on cortisol levels associated with anesthesia/surgery see[54]. Importantly, our findings in rodents support the notion that GABAergic anesthetics act via specific molecular mechanisms to induce stress-like responses, rather than that GA-caused increases in glucocorticoid levels are the result of the systemic stress response because of uncontrolled physiological parameters during anesthesia. Thus, in neonatal rats, the gamma aminobutyric acid (GABA) type A receptor (GABAAR) antagonist bicuculline at a low dose (0.01 mg/kg) or aromatase inhibitor formestane, administered prior to anesthesia with sevoflurane, prevented the sevoflurane-increased corticosterone secretion without an obvious effect on the sedation depth induced by the anesthetic (unpublished observations).

The inhibitory control of the corticotropin-releasing hormone-secreting hypothalamic paraventricular neurons by GABAAR-mediated signaling and the positive modulation of this signaling by neuroactive steroids is one of the fundamental mechanisms of downregulating the stress response[55,56]. Due to relatively high and low expressions of the Cl- transporters Na+-K+-2Cl- (NKCC1) and K+-2Cl- (KCC2), respectively, immature neurons have elevated intracellular concentrations of Cl-, the main charge carriers through GABAAR channels[57-61], a major substrate for the otherwise inhibitory effects of GABAergic anesthetics[22-25]. Activation of GABAARs in immature neurons causes Cl- efflux, membrane depolarization, activation of the voltage-gated Ca++ channels, and relief of the Mg++-block of Ca++ permeable N-methyl-D-aspartate receptors[58-62]. The GABA-initiated Ca++ influxes regulate a wide spectrum of developmental processes from gene expression to synapse formation[61,62]. During the second postnatal week, GABAAR-mediated signaling in the brain undergoes a fundamental transition from predominantly stimulating/excitatory to inhibitory, which is caused by a concomitant developmental downregulation of NKCC1 and, most importantly, upregulation of neuronal-specific KCC2[61,62]. It is plausible that in the neonatal brain such GABAAR inhibitory signaling-mediated control of the stress response system is weakened or GABAergic anesthetics may even stimulate the stress response through positive modulation of depolarizing/excitatory GABAAR signaling at this age.

Consistent with stressor-like effects of GABAergic anesthetics, we found that a single exposure of neonatal rats to the GABAergic GAs sevoflurane or propofol was sufficient to cause multifold increases in corticosterone secretion and electroencephalography-detectable seizures at the time of anesthesia[63-67]. The anesthetic-caused increases in excitatory GABAAR signaling and corticosterone levels may be required for neonatal GABAergic anesthetic-induced seizures to occur[68,69]. Importantly, a single exposure to sevoflurane or propofol early in life induced neuroendocrine abnormalities[64,66,70-73] similar to those induced by repeated, but not single, maternal separations, a widely used rodent model of developmental effects of early-life stress in humans[74-78]. The GABAergic anesthetic-induced long-term neuroendocrine abnormalities, which were more robust in males, included increased anxiety-like behavior and exacerbated corticosterone responses to stress[64-67]. In addition, the rats, neonatally exposed to anesthesia, had elevated Crh mRNA levels in the hypothalamus, as well as up- and downregulated hypothalamic and hippocampal mRNA levels of Nkcc1 and Kcc2, respectively[66,67,73]. Notably, delays in the GABAAR signaling transition to inhibitory have been linked in animal models and humans to a number of cognitive neuropsychiatric disorders, such as schizophrenia, autism spectrum disorder (ASD), and Rett syndrome[79-85]. For example, in Rett syndrome, a severe form of ASD, the methyl CpG binding protein 2 deficiency-induced KCC2 downregulation may play an important causal role[84,85].

When used at a low dose, bumetanide, a loop diuretic, is the most selective of the available inhibitors of NKCC1 activity[86,87]. Our lab and others have demonstrated that pretreatment of neonatal rats with bumetanide prior to anesthetic exposure ameliorated many of the acute and lasting developmental effects of GABAergic anesthetics, including: (1) Seizures; (2) Downregulated Kcc2 levels; (3) Elevated levels of Crh mRNA; (4) Exacerbated corticosterone responses to acute stress; and (5) Behavioral abnormalities[63-66,88]. Bumetanide’s ameliorating effects suggest that anesthetic-exacerbated GABAAR-mediated stimulation/excitation in the neonatal rodent brain is an initial step in anesthetic-induced developmental abnormalities. Importantly, bumetanide exhibits promising therapeutic effects against ASD, schizophrenia, and Fragile X syndrome in animal models and humans[89-93], suggesting that these diseases and the GABAergic anesthetic-induced abnormalities may share similar mediating mechanisms. Our current understanding of rodent and human ontogeny supports the possibility that similar GA-sensitive mechanisms operate in rodents and humans early in life. Based on the intensity of synaptogenesis, a 1-wk-old rat can be compared to a 2- to 3-year-old human[94-98]. In animals, neonatal anesthetic exposure[63,65,72,99,100] and early-life stress[101-103] have profound long-term effects on synaptic morphology and function, suggesting that humans are vulnerable well into the postnatal period. Similar to the rodent brain during the first 2 postnatal weeks, the human brain is more excitable during the first year of life than at any other time, with seizures occurring in 3.5 per 1000 live births[104-106]. Human neonatal seizures are resistant to GABAergic antiepileptic drugs (AEDs) because of depolarizing/excitatory GABAAR signaling at this age[107,108]. Many human neonatal seizures can be detected only through electroencephalographies because they are not accompanied by clinical signs such as convulsions[109], which helps explain why epileptic seizures are not routinely reported in anesthetized human infants. In neonatal seizures with clinical manifestations, GABAAR-enhancing AEDs depress convulsions but may exacerbate electrographic cortical seizure activity (electroclinical uncoupling)[110]. The NKCC1 inhibitor bumetanide, administered alone or in combination with GABAergic AEDs, may alleviate neonatal seizures in rodents and humans[111,112]. In humans, the KCC2 protein levels at birth are only about 20% of adult levels and significantly increase during the first postnatal year[86]. This late KCC2 increase during brain development may make KCC2 a highly susceptible molecular target for modulation by environmental factors, including GAs.

The heightened corticosterone responses to acute stress months after exposure to the GABAergic anesthetics early in life suggest that stressors in post-anesthesia life can further exacerbate developmental abnormalities, initially programmed by GABAergic anesthetics. This concept is supported by our findings that adult rats exposed neonatally for a relatively short time to anesthesia with etomidate or sevoflurane followed by a subsequent single episode of maternal separation exhibited developmental abnormalities significantly greater than those exposed to only one of the two interventions[66,73,113]. In further support of the idea that the long-term adverse outcomes of early-life anesthesia may result from a combination of the effects of GAs at the time of anesthesia and the effects of “post-anesthesia” environmental factors, several laboratories have demonstrated that the adverse developmental effects of neonatal anesthesia in rodents may be alleviated, not only by pharmacological interventions before exposure to anesthesia, but also by post-weaning housing of the exposed animals in an enriched environment[114-116]. Environmental factors may alleviate or exacerbate the effects of neonatal anesthesia. In other words, two subjects exposed to the same anesthesia regimen may have different long-term outcomes based on post-anesthesia life experiences. In support of this possibility, Zhang et al[117] reported that rats neonatally exposed to sevoflurane and then housed from the time of weaning in isolation in enrichment-deprived environments exhibited reduced levels of brain-derived neurotrophic factor (BDNF), synaptic protein markers, and survival of new granule cells in the hippocampus, as well as behavioral abnormalities. On the other hand, rats that were exposed to the same regimen of anesthesia with sevoflurane and housed in groups in enriched environments had the same outcomes as their counterparts who were not exposed to sevoflurane. These findings further support the possibility that GABAergic anesthetics administered during early life can be considered environmental stressors that predispose the exposed subjects to stress vulnerability later in life. Identifying environmental factors that predispose humans to abnormal stress reactivity later in life and the mechanisms underlying their effects is of important clinical and basic neuroscience concern because dysregulated stress response systems have been linked to the pathophysiology of several neuropsychiatric disorders[118].

Environmental enrichment, which may alleviate the neurodevelopmental effects of early-life exposure to GAs in rodents, is not an issue for the majority of human patients because this is typical for most children. If similar mechanisms operate in humans, then brain development in healthy human patients who experience normal stress levels may be minimally affected, if at all, after exposure to general anesthesia early in life. However, most children who require general anesthesia during the early postnatal period inevitably experience a variety of stressors during life post-anesthesia exposure (e.g., diseases, pain, hunger, psychological stress). Such patients may be at risk of developing early-life, anesthetic-programmed neuroendocrine and neurocognitive abnormalities. The exacerbating effects of environmental stressors on long-term adverse outcomes of early-life exposure to GAs may be why several recent studies have not found negative neurocognitive consequences of relatively short (≤ 1 h) anesthesia exposures in healthy children[6,7,28], while studies in very sick children have[29]. It will be important to take this factor into consideration when planning new clinical studies. Further investigation of the interaction of adverse effects of early-life exposure to GAs and post-anesthesia stressors is important because it may identify not only the most vulnerable patients, but also those who are at diminished risk and would not benefit from delaying needed anesthesia-required interventions.

