Background: In contemporary medical practice, general anesthesia plays an essential role in pediatric surgical procedures. While modern anesthetic protocols have demonstrated safety and efficacy across various pathological conditions, concerns persist regarding the potential neurotoxic effects associated with early exposure to general anesthesia. Summary: Current research primarily examines the neurocognitive developmental impacts, with limited focus on neurobehavioral developmental disorders. This review presents a comprehensive analysis of clinical trial results related to five critical neurobehavioral developmental disorders: fine motor disability, attention-deficit hyperactivity disorder, impulse control disorders, autism spectrum disorder, and developmental coordination disorder. Furthermore, this review synthesizes insights from basic research on the potential toxicological mechanisms of general anesthetic agents that could influence clinical neurobehavioral changes. These findings provide valuable guidance for the prudent and safe utilization of anesthetic agents in pediatric patients. Key Messages: This review explores the potential connections between general anesthesia and five neurobehavioral disorders, highlighting the importance of cautious anesthetic use in children in light of current research findings.

General anesthesia has become an integral part of surgical procedures in contemporary clinical practice. It not only greatly improves patient comfort during surgery by effectively reducing intraoperative pain but also opens up the possibility for surgeons to perform more complex and time-consuming procedures. Modern anesthetic protocols and strategies have been proven to be safe and recommended in many pathological conditions, but certain commonly used intravenous and inhalation general anesthetic agents were found to cause neuronal apoptosis and impair synaptogenesis in immature animals (including nonhuman primates), affecting brain development and leading to cognitive decline as well as a variety of long-lasting (or even permanent) behavioral deficits, which can severely impact normal life [1, 2]. Being very vulnerable at the critical stage of synaptogenesis, the brain is highly susceptible to general anesthesia [3]. The “window of vulnerability” in humans has been reported to be between the last third trimester of pregnancy and 2–3 years of age [4‒6]. The undesirable impact of general anesthesia imposed on the neurobehavioral development of these groups of populations is worth being studied.

With the development of society and the continuous improvement of medical care, a considerable number of children undergo general anesthesia each year in the USA alone [7], and given the population density of China was 4 times that of the USA, it was expected that the number of infants and children receiving surgical procedures under general anesthesia in China would be several times higher than that in the USA. Considering the large population, despite the current uncertainty about the long-term neurotoxicity of early exposure to general anesthesia, the US Food and Drug Administration (FDA) has warned that the multiple (two or more) or prolonged (more than 3 h) use of general anesthetics and sedative medications during surgical procedures in pregnant women in their last trimester of pregnancy, and children under the age of 3 years may affect the brain development of both the fetus and the child [8]. Meanwhile, guidelines from the UK and Ireland [9] and a statement from the European Institutions [10], though recommending avoiding unnecessary general anesthesia, do not suggest adjustments to clinical practice. Determining the risk of neurotoxicity from general anesthesia is therefore an urgent public health issue.

To date, clinical reports have generally shown that children exposed to general anesthesia during procedures at least bear moderate risk of neurodevelopmental impairment but do not show cognitive decline on intelligence tests compared with their peers not exposed to general anesthesia [11‒13]. However, the neurotoxic impact induced by general anesthesia can still be monitored through more comprehensive cognitive assessments during later childhood development. As more clinical investigations on general anesthesia have been conducted, we are beginning to observe that exposure to general anesthesia during infancy may exert adverse effects on children’s long-term behavioral development compared with those who have not experienced such exposure, ranging from internalizing behavioral problems (e.g., anxiety, depression, or somatization symptoms) to externalizing behavioral problems (e.g., impulsive and disruptive behaviors) which are exacerbated by impairments in executive functioning [14, 15]. Of greater concern, in addition to the neurobehavioral deficits observed, a growing body of research points to the possibility that early exposure to general anesthesia may also induce social and emotional deficits, such as attention-deficit hyperactivity disorder (ADHD) and autism spectrum disorder (ASD). Furthermore, a large body of preclinical evidence suggests that a variety of animals (from nematodes to nonhuman primates) exposed to general anesthesia during critical stages of brain development experience long-term neurobehavioral changes [2]. Thus, it is reasonable to hypothesize that exposure to general anesthesia at an early age may be associated with neurodevelopmental behavioral disorders in children.

This review retrospects a large number of clinical studies of early exposure to general anesthesia affecting neurobehavioral development in children, summaries the toxicological mechanisms of general anesthetics that have been closely associated with clinically significant neurobehavioral changes in basic research, and focuses on fine motor disability, ADHD, impulse control disorders (ICDs), ASD, and developmental coordination disorder. Five key neurobehavioral disorders are discussed, and future perspectives are presented to provide references for more rational and safer clinical use of general anesthesia in infants and young children.

The possibility that fetuses, infants, and young children exposed to general anesthesia may suffer from persistent neurotoxicity, which may induce long-term neurobehavioral disorders and developmental deficits, requires broad attention and in-depth research from both the academic and societal perspectives. We will explore the long-term effects of early exposure to general anesthesia on the neurobehavioral functions of children with five representative neurobehavioral disorders, including fine motor disability, ADHD, ICDs, ASD, and DCD.

Fine Motor Disability

Fine motor disability is the abnormality in the coordination of small muscle groups that hinders the body’ to perform tasks that require some degree of manual dexterity [16, 17]. Patients usually present with symptoms such as poor finger movements, impaired hand-eye coordination, and lack of accuracy in hand manipulation. The execution of fine motor skills requires the synergistic action of multiple brain regions, with even the simplest fine motor skills involving effective connections among the premotor cortex, motor cortex, cerebellum, basal ganglia, pyramidal tract, and peripheral nerves, not to mention visuospatial, sensory, and executive functions, which are more complex skills [18, 19]. Fine motor deficit usually appears as one of the symptoms of an underlying disease process and is rarely discussed as an independent disorder, but negative consequences it brings can affect the normal life of the child. Therefore, special attention should be paid to any risk factors that may induce fine motor deficit.

Given that a significant number of children undergo general anesthesia each year, whether general anesthesia has an impact on the long-term fine motor skills of children has become a widespread concern in the society. A series of clinical studies have been conducted to provide valuable references. In 2018, the Mayo Anesthesia Safety in Kids (MASK) study retrospectively analyzed nearly 1,000 cases, with changes in intelligence being the primary outcome, while behavioral changes being the secondary outcome. Evaluating intellectual impact in children predominantly relies on standardized full-scale intelligence quotient (IQ) scores derived from the Wechsler Intelligence Scale. In contrast, determining the deficits in fine motor skill necessitates a comprehensive neuropsychological assessment combined with parental reports to ensure a thorough evaluation. The study ultimately found that there were no significant differences in intelligence between children who received general anesthesia twice or more and those who experienced single or no anesthetic exposure before 3 years of age. Whereas, the multiple exposure group, as they grew into adolescence, showed a decrease in problem-solving speed as well as compromised fine motor skills comparing with none or single exposure group [12]. The conclusion drawn from the secondary indicators is preliminary and exploratory, requiring more accurate analytical methods to further validate. Therefore, the team conducted a more thorough and comprehensive statistical analysis of nearly 1,000 cases using factor analysis and cluster analysis in 2019. The results of the analysis remain consistent with the observation that early repeated exposures to general anesthesia does not lead to cognitive deficits but may cause impairments in specific behavioral patterns, such as reduced problem processing speed and fine motor skills [20]. In 2020, a cohort study fully assessed the neurodevelopmental outcomes of 13,433 children between the ages of 7 and 16 across multiple dimensions of learning, cognition, motor, social behavior, and language using dozens of different tests and scales. Similarly, the study revealed that children exposed to general anesthesia at an early age did not show significant differences in neurocognitive aspects but exhibited specific impairments in motor skills and fine motor abilities (hand dexterity) [21]. Another cohort study in the same year evaluated child development with the Japanese translation of the Ages and Stages Questionnaires, third edition (J-ASQ-3) and demonstrated that infants who underwent three or more general anesthesia exposure had neurodevelopmental delays at 1 year of age, which may result in a significantly increased risk of delays in the development of communicative, gross motor, fine motor, problem-solving, and interpersonal skills, especially gross motor skills [22]. The above clinical studies indicated that early exposure to general anesthesia may cause fine motor disability to some children.

A series of preclinical studies have also been initiated to delve into the mechanisms of general anesthesia-induced neurotoxicity on fine motor skills. Sevoflurane, as one of the most commonly used general anesthetic agents in pediatric surgery, is a preferred target for exploring the neurotoxicity of pediatric general anesthetics. Published in 2022, the study on the possibility that sevoflurane exposure may cause dendritic spine dysplasia and impaired fine motor ability in developing mice via the PI3K/AKT/mTOR pathway has attracted much attention. In this experimental investigation, neonatal C57BL/6 mice at 7 days of age received intraperitoneal injections 30 min prior to exposure to either 2% sevoflurane or 100% oxygen. Specifically, the mice were administered either the PI3K/AKT/mTOR activator IGF-1 or the mTOR phosphorylation inhibitor KU0063794. Subsequently, the mice were exposed to either 2% sevoflurane or 100% oxygen for 3 h. Within 6 h after the cessation of sevoflurane or pure oxygen exposure, the hippocampus was rapidly collected for subsequent Western blotting experiments to detect protein expression of PI3K/AKT/mTOR pathway phosphorylation. In addition to this, the team performed Golgi staining of the brain tissue of the mice at postnatal day 14 and observed the morphological features of the dendritic spine of hippocampal neurons using oil immersion microscopy. As the mice grew into adolescence (at postnatal day 48), fine motor skills were measured by the balance beam test. The results of the study illustrated that early exposure to sevoflurane reduced the proportion of fine dendritic spine and increased the proportion of mushroom-shaped dendritic spine but did not change the density of dendritic spine. In addition, early sevoflurane exposure inhibited phosphorylation of the PI3K/AKT/mTOR pathway in the hippocampus of young mice, ultimately leading to long-term fine motor disability. This impairment can be rescued by pretreatment with IGF-1, whereas pretreatment with KU0063794 exacerbated the damage [23]. In conclusion, this study investigated the role of changes in dendritic spine plasticity in sevoflurane-induced neurodevelopmental toxicity and provided new insights into the neurotoxic effects of sevoflurane. A study published in 2021 explored general anesthetics and mechanisms of postoperative long-range fine motor injury using juvenile nonhuman primates and rodents, and for the first time, highlighted the effects and mechanisms of m6A methylation on fine motor injury in sevoflurane anesthesia [24]. m6A modification is the most abundant form of RNA methylation modification, and YTHDF1 is one of the recognition proteins for m6A methylation and has been testified to be involved in regulating the formation and development of neurocognition [25‒27]. This study found that the expression of YTHDF1 was significantly downregulated in the brains of juvenile primates and rodents after sevoflurane anesthesia. Furthermore, single-cell sequencing results manifested that the decline in YTHDF1 expression was most pronounced in sp8-positive interneurons. The results of RNA-binding protein immunoprecipitation assay and m6A-seq assay showed that m6A in the synaptophysin mRNA where existed YTHDF1-binding sites was highly enriched. Considering that the main function of YTHDF1 is to recognize methylation sites on RNA, a strong correlation between synaptophysin and neurodevelopmental toxicity of general anesthetic agents was found in previous studies [28], and the overexpression of YTHDF1 rescued sevoflurane-induced fine motor dysfunction, cognitive deficits, and changes in synaptophysin in young mice was detected in this study, it was elucidated that YTHDF1 regulated the expression of its downstream target gene synaptophysin in an m6A methylation-dependent manner, and early sevoflurane exposure interfered with the interaction of YTHDF1 with the downstream genes, which led to fine motor skill deficits [24]. This finding opened up a whole new area of research to unveil the potential mechanisms of the effects of general anesthesia on the neurodevelopment of the young brain.