Histone acetylation and DNA methylation are important epigenetic mechanisms whereby environmental factors, in particular stress, affect brain development and function[119-121]. Histone acetylation facilitates gene transcription by enabling chromatin relaxation, while histone deacetylation results in stronger histone interaction with DNA, more compact chromatin structure, and repression of gene transcription. Histone acetylation is regulated by adding and removing acetyl groups to the N-terminal of histone tails by acetyltransferases and histone deacetylases, respectively[122-126]. Jia et al[99] found that repeated exposure of neonatal rats to sevoflurane led to increased levels of histone deacetylases 3 and 8 and reduced levels of acetylated histones H3 and H4 in the hippocampus. The sevoflurane-exposed rats had lower hippocampal density of dendritic spines and synaptic protein markers and exhibited impaired hippocampus function-based behavior. These effects of sevoflurane were alleviated by treatments with the histone deacetylase inhibitor sodium butyrate, suggesting a potential role of histone acetylation as one of the mediators of the developmental effects of GAs. The role of histone acetylation in GA-induced abnormalities in neonatal rodents has also been reported by other laboratories[127,128]. Furthermore, histone acetylation may be involved in the mediation of learning and memory dysfunction in offspring after pregnant rats exposed to sevoflurane, isoflurane, or propofol[129-131].

Ju et al[100] have found that repeated exposure of neonatal rats to sevoflurane resulted in increased expression of hippocampal DNA methyltransferases 3A/B (DNMT3A/B), but not DNA methyltransferase 1 (DNMT1). These enzymes catalyze DNA methylation at the 5′ position of cytosine residues adjacent to guanines (CpG sites), typically leading to long-term transcriptional repression. The activity of DNMT1 is responsible for maintenance of the remaining 5mC marks during cell divisions because of DNMT1 selectivity to hemi-methylated DNA, while DNMT3A/B are de novo DNMTs, which are induced by internal and external (environmental) stimuli[132,133]. In addition to increased expression of DNMT3A/B, the rats neonatally exposed to sevoflurane had downregulated methyl CpG binding protein 2, hypermethylated the Bdnf gene, downregulated BDNF levels, and exhibited behavioral deficiencies[100]. The ability of sevoflurane to induce these abnormalities was significantly diminished in rats that were pretreated with a non-selective DNMT inhibitor, 5-aza-2'-deoxycytidin, prior to sevoflurane exposure[100]. On the other hand, Wu et al[134] found that exposure of neonatal rats to isoflurane, another inhaled anesthetic that has positive modulation of GABAAR activity like sevoflurane, induced a significant increase in the expression of hippocampal DNMT1. More experimental studies are needed before we can draw conclusions on the specific epigenetic mechanisms involved in the mediation of the adverse effects of a given GA or a class of GAs.

It has been established in a wide range of species, including humans, that environmental factors, in particular stress, acting via epigenetic mechanisms may affect not only the exposed subjects, but also future generations[135-139]. In contrast to studies of famine or war survivors, in which large groups of people in relatively compact living areas within a specific time period were affected[135,137,140], it is not trivial to link someone’s neurodevelopmental abnormalities to his/her parent’s relatively short exposure to GAs. Still, the emerging laboratory and clinical data demonstrate that the adverse effects of GAs may include epigenetic modifications, not only in the somatic cells of the exposed subjects, but also in their germ cell epigenome. Of most direct support of the possibility of epigenetic germ cell effects of general anesthesia in humans, Donkin et al[141] found that the DNA methylation status of 1509 genes in the sperm of male patients was changed 1 wk after bariatric surgery. The findings of epigenetic changes in spermatozoa just 1 wk after anesthesia/surgery suggest that the anesthesia/surgery may be an important cause of such changes and that even the mature human sperm is susceptible to epigenetic reprogramming by environmental factors. The latter is supported by the presence of DNMTs in mature human sperm[142]. Importantly, of the 1509 genes altered at 1 wk after the anesthesia/surgery, 1004 genes remained altered 1 year later[141]. Alarmingly, several small pilot clinical assessments found that anesthesia care providers may have altered female/male offspring ratios, also suggesting that persistent exposure to traces of the escaped GAs from scavenging in operating rooms might affect germ cells, and, hence, the next generation(s)[143-147].

To test whether neonatal exposure to anesthesia with sevoflurane can affect not only the exposed animals, but also their future offspring, we exposed male and female postnatal day (P) 5 rats (generation F0) to 6 h anesthesia with 2.1% sevoflurane[67]. On P90, the exposed and control rats were used as breeders to produce the second generation of rats (generation F1). We have found that adult offspring of parents who were neonatally exposed to sevoflurane exhibited neurobehavioral abnormalities. Irrespective of whether sires, dams, or both parents were exposed to neonatal sevoflurane, only F1 males, but not F1 females, were affected. F1 males exhibited reduced Kcc2 Cl- exporter expression and behavioral abnormalities. Bisulfate sequencing revealed CpG dinucleotide hypermethylation in the Kcc2 promoter in the F0 sperm and ovary and in the hypothalamus and hippocampus of F1 males[67]. The correlation of impaired hippocampal Kcc2 expression and hippocampus-dependent behavior in F1 males points to the involvement of epigenetic Kcc2 modulation in the mediation of the intergenerational effects of sevoflurane[67]. Because DNA methylation is often associated with transcriptional repression, these data suggest that methylation of the Kcc2 promoter in F0 gametes may contribute to sevoflurane-induced intergenerational impairment in Kcc2 expression. The role of DNA methylation in the intergenerational effects of neonatal sevoflurane exposure in rats was also reported by Chastain-Potts et al[148]. Surprisingly, despite similar, but not identical, neurobehavioral abnormalities in the exposed parents and their male offspring, including downregulation of the Kcc2 expression, the male offspring, in contrast to their exposed parents, exhibited corticosterone responses to stress that were the same as the corticosterone responses to stress in the male offspring of control parents[67]. These findings, taken together with the effects of GABAergic anesthetics in the exposed animals, suggest that GA-induced modulation of GABAAR signaling through reduced expression of Kcc2 may be required but not sufficient to induce exacerbated responses to stress in adulthood.

Importantly, stress-like effects of GABAergic anesthetics may not be limited to the early postnatal period. Thus, exposure of young adult rats to sevoflurane on 3 alternating days starting on P56 resulted in similar increases in serum levels of corticosterone in male and female rats 1 h after the last exposure to the anesthetic[149]. However, long term, the exposed female rats were the same as the controls; they were not affected. On the other hand, contrary to the currently generally accepted view and our initial hypothesis that adult rats are resilient to the long-term adverse effects of GAs, the exposed young adult male rats developed neuroendocrine and behavioral abnormalities. More than 3 months after exposure to sevoflurane, they exhibited not only an exacerbated hypothalamic pituitary adrenal axis response to stress, but their serum levels of luteinizing and testosterone hormones were also significantly increased, as was their expression of the hypothalamic gonadotropin-releasing hormone gene. The hypothalamic pituitary testicular axis functioning in the exposed adult male rats was also altered at the level of expressions of the hypothalamic aromatase and estrogen receptor α and β genes. The expressions of the aromatase and estrogen receptor α genes were significantly increased, while the expression of the estrogen receptor β gene was slightly, but significantly, decreased[149]. These findings demonstrate that anesthesia with sevoflurane alters (disrupts) not only the functioning of the hypothalamic pituitary adrenal axis, but also the hypothalamic pituitary testicular axis functioning[53]. We have previously demonstrated the role of estradiol in the acute adverse effects of sevoflurane in neonatal rats[69].

Similar to the effects of neonatal exposure to sevoflurane, sevoflurane administered to young adult male rats induced significant impairment in expressions of the hypothalamic and hippocampal Kcc2 genes. Similar to adult rats that were neonatally exposed to sevoflurane, male and female rats exposed to sevoflurane in young adulthood had hypermethylated Kcc2 gene in spermatozoa and ovarian tissue, respectively[149]. Interestingly, we have analyzed the same CpG sites in the Kcc2 promoter region in the germ cells of rats that were exposed to sevoflurane neonatally or in young adulthood and found similar changes in their methylation regardless of the age of sevoflurane exposure[67,149]. These findings suggest that sevoflurane may induce germ cell effects via similar mediating mechanisms when administered to rats over a wide range of ages, from neonates to young adults. The similarities between the effects of neonatal and young adult exposure to sevoflurane were also evident in the offspring of the exposed parents. Thus, the Kcc2 gene was hypermethylated and exhibited reduced expression in the hypothalamus and hippocampus of the F1 male, but not female, offspring of the exposed parents. These changes in the Kcc2 gene in the hypothalamus and hippocampus of adult F1 male offspring were accompanied by behavioral deficiencies in the elevated plus maze and prepulse inhibition (PPI) of acoustic startle response tests, but their corticosterone responses to stress were not different from the controls[67,149]. The sevoflurane-exposed young adult male and female rats had similar acute increases in serum corticosterone levels and changes in DNA methylation status of the Kcc2 gene in spermatozoa and ovarian tissue and passed neurobehavioral abnormalities to their male offspring, despite the finding that the exposed dams lacked the somatic effects[149]. These findings allow us to hypothesize that sevoflurane-induced corticosterone secretion at the time of anesthesia is involved in the anesthetic-induced germ cell effects and by extension the intergenerational effects. Also, the findings that the exposed but long-term physiologically unaffected dams, similar to the exposed and affected sires, pass deleterious effects of sevoflurane to their unexposed male offspring suggest that compared to the somatic cells, the germ cells are more sensitive to the deleterious effects of sevoflurane. This raises the possibility that male offspring may be affected even when the anesthesia level/duration is insufficient to induce significant abnormalities in their exposed parents. Future studies will be needed to test these hypotheses that may have important translational applicability.

We have tested the role of the neuron-specific Kcc2 gene as a mediator of the germ cell and intergenerational effects of sevoflurane only because we have extensive background data supporting its involvement in the mediation of the somatic effects of neonatal and young adult exposure to sevoflurane in the exposed animals[64-67,149]. The findings that the Kcc2 gene was affected in the parental somatic (brain) cells and germ cells, two effects that may not necessarily have similar mediating mechanisms, was surprising, on the one hand, but on the other hand suggested that the Kcc2 gene could be one of many genes involved in passing on the intergenerational effects of the anesthetic. Indeed, using genome-wide reduced representation bisulfite sequencing, we found more than 2000 differentially methylated DNA regions in the sperm of adult rats neonatally exposed to sevoflurane compared to unexposed controls (unpublished observations). Future detailed investigation of various genes that are involved in the mediation of the intergenerational effects of sevoflurane and other GAs may help to identify a full spectrum of the biological intergenerational effects of GAs and may lead to new, unexpected parental GA-induced phenotypes in offspring. It is likely that the intergenerational effects of exposure to GAs in young adulthood are not limited to sevoflurane, as Tang et al[150] have shown behavioral abnormalities in the offspring of mice that were exposed to enflurane at 11 wk of age.

The weakness of most rodent behavioral paradigms is that they are difficult to directly replicate in humans. The results of our studies demonstrate that the PPI of startle was impaired in F0 rats exposed to sevoflurane as neonates or in young adulthood, as well as in their F1 male offspring[64-67,149]. Also, male and female rats, exposed to sevoflurane neonatally, and male rats exposed to sevoflurane in young adulthood, exhibited heightened corticosterone responses to stress long term[64-67,149]. In humans, PPI can be measured safely via changes in the eyeblink reflex using nearly identical parameters as in rodents[151-155], while heightened stress-related cortisol levels can be readily measured in saliva[156,157]. If GAs induce similar effects in humans, the PPI of startle and saliva levels of cortisol may be used as objective, translatable, and easily and safely measurable biological markers of the adverse effects of GAs in humans. Identification of such a biomarker(s) will facilitate investigation of the underlying mechanisms of the adverse effects of GAs in humans to guide development of safer anesthetic approaches. Of particular relevance, one of the effective, tested therapeutic agents to alleviate the developmental effects of GABAergic anesthetics in rodent models, the NKCC1 inhibitor bumetanide[63-66,88], is approved for the treatment of various pediatric conditions and exhibits promising therapeutic effects in human studies of neurodevelopmental disorders[89-93].

Considering relatively well-studied molecular targets for GA actions and emerging evidence of similarities between the adverse outcomes of exposure to GABAergic anesthetics and psychiatric disorders, understanding the molecular mechanisms of the adverse effects of GAs may help to elucidate the mechanistic basis and etiology of complex neurodevelopmental disorders.

CONCLUSION

The results of recent studies raise many intriguing questions related to the types of adverse effects of GABAergic GAs and their underlying mechanisms, the answers to which may have important translational applicability for establishing safer general anesthesia, in particular, and for better understanding of the nature of neuropsychiatric disorders, in general. They suggest that GABAergic GAs, in particular sevoflurane, may act as stressors and endocrine disruptors in neonates and young adults. These stress-like effects of GABAergic GAs may be involved in the mediation of two distinct types of long-term adverse effects of GAs in the exposed rodents: neuroendocrine effects (the somatic effects) and epigenetic reprogramming of their germ cells (the germ cell effects). The latter may pass neurobehavioral abnormalities to male offspring. The intergenerational effects of sevoflurane are similar, but not identical, when administered to neonatal and young adult rats, suggesting that similar mediating mechanisms are involved over a wide range of ages at the time of anesthesia. The initial data suggest that compared to the somatic cells, the germ cells are more sensitive to the deleterious effects of sevoflurane, raising the possibility that the offspring may be affected even when levels of anesthesia are not harmful to the exposed parents. The long-term adverse effects of GAs in the exposed young adult male rats suggest that current views on the window of vulnerability to the adverse effects of GAs in rodents (up to the first 2 postnatal weeks)[158,159], and, hence, the United States Food and Drug Administration recommendations to avoid GAs in children younger than 3[160], may need to be reconsidered to include more advanced ages. All of these possibilities may have important translational applicability if confirmed; further rigorous experimental testing is required.

Footnotes

Manuscript source: Invited manuscript

Corresponding Author's Membership in Professional Societies: American Society of Anesthesiologists.

Specialty type: Psychiatry

Country/Territory of origin: United States

Peer-review reports scientific quality classification

Grade A (Excellent): A

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): D

Grade E (Poor): 0

P-Reviewer: Hosak L, Ishizawa K, Shiina A S-Editor: Dou Y L-Editor: A E-Editor: Liu JH