The integrity of the myelin sheath is a key structure for the brain’s processing speed and motor coordination function. Thus, developmental defects in myelin sheaths will likely result in fine motor deficits. As mentioned earlier, a series of clinical studies have found that early exposure to general anesthetics was associated with postoperative fine motor dysfunction long after the surgery. Consequently, the hypothesis that general anesthetics inducing developmental defects in myelin sheaths may lead to fine motor deficits was raised. In 2019, the concept of myelin developmental toxicity of general anesthetics was first introduced in a study of the neurodevelopmental toxicity of general anesthetics. The study found that young rhesus monkeys and young rats exposed to sevoflurane both had reduced expression of Ermin protein-coding gene (ERMN), a key gene for myelin development, in the prefrontal cortex. The result was validated in the classical model of rats exposed to sevoflurane. The myelin development of those rats was found to be impaired, and they showed long-term neurobehavioral changes [29]. This finding attracted widespread attention, setting off a wave of research into the effects of general anesthetics on myelin development [30‒32], which have brought the concept of myelin developmental toxicity of general anesthetics to the forefront. Meanwhile, the idea that well- or poorly-developed myelination in the brain is closely linked to fine motor skills has also been widely discussed [33, 34]. For example, in 2021, Cullen et al. [34] put forward that mice with conditional knockout of the Kif3a gene in oligodendrocyte (OL) precursor cells demonstrate fine motor dysfunction, manifested by a significant reduction in stride length and an increase in step frequency during gait analysis. Additionally, these mice exhibit increased error counts in specific balance and coordination tasks. OL precursor cells have the capacity to proliferate and differentiate into mature OLs, which migrate along axons to form myelin sheaths. Kif3a knockout mice have impaired primary cilia assembly and OL production, leading to defective myelin development and ultimately fine motor coordination [34]. Taken together, these clinical and preclinical studies are important in exploring the relationship between early general anesthesia exposure and long-term fine motor deficits, as well as possible underlying mechanisms.

ADHD

ADHD, also known as mildly dysfunctional brain syndrome, is a neuropsychological developmental disorder characterized by hyperactivity, impulsivity, inattention, lack of vigilance, and an inability to adapt to rapid change [35]. It has a fairly high prevalence rate. Approximately 6%–9% of children and adolescents were affected globally, with a prevalence of 5.29% in population aged under 18 [36], among which approximately 50%–80% have ADHD symptoms till their adulthood, manifesting severe behavioral and psychiatric problems. It is therefore crucial to identify people with ADHD timely and provide necessary supports and treatments to reduce the burden on patients, their families, and the society.

The idea that there was a link between early exposure to general anesthesia, and the incidence of ADHD was first proposed 12 years ago when Sprung et al.’s [37] team found that children exposed to general anesthesia multiple times (two or more times) in the early years were at a three-fold increased risk of developing ADHD, while there was no significant association between a single exposure to general anesthesia and the development of ADHD. Similar conclusions were drawn from another paper published in 2015. Through Cox regression analysis of data collected from 1,146 children who had received general anesthesia and 3,438 participants in age- and gender-matched control group, the study evidenced that the risk of being diagnosed with ADHD in children who were exposed to general anesthesia once and for less than 3 h did not increase. However, for children who were exposed to general anesthesia multiple times and for more than 3 h, the risk of developing ADHD in the future significantly increased [38]. Nevertheless, the results of numerous studies on the association between single general anesthesia exposure and the subsequent development of ADHD have been inconsistent. Studies with large sample sizes have demonstrated an increased risk of future development of ADHD in children exposed to general anesthesia on a single occasion [39], whereas studies with small sample sizes have found no such increased risk [40, 41]. The difference may lie in the smaller risk ratio. Specifically, only when the sample size of the study was large enough can the relevance between general anesthesia and ADHD be clearly determined. Therefore, a detailed analysis of studies with large sample sizes would be provided. An article published in 2017 examined the relationship between exposure to surgery and anesthesia at a young age and the diagnosis of mental disorders in children. It conducted survival analysis on 38,493 children who were exposed to general anesthesia and surgery at least once, and 192,465 children who have not been exposed to general anesthesia before the age of 5, with two groups being matched in age and gender. Ultimately, the result indicated that children who underwent surgery and anesthesia before the age of 5 had a slightly increased risk of psychological disorders, including ADHD, but the increased risk was not dependent on the specific timing of the surgery and anesthesia [39]. Another study similarly noted that children undergoing general anesthesia before the age of 5 years were at an increased risk of developing ADHD and that multiple general anesthesia exposures further intensified this potential risk. It also found that race was a key moderator of this risk that regardless of the age of first exposure, non-white children being more likely to develop ADHD after general anesthesia than white children [42]. A recent publication filled the research gap in children aged 0–3 years. Using a population-based cohort study and propensity score matching methodology, the study aimed to investigate the potential association between general anesthesia exposure and the development of ADHD in children between 0 and 3 years of age. The result revealed that exposure to general anesthesia before the age of 3 years significantly raised the risk of developing ADHD in the future. This finding was consistent with the results of the aforementioned study, which used five-year-old children as subjects. Meanwhile, the study identified several risk factors for ADHD, including times of exposure to general anesthesia, duration of exposure, gender, and central nervous system surgery [43]. In addition to the clinical studies reviewed above, an experimental animal study reported that early ketamine exposure caused hyperactive behavior similar to ADHD, which further supported the idea that repeated exposures to general anesthesia in children was associated with the development of ADHD [44]. Therefore, in clinical practice, appropriate general anesthesia strategies, preventive measures, and follow-up measures should be adopted for children undergoing surgical procedures that include general anesthesia. Furthermore, the potential mechanisms by which early exposure to general anesthesia leads to ADHD should be elucidated and ways to minimize the possible threat posed by general anesthesia should be explored.

From a molecular genetic perspective, ADHD is one of the most heritable psychiatric disorders. Children whose parents have ADHD are subjected to a two- to eight-fold increased risk of developing ADHD, and siblings of children with ADHD also face an increased risk of ADHD [45]. Whereas the specific genetic mechanisms of ADHD may involve interactions between multiple genes and environmental factors, which are complex and remain to be disclosed. Faraone’s team has been dedicated to exploring the molecular genetics of ADHD and has identified candidate genes associated with ADHD, such as DRD4, DRD5, SLC6A3, SNAP-25, HTR1B, DBH, MAOA, SLC6A2, TPH-2, SLC6A4, CHRNA4, GRIN2A, and ADRA2A. Despite their weak effects, these candidate genes confirmed that genetic susceptibility to ADHD stemmed from the combined action of multiple small effector genes. These candidate genes contained those associated with dopamine, norepinephrine, 5-hydroxytryptamine-related receptors and transporter proteins, brain-derived neurotrophic factors, and nicotinic acetylcholine receptor α4 subunit genes, constructing a map of genes related to the regulation of neurotransmitter pathways associated with ADHD [46]. Nonetheless, the current understanding of the molecular genetics of ADHD is still limited, and studies with more robust statistical methods to detect small effector genes, large-scale collaborative studies, and genome-wide association studies can be conducted to further uncover the molecular genetics of ADHD.

From a molecular biology perspective, the pathogenesis of ADHD involves the central dopamine neuronal pathway, the noradrenergic neuronal pathway, the serotonergic neuronal pathway, the glutamatergic neuronal pathway, as well as brain-derived neurotrophic factors. Numerous genetic studies have indirectly substantiated the reliability of these systems’ association with ADHD [46]. Recent studies have shown that abnormalities in the regulation of the neural network comprising the frontal cortex of the brain and its downstream nervous system were closely linked to the development of ADHD [47]. In the neural network, the critical functions of dopaminergic neurons in the central nervous system were implied through 5 different dopamine receptors. Damage to specific brain regions, such as the ventral tegmental area of the midbrain, can result in diminished dopamine neurotransmission within the prefrontal cortex. This reduction in neurotransmission can lead to an increase in dopamine receptor sensitivity, potentially triggering ADHD. Some studies have revealed that different dopamine receptor subtypes were closely linked to the pathogenesis of ADHD. A deeper understanding of the relationship between these five different dopamine receptors and ADHD will help elucidate the different roles of these receptors and inspire more therapeutic approaches for ADHD. In 2012, Wu et al. [48] published an article on the role of dopamine receptors in ADHD, which pointed out that the activity of D2 and D3 receptor subtypes was increased in the striatum of adolescents with ADHD, and D2, D4, and D5 receptor subtypes were closely related to the onset of ADHD. Methylphenidate and amphetamine are currently used clinically to treat ADHD, and they act by stimulating D1 receptors in the prefrontal cortex and D2 receptors in the striatum. In addition to clinical studies, an animal study also found that mice with conditional knockout of circadian clock genes exhibited decreased activity of dopaminergic neurons in the ventral striatum, hypothalamus, and prefrontal cortex. These mice displayed behavioral manifestations similar to ADHD. Furthermore, it was observed that damage to dopaminergic neurons exacerbated symptoms of ADHD [49]. This finding further testified the involvement of central dopaminergic neuronal pathways in the pathogenesis of ADHD. Recent studies have illustrated that the central dopamine pathway was involved in general anesthesia. Therefore, early exposure to general anesthesia may lead to alterations in the structure and function of the central dopaminergic system, which may predispose patients to ADHD. The conclusions drawn from this study were actually in line with similar propositions put forth as early as 2008 [50], suggesting that dysregulation of the catecholamine system may lead to ADHD. ADHD was considered a consequence of abnormality in the brain’s frontal lobe regions, particularly the prefrontal cortex and associated subcortical structures and circuits. The underlying anomalies were attributed to disruptions in catecholamine neurotransmission. Patients with ADHD exhibited decreased levels of dopamine and norepinephrine, primarily due to impairments in their respective neurotransmitter transport systems. The efficacy of catecholaminergic stimulants further confirmed that neurotransmitter abnormalities in ADHD patients primarily stemmed from the catecholamine system. This system comprises the dopamine system as well as the norepinephrine and adrenaline systems, which aligns with the implications of the gene map related to neurotransmitter pathway regulation associated with ADHD mentioned earlier [50]. Besides, abnormalities in glutamatergic pyramidal neurons, a major cellular component of the prefrontal cortex and associated subcortical structures, are often cited as a cause of ADHD. Prefrontal cortex exerts its functions largely on glutamatergic neurotransmission. In 2017, an article stated that the impairment of AMPA receptors in pyramidal neurons in the prefrontal cortex of animal models of ADHD led to dysfunctional glutamatergic transmission and ultimately to behavioral abnormalities. Methamphetamine can normalize ADHD-like behavior by improving AMPA receptor function and activating pyramidal neurons in the prefrontal cortex. These findings demonstrated that dysfunction of AMPA receptor in the prefrontal cortex may be responsible for adolescent ADHD behaviors, and enhancing the prefrontal cortex activity may be one of the therapeutic strategies for ADHD [51]. The neurotoxic effects of general anesthesia cause impaired neuronal development in the prefrontal cortex, resulting in cognitive and behavioral developmental deficits. It is therefore reasonable to speculate that AMPAR may be involved in the pathogenesis of ADHD from general anesthesia exposure. Another study manifested that ketamine and other NMDAR antagonists can induce ADHD-like behaviors in mice, accompanied by alterations in dendritic spine morphology in the striatum [52]. Symptoms of ADHD in mice can be altered by the activation of pyramidal NMDAR [51]. Both AMPAR and NMDAR belong to the family of glutamatergic receptors, indicating that the glutamatergic system may lie behind the pathogenesis of general anesthesia exposure-induced ADHD. In addition to this, BDNF is also closely related to the development of ADHD. The formation and plasticity of neuronal spine may underlie the pathogenesis of ADHD and are regulated by brain-derived neurotrophic factors [53]. During the development of the frontal lobe of the brain, the reduction of BDNF may induce ADHD [54]. Moreover, the severity of ADHD is strongly associated with reduced BDNF concentrations, especially in the midbrain [55]. Anesthesia may diminish BDNF, whereas methamphetamine increases plasma BDNF concentrations in children with ADHD [56]. Therefore, general anesthesia may weaken the expression of BDNF and interfere with the normal process of synaptogenesis, thus inducing ADHD. This conclusion provides us with important clues for further investigation of the pathological mechanisms of ADHD. Taken together, these clinical and preclinical studies are significant for combing the correlation between early exposure to general anesthesia and ADHD, as well as possible underlying mechanisms.

ICDs

ICDs are seldom discussed as an independent disorder and are often considered as clinical diagnostic and pathophysiological markers of some neurobehavioral disorders, such as ADHD, borderline personality disorder, obsessive-compulsive disorder, trichotillomania, pathological gambling, and chronic substance abuse [57, 58]. Therefore, the research on ICDs is often in conjunction with ADHD. ADHD, a chronic neurodevelopmental disorder, is one of the most prevalent neurobehavioral disorders in children, presenting a growing global concern. The core symptoms of ADHD include inattention, hyperactivity, and impulsivity [35], with impulsivity being a critical symptom for diagnosing the disorder. Impulsivity is characterized by difficulties in inhibiting behaviors and delayed aversion [59], such as impulsive responses before task completion, challenges in waiting for turns, or a tendency to interrupt and disturb others. Adolescents and adults with ADHD-like impulsive behaviors have been found to have a significantly increased propensity to engage in undesirable behaviors such as drug addiction, pathological gambling, and suicide, resulting in significant negative consequences for the individual, family, and society [60].

Currently, a multitude of clinical investigations have highlighted a correlation between early repeated exposures to general anesthesia and the subsequent development of ADHD, but few studies concentrated specifically on the relationship between early exposure to general anesthesia and ADHD-like impulse control deficits. Given the potential implications for public mental health and social stability, it is crucial to establish the connection between early exposure to general anesthesia and ADHD-like ICDs and further explore the underlying mechanisms.

As the most widely used general anesthetic in clinical practice for infants and young children, sevoflurane exerts its efficacy by both inhibiting neuronal activity through its action on gamma-aminobutyric acid (GABA) receptors and attenuating neuronal excitatory effects via the reduction of NMDA receptor activity [61]. Multiple studies have demonstrated that early exposure to sevoflurane may engender synaptic alterations and neuronal morphological abnormalities in various brain regions during development, particularly in the prefrontal cortex [1]. In 2012, Briner et al. [62] put forward that general anesthetics could increase dendritic spine density in the prefrontal cortex of rats during the “synaptogenesis” process. During critical developmental periods, neurons in the prefrontal cortex are particularly vulnerable to the neurotoxic effects induced by sevoflurane exposure [63], and these effects are enduring [64]. Of further significance, current research has indicated the involvement of the prefrontal cortex in the regulation of impulsive behavior [65], with patients exhibiting ADHD-like impulsive behaviors often displaying hyperactivity in the prefrontal cortex [66]. The prefrontal cortex integrates input from various brain regions, encodes and regulates higher cognitive functions, including inhibitory responses and impulse control, to achieve corresponding physiological functions [67, 68]. The normal functioning of the brain relies on a delicate dynamic balance between excitatory and inhibitory signals. Any disruption in this balance within the neural circuits of the brain during development can easily precipitate related neurological disorders. In 2020, Xie et al. [64] investigated the induction of ADHD-like impulsive behavior in adult mice following early repeated exposures to general anesthesia, along with the underlying mechanisms. In their study, postnatal day 6 (a critical period for brain development) C57 mice were exposed to 3% sevoflurane for 2 h per day over 3 consecutive days. Mice in the control group were exposed to air at an equivalent flow rate, while all other experimental conditions remained consistent with the sevoflurane group. Subsequently, the cliff avoidance reactions of 8-week mice were tested to assess their impulsivity. The results revealed that compared to mice in the air control group, those in the sevoflurane group exhibited a significant increase in the number of cliff jumps during the testing period, along with a significant reduction in the latency to the first cliff jump. Moreover, in order to eliminate impulsive behavior caused by anxiety or fear, the research team performed elevated plus maze tests (EPM) following the cliff avoidance reaction tests. No significant differences were detected between the two groups in terms of either the time spent exploring the open arms or the number of entries into the open arms. Additionally, compared to mice in the air control group, those in the sevoflurane group exhibited shorter durations of freezing behavior, indicative of fear, during the EPM tests. This suggested that early repeated exposures to sevoflurane after birth can induce ADHD-like impulsive behavior in adulthood. Furthermore, the study utilized immunofluorescence staining experiments to further investigate whether the medial prefrontal cortex region was involved in sevoflurane-induced ADHD-like impulsive behavior. The experimental results revealed a significant increase in c-Fos signal expression in the medial prefrontal cortex region of mice in the sevoflurane group compared to the air control group, with a greater number of co-localized cells showing both c-Fos signal and excitatory neuron marker (CaMKIIα+) signal. This proved that the ADHD-like impulsive behavior induced by early sevoflurane exposure was mediated by excitatory neurons in the prefrontal cortex region. Finally, the team utilized designer receptors exclusively activated by designer drugs technology to activate excitatory neurons in the prefrontal cortex region of mice in the air control group and simultaneously inhibit excitatory neurons in the prefrontal cortex region of mice in the sevoflurane group. The results demonstrated that mice with activated excitatory neurons in the prefrontal cortex exhibited ADHD-like impulsive behavior, while inhibiting excitatory neurons in the prefrontal cortex alleviated the ADHD-like impulsive behavior induced by sevoflurane exposure. This further suggests a close association between the activity of excitatory neurons in the medial prefrontal cortex region and the ADHD-like impulsive behavior induced by sevoflurane exposure. In summary, this study manifested that early repeated exposures to sevoflurane may disrupt the development of medial prefrontal cortex neurons, leading to an increased ratio of excitatory to inhibitory signals in the medial prefrontal cortex neurons area, thereby triggering ADHD-like impulsivity behaviors [64]. The research undertaken by Xie’s team represents a rare focus on early-life general anesthesia exposure and its relationship with ICDs, pioneering an examination into the functional involvement of the medial prefrontal cortex in ADHD-like impulsive behaviors and filling the current research gap. However, the medial prefrontal cortex is intricately connected to numerous neural circuits characterized by complex afferent and efferent projections, which have been inextricably associated with the pathogenesis of psychiatric disorders [69]. The precise neural circuits underlying impulsive behavior control disorders remain elusive. As a consequence, future investigations may use chemogenetics or optogenetics techniques to reveal the interactions between medial prefrontal cortex projection neurons and downstream target brain regions, elucidating potential pathogenic pathways behind ADHD-like impulsive behaviors.

In addition, it is also evident that both dopamine and serotonin systems play indispensable roles in ADHD-like impulsive behaviors. While the noradrenergic system is implicated in the onset and treatment of ADHD, its regulatory role in impulsive behavior remains poorly understood. Methylphenidate, the most commonly prescribed medication for ADHD, acts through the modulation of monoaminergic neurotransmitter imbalances, particularly dopamine or norepinephrine dysregulation. Most studies have substantiated that atomoxetine has a significant impact on impulsive behavior tests in rodents, such as the stop-signal task, delay discounting task, and 5-choice serial reaction time task. Yet, the specific neural mechanisms involved remained to be unearthed [70, 71]. This observation superficially reinforced the intricate relationship between methylphenidate’s dopaminergic modulation and impulse regulation. Beyond mere pharmacological inquiry, several clinical investigations have unveiled anomalies in dopamine transporter function and dopamine dehydrogenase activity within the prefrontal cortex of individuals with ADHD [72, 73]. Additionally, supporting evidence from animal models underscores the significant correlation between the dopamine system and impulsive behaviors. Both dopamine transporter-deficient mice (DAT KO) and rats subjected to neonatal 6-hydroxydopamine lesions demonstrate impulsive traits across behavioral paradigms [74]. Collectively, clinical and preclinical findings highlighted the irreplaceable contribution of the dopamine system to ADHD-like impulsivity. Similarly, the serotonin system emerges as a pivotal player in ADHD-like impulsivity. On the other hand, Daniel and his colleagues investigated the impact of reduced levels of serotonin (5-HT) in the central nervous system following rapid tryptophan depletion (RTD) on impulsive aggressive behavior in boys with ADHD. The study employed a double-blind crossover design, where 22 male adolescents with ADHD underwent RTD testing on 1 day and were given a tryptophan-balanced placebo on another day. Competitive reaction time games were used within 270 min after ingesting the RTD/placebo to stimulate aggressive behavior. The results revealed that RTD significantly increased aggression in individuals with low impulsivity [75]. Furthermore, investigations reflecting the therapeutic efficacy of selective serotonin reuptake inhibitors in ADHD treatment have yielded equivocal outcomes, elucidating serotonin’s nuanced role in ADHD etiology. However, selective serotonin reuptake inhibitors demonstrate therapeutic efficacy in alleviating other ICDs such as intermittent explosive disorder [76]. Notably, substantial evidence supports that 5-HT modulates DA levels, and their interaction alters the equilibrium between them, inducing different levels or types of impulsive behavior. This may also be one of the key factors in the neurochemical basis of ADHD [77]. In summary, the intricate interplay between the dopamine and serotonin systems emerges as a cornerstone in elucidating the mechanisms underpinning ADHD-like impulsive behaviors. Taken together, these related studies serve as valuable resources for exploring the correlation between early exposure to general anesthetics and ADHD-like impulsive behavior, as well as possible underlying mechanisms.

ASD

ASD is a neurodevelopmental disorder characterized by deficits in social communication, restricted interests, and repetitive behaviors [78]. According to the World Health Organization, the estimated international prevalence of ASD was 0.6% [79]. The steadily climbing prevalence over the past decades has garnered widespread attention globally.