References
1.  Weiser TG, Haynes AB, Molina G, Lipsitz SR, Esquivel MM, Uribe-Leitz T, Fu R, Azad T, Chao TE, Berry WR, Gawande AA. Size and distribution of the global volume of surgery in 2012. Bull World Health Organ. 2016;94:201-209F.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 415]  [Cited by in F6Publishing: 351]  [Article Influence: 43.9]  [Reference Citation Analysis (0)]
2.  Brown EN, Lydic R, Schiff ND. General anesthesia, sleep, and coma. N Engl J Med. 2010;363:2638-2650.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 886]  [Cited by in F6Publishing: 718]  [Article Influence: 51.3]  [Reference Citation Analysis (0)]
3.  Vutskits L, Xie Z. Lasting impact of general anaesthesia on the brain: mechanisms and relevance. Nat Rev Neurosci. 2016;17:705-717.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 260]  [Cited by in F6Publishing: 311]  [Article Influence: 44.4]  [Reference Citation Analysis (0)]
4.  Crosby C, Culley DJ, Baxter MG, Yukhananov R, Crosby G. Spatial memory performance 2 weeks after general anesthesia in adult rats. Anesth Analg. 2005;101:1389-1392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 60]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
5.  Hu D, Flick RP, Zaccariello MJ, Colligan RC, Katusic SK, Schroeder DR, Hanson AC, Buenvenida SL, Gleich SJ, Wilder RT, Sprung J, Warner DO. Association between Exposure of Young Children to Procedures Requiring General Anesthesia and Learning and Behavioral Outcomes in a Population-based Birth Cohort. Anesthesiology. 2017;127:227-240.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 173]  [Article Influence: 24.7]  [Reference Citation Analysis (0)]
6.  Sun LS, Li G, Miller TL, Salorio C, Byrne MW, Bellinger DC, Ing C, Park R, Radcliffe J, Hays SR, DiMaggio CJ, Cooper TJ, Rauh V, Maxwell LG, Youn A, McGowan FX. Association Between a Single General Anesthesia Exposure Before Age 36 Months and Neurocognitive Outcomes in Later Childhood. JAMA. 2016;315:2312-2320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 557]  [Cited by in F6Publishing: 534]  [Article Influence: 66.8]  [Reference Citation Analysis (0)]
7.  Davidson AJ, Disma N, de Graaff JC, Withington DE, Dorris L, Bell G, Stargatt R, Bellinger DC, Schuster T, Arnup SJ, Hardy P, Hunt RW, Takagi MJ, Giribaldi G, Hartmann PL, Salvo I, Morton NS, von Ungern Sternberg BS, Locatelli BG, Wilton N, Lynn A, Thomas JJ, Polaner D, Bagshaw O, Szmuk P, Absalom AR, Frawley G, Berde C, Ormond GD, Marmor J, McCann ME; GAS consortium. Neurodevelopmental outcome at 2 years of age after general anaesthesia and awake-regional anaesthesia in infancy (GAS): an international multicentre, randomised controlled trial. Lancet. 2016;387:239-250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 617]  [Cited by in F6Publishing: 580]  [Article Influence: 72.5]  [Reference Citation Analysis (0)]
8.  Graham MR, Brownell M, Chateau DG, Dragan RD, Burchill C, Fransoo RR. Neurodevelopmental Assessment in Kindergarten in Children Exposed to General Anesthesia before the Age of 4 Years: A Retrospective Matched Cohort Study. Anesthesiology. 2016;125:667-677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 115]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
9.  Ing CH, DiMaggio CJ, Malacova E, Whitehouse AJ, Hegarty MK, Feng T, Brady JE, von Ungern-Sternberg BS, Davidson AJ, Wall MM, Wood AJ, Li G, Sun LS. Comparative analysis of outcome measures used in examining neurodevelopmental effects of early childhood anesthesia exposure. Anesthesiology. 2014;120:1319-1332.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 118]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
10.  Filan PM, Hunt RW, Anderson PJ, Doyle LW, Inder TE. Neurologic outcomes in very preterm infants undergoing surgery. J Pediatr. 2012;160:409-414.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 76]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
11.  Sprung J, Flick RP, Katusic SK, Colligan RC, Barbaresi WJ, Bojanić K, Welch TL, Olson MD, Hanson AC, Schroeder DR, Wilder RT, Warner DO. Attention-deficit/hyperactivity disorder after early exposure to procedures requiring general anesthesia. Mayo Clin Proc. 2012;87:120-129.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 309]  [Cited by in F6Publishing: 325]  [Article Influence: 27.1]  [Reference Citation Analysis (0)]
12.  Hansen TG, Pedersen JK, Henneberg SW, Pedersen DA, Murray JC, Morton NS, Christensen K. Academic performance in adolescence after inguinal hernia repair in infancy: a nationwide cohort study. Anesthesiology. 2011;114:1076-1085.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 237]  [Cited by in F6Publishing: 231]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
13.  Sun L. Early childhood general anaesthesia exposure and neurocognitive development. Br J Anaesth. 2010;105 Suppl 1:i61-i68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 260]  [Cited by in F6Publishing: 278]  [Article Influence: 21.4]  [Reference Citation Analysis (0)]
14.  Ing C, Sun M, Olfson M, DiMaggio CJ, Sun LS, Wall MM, Li G. Age at Exposure to Surgery and Anesthesia in Children and Association With Mental Disorder Diagnosis. Anesth Analg. 2017;125:1988-1998.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 73]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
15.  Block RI, Thomas JJ, Bayman EO, Choi JY, Kimble KK, Todd MM. Are anesthesia and surgery during infancy associated with altered academic performance during childhood? Anesthesiology. 2012;117:494-503.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 130]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
16.  DiMaggio C, Sun LS, Li G. Early childhood exposure to anesthesia and risk of developmental and behavioral disorders in a sibling birth cohort. Anesth Analg. 2011;113:1143-1151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 385]  [Cited by in F6Publishing: 378]  [Article Influence: 29.1]  [Reference Citation Analysis (0)]
17.  DiMaggio C, Sun LS, Kakavouli A, Byrne MW, Li G. A retrospective cohort study of the association of anesthesia and hernia repair surgery with behavioral and developmental disorders in young children. J Neurosurg Anesthesiol. 2009;21:286-291.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 374]  [Cited by in F6Publishing: 364]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
18.  Heindel JJ, Balbus J, Birnbaum L, Brune-Drisse MN, Grandjean P, Gray K, Landrigan PJ, Sly PD, Suk W, Cory Slechta D, Thompson C, Hanson M. Developmental Origins of Health and Disease: Integrating Environmental Influences. Endocrinology. 2015;156:3416-3421.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 242]  [Cited by in F6Publishing: 239]  [Article Influence: 26.6]  [Reference Citation Analysis (0)]
19.  Barker DJ. The origins of the developmental origins theory. J Intern Med. 2007;261:412-417.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1749]  [Cited by in F6Publishing: 1694]  [Article Influence: 99.6]  [Reference Citation Analysis (0)]
20.  Gluckman PD, Hanson MA, Mitchell MD. Developmental origins of health and disease: reducing the burden of chronic disease in the next generation. Genome Med. 2010;2:14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 107]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
21.  Baird J, Jacob C, Barker M, Fall CH, Hanson M, Harvey NC, Inskip HM, Kumaran K, Cooper C. Developmental Origins of Health and Disease: A Lifecourse Approach to the Prevention of Non-Communicable Diseases. Healthcare (Basel). 2017;5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 101]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
22.  Antkowiak B, Rudolph U. New insights in the systemic and molecular underpinnings of general anesthetic actions mediated by γ-aminobutyric acid A receptors. Curr Opin Anaesthesiol. 2016;29:447-453.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 21]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
23.  Kotani N, Akaike N. The effects of volatile anesthetics on synaptic and extrasynaptic GABA-induced neurotransmission. Brain Res Bull. 2013;93:69-79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 38]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
24.  Khan KS, Hayes I, Buggy DJ. Pharmacology of anaesthetic agents II: inhalation anaesthetic agents. Continuing Education in Anaesthia Critical Care & Pain. 2014;14:106-111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 48]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
25.  Brohan J, Goudra BG. The Role of GABA Receptor Agonists in Anesthesia and Sedation. CNS Drugs. 2017;31:845-856.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 74]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
26.  Wilder RT, Flick RP, Sprung J, Katusic SK, Barbaresi WJ, Mickelson C, Gleich SJ, Schroeder DR, Weaver AL, Warner DO. Early exposure to anesthesia and learning disabilities in a population-based birth cohort. Anesthesiology. 2009;110:796-804.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 993]  [Cited by in F6Publishing: 985]  [Article Influence: 65.7]  [Reference Citation Analysis (0)]
27.  Kalkman CJ, Peelen L, Moons KG, Veenhuizen M, Bruens M, Sinnema G, de Jong TP. Behavior and development in children and age at the time of first anesthetic exposure. Anesthesiology. 2009;110:805-812.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 260]  [Cited by in F6Publishing: 257]  [Article Influence: 17.1]  [Reference Citation Analysis (0)]
28.  McCann ME, de Graaff JC, Dorris L, Disma N, Withington D, Bell G, Grobler A, Stargatt R, Hunt RW, Sheppard SJ, Marmor J, Giribaldi G, Bellinger DC, Hartmann PL, Hardy P, Frawley G, Izzo F, von Ungern Sternberg BS, Lynn A, Wilton N, Mueller M, Polaner DM, Absalom AR, Szmuk P, Morton N, Berde C, Soriano S, Davidson AJ; GAS Consortium. Neurodevelopmental outcome at 5 years of age after general anaesthesia or awake-regional anaesthesia in infancy (GAS): an international, multicentre, randomised, controlled equivalence trial. Lancet. 2019;393:664-677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 352]  [Cited by in F6Publishing: 367]  [Article Influence: 73.4]  [Reference Citation Analysis (0)]