ASD is a highly heritable disorder. Genetic factors play an undeniable role in the etiology of ASD, but the phenotypic expression of genetic susceptibility shows considerable heterogeneity among individuals with ASD [80‒82]. Moreover, in specific patient populations, genetic factors may be modulated by obstetric and perinatal factors as well as early neonatal environmental exposures [83]. Therefore, the correlation between cesarean section delivery and ASD has become a direction of academic interests. In addition, cesarean section is a surgical procedure used in childbirth to prevent risks of compromising the health of the mother or baby during the process of vaginal delivery, and the global incidence of cesarean section takes a mounting trend, with an estimated annual growth of 4% [84]. The proportion of cesarean section in China has rapidly increased to approximately 40% [85]. All these add to the imperative to clarify the relationship between cesarean section and ASD. A meta-analysis indicated that cesarean section was associated with a 23% increased risk of ASD compared to vaginal delivery [85]. Regional anesthesia is often the preferred choice for cesarean section surgeries, while general anesthesia is primarily used for emergency deliveries or in cases where there is a maternal preference, insufficient regional anesthesia, or failed attempts at regional anesthesia [86]. Whereas, in recent years, due to elevated risks to the mother, the frequency of using regional anesthesia in cesarean section surgeries has been decreasing [87]. Meanwhile, more animal experiments showed that several general anesthesia techniques can lead to abnormal apoptosis of neurons and behavioral defects. Investigating the correlation between general anesthesia and the risk of ASD has been particularly necessary. In 2015, Chien and colleagues [88] evaluated the subsequent risk of ASD in newborns under general anesthesia during cesarean section by comparing it with the risk of ASD under natural delivery or regional anesthesia. The results showed that compared to vaginal delivery, newborns delivered via cesarean section under general anesthesia bore a 52% increased risk of developing ASD. Additionally, the incidence of autism was higher in them compared to those delivered via cesarean section with RA. Therefore, the use of general anesthesia during cesarean section was associated with an increased risk of neurodevelopmental disorders in children, such as ASD. It is worth noting that this study did not demonstrate a causal relationship between newborns delivered via cesarean section under general anesthesia and the subsequent risk of developing ASD; it only showed a statistical association between them [88]. In 2021, Yang and colleagues [89] conducted a study examining the relationship between different anesthesia methods and ASD, revealing a potential association between general anesthesia and increased risk of ASD. The study employed multivariable conditional logistic regression analysis to process data from 950 ASD patients and 764 controls. After adjusting for confounding factors, the team found that newborns delivered via cesarean section with general anesthesia had a significantly higher risk of developing ASD compared to those delivered via cesarean section with regional anesthesia. This suggests that, after considering other potential influencing factors, general anesthesia may be an independent risk factor for ASD [89]. While there may be conflicting findings in the above studies, they provided valuable insights into the complex relationship between exposure to general anesthesia during pregnancy and the subsequent development of ASD in children, and the possible guidance for future clinical practices aimed at lowering the potential risk of ASD.

In addition to obstetric and perinatal factors influencing ASD, early neonatal exposure may also modulate ASD development. Among these, exposure to general anesthesia is one of the common exposures in newborns. Current research suggests that early childhood exposure to general anesthesia may lead to neurodevelopmental disorders such as ASD. Nevertheless, this potential link was rarely studied, and the results were not convincing due to small sample sizes [90, 91]. In 2023, Pikwer et al. [92] conducted a large-scale study on the relationship between early childhood exposure to general anesthesia and the risk of ASD. This study represents the first population-based research of its kind, encompassing 401,750 children aged 0–5 years who underwent general anesthesia, and a matched control group of 1,187,796 children. After the thorough consideration of potential confounding factors, the study revealed that the risk of developing ASD in children exposed to general anesthesia early in life was nearly doubled compared to those not exposed to general anesthesia. Additionally, it was observed that children exposed to general anesthesia but not undergoing surgery had a higher risk of developing autism subsequently compared to those who received both general anesthesia and surgery. This implied that general anesthesia may represent an independent risk factor distinct from the surgical procedure itself. Furthermore, the study found that children exposed to both general anesthesia and surgery still had a significantly elevated risk of developing ASD compared to those who did not receive them. This indicated a significant association between early exposure to general anesthesia and the subsequent development of ASD, but the association is statistical instead of casual and the absolute risk is small [92]. Whether there are other unknown factors influencing this outcome requires further observational and research efforts.

ASD is a complex neurodevelopmental disorder with a wide range of risk factors. Despite the cumulative research on ASD in recent decades [93], a precise consensus regarding its core pathogenic mechanisms and effective therapeutic approaches has yet to be established. Among various hypotheses in ASD research, the genetic hypothesis has been widely recognized. Nevertheless, ASD is still believed to be a multifactorial and complex disease, and that in addition to the interplay of genetic and environmental factors, various mechanisms such as synaptic developmental disorders and dysregulation of the excitatory/inhibitory (E/I) balance in the brain may also be involved in the pathogenesis of ASD. Regarding the synaptic developmental disorder hypothesis, numerous genetic studies have indirectly affirmed its reliability. These studies have found associations between frequent variant genes in ASD and aspects such as synaptic structure establishment, neurotransmitter receptor function, and synaptic plasticity regulation. For example, as early as 2011, the team led by Gilman identified a large ASD-related functional gene network that primarily comprises genes involved in synaptic formation and function, including IP3R2, NRXN1, NLGN3, and the SHANK protein family [94]. Additionally, recent preclinical studies have found alterations in astrocytes in the pathophysiological processes of ASD [95, 96]. As one of the primary cell types in the central nervous system, astrocytes play crucial roles in supporting neural network construction, participating in synaptic formation and removal, maintaining neuronal function, and stabilizing the brain’s internal environment. An increase in Ca2+ serves as a hallmark of astrocyte activation, a process primarily driven by the second type of 1,4,5-triphosphate receptor (IP3R2). Interestingly, the IP3R2 gene has long been found to exhibit abnormalities in patients with ASDs [94]. In 2021, Wang et al. [97] probed the relationship between calcium signaling pathways in astrocytes and ASD-like behaviors through methods such as astrocyte-specific IP3R2 conditional knockout mouse models and drug interventions. Their study found that mice with conditional knockout of IP3R2 specific to astrocytes exhibited atypical social and repetitive behaviors. To further delve into the neurobiological mechanisms, the team utilized in vivo microdialysis techniques to analyze the levels of neurotransmitter transmission in the medial prefrontal cortex of rodents involved in social behavior and observed significantly reduced ATP levels in knockout mice. Additionally, treatment with ATP mediated by P2X2 receptors can rescue social behavior in mice [97]. From this, it could be seen that impaired calcium release led to astrocyte activation failure, synaptic changes, and subsequently, the manifestation of ASD-like behaviors in mice. In 2019, Zhou et al. [98] investigated the specific mechanistic pathways by which sevoflurane, a representative general anesthetic for children, causes astrocyte damage and consequently behavioral disorders. This study indicated that sevoflurane exposure had both immediate and long-term disruptive effects on Ca2+ homeostasis in developing astrocytes, including reducing basal Ca2+ levels and altering spontaneous and neurotransmitter-induced Ca2+ signals in astrocytes. Moreover, changes in Ca2+ signaling led to downregulation of ezrin protein expression, affecting the morphogenesis of astrocytes, especially in the development of fine processes [98]. Ezrin, as a membrane-associated protein binding to actin, plays a critical role in the fine processes of astrocytes [99, 100]. The morphological changes in astrocytes are associated with synaptic overgrowth and functional impairment in the cortical regions of the brain, affecting the interaction between astrocytes and neurons. This has a negative impact on the stability and maturation of synapses, ultimately resulting in social behavioral deficits in mice. Further research revealed that specific enhancement of ezrin protein expression in astrocytes can mitigate the morphological and functional damage caused by sevoflurane exposure and completely correct behavioral defects in developing mice [98], suggesting that astrocytes may be an important target for general anesthetics, and their morphological integrity is crucial for synaptic formation and neural behavioral development. Additionally, there is evidence that early exposure to general anesthesia can lead to social impairments, which are core symptoms of ASD [101]. To sum up, early exposure to general anesthetics disrupts Ca2+ homeostasis in astrocytes, leading to downregulation of ezrin protein expression, which subsequently affects the morphology and function of astrocytes, alters synaptic formation, and may ultimately result in ASD-like behaviors. This discovery presents novel avenues for the neurodevelopmental and behavioral disturbances induced by anesthetics.

As mentioned earlier, GABA is the most common inhibitory neurotransmitter, while glutamate is the most common excitatory neurotransmitter. The two maintain a delicate balance in the brain, collectively sustaining the E/I balance of the central nervous system to support normal neural network function and behavioral performance. Disruption of this balance may induce serious behavioral disorders. Recent research illustrated that E/I imbalance may be a key factor underlying ASD characteristic behaviors and cognitive abnormalities [102‒104]. In related studies, abnormal expression of glutamate and GABA receptors in the postmortem brains of ASD patients has been observed [105]. Changes in the levels of glutamate and glutamine in the cortex and basal ganglia of ASD children, as well as in the basal ganglia of adult patients, using proton magnetic resonance spectroscopy have also been identified [106‒109]. Additionally, reductions in GABA levels in several brain regions of children with ASD have been reported [110, 111]. While studies supported the hypothesis of E/I imbalance in ASD, the heterogeneity of the E/I ratio revealed by them was not always consistent, and the biological mechanisms behind the imbalance remain unclear. In 2018, Horder et al. [112] investigated abnormalities in glutamate and GABA in ASD. The study compared glutamate and GABA levels in the striatum of adult human ASD patients and 6 different rodent models of ASD using MRS technology. Reduced glutamate concentration in the striatum of adult ASD patients was inspected, which was correlated with the severity of social symptoms. Meanwhile, reduced glutamate concentration in the striatum was also observed in certain rodent models [112]. The findings of this study supported the hypothesis of E/I imbalance in ASD and suggested that the reduction of glutamate in the striatum may be one of the key pathological mechanisms of ASD. The association between general anesthesia and ASD can be inferred from Satomoto et al.’s [113] report in 2009 that neonatal exposure to sevoflurane may lead to social behavioral abnormalities, such as ASD. Briefly, general anesthetics may lead to overactivation or impairment of GABAergic neurons, resulting in E/I imbalance, which in turn can cause social behavioral abnormalities such as ASD.

In a nutshell, the genetic hypothesis, the synaptic developmental disorder hypothesis, and the E/I imbalance hypothesis interact with each other and collectively contribute to the pathophysiological processes of ASD. This aligns with the widely accepted view that ASD is a complex disorder caused by multiple factors.

DCD

DCD is a neurobehavioral developmental disorder characterized by slow, awkward, or inaccurate movements, as well as learning difficulties (new motor tasks or adapting previously learned gestures to modifications or additional constraints), which severely affects a child’s ability to perform coordinated movements [114]. The prevalence of DCD is estimated to be around 4.3% [115], and such high prevalence has aroused worry and concern among families and society.

Currently, the diagnosis of DCD is based on abnormal performance in manual dexterity, ballistic motor skills deficiencies, and balance skills deficit [114]. Recent studies have shown that abnormal brain function in children with DCD involves multiple brain regions, including but not limited to the cerebellum, basal ganglia, parietal lobe, and frontal lobe [116]. As a critical stage of brain development, infancy and early childhood are highly susceptible to general anesthetics which may ignite a range of cognitive and behavioral developmental disorders [3]. Clarification of the correlation between early exposure to general anesthesia and DCD has become requisite.

More than 2 decades ago, Ivry’s [117] team had already identified cerebellar injury as an important cause of DCD that was intimately linked to uncoordinated behavior, clumsiness, and postural control. This finding provided insight into the pathogenesis of DCD. Five years later, Zwicker et al.’s [118] team further verified the pivotal function of the cerebellum in DCD by reviewing and comparatively analyzing data from relevant animal and human studies. It has been suggested that early exposure to general anesthesia may lead to neuronal degenerative necrosis in several brain regions, such as the cingulate cortex, hippocampus, thalamus, and parietal lobe, but the effects on the cerebellum have not been clearly explicated [119]. Therefore, whether early exposure to general anesthesia provokes apoptotic degeneration of cerebellar neurons and the subsequent development of DCD has become an interesting line of research. In 2017, Xiao et al. [120] conducted a study on the effects of early isoproterenol exposure on cerebellar development in mice. Immunofluorescence experiments were performed to observe and assess Purkinje cell dendritogenesis and Bergmann glial cell development in the cerebellum of isoproterenol-anesthetized mice. The experimental results showed that Purkinje cells and their dendritic length were significantly reduced in the cerebellum, and the filopodia formation and cellular phenotypic differentiation of Bergmann glial cells were also disturbed. Among them, Purkinje cells were the only output neurons in cerebellar circuits, which were the central cell populations integrating motor coordination and learning [121]. In contrast, Bergmann glial cells specifically promoted the development of dendrites of Purkinje cells [122]. Both of them were fundamental in maintaining cerebellar functions. This specified that early exposure to general anesthesia may lead to abnormal development of key neurons in the cerebellum, ultimately resulting in DCD.

In addition, some neuroimaging findings manifested that there were alterations in the white matter microstructure of the brain in children with DCDs, especially in the corticospinal tract, posterior thalamic radiation, and sensory-motor tracts in the parietal subregion of the corpus callosum [114]. In 2017, Wilson et al. [123] analyzed the diffusion-weighted magnetic resonance imaging data and found that sensory-motor-related white matter organization in children with DCD was decreased, along with changes in the structural connectivity of brain networks. These findings denoted that cerebral white matter damage may be an important contributor to DCD. The neurotoxic effects of multiple general anesthetic exposures in early life leading to developmental deficits in the white matter of the brain have long been the focus of academic research. In 2023, Kim et al. [124] conducted a longitudinal structural magnetic resonance imaging analysis of the effects of multiple early general anesthesia exposures on the development of the brain in rhesus monkeys and demonstrated that multiple general anesthesia exposures during infancy may delay the maturation of white matter and impair its microstructural integrity. Taken together, early general anesthesia exposure may trigger developmental deficits in cerebral white matter, and there may be a correlation between damage to cerebral white matter and DCDs. However, there is a lack of specialized studies revealing the exact link between early general anesthesia exposure and DCD, which illuminates a direction for future research.

In addition to cerebellar developmental disorders and damage to the white matter of the brain, damage to brain regions such as the basal ganglia and parietal lobe may also be associated with DCD [125]. A review has now explored in detail the association of parietal lobes with DCD and explicated that children with DCD have deficits in visuospatial processing, face recognition and executive functioning, all associated with the parietal lobe [116]. Nonetheless, the developmental toxicity imposed by general anesthesia exposure on the parietal lobe is still an area that deserves in-depth exploration, and further studies are required to reveal its potential mechanisms and effects.

Apart from structural and functional damage to brain regions, insufficient activation of functional networks between brain regions has been associated with DCD [114]. In 2016, in children with DCD, Biotteau et al.’s [116] team found reduced cerebellar dysfunction and parietal activation patterns. Frontal-parietal and frontal-cerebellar neural networks play a crucial role in motor planning, motor control (especially internal modeling), visuomotor mapping, and automation [126]. At the same time, these regions overlap with the mirror neuron system and are involved in developing learned behaviors [127]. Therefore, underactivation of functional networks between these brain regions may be one of the reasons for developing DCD. Meanwhile, general anesthetics disrupt frontal-parietal brain communication [128]. Accordingly, we speculated that early exposure to general anesthesia may cause dysfunctional networks among different brain regions and lead to DCD. Whereas there lacks definitive research evidence, which awaits further exploration. In summation, early exposure to general anesthesia is strongly associated with the DCD, and the neurotoxic effects of general anesthesia leading to neurobehavioral developmental disorders remain to be unveiled.

To avoid the neurological toxicity effects and long-term negative consequences resulting from early exposure to general anesthesia, screening and monitoring of vulnerable populations should be prioritized, particularly children with specific genetic backgrounds or a history of neurological disorders. Simultaneously, it is essential to develop and employ more precise assessment tools to identify patients at higher risk of neurological toxicity when being exposed to general anesthetics. Tailored anesthesia plans that take into consideration the unique characteristics of each patient should be crafted. Furthermore, long-term neurobehavioral tracking of patients receiving general anesthesia is necessary to timely identify and take interventions for developmental behavioral disorders.

For researchers, animal models with high similarity to human neurobiology such as macaques should be developed first to provide basic tools for studying the neurotoxicity and mechanisms of general anesthetics. Second, the molecular and cellular mechanisms that drive the neurotoxic effects of general anesthetics should be explored, including their impact on critical processes like neuronal survival, differentiation, synaptogenesis, and function. Finally, based on a comprehensive understanding of neurotoxicity mechanisms, new intervention measures and drugs to alleviate or prevent the adverse effects of general anesthetics should be researched and developed.

In this review, a vast array of literature on the effects of general anesthesia on long-term neurobehavioral function in children was reviewed. Through analyzing those researches, it summarized the toxicological mechanisms closely linked to clinical neurobehavioral changes that may be induced by general anesthetic agents in basic research, offering clinical insights for the appropriate and safe use of these drugs in infants and young children. Moreover, it identified existing research gaps and shed light on future research directions which are expected to provide scientific evidence for the formulation of safer anesthesia practice policies and guidelines.

The authors have no conflicts of interest to declare.

This work was supported by the National Natural Science Foundation of China (Grant No. 82371276, 82171173); the Natural Science Foundation of Shanghai (Grant No. 22ZR1437200); and Clinical Research Program of Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine (Grant No. JYLJ202308).

Jinnan Xu conceptualized and outlined the review article. Jinnan Xu, Fan Wang, Yi Gao, Chuanyu Qi, and Tiannan Chen performed material preparation and literature search. Jinnan Xu wrote the first draft, and Jia Yan commented and edited. All authors approved the final draft.