29.  Banerjee P, Rossi MG, Anghelescu DL, Liu W, Breazeale AM, Reddick WE, Glass JO, Phillips NS, Jacola LM, Sabin ND, Inaba H, Srivastava D, Robison LL, Pui CH, Hudson MM, Krull KR. Association Between Anesthesia Exposure and Neurocognitive and Neuroimaging Outcomes in Long-term Survivors of Childhood Acute Lymphoblastic Leukemia. JAMA Oncol. 2019;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 54]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
30.  Lin EP, Lee JR, Lee CS, Deng M, Loepke AW. Do anesthetics harm the developing human brain? An integrative analysis of animal and human studies. Neurotoxicol Teratol. 2017;60:117-128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 93]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
31.  Deoni SC, Adams SH, Li X, Badger TM, Pivik RT, Glasier CM, Ramakrishnaiah RH, Rowell AC, Ou X. Cesarean Delivery Impacts Infant Brain Development. AJNR Am J Neuroradiol. 2019;40:169-177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
32.  Culley DJ, Baxter M, Yukhananov R, Crosby G. The memory effects of general anesthesia persist for weeks in young and aged rats. Anesth Analg. 2003;96:1004-1009, table of contents.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 190]  [Cited by in F6Publishing: 201]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
33.  Stratmann G, Sall JW, May LD, Bell JS, Magnusson KR, Rau V, Visrodia KH, Alvi RS, Ku B, Lee MT, Dai R. Isoflurane differentially affects neurogenesis and long-term neurocognitive function in 60-day-old and 7-day-old rats. Anesthesiology. 2009;110:834-848.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 254]  [Cited by in F6Publishing: 272]  [Article Influence: 18.1]  [Reference Citation Analysis (0)]
34.  Zhu C, Gao J, Karlsson N, Li Q, Zhang Y, Huang Z, Li H, Kuhn HG, Blomgren K. Isoflurane anesthesia induced persistent, progressive memory impairment, caused a loss of neural stem cells, and reduced neurogenesis in young, but not adult, rodents. J Cereb Blood Flow Metab. 2010;30:1017-1030.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 217]  [Cited by in F6Publishing: 225]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
35.  Ly L, Chan D, Aarabi M, Landry M, Behan NA, MacFarlane AJ, Trasler J. Intergenerational impact of paternal lifetime exposures to both folic acid deficiency and supplementation on reproductive outcomes and imprinted gene methylation. Mol Hum Reprod. 2017;23:461-477.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 64]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
36.  Rompala GR, Homanics GE. Intergenerational Effects of Alcohol: A Review of Paternal Preconception Ethanol Exposure Studies and Epigenetic Mechanisms in the Male Germline. Alcohol Clin Exp Res. 2019;43:1032-1045.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 34]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
37.  Rodgers AB, Morgan CP, Bronson SL, Revello S, Bale TL. Paternal stress exposure alters sperm microRNA content and reprograms offspring HPA stress axis regulation. J Neurosci. 2013;33:9003-9012.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 527]  [Cited by in F6Publishing: 497]  [Article Influence: 45.2]  [Reference Citation Analysis (0)]
38.  Chan JC, Nugent BM, Bale TL. Parental Advisory: Maternal and Paternal Stress Can Impact Offspring Neurodevelopment. Biol Psychiatry. 2018;83:886-894.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 128]  [Article Influence: 21.3]  [Reference Citation Analysis (0)]
39.  Goldberg LR, Gould TJ. Multigenerational and transgenerational effects of paternal exposure to drugs of abuse on behavioral and neural function. Eur J Neurosci. 2019;50:2453-2466.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 49]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
40.  Chastain LG, Sarkar DK. Alcohol effects on the epigenome in the germline: Role in the inheritance of alcohol-related pathology. Alcohol. 2017;60:53-66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 38]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
41.  Rattan S, Flaws JA. The epigenetic impacts of endocrine disruptors on female reproduction across generations†. Biol Reprod. 2019;101:635-644.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 47]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
42.  Gillette R, Son MJ, Ton L, Gore AC, Crews D. Passing experiences on to future generations: endocrine disruptors and transgenerational inheritance of epimutations in brain and sperm. Epigenetics. 2018;13:1106-1126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 41]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
43.  Yehuda R, Daskalakis NP, Lehrner A, Desarnaud F, Bader HN, Makotkine I, Flory JD, Bierer LM, Meaney MJ. Influences of maternal and paternal PTSD on epigenetic regulation of the glucocorticoid receptor gene in Holocaust survivor offspring. Am J Psychiatry. 2014;171:872-880.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 304]  [Cited by in F6Publishing: 242]  [Article Influence: 24.2]  [Reference Citation Analysis (0)]
44.  Pembrey ME, Bygren LO, Kaati G, Edvinsson S, Northstone K, Sjöström M, Golding J; ALSPAC Study Team. Sex-specific, male-line transgenerational responses in humans. Eur J Hum Genet. 2006;14:159-166.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 820]  [Cited by in F6Publishing: 672]  [Article Influence: 37.3]  [Reference Citation Analysis (0)]
45.  Mihic SJ, Ye Q, Wick MJ, Koltchine VV, Krasowski MD, Finn SE, Mascia MP, Valenzuela CF, Hanson KK, Greenblatt EP, Harris RA, Harrison NL. Sites of alcohol and volatile anaesthetic action on GABA(A) and glycine receptors. Nature. 1997;389:385-389.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 972]  [Cited by in F6Publishing: 1011]  [Article Influence: 37.4]  [Reference Citation Analysis (0)]
46.  Beckstead MJ, Phelan R, Mihic SJ. Antagonism of inhalant and volatile anesthetic enhancement of glycine receptor function. J Biol Chem. 2001;276:24959-24964.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 34]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
47.  Beckstead MJ, Phelan R, Trudell JR, Bianchini MJ, Mihic SJ. Anesthetic and ethanol effects on spontaneously opening glycine receptor channels. J Neurochem. 2002;82:1343-1351.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
48.  Rees DC, Knisely JS, Breen TJ, Balster RL. Toluene, halothane, 1,1,1-trichloroethane and oxazepam produce ethanol-like discriminative stimulus effects in mice. J Pharmacol Exp Ther. 1987;243:931-937.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Bowen SE, Balster RL. Desflurane, enflurane, isoflurane and ether produce ethanol-like discriminative stimulus effects in mice. Pharmacol Biochem Behav. 1997;57:191-198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 24]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
50.  Khilnani P, Munoz R, Salem M, Gelb C, Todres ID, Chernow B. Hormonal responses to surgical stress in children. J Pediatr Surg. 1993;28:1-4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
51.  Rains PC, Rampersad N, De Lima J, Murrell D, Kinchington D, Lee JW, Maguire AM, Donaghue KC. Cortisol response to general anaesthesia for medical imaging in children. Clin Endocrinol (Oxf). 2009;71:834-839.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 17]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
52.  Hsu AA, von Elten K, Chan D, Flynn T, Walker K, Barnhill J, Naun C, Pedersen AM, Ponaman M, Fredericks GJ, Crudo DF, Pinsker JE. Characterization of the cortisol stress response to sedation and anesthesia in children. J Clin Endocrinol Metab. 2012;97:E1830-E1835.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 26]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
53.  Aggo AT, Fyneface-Ogan S, Mato CN. The differential impact of two anesthetic techniques on cortisol levels in Nigerian surgical patients. Niger J Clin Pract. 2012;15:68-74.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
54.  Prete A, Yan Q, Al-Tarrah K, Akturk HK, Prokop LJ, Alahdab F, Foster MA, Lord JM, Karavitaki N, Wass JA, Murad MH, Arlt W, Bancos I. The cortisol stress response induced by surgery: A systematic review and meta-analysis. Clin Endocrinol (Oxf). 2018;89:554-567.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 76]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
55.  Mody I, Maguire J. The reciprocal regulation of stress hormones and GABA(A) receptors. Front Cell Neurosci. 2011;6:4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 62]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
56.  Kakizawa K, Watanabe M, Mutoh H, Okawa Y, Yamashita M, Yanagawa Y, Itoi K, Suda T, Oki Y, Fukuda A. A novel GABA-mediated corticotropin-releasing hormone secretory mechanism in the median eminence. Sci Adv. 2016;2:e1501723.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 26]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
57.  Salmon CK, Pribiag H, Gizowski C, Farmer WT, Cameron S, Jones EV, Mahadevan V, Bourque CW, Stellwagen D, Woodin MA, Murai KK. Depolarizing GABA Transmission Restrains Activity-Dependent Glutamatergic Synapse Formation in the Developing Hippocampal Circuit. Front Cell Neurosci. 2020;14:36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
58.  Ben-Ari Y. The GABA excitatory/inhibitory developmental sequence: a personal journey. Neuroscience. 2014;279:187-219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 198]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
59.  Khazipov R, Valeeva G, Khalilov I. Depolarizing GABA and developmental epilepsies. CNS Neurosci Ther. 2015;21:83-91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 58]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
60.  Ito S. GABA and glycine in the developing brain. J Physiol Sci. 2016;66:375-379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 37]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
61.  Farrant M, Kaila K. The cellular, molecular and ionic basis of GABA(A) receptor signalling. Prog Brain Res. 2007;160:59-87.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 255]  [Cited by in F6Publishing: 262]  [Article Influence: 15.4]  [Reference Citation Analysis (0)]
62.  Dehorter N, Vinay L, Hammond C, Ben-Ari Y. Timing of developmental sequences in different brain structures: physiological and pathological implications. Eur J Neurosci. 2012;35:1846-1856.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 66]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
63.  Edwards DA, Shah HP, Cao W, Gravenstein N, Seubert CN, Martynyuk AE. Bumetanide alleviates epileptogenic and neurotoxic effects of sevoflurane in neonatal rat brain. Anesthesiology. 2010;112:567-575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 109]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
64.  Tan S, Xu C, Zhu W, Willis J, Seubert CN, Gravenstein N, Sumners C, Martynyuk AE. Endocrine and neurobehavioral abnormalities induced by propofol administered to neonatal rats. Anesthesiology. 2014;121:1010-1017.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 29]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
65.  Xu C, Tan S, Zhang J, Seubert CN, Gravenstein N, Sumners C, Vasilopoulos T, Martynyuk AE. Anesthesia with sevoflurane in neonatal rats: Developmental neuroendocrine abnormalities and alleviating effects of the corticosteroid and Cl(-) importer antagonists. Psychoneuroendocrinology. 2015;60:173-181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 36]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
66.  Ju LS, Yang JJ, Gravenstein N, Seubert CN, Morey TE, Sumners C, Vasilopoulos T, Yang JJ, Martynyuk AE. Role of environmental stressors in determining the developmental outcome of neonatal anesthesia. Psychoneuroendocrinology. 2017;81:96-104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 20]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
67.  Ju LS, Yang JJ, Morey TE, Gravenstein N, Seubert CN, Resnick JL, Zhang JQ, Martynyuk AE. Role of epigenetic mechanisms in transmitting the effects of neonatal sevoflurane exposure to the next generation of male, but not female, rats. Br J Anaesth. 2018;121:406-416.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 41]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
68.  Willis J, Zhu W, Perez-Downes J, Tan S, Xu C, Seubert C, Gravenstein N, Martynyuk A. Propofol-induced electroencephalographic seizures in neonatal rats: the role of corticosteroids and γ-aminobutyric acid type A receptor-mediated excitation. Anesth Analg. 2015;120:433-439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
69.  Zhang J, Xu C, Puentes DL, Seubert CN, Gravenstein N, Martynyuk AE. Role of Steroids in Hyperexcitatory Adverse and Anesthetic Effects of Sevoflurane in Neonatal Rats. Neuroendocrinology. 2016;103:440-451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 15]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
70.  Cao W, Pavlinec C, Gravenstein N, Seubert CN, Martynyuk AE. Roles of aldosterone and oxytocin in abnormalities caused by sevoflurane anesthesia in neonatal rats. Anesthesiology. 2012;117:791-800.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 36]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
71.  Seubert CN, Zhu W, Pavlinec C, Gravenstein N, Martynyuk AE. Developmental effects of neonatal isoflurane and sevoflurane exposure in rats. Anesthesiology. 2013;119:358-364.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 37]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
72.  Xu C, Seubert CN, Gravenstein N, Martynyuk AE. Propofol, but not etomidate, increases corticosterone levels and induces long-term alteration in hippocampal synaptic activity in neonatal rats. Neurosci Lett. 2016;618:1-5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
73.  Yang J, Ju L, Jia M, Zhang H, Sun X, Ji M, Yang J, Martynyuk AE. Subsequent maternal separation exacerbates neurobehavioral abnormalities in rats neonatally exposed to sevoflurane anesthesia. Neurosci Lett. 2017;661:137-142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
74.  Majcher-Maślanka I, Solarz A, Chocyk A. Maternal separation disturbs postnatal development of the medial prefrontal cortex and affects the number of neurons and glial cells in adolescent rats. Neuroscience. 2019;423:131-147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 25]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
75.  Heydari A, Esmaeilpour K, Sheibani V. Maternal separation impairs long term-potentiation in CA3-CA1 synapses in adolescent female rats. Behav Brain Res. 2019;376:112239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
76.  Brunson KL, Kramár E, Lin B, Chen Y, Colgin LL, Yanagihara TK, Lynch G, Baram TZ. Mechanisms of late-onset cognitive decline after early-life stress. J Neurosci. 2005;25:9328-9338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 321]  [Cited by in F6Publishing: 347]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
77.  Furukawa M, Tsukahara T, Tomita K, Iwai H, Sonomura T, Miyawaki S, Sato T. Neonatal maternal separation delays the GABA excitatory-to-inhibitory functional switch by inhibiting KCC2 expression. Biochem Biophys Res Commun. 2017;493:1243-1249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 37]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
78.  Patchev VK, Montkowski A, Rouskova D, Koranyi L, Holsboer F, Almeida OF. Neonatal treatment of rats with the neuroactive steroid tetrahydrodeoxycorticosterone (THDOC) abolishes the behavioral and neuroendocrine consequences of adverse early life events. J Clin Invest. 1997;99:962-966.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 103]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
79.  Merner ND, Chandler MR, Bourassa C, Liang B, Khanna AR, Dion P, Rouleau GA, Kahle KT. Regulatory domain or CpG site variation in SLC12A5, encoding the chloride transporter KCC2, in human autism and schizophrenia. Front Cell Neurosci. 2015;9:386.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 73]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
80.  Ben-Ari Y, Khalilov I, Kahle KT, Cherubini E. The GABA excitatory/inhibitory shift in brain maturation and neurological disorders. Neuroscientist. 2012;18:467-486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 391]  [Cited by in F6Publishing: 407]  [Article Influence: 33.9]  [Reference Citation Analysis (0)]
81.  Merner ND, Mercado A, Khanna AR, Hodgkinson A, Bruat V, Awadalla P, Gamba G, Rouleau GA, Kahle KT. Gain-of-function missense variant in SLC12A2, encoding the bumetanide-sensitive NKCC1 cotransporter, identified in human schizophrenia. J Psychiatr Res. 2016;77:22-26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 37]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
82.  Genç F, Kara M, Ünal Y, Uygur Küçükseymen E, Biçer Gömceli Y, Kaynar T, Tosun K, Kutlu G. Methylation of cation-chloride cotransporters NKCC1 and KCC2 in patients with juvenile myoclonic epilepsy. Neurol Sci. 2019;40:1007-1013.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
83.  Han P, Welsh CT, Smith MT, Schmidt RE, Carroll SL. Complex Patterns of GABAergic Neuronal Deficiency and Type 2 Potassium-Chloride Cotransporter Immaturity in Human Focal Cortical Dysplasia. J Neuropathol Exp Neurol. 2019;78:365-372.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
84.  Tang X, Kim J, Zhou L, Wengert E, Zhang L, Wu Z, Carromeu C, Muotri AR, Marchetto MC, Gage FH, Chen G. KCC2 rescues functional deficits in human neurons derived from patients with Rett syndrome. Proc Natl Acad Sci USA. 2016;113:751-756.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 148]  [Article Influence: 18.5]  [Reference Citation Analysis (0)]
85.  Tang X, Drotar J, Li K, Clairmont CD, Brumm AS, Sullins AJ, Wu H, Liu XS, Wang J, Gray NS, Sur M, Jaenisch R. Pharmacological enhancement of KCC2 gene expression exerts therapeutic effects on human Rett syndrome neurons and Mecp2 mutant mice. Sci Transl Med. 2019;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 73]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
86.  Dzhala VI, Talos DM, Sdrulla DA, Brumback AC, Mathews GC, Benke TA, Delpire E, Jensen FE, Staley KJ. NKCC1 transporter facilitates seizures in the developing brain. Nat Med. 2005;11:1205-1213.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 682]  [Cited by in F6Publishing: 677]  [Article Influence: 35.6]  [Reference Citation Analysis (0)]
87.  Ben-Ari Y. NKCC1 Chloride Importer Antagonists Attenuate Many Neurological and Psychiatric Disorders. Trends Neurosci. 2017;40:536-554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 130]  [Article Influence: 18.6]  [Reference Citation Analysis (0)]
88.  Liu G, Zhu T, Zhang A, Li F, Qian W, Qian B. Heightened stress response and cognitive impairment after repeated neonatal sevoflurane exposures might be linked to excessive GABAAR-mediated depolarization. J Anesth. 2016;30:834-841.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
89.  Du L, Shan L, Wang B, Li H, Xu Z, Staal WG, Jia F. A Pilot Study on the Combination of Applied Behavior Analysis and Bumetanide Treatment for Children with Autism. J Child Adolesc Psychopharmacol. 2015;25:585-588.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 52]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
90.  Lemonnier E, Robin G, Degrez C, Tyzio R, Grandgeorge M, Ben-Ari Y. Treating Fragile X syndrome with the diuretic bumetanide: a case report. Acta Paediatr. 2013;102:e288-e290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 48]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
91.  Lemonnier E, Lazartigues A, Ben-Ari Y. Treating Schizophrenia With the Diuretic Bumetanide: A Case Report. Clin Neuropharmacol. 2016;39:115-117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 43]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
92.  Lemonnier E, Villeneuve N, Sonie S, Serret S, Rosier A, Roue M, Brosset P, Viellard M, Bernoux D, Rondeau S, Thummler S, Ravel D, Ben-Ari Y. Effects of bumetanide on neurobehavioral function in children and adolescents with autism spectrum disorders. Transl Psychiatry. 2017;7:e1056.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 102]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
93.  Hadjikhani N, Åsberg Johnels J, Lassalle A, Zürcher NR, Hippolyte L, Gillberg C, Lemonnier E, Ben-Ari Y. Bumetanide for autism: more eye contact, less amygdala activation. Sci Rep. 2018;8:3602.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 52]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
94.  Semple BD, Blomgren K, Gimlin K, Ferriero DM, Noble-Haeusslein LJ. Brain development in rodents and humans: Identifying benchmarks of maturation and vulnerability to injury across species. Prog Neurobiol. 2013;106-107:1-16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1413]  [Cited by in F6Publishing: 1299]  [Article Influence: 118.1]  [Reference Citation Analysis (0)]