1.
Fredriksson
A
,
Pontén
E
,
Gordh
T
,
Eriksson
P
.
Neonatal exposure to a combination of N-methyl-D-aspartate and gamma-aminobutyric acid type A receptor anesthetic agents potentiates apoptotic neurodegeneration and persistent behavioral deficits
.
Anesthesiology
.
2007
;
107
(
3
):
427
36
.
2.
Walters
JL
,
Paule
MG
.
Review of preclinical studies on pediatric general anesthesia-induced developmental neurotoxicity
.
Neurotoxicol Teratol
.
2017
;
60
:
2
23
.
3.
Jevtovic-Todorovic
V
.
Exposure of developing brain to general anesthesia: what is the animal evidence
.
Anesthesiology
.
2018
;
128
(
4
):
832
9
.
4.
Morriss
FH
Jr
,
Saha
S
,
Bell
EF
,
Colaizy
TT
,
Stoll
BJ
,
Hintz
SR
, et al
.
Surgery and neurodevelopmental outcome of very low-birth-weight infants
.
JAMA Pediatr
.
2014
;
168
(
8
):
746
54
.
5.
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
(
4
):
667
77
.
6.
Clausen
NG
,
Pedersen
DA
,
Pedersen
JK
,
Moller
SE
,
Grosen
D
,
Wehby
GL
, et al
.
Oral clefts and academic performance in adolescence: the impact of anesthesia-related neurotoxicity, timing of surgery, and type of oral clefts
.
Cleft Palate Craniofac J
.
2017
;
54
(
4
):
371
80
.
7.
Rabbitts
JA
,
Groenewald
CB
.
Epidemiology of pediatric surgery in the United States
.
Paediatr Anaesth
.
2020
;
30
(
10
):
1083
90
.
8.
Davidson
AJ
,
Sun
LS
.
Clinical evidence for any effect of anesthesia on the developing brain
.
Anesthesiology
.
2018
;
128
(
4
):
840
53
.
9.
Davidson
A
,
Vutskits
L
.
The new FDA drug safety communication on the use of general anesthetics in young children: what should we make of it
.
Paediatr Anaesth
.
2017
;
27
(
4
):
336
7
.
10.
Hansen
TG
.
Use of anesthetics in young children consensus statement of the European society of anaesthesiology (ESA), the European society for paediatric anaesthesiology (ESPA), the European association of cardiothoracic anaesthesiology (EACTA), and the European safe tots anaesthesia research initiative (EuroSTAR)
.
Paediatr Anaesth
.
2017
;
27
(
6
):
558
9
.
11.
Sun
LS
,
Li
G
,
Miller
TL
,
Salorio
C
,
Byrne
MW
,
Bellinger
DC
, et al
.
Association between a single general anesthesia exposure before age 36 months and neurocognitive outcomes in later childhood
.
JAMA
.
2016
;
315
(
21
):
2312
20
.
12.
Warner
DO
,
Zaccariello
MJ
,
Katusic
SK
,
Schroeder
DR
,
Hanson
AC
,
Schulte
PJ
, et al
.
Neuropsychological and behavioral outcomes after exposure of young children to procedures requiring general anesthesia: the Mayo anesthesia safety in Kids (MASK) study
.
Anesthesiology
.
2018
;
129
(
1
):
89
105
.
13.
McCann
ME
,
de Graaff
JC
,
Dorris
L
,
Disma
N
,
Withington
D
,
Bell
G
, et al
.
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
(
10172
):
664
77
.
14.
Ing
C
,
Landau
R
,
DeStephano
D
,
Miles
CH
,
von Ungern-Sternberg
BS
,
Li
G
, et al
.
Prenatal exposure to general anesthesia and childhood behavioral deficit
.
Anesth Analg
.
2021
;
133
(
3
):
595
605
.
15.
Reighard
C
,
Junaid
S
,
Jackson
WM
,
Arif
A
,
Waddington
H
,
Whitehouse
AJO
, et al
.
Anesthetic exposure during childhood and neurodevelopmental outcomes: a systematic review and meta-analysis
.
JAMA Netw Open
.
2022
;
5
(
6
):
e2217427
.
16.
Fraser
SA
,
Li
KZ
,
Penhune
VB
.
Dual-task performance reveals increased involvement of executive control in fine motor sequencing in healthy aging
.
J Gerontol B Psychol Sci Soc Sci
.
2010
;
65
(
5
):
526
35
.
17.
Bos
AF
,
Van Braeckel
KN
,
Hitzert
MM
,
Tanis
JC
,
Roze
E
.
Development of fine motor skills in preterm infants
.
Dev Med Child Neurol
.
2013
;
55
(
Suppl 4
):
1
4
.
18.
Morris
R
,
Whishaw
IQ
.
Arm and hand movement: current knowledge and future perspective
.
Front Neurol
.
2015
;
6
:
19
.
19.
Ludwig
PE
,
Reddy
V
,
Varacallo
M
. Neuroanatomy, Central Nervous System (CNS). StatPearls. Treasure Island (FL) ineligible companies. Disclosure: vamsi Reddy declares no relevant financial relationships with ineligible companies. Disclosure: matthew Varacallo declares no relevant financial relationships with ineligible companies. StatPearls Publishing Copyright © 2024. StatPearls Publishing LLC; 2024.
20.
Zaccariello
MJ
,
Frank
RD
,
Lee
M
,
Kirsch
AC
,
Schroeder
DR
,
Hanson
AC
, et al
.
Patterns of neuropsychological changes after general anaesthesia in young children: secondary analysis of the Mayo Anesthesia Safety in Kids study
.
Br J Anaesth
.
2019
;
122
(
5
):
671
81
.
21.
Walkden
GJ
,
Gill
H
,
Davies
NM
,
Peters
AE
,
Wright
I
,
Pickering
AE
.
Early childhood general anesthesia and neurodevelopmental outcomes in the avon longitudinal study of parents and children birth cohort
.
Anesthesiology
.
2020
;
133
(
5
):
1007
20
.
22.
Kobayashi
Y
,
Tokuda
N
,
Adachi
S
,
Takeshima
Y
,
Hirose
M
,
Shima
M
, et al
.
Association between surgical procedures under general anesthesia in infancy and developmental outcomes at 1 year: the Japan Environment and Children’s Study
.
Environ Health Prev Med
.
2020
;
25
(
1
):
32
.
23.
Zhong
L
,
Ma
X
,
Niu
Y
,
Zhang
L
,
Xue
Z
,
Yan
J
, et al
.
Sevoflurane exposure may cause dysplasia of dendritic spines and result in fine motor dysfunction in developing mouse through the PI3K/AKT/mTOR pathway
.
Front Neurosci
.
2022
;
16
:
1006175
.
24.
Zhang
L
,
Cheng
Y
,
Xue
Z
,
Li
J
,
Wu
N
,
Yan
J
, et al
.
Sevoflurane impairs m6A-mediated mRNA translation and leads to fine motor and cognitive deficits
.
Cell Biol Toxicol
.
2022
;
38
(
2
):
347
69
.
25.
Shi
H
,
Zhang
X
,
Weng
YL
,
Lu
Z
,
Liu
Y
,
Lu
Z
, et al
.
m(6)A facilitates hippocampus-dependent learning and memory through YTHDF1
.
Nature
.
2018
;
563
(
7730
):
249
53
.
26.
Weng
YL
,
Wang
X
,
An
R
,
Cassin
J
,
Vissers
C
,
Liu
Y
, et al
.
Epitranscriptomic m(6)A regulation of axon regeneration in the adult mammalian nervous system
.
Neuron
.
2018
;
97
(
2
):
313
25.e6
.
27.
Zhuang
M
,
Li
X
,
Zhu
J
,
Zhang
J
,
Niu
F
,
Liang
F
, et al
.
The m6A reader YTHDF1 regulates axon guidance through translational control of Robo3.1 expression
.
Nucleic Acids Res
.
2019
;
47
(
9
):
4765
77
.
28.
Xu
G
,
Lu
H
,
Dong
Y
,
Shapoval
D
,
Soriano
SG
,
Liu
X
, et al
.
Coenzyme Q10 reduces sevoflurane-induced cognitive deficiency in young mice
.
Br J Anaesth
.
2017
;
119
(
3
):
481
91
.
29.
Zhang
L
,
Xue
Z
,
Liu
Q
,
Liu
Y
,
Xi
S
,
Cheng
Y
, et al
.
Disrupted folate metabolism with anesthesia leads to myelination deficits mediated by epigenetic regulation of ERMN
.
EBioMedicine
.
2019
;
43
:
473
86
.
30.
Li
Q
,
Mathena
RP
,
Xu
J
,
Eregha
ON
,
Wen
J
,
Mintz
CD
.
Early postnatal exposure to isoflurane disrupts oligodendrocyte development and myelin formation in the mouse Hippocampus
.
Anesthesiology
.
2019
;
131
(
5
):
1077
91
.
31.
Wu
Z
,
Xue
H
,
Gao
Q
,
Zhao
P
.
Effects of early postnatal sevoflurane exposure on oligodendrocyte maturation and myelination in cerebral white matter of the rat
.
Biomed Pharmacother
.
2020
;
131
:
110733
.
32.
Zuo
Y
,
Li
B
,
Xie
J
,
Ma
Z
,
Thirupathi
A
,
Yu
P
, et al
.
Sevoflurane anesthesia during pregnancy in mice induces cognitive impairment in the offspring by causing iron deficiency and inhibiting myelinogenesis
.
Neurochem Int
.
2020
;
135
:
104693
.
33.
Wartenberg
KE
,
Lorenzano
S
.
White matter lesion volume reduces fine motor skills: is primary stroke prophylaxis warranted
.
Neurology
.
2015
;
84
(
19
):
1914
5
.
34.
Cullen
CL
,
O'Rourke
M
,
Beasley
SJ
,
Auderset
L
,
Zhen
Y
,
Pepper
RE
, et al
.
Kif3a deletion prevents primary cilia assembly on oligodendrocyte progenitor cells, reduces oligodendrogenesis and impairs fine motor function
.
Glia
.
2021
;
69
(
5
):
1184
203
.
35.
Zheng
J
,
Gao
Y
,
Xu
X
,
Kang
K
,
Liu
H
,
Wang
H
, et al
.
Correlation of bispectral index and Richmond agitation sedation scale for evaluating sedation depth: a retrospective study
.
J Thorac Dis
.
2018
;
10
(
1
):
190
5
.
36.
Hayward
A
,
Tomlinson
A
,
Neill
JC
.
Low attentive and high impulsive rats: a translational animal model of ADHD and disorders of attention and impulse control
.
Pharmacol Ther
.
2016
;
158
:
41
51
.
37.
Sprung
J
,
Flick
RP
,
Katusic
SK
,
Colligan
RC
,
Barbaresi
WJ
,
Bojanić
K
, et al
.
Attention-deficit/hyperactivity disorder after early exposure to procedures requiring general anesthesia
.
Mayo Clin Proc
.
2012
;
87
(
2
):
120
9
.
38.
Tsai
CJ
,
Lee
CT
,
Liang
SH
,
Tsai
PJ
,
Chen
VC
,
Gossop
M
.
Risk of ADHD after multiple exposures to general anesthesia: a nationwide retrospective cohort study
.
J Atten Disord
.
2018
;
22
(
3
):
229
39
.
39.
Ing
C
,
Sun
M
,
Olfson
M
,
DiMaggio
CJ
,
Sun
LS
,
Wall
MM
, et al
.
Age at exposure to surgery and anesthesia in children and association with mental disorder diagnosis
.
Anesth Analg
.
2017
;
125
(
6
):
1988
98
.
40.
Ko
WR
,
Liaw
YP
,
Huang
JY
,
Zhao
DH
,
Chang
HC
,
Ko
PC
, et al
.
Exposure to general anesthesia in early life and the risk of attention deficit/hyperactivity disorder development: a nationwide, retrospective matched-cohort study
.
Paediatr Anaesth
.
2014
;
24
(
7
):
741
8
.
41.
Warner
DO
,
Hu
D
,
Zaccariello
MJ
,
Schroeder
DR
,
Hanson
AC
,
Wilder
RT
, et al
.
Association between behavioral and learning outcomes and single exposures to procedures requiring general anesthesia before age 3: secondary analysis of data from olmsted county, MN
.
Anesth Analg
.
2021
;
133
(
1
):
160
7
.
42.
Shi
Y
,
Dykhoff
HJ
,
Guevara
LRH
,
Sangaralingham
LR
,
Schroeder
DR
,
Flick
RP
, et al
.