95.  Schachtele SJ, Losh J, Dailey ME, Green SH. Spine formation and maturation in the developing rat auditory cortex. J Comp Neurol. 2011;519:3327-3345.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
96.  Lenroot RK, Giedd JN. Brain development in children and adolescents: insights from anatomical magnetic resonance imaging. Neurosci Biobehav Rev. 2006;30:718-729.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1131]  [Cited by in F6Publishing: 1102]  [Article Influence: 61.2]  [Reference Citation Analysis (0)]
97.  Petanjek Z, Judaš M, Šimic G, Rasin MR, Uylings HB, Rakic P, Kostovic I. Extraordinary neoteny of synaptic spines in the human prefrontal cortex. Proc Natl Acad Sci USA. 2011;108:13281-13286.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 850]  [Cited by in F6Publishing: 836]  [Article Influence: 64.3]  [Reference Citation Analysis (0)]
98.  Liu X, Somel M, Tang L, Yan Z, Jiang X, Guo S, Yuan Y, He L, Oleksiak A, Zhang Y, Li N, Hu Y, Chen W, Qiu Z, Pääbo S, Khaitovich P. Extension of cortical synaptic development distinguishes humans from chimpanzees and macaques. Genome Res. 2012;22:611-622.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 163]  [Cited by in F6Publishing: 158]  [Article Influence: 13.2]  [Reference Citation Analysis (0)]
99.  Jia M, Liu WX, Yang JJ, Xu N, Xie ZM, Ju LS, Ji MH, Martynyuk AE, Yang JJ. Role of histone acetylation in long-term neurobehavioral effects of neonatal Exposure to sevoflurane in rats. Neurobiol Dis. 2016;91:209-220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 38]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
100.  Ju LS, Jia M, Sun J, Sun XR, Zhang H, Ji MH, Yang JJ, Wang ZY. Hypermethylation of Hippocampal Synaptic Plasticity-Related genes is Involved in Neonatal Sevoflurane Exposure-Induced Cognitive Impairments in Rats. Neurotox Res. 2016;29:243-255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 39]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
101.  Bock J, Braun K. The impact of perinatal stress on the functional maturation of prefronto-cortical synaptic circuits: implications for the pathophysiology of ADHD? Prog Brain Res. 2011;189:155-169.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 26]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
102.  Ohta KI, Suzuki S, Warita K, Kaji T, Kusaka T, Miki T. Prolonged maternal separation attenuates BDNF-ERK signaling correlated with spine formation in the hippocampus during early brain development. J Neurochem. 2017;141:179-194.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 32]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
103.  Danielewicz J, Hess G. Early life stress alters synaptic modification range in the rat lateral amygdala. Behav Brain Res. 2014;265:32-37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 14]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
104.  Lanska MJ, Lanska DJ, Baumann RJ, Kryscio RJ. A population-based study of neonatal seizures in Fayette County, Kentucky. Neurology. 1995;45:724-732.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 164]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
105.  van Rooij LG, Hellström-Westas L, de Vries LS. Treatment of neonatal seizures. Semin Fetal Neonatal Med. 2013;18:209-215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 47]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
106.  Volpe J. Neurology of the newborn. 5th ed.  Philadelphia, PA: Saunders, 2008.  [PubMed]  [DOI]  [Cited in This Article: ]
107.  Painter MJ, Scher MS, Stein AD, Armatti S, Wang Z, Gardiner JC, Paneth N, Minnigh B, Alvin J. Phenobarbital compared with phenytoin for the treatment of neonatal seizures. N Engl J Med. 1999;341:485-489.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 419]  [Cited by in F6Publishing: 359]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
108.  Boylan GB, Rennie JM, Chorley G, Pressler RM, Fox GF, Farrer K, Morton M, Binnie CD. Second-line anticonvulsant treatment of neonatal seizures: a video-EEG monitoring study. Neurology. 2004;62:486-488.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 121]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
109.  Boylan GB, Stevenson NJ, Vanhatalo S. Monitoring neonatal seizures. Semin Fetal Neonatal Med. 2013;18:202-208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 82]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
110.  Connell J, Oozeer R, de Vries L, Dubowitz LM, Dubowitz V. Clinical and EEG response to anticonvulsants in neonatal seizures. Arch Dis Child. 1989;64:459-464.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 79]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
111.  Puskarjov M, Kahle KT, Ruusuvuori E, Kaila K. Pharmacotherapeutic targeting of cation-chloride cotransporters in neonatal seizures. Epilepsia. 2014;55:806-818.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 105]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
112.  Kahle KT, Barnett SM, Sassower KC, Staley KJ. Decreased seizure activity in a human neonate treated with bumetanide, an inhibitor of the Na(+)-K(+)-2Cl(-) cotransporter NKCC1. J Child Neurol. 2009;24:572-576.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 101]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
113.  Yang J, Ju L, Yang C, Xue J, Setlow B, Morey TE, Gravenstein N, Seubert CN, Vasilopoulos T, Martynyuk AE. Effects of combined brief etomidate anesthesia and postnatal stress on amygdala expression of Cl- cotransporters and corticotropin-releasing hormone and alcohol intake in adult rats. Neurosci Lett. 2018;685:83-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
114.  Shih J, May LD, Gonzalez HE, Lee EW, Alvi RS, Sall JW, Rau V, Bickler PE, Lalchandani GR, Yusupova M, Woodward E, Kang H, Wilk AJ, Carlston CM, Mendoza MV, Guggenheim JN, Schaefer M, Rowe AM, Stratmann G. Delayed environmental enrichment reverses sevoflurane-induced memory impairment in rats. Anesthesiology. 2012;116:586-602.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 133]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
115.  Zheng H, Dong Y, Xu Z, Crosby G, Culley DJ, Zhang Y, Xie Z. Sevoflurane anesthesia in pregnant mice induces neurotoxicity in fetal and offspring mice. Anesthesiology. 2013;118:516-526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 147]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
116.  Chinn GA, Sasaki Russell JM, Banh ET, Lee SC, Sall JW. Voluntary Exercise Rescues the Spatial Memory Deficit Associated With Early Life Isoflurane Exposure in Male Rats. Anesth Analg. 2019;129:1365-1373.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
117.  Zhang MQ, Ji MH, Zhao QS, Jia M, Qiu LL, Yang JJ, Peng YG, Yang JJ, Martynyuk AE. Neurobehavioural abnormalities induced by repeated exposure of neonatal rats to sevoflurane can be aggravated by social isolation and enrichment deprivation initiated after exposure to the anaesthetic. Br J Anaesth. 2015;115:752-760.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 38]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
118.  Atrooz F, Liu H, Salim S. Stress, psychiatric disorders, molecular targets, and more. Prog Mol Biol Transl Sci. 2019;167:77-105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 9]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
119.  D'Mello SR. Regulation of Central Nervous System Development by Class I Histone Deacetylases. Dev Neurosci. 2019;41:149-165.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
120.  McClelland S, Korosi A, Cope J, Ivy A, Baram TZ. Emerging roles of epigenetic mechanisms in the enduring effects of early-life stress and experience on learning and memory. Neurobiol Learn Mem. 2011;96:79-88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 68]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
121.  Powell WT, LaSalle JM. Epigenetic mechanisms in diurnal cycles of metabolism and neurodevelopment. Hum Mol Genet. 2015;24:R1-R9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 24]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
122.  Rudenko A, Tsai LH. Epigenetic modifications in the nervous system and their impact upon cognitive impairments. Neuropharmacology. 2014;80:70-82.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 70]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
123.  Bannister AJ, Kouzarides T. Regulation of chromatin by histone modifications. Cell Res. 2011;21:381-395.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3359]  [Cited by in F6Publishing: 3470]  [Article Influence: 266.9]  [Reference Citation Analysis (0)]
124.  Fischer A, Sananbenesi F, Mungenast A, Tsai LH. Targeting the correct HDAC(s) to treat cognitive disorders. Trends Pharmacol Sci. 2010;31:605-617.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 266]  [Cited by in F6Publishing: 279]  [Article Influence: 19.9]  [Reference Citation Analysis (0)]
125.  Guan JS, Haggarty SJ, Giacometti E, Dannenberg JH, Joseph N, Gao J, Nieland TJ, Zhou Y, Wang X, Mazitschek R, Bradner JE, DePinho RA, Jaenisch R, Tsai LH. HDAC2 negatively regulates memory formation and synaptic plasticity. Nature. 2009;459:55-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1125]  [Cited by in F6Publishing: 1172]  [Article Influence: 78.1]  [Reference Citation Analysis (0)]
126.  McQuown SC, Barrett RM, Matheos DP, Post RJ, Rogge GA, Alenghat T, Mullican SE, Jones S, Rusche JR, Lazar MA, Wood MA. HDAC3 is a critical negative regulator of long-term memory formation. J Neurosci. 2011;31:764-774.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 361]  [Cited by in F6Publishing: 373]  [Article Influence: 28.7]  [Reference Citation Analysis (0)]
127.  Dalla Massara L, Osuru HP, Oklopcic A, Milanovic D, Joksimovic SM, Caputo V, DiGruccio MR, Ori C, Wang G, Todorovic SM, Jevtovic-Todorovic V. General Anesthesia Causes Epigenetic Histone Modulation of c-Fos and Brain-derived Neurotrophic Factor, Target Genes Important for Neuronal Development in the Immature Rat Hippocampus. Anesthesiology. 2016;124:1311-1327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 55]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
128.  Joksimovic SM, Osuru HP, Oklopcic A, Beenhakker MP, Jevtovic-Todorovic V, Todorovic SM. Histone Deacetylase Inhibitor Entinostat (MS-275) Restores Anesthesia-induced Alteration of Inhibitory Synaptic Transmission in the Developing Rat Hippocampus. Mol Neurobiol. 2018;55:222-228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 13]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
129.  Luo F, Hu Y, Zhao W, Zuo Z, Yu Q, Liu Z, Lin J, Feng Y, Li B, Wu L, Xu L. Maternal Exposure of Rats to Isoflurane during Late Pregnancy Impairs Spatial Learning and Memory in the Offspring by Up-Regulating the Expression of Histone Deacetylase 2. PLoS One. 2016;11:e0160826.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
130.  Lin J, Wang S, Feng Y, Zhao W, Zhao W, Luo F, Feng N. Propofol exposure during early gestation impairs learning and memory in rat offspring by inhibiting the acetylation of histone. J Cell Mol Med. 2018;22:2600-2611.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
131.  Wu Z, Li X, Zhang Y, Tong D, Wang L, Zhao P. Effects of Sevoflurane Exposure During Mid-Pregnancy on Learning and Memory in Offspring Rats: Beneficial Effects of Maternal Exercise. Front Cell Neurosci. 2018;12:122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 34]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
132.  Kinney SR, Pradhan S. Regulation of expression and activity of DNA (cytosine-5) methyltransferases in mammalian cells. Prog Mol Biol Transl Sci. 2011;101:311-333.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 61]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
133.  Lyko F. The DNA methyltransferase family: a versatile toolkit for epigenetic regulation. Nat Rev Genet. 2018;19:81-92.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 596]  [Cited by in F6Publishing: 744]  [Article Influence: 106.3]  [Reference Citation Analysis (0)]
134.  Wu J, Bie B, Naguib M. Epigenetic Manipulation of Brain-derived Neurotrophic Factor Improves Memory Deficiency Induced by Neonatal Anesthesia in Rats. Anesthesiology. 2016;124:624-640.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 56]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
135.  Lehrner A, Bierer LM, Passarelli V, Pratchett LC, Flory JD, Bader HN, Harris IR, Bedi A, Daskalakis NP, Makotkine I, Yehuda R. Maternal PTSD associates with greater glucocorticoid sensitivity in offspring of Holocaust survivors. Psychoneuroendocrinology. 2014;40:213-220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 71]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
136.  Khashan AS, Abel KM, McNamee R, Pedersen MG, Webb RT, Baker PN, Kenny LC, Mortensen PB. Higher risk of offspring schizophrenia following antenatal maternal exposure to severe adverse life events. Arch Gen Psychiatry. 2008;65:146-152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 394]  [Cited by in F6Publishing: 355]  [Article Influence: 22.2]  [Reference Citation Analysis (0)]
137.  Pembrey M, Saffery R, Bygren LO; Network in Epigenetic Epidemiology; Network in Epigenetic Epidemiology. Human transgenerational responses to early-life experience: potential impact on development, health and biomedical research. J Med Genet. 2014;51:563-572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 204]  [Cited by in F6Publishing: 190]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
138.  Bygren LO, Kaati G, Edvinsson S. Longevity determined by paternal ancestors' nutrition during their slow growth period. Acta Biotheor. 2001;49:53-59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 230]  [Cited by in F6Publishing: 233]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
139.  Kaati G, Bygren LO, Edvinsson S. Cardiovascular and diabetes mortality determined by nutrition during parents' and grandparents' slow growth period. Eur J Hum Genet. 2002;10:682-688.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 620]  [Cited by in F6Publishing: 480]  [Article Influence: 21.8]  [Reference Citation Analysis (0)]
140.  van Os J, Selten JP. Prenatal exposure to maternal stress and subsequent schizophrenia. The May 1940 invasion of The Netherlands. Br J Psychiatry. 1998;172:324-326.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 353]  [Cited by in F6Publishing: 374]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
141.  Donkin I, Versteyhe S, Ingerslev LR, Qian K, Mechta M, Nordkap L, Mortensen B, Appel EV, Jørgensen N, Kristiansen VB, Hansen T, Workman CT, Zierath JR, Barrès R. Obesity and Bariatric Surgery Drive Epigenetic Variation of Spermatozoa in Humans. Cell Metab. 2016;23:369-378.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 357]  [Cited by in F6Publishing: 313]  [Article Influence: 39.1]  [Reference Citation Analysis (0)]
142.  Marques CJ, João Pinho M, Carvalho F, Bièche I, Barros A, Sousa M. DNA methylation imprinting marks and DNA methyltransferase expression in human spermatogenic cell stages. Epigenetics. 2011;6:1354-1361.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 94]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
143.  Wyatt R, Wilson AM. Children of anaesthetists. Br Med J. 1973;1:675.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 21]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
144.  Gupta D, Kaminski E, McKelvey G, Wang H. Firstborn offspring sex ratio is skewed towards female offspring in anesthesia care providers: A questionnaire-based nationwide study from United States. J Anaesthesiol Clin Pharmacol. 2013;29:221-227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 8]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
145.  Nagella AB, Ravishankar M, Hemanth Kumar VR. Anaesthesia practice and reproductive outcomes: Facts unveiled. Indian J Anaesth. 2015;59:706-714.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
146.  Nagella AB, Ravishankar M, Hemanth Kumar VR. Anaesthesia practice and reproductive outcomes: Facts unveiled. Indian J Anaesth. 2016;60:225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
147.  Gupta D. Firstborn female offsprings are significantly more common among Indian anaesthesiologists as compared to national child sex ratio. Indian J Anaesth. 2016;60:224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
148.  Chastain-Potts SE, Tesic V, Tat QL, Cabrera OH, Quillinan N, Jevtovic-Todorovic V. Sevoflurane Exposure Results in Sex-Specific Transgenerational Upregulation of Target IEGs in the Subiculum. Mol Neurobiol. 2020;57:11-22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
149.  Ju LS, Yang JJ, Xu N, Li J, Morey TE, Gravenstein N, Seubert CN, Setlow B, Martynyuk AE. Intergenerational Effects of Sevoflurane in Young Adult Rats. Anesthesiology. 2019;131:1092-1109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 21]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
150.  Tang CK, Chalon J, Markham JP, Ramanathan S, Turndorf H. Exposure of sires to enflurane affects learning function of murine progeny. Anesth Analg. 1984;63:729-730.  [PubMed]  [DOI]  [Cited in This Article: ]
151.  Swerdlow NR, Weber M, Qu Y, Light GA, Braff DL. Realistic expectations of prepulse inhibition in translational models for schizophrenia research. Psychopharmacology (Berl). 2008;199:331-388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 427]  [Cited by in F6Publishing: 415]  [Article Influence: 25.9]  [Reference Citation Analysis (0)]
152.  Greenwood TA, Light GA, Swerdlow NR, Calkins ME, Green MF, Gur RE, Gur RC, Lazzeroni LC, Nuechterlein KH, Olincy A, Radant AD, Seidman LJ, Siever LJ, Silverman JM, Stone WS, Sugar CA, Tsuang DW, Tsuang MT, Turetsky BI, Freedman R, Braff DL. Gating Deficit Heritability and Correlation With Increased Clinical Severity in Schizophrenia Patients With Positive Family History. Am J Psychiatry. 2016;173:385-391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 39]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
153.  Graham FK. Presidential Address, 1974. The more or less startling effects of weak prestimulation. Psychophysiology. 1975;12:238-248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 856]  [Cited by in F6Publishing: 848]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
154.  Braff DL, Geyer MA. Sensorimotor gating and schizophrenia. Human and animal model studies. Arch Gen Psychiatry. 1990;47:181-188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 977]  [Cited by in F6Publishing: 979]  [Article Influence: 28.8]  [Reference Citation Analysis (0)]
155.  Morales-Muñoz I, Jurado-Barba R, Ponce G, Martínez-Gras I, Jiménez-Arriero MÁ, Moratti S, Rubio G. Characterizing cannabis-induced psychosis: a study with prepulse inhibition of the startle reflex. Psychiatry Res. 2014;220:535-540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 13]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
156.  Tervahartiala K, Karlsson L, Pelto J, Kortesluoma S, Hyttinen S, Ahtola A, Junttila N, Karlsson H. Toddlers' diurnal cortisol levels affected by out-of-home, center-based childcare and at-home, guardian-supervised childcare: comparison between different caregiving contexts. Eur Child Adolesc Psychiatry. 2019;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
157.  Landram MJ, Koch AJ, Mayhew JL. Salivary stress hormone response and performance in full competition after linear or undulating periodization training in elite powerlifters. J Sports Med Phys Fitness. 2020;60:152-159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
158.  Sanders RD, Hassell J, Davidson AJ, Robertson NJ, Ma D. Impact of anaesthetics and surgery on neurodevelopment: an update. Br J Anaesth. 2013;110 Suppl 1:i53-i72.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 147]  [Cited by in F6Publishing: 146]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
159.  Stratmann G. Review article: Neurotoxicity of anesthetic drugs in the developing brain. Anesth Analg. 2011;113:1170-1179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 133]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
160.  US Food and Drug Administration. FDA drug safety communication: FDA review results in new warnings about using general anesthetics and sedation drugs in young children and pregnant women. [accessed 2017 Aug 25].  Available from: https://www.fda.gov/Drugs /DrugSafety/ucm532356.htm.  [PubMed]  [DOI]  [Cited in This Article: ]