Moderators of the association between attention-deficit/hyperactivity disorder and exposure to anaesthesia and surgery in children
.
Br J Anaesth
.
2021
;
127
(
5
):
722
8
.
43.
Sun
M
,
Chen
WM
,
Fu
S
,
Wu
SY
,
Zhang
J
.
Early childhood general anesthesia and risk of attention deficit hyperactivity disorder
.
J Child Psychol Psychiatry
.
2024
;
65
(
2
):
165
75
.
44.
Fredriksson
A
,
Archer
T
.
Neurobehavioural deficits associated with apoptotic neurodegeneration and vulnerability for ADHD
.
Neurotox Res
.
2004
;
6
(
6
):
435
56
.
45.
Tistarelli
N
,
Fagnani
C
,
Troianiello
M
,
Stazi
MA
,
Adriani
W
.
The nature and nurture of ADHD and its comorbidities: a narrative review on twin studies
.
Neurosci Biobehav Rev
.
2020
;
109
:
63
77
.
46.
Faraone
SV
,
Mick
E
.
Molecular genetics of attention deficit hyperactivity disorder
.
Psychiatr Clin North Am
.
2010
;
33
(
1
):
159
80
.
47.
Arnsten
AF
,
Rubia
K
.
Neurobiological circuits regulating attention, cognitive control, motivation, and emotion: disruptions in neurodevelopmental psychiatric disorders
.
J Am Acad Child Adolesc Psychiatry
.
2012
;
51
(
4
):
356
67
.
48.
Wu
J
,
Xiao
H
,
Sun
H
,
Zou
L
,
Zhu
LQ
.
Role of dopamine receptors in ADHD: a systematic meta-analysis
.
Mol Neurobiol
.
2012
;
45
(
3
):
605
20
.
49.
Huang
J
,
Zhong
Z
,
Wang
M
,
Chen
X
,
Tan
Y
,
Zhang
S
, et al
.
Circadian modulation of dopamine levels and dopaminergic neuron development contributes to attention deficiency and hyperactive behavior
.
J Neurosci
.
2015
;
35
(
6
):
2572
87
.
50.
Prince
J
.
Catecholamine dysfunction in attention-deficit/hyperactivity disorder: an update
.
J Clin Psychopharmacol
.
2008
;
28
(
3 Suppl 2
):
S39
45
.
51.
Cheng
J
,
Liu
A
,
Shi
MY
,
Yan
Z
.
Disrupted glutamatergic transmission in prefrontal cortex contributes to behavioral abnormality in an animal model of ADHD
.
Neuropsychopharmacology
.
2017
;
42
(
10
):
2096
104
.
52.
Carli
M
,
Invernizzi
RW
.
Serotoninergic and dopaminergic modulation of cortico-striatal circuit in executive and attention deficits induced by NMDA receptor hypofunction in the 5-choice serial reaction time task
.
Front Neural Circuits
.
2014
;
8
:
58
.
53.
Bath
KG
,
Schilit
A
,
Lee
FS
.
Stress effects on BDNF expression: effects of age, sex, and form of stress
.
Neuroscience
.
2013
;
239
:
149
56
.
54.
Bramham
CR
,
Panja
D
.
BDNF regulation of synaptic structure, function, and plasticity
.
Neuropharmacology
.
2014
;
76 Pt C
:
601
2
.
55.
Liu
DY
,
Shen
XM
,
Yuan
FF
,
Guo
OY
,
Zhong
Y
,
Chen
JG
, et al
.
The physiology of BDNF and its relationship with ADHD
.
Mol Neurobiol
.
2015
;
52
(
3
):
1467
76
.
56.
Amiri
A
,
Torabi Parizi
G
,
Kousha
M
,
Saadat
F
,
Modabbernia
MJ
,
Najafi
K
, et al
.
Changes in plasma Brain-derived neurotrophic factor (BDNF) levels induced by methylphenidate in children with Attention deficit-hyperactivity disorder (ADHD)
.
Prog Neuro-Psychopharmacol Biol Psychiatry
.
2013
;
47
:
20
4
.
57.
Chamberlain
SR
,
Sahakian
BJ
.
The neuropsychiatry of impulsivity
.
Curr Opin Psychiatry
.
2007
;
20
(
3
):
255
61
.
58.
Aron
AR
.
From reactive to proactive and selective control: developing a richer model for stopping inappropriate responses
.
Biol Psychiatry
.
2011
;
69
(
12
):
e55
68
.
59.
Besson
M
,
Pelloux
Y
,
Dilleen
R
,
Theobald
DE
,
Lyon
A
,
Belin-Rauscent
A
, et al
.
Cocaine modulation of frontostriatal expression of Zif268, D2, and 5-HT2c receptors in high and low impulsive rats
.
Neuropsychopharmacology
.
2013
;
38
(
10
):
1963
73
.
60.
Yang
MT
,
Lu
DH
,
Chen
JC
,
Fu
WM
.
Inhibition of hyperactivity and impulsivity by carbonic anhydrase inhibitors in spontaneously hypertensive rats, an animal model of ADHD
.
Psychopharmacology
.
2015
;
232
(
20
):
3763
72
.
61.
Cabrera
OH
,
Tesic
V
,
Tat
QL
,
Chastain
S
,
Quillinan
N
,
Jevtovic-Todorovic
V
.
Sevoflurane-induced dysregulation of cation-chloride cotransporters NKCC1 and KCC2 in neonatal mouse brain
.
Mol Neurobiol
.
2020
;
57
(
1
):
1
10
.
62.
Briner
A
,
De Roo
M
,
Dayer
A
,
Muller
D
,
Habre
W
,
Vutskits
L
.
Volatile anesthetics rapidly increase dendritic spine density in the rat medial prefrontal cortex during synaptogenesis
.
Anesthesiology
.
2010
;
112
(
3
):
546
56
.
63.
Song
R
,
Ling
X
,
Peng
M
,
Xue
Z
,
Cang
J
,
Fang
F
.
Maternal sevoflurane exposure causes abnormal development of fetal prefrontal cortex and induces cognitive dysfunction in offspring
.
Stem Cells Int
.
2017
;
2017
:
6158468
.
64.
Xie
L
,
Liu
Y
,
Hu
Y
,
Wang
B
,
Zhu
Z
,
Jiang
Y
, et al
.
Neonatal sevoflurane exposure induces impulsive behavioral deficit through disrupting excitatory neurons in the medial prefrontal cortex in mice
.
Transl Psychiatry
.
2020
;
10
(
1
):
202
.
65.
Loos
M
,
Mueller
T
,
Gouwenberg
Y
,
Wijnands
R
,
van der Loo
RJ
;
Neuro-BSIK Mouse Phenomics Consortium
, et al
.
Neuregulin-3 in the mouse medial prefrontal cortex regulates impulsive action
.
Biol Psychiatry
.
2014
;
76
(
8
):
648
55
.
66.
Sebastian
A
,
Jung
P
,
Krause-Utz
A
,
Lieb
K
,
Schmahl
C
,
Tüscher
O
.
Frontal dysfunctions of impulse control - a systematic review in borderline personality disorder and attention-deficit/hyperactivity disorder
.
Front Hum Neurosci
.
2014
;
8
:
698
.
67.
Euston
DR
,
Gruber
AJ
,
McNaughton
BL
.
The role of medial prefrontal cortex in memory and decision making
.
Neuron
.
2012
;
76
(
6
):
1057
70
.
68.
Warden
MR
,
Selimbeyoglu
A
,
Mirzabekov
JJ
,
Lo
M
,
Thompson
KR
,
Kim
SY
, et al
.
A prefrontal cortex-brainstem neuronal projection that controls response to behavioural challenge
.
Nature
.
2012
;
492
(
7429
):
428
32
.
69.
Riga
D
,
Matos
MR
,
Glas
A
,
Smit
AB
,
Spijker
S
,
Van den Oever
MC
.
Optogenetic dissection of medial prefrontal cortex circuitry
.
Front Syst Neurosci
.
2014
;
8
:
230
.
70.
Caballero-Puntiverio
M
,
Fitzpatrick
CM
,
Woldbye
DP
,
Andreasen
JT
.
Effects of amphetamine and methylphenidate on attentional performance and impulsivity in the mouse 5-Choice Serial Reaction Time Task
.
J Psychopharmacol
.
2017
;
31
(
2
):
272
83
.
71.
Maguire
DR
,
France
CP
.
Effects of amphetamine, methylphenidate, atomoxetine, and morphine in rats responding under an adjusting stop signal reaction time task
.
Psychopharmacology
.
2019
;
236
(
6
):
1959
72
.
72.
Bonvicini
C
,
Faraone
SV
,
Scassellati
C
.
Common and specific genes and peripheral biomarkers in children and adults with attention-deficit/hyperactivity disorder
.
World J Biol Psychiatry
.
2018
;
19
(
2
):
80
100
.
73.
Bacanlı
A
,
Unsel-Bolat
G
,
Suren
S
,
Yazıcı
KU
,
Callı
C
,
Aygunes Jafari
D
, et al
.
Effects of the dopamine transporter gene on neuroimaging findings in different attention deficit hyperactivity disorder presentations
.
Brain Imaging Behav
.
2021
;
15
(
2
):
1103
14
.
74.
Mereu
M
,
Contarini
G
,
Buonaguro
EF
,
Latte
G
,
Managò
F
,
Iasevoli
F
, et al
.
Dopamine transporter (DAT) genetic hypofunction in mice produces alterations consistent with ADHD but not schizophrenia or bipolar disorder
.
Neuropharmacology
.
2017
;
121
:
179
94
.
75.
Zepf
FD
,
Stadler
C
,
Demisch
L
,
Schmitt
M
,
Landgraf
M
,
Poustka
F
.
Serotonergic functioning and trait-impulsivity in attention-deficit/hyperactivity-disordered boys (ADHD): influence of rapid tryptophan depletion
.
Hum Psychopharmacol
.
2008
;
23
(
1
):
43
51
.
76.
Cremers
H
,
Lee
R
,
Keedy
S
,
Phan
KL
,
Coccaro
E
.
Effects of escitalopram administration on face processing in intermittent explosive disorder: an fMRI study
.
Neuropsychopharmacology
.
2016
;
41
(
2
):
590
7
.
77.
Gendle
MH
,
Young
EL
,
Romano
AC
.
Effects of oral 5-hydroxytryptophan on a standardized planning task: insight into possible dopamine/serotonin interactions in the forebrain
.
Hum Psychopharmacol
.
2013
;
28
(
3
):
270
3
.
78.
Widiger
TA
,
Hines
A
.
The diagnostic and statistical manual of mental disorders, fifth edition alternative model of personality disorder
.
Personal Disord
.
2022
;
13
(
4
):
347
55
.
79.
Salari
N
,
Rasoulpoor
S
,
Rasoulpoor
S
,
Shohaimi
S
,
Jafarpour
S
,
Abdoli
N
, et al
.
The global prevalence of autism spectrum disorder: a comprehensive systematic review and meta-analysis
.
Ital J Pediatr
.
2022
;
48
(
1
):
112
.
80.
Risch
N
,
Hoffmann
TJ
,
Anderson
M
,
Croen
LA
,
Grether
JK
,
Windham
GC
.
Familial recurrence of autism spectrum disorder: evaluating genetic and environmental contributions
.
Am J Psychiatry
.
2014
;
171
(
11
):
1206
13
.
81.
Sandin
S
,
Lichtenstein
P
,
Kuja-Halkola
R
,
Larsson
H
,
Hultman
CM
,
Reichenberg
A
.
The familial risk of autism
.
Jama
.
2014
;
311
(
17
):
1770
7
.
82.
Kim
H
,
Keifer
C
,
Rodriguez-Seijas
C
,
Eaton
N
,
Lerner
M
,
Gadow
K
.
Quantifying the optimal structure of the autism phenotype: a comprehensive comparison of dimensional, categorical, and hybrid models
.
J Am Acad Child Adolesc Psychiatry
.
2019
;
58
(
9
):
876
86.e2
.
83.
Wang
C
,
Geng
H
,
Liu
W
,
Zhang
G
.
Prenatal, perinatal, and postnatal factors associated with autism: a meta-analysis
.
Medicine
.
2017
;
96
(
18
):
e6696
.
84.
The
Lancet
.
Stemming the global caesarean section epidemic
.
Lancet
.
2018
;
392
(
10155
):
1279
.
85.
Feng
XL
,
Wang
Y
,
An
L
,
Ronsmans
C
.
Cesarean section in the People’s Republic of China: current perspectives
.
Int J Womens Health
.
2014
;
6
:
59
74
.
86.
Shroff
R
,
Thompson
AC
,
McCrum
A
,
Rees
SG
.
Prospective multidisciplinary audit of obstetric general anaesthesia in a district general hospital
.
J Obstet Gynaecol
.
2004
;
24
(
6
):
641
6
.
87.
Ikeda
T
,
Kato
A
,
Bougaki
M
,
Araki
Y
,
Ohata
T
,
Kawashima
S
, et al
.
A retrospective review of 10-year trends in general anesthesia for cesarean delivery at a university hospital: the impact of a newly launched team on obstetric anesthesia practice
.
BMC Health Serv Res
.
2020
;
20
(
1
):
421
.
88.
Chien
LN
,
Lin
HC
,
Shao
YH
,
Chiou
ST
,
Chiou
HY
.
Risk of autism associated with general anesthesia during cesarean delivery: a population-based birth-cohort analysis
.
J Autism Dev Disord
.
2015
;
45
(
4
):
932
42
.
89.
Yang
Y
,
Lin
J
,
Lu
X
,
Xun
G
,
Wu
R
,
Li
Y
, et al
.
Anesthesia, sex and miscarriage history may influence the association between cesarean delivery and autism spectrum disorder
.
BMC Pediatr
.
2021
;
21
(
1
):
62
.
90.
Creagh
O
,
Torres
H
,
Rivera
K
,
Morales-Franqui
M
,
Altieri-Acevedo
G
,
Warner
D
.
Previous exposure to anesthesia and autism spectrum disorder (ASD): a Puerto Rican population-based sibling cohort study
.
Bol Asoc Med P R
.
2015
;
107
(
3
):
29
37
.
91.
Castellheim
A
,
Lundström
S
,
Molin
M
,
Kuja-Halkola
R
,
Gillberg
C
,
Gillberg
C
.
The role of general anesthesia on traits of neurodevelopmental disorders in a Swedish cohort of twins
.
J Child Psychol Psychiatry
.
2018
;
59
(
9
):
966
72
.
92.
Pikwer
A
,
Yang
B
,
Granström
M
,
Mattsson
N
,
Sadr-Azodi
O
.
General anesthesia in early childhood and possible association with autism: a population-based matched cohort study
.
Minerva Anestesiol
.
2023
;
89
(
1–2
):
22
31
.
93.
Christensen
DL
,
Maenner
MJ
,
Bilder
D
,
Constantino
JN
,
Daniels
J
,
Durkin
MS
, et al
.
Prevalence and characteristics of autism spectrum disorder among children aged 4 years - early autism and developmental disabilities monitoring network, seven sites, United States, 2010, 2012, and 2014
.
MMWR Surveill Summ
.
2019
;
68
(
2
):
1
19
.
94.
Gilman
SR
,
Iossifov
I
,
Levy
D
,
Ronemus
M
,
Wigler
M
,
Vitkup
D
.
Rare de novo variants associated with autism implicate a large functional network of genes involved in formation and function of synapses
.
Neuron
.
2011
;
70
(
5
):
898
907
.
95.
Edmonson
C
,
Ziats
MN
,
Rennert
OM
.
Altered glial marker expression in autistic post-mortem prefrontal cortex and cerebellum
.
Mol Autism
.
2014
;
5
(
1
):
3
.
96.
Petrelli
F
,
Pucci
L
,
Bezzi
P
.
Astrocytes and microglia and their potential link with autism spectrum disorders
.
Front Cell Neurosci
.
2016
;
10
:
21
.
97.
Wang
Q
,
Kong
Y
,
Wu
DY
,
Liu
JH
,
Jie
W
,
You
QL
, et al
.
Impaired calcium signaling in astrocytes modulates autism spectrum disorder-like behaviors in mice
.
Nat Commun
.
2021
;
12
(
1
):
3321
.
98.
Zhou
B
,
Chen
L
,
Liao
P
,
Huang
L
,
Chen
Z
,
Liao
D
, et al
.
Astroglial dysfunctions drive aberrant synaptogenesis and social behavioral deficits in mice with neonatal exposure to lengthy general anesthesia
.
PLoS Biol
.
2019
;
17
(
8
):
e3000086
.
99.
Derouiche
A
,
Frotscher
M
.
Peripheral astrocyte processes: monitoring by selective immunostaining for the actin-binding ERM proteins
.
Glia
.
2001
;
36
(
3
):
330
41
.
100.
Lavialle
M
,
Aumann
G
,
Anlauf
E
,
Pröls
F
,
Arpin
M
,
Derouiche
A
.
Structural plasticity of perisynaptic astrocyte processes involves ezrin and metabotropic glutamate receptors
.
Proc Natl Acad Sci USA
.
2011
;
108
(
31
):
12915
9
.
101.
Chen
Q
,
Chu
W
,
Sheng
R
,
Song
S
,
Yang
J
,
Ji
F
, et al
.
Maternal anesthesia with sevoflurane during the mid-gestation induces social interaction deficits in offspring C57BL/6 mice
.
Biochem Biophys Res Commun
.
2021
;
553
:
65
71
.
102.
Rubenstein
JL
,
Merzenich
MM
.
Model of autism: increased ratio of excitation/inhibition in key neural systems
.
Genes Brain Behav
.
2003
;
2
(
5
):
255
67
.
103.
Yizhar
O
,
Fenno
LE
,
Prigge
M
,
Schneider
F
,
Davidson
TJ
,
O'Shea
DJ
, et al
.
Neocortical excitation/inhibition balance in information processing and social dysfunction
.
Nature
.
2011
;
477
(
7363
):
171
8
.
104.
Coghlan
S
,
Horder
J
,
Inkster
B
,
Mendez
MA
,
Murphy
DG
,
Nutt
DJ
.
GABA system dysfunction in autism and related disorders: from synapse to symptoms
.
Neurosci Biobehav Rev
.
2012
;
36
(
9
):
2044
55
.
105.
Blatt
GJ
,
Fatemi
SH
.
Alterations in GABAergic biomarkers in the autism brain: research findings and clinical implications
.
Anat Rec
.
2011
;
294
(
10
):
1646
52
.
106.
Bernardi
S
,
Anagnostou
E
,
Shen
J
,
Kolevzon
A
,
Buxbaum
JD
,
Hollander
E
, et al
.
In vivo 1H-magnetic resonance spectroscopy study of the attentional networks in autism
.
Brain Res
.
2011
;
1380
:
198
205
.
107.
Bejjani
A
,
O'Neill
J
,
Kim
JA
,
Frew
AJ
,
Yee
VW
,
Ly
R
, et al
.
Elevated glutamatergic compounds in pregenual anterior cingulate in pediatric autism spectrum disorder demonstrated by 1H MRS and 1H MRSI
.
PLoS One
.
2012
;
7
(
7
):
e38786
.
108.
Kubas
B
,
Kułak
W
,
Sobaniec
W
,
Tarasow
E
,
Lebkowska
U
,
Walecki
J
.
Metabolite alterations in autistic children: a 1H MR spectroscopy study
.
Adv Med Sci
.
2012
;
57
(
1
):
152
6
.
109.
Naaijen
J
,
Forde
NJ
,
Lythgoe
DJ
,
Akkermans
SE
,
Openneer
TJ
,
Dietrich
A
, et al
.
Fronto-striatal glutamate in children with Tourette's disorder and attention-deficit/hyperactivity disorder
.
Neuroimage Clin
.
2017
;
13
:
16
23
.
110.
Gaetz
W
,
Bloy
L
,
Wang
DJ
,
Port
RG
,
Blaskey
L
,
Levy
SE
, et al
.
GABA estimation in the brains of children on the autism spectrum: measurement precision and regional cortical variation
.
Neuroimage
.
2014
;
86
:
1
9
.
111.
Puts
NAJ
,
Wodka
EL
,
Harris
AD
,
Crocetti
D
,
Tommerdahl
M
,
Mostofsky
SH
, et al
.
Reduced GABA and altered somatosensory function in children with autism spectrum disorder
.
Autism Res
.
2017
;
10
(
4
):
608
19
.
112.
Horder
J
,
Petrinovic
MM
,
Mendez
MA
,
Bruns
A
,
Takumi
T
,
Spooren
W
, et al
.
Glutamate and GABA in autism spectrum disorder-a translational magnetic resonance spectroscopy study in man and rodent models
.
Transl Psychiatry
.
2018
;
8
(
1
):
106
.
113.
Satomoto
M
,
Satoh
Y
,
Terui
K
,
Miyao
H
,
Takishima
K
,
Ito
M
, et al
.
Neonatal exposure to sevoflurane induces abnormal social behaviors and deficits in fear conditioning in mice
.
Anesthesiology
.
2009
;
110
(
3
):
628
37
.
114.
Blank
R
,
Barnett
AL
,
Cairney
J
,
Green
D
,
Kirby
A
,
Polatajko
H
, et al
.
International clinical practice recommendations on the definition, diagnosis, assessment, intervention, and psychosocial aspects of developmental coordination disorder
.
Dev Med Child Neurol
.
2019
;
61
(
3
):
242
85
.
115.
Cardoso
AA
,
Magalhães
LC
,
Rezende
MB
.
Motor skills in Brazilian children with developmental coordination disorder versus children with motor typical development
.
Occup Ther Int
.
2014
;
21
(
4
):
176
85
.
116.
Biotteau
M
,
Chaix
Y
,
Blais
M
,
Tallet
J
,
Péran
P
,
Albaret
JM
.
Neural signature of DCD: a critical review of MRI neuroimaging studies
.
Front Neurol
.
2016
;
7
:
227
.
117.
Ivry
RB
.
Cerebellar involvement in clumsiness and other developmental disorders
.
Neural Plast
.
2003
;
10
(
1–2
):
141
53
.
118.
Zwicker
JG
,
Missiuna
C
,
Boyd
LA
.
Neural correlates of developmental coordination disorder: a review of hypotheses
.
J Child Neurol
.
2009
;
24
(
10
):
1273
81
.
119.
Jevtovic-Todorovic
V
,
Hartman
RE
,
Izumi
Y
,
Benshoff
ND
,
Dikranian
K
,
Zorumski
CF
, et al
.
Early exposure to common anesthetic agents causes widespread neurodegeneration in the developing rat brain and persistent learning deficits
.
J Neurosci
.
2003
;
23
(
3
):
876
82
.
120.
Xiao
R
,
Yu
D
,
Li
X
,
Huang
J
,
Jing
S
,
Bao
X
, et al
.
Propofol exposure in early life induced developmental impairments in the mouse cerebellum
.
Front Cell Neurosci
.
2017
;
11
:
373
.
121.
Lee
RX
,
Huang
JJ
,
Huang
C
,
Tsai
ML
,
Yen
CT
.
Plasticity of cerebellar Purkinje cells in behavioral training of body balance control
.
Front Syst Neurosci
.
2015
;
9
:
113
.
122.
Bellamy
TC
.
Interactions between Purkinje neurones and Bergmann glia
.
Cerebellum
.
2006
;
5
(
2
):
116
26
.
123.
Wilson
PH
,
Smits-Engelsman
B
,
Caeyenberghs
K
,
Steenbergen
B
,
Sugden
D
,
Clark
J
, et al
.
Cognitive and neuroimaging findings in developmental coordination disorder: new insights from a systematic review of recent research
.
Dev Med Child Neurol
.
2017
;
59
(
11
):
1117
29
.
124.
Kim
J
,
Barcus
R
,
Lipford
ME
,
Yuan
H
,
Ririe
DG
,
Jung
Y
, et al
.
Effects of multiple anesthetic exposures on rhesus macaque brain development: a longitudinal structural MRI analysis
.
Cereb Cortex
.
2024
;
34
(
1
):
bhad463
.
125.
Adi-Japha
E
,
Brestel
G
.
Motor skill learning with impaired transfer by children with developmental coordination disorder
.
Res Dev Disabil
.
2020
;
103
:
103671
.
126.
Subara-Zukic
E
,
Cole
MH
,
McGuckian
TB
,
Steenbergen
B
,
Green
D
,
Smits-Engelsman
BC
, et al
.
Behavioral and neuroimaging research on developmental coordination disorder (DCD): a combined systematic review and meta-analysis of recent findings
.
Front Psychol
.
2022
;
13
:
809455
.
127.
Reynolds
JE
,
Licari
MK
,
Billington
J
,
Chen
Y
,
Aziz-Zadeh
L
,
Werner
J
, et al
.
Mirror neuron activation in children with developmental coordination disorder: a functional MRI study
.
Int J Dev Neurosci
.
2015
;
47
(
Pt B
):
309
19
.
128.
Lee
U
,
Ku
S
,
Noh
G
,
Baek
S
,
Choi
B
,
Mashour
GA
.
Disruption of frontal-parietal communication by ketamine, propofol, and sevoflurane
.
Anesthesiology
.
2013
;
118
(
6
):
1264
75
.