Abstract
Maternal smoking during pregnancy is associated with an ensemble of neurodevelopmental consequences in children and therefore constitutes a pressing public health concern. Adding to this burden, contemporary epidemiological and especially animal model research suggests that grandmaternal smoking is similarly associated with neurodevelopmental abnormalities in grandchildren, indicative of intergenerational transmission of the neurodevelopmental impacts of maternal smoking. Probing the mechanistic bases of neurodevelopmental anomalies in the children of maternal smokers and the intergenerational transmission thereof, emerging research intimates that epigenetic changes, namely DNA methylome perturbations, are key factors. Altogether, these findings warrant future research to fully elucidate the etiology of neurodevelopmental impairments in the children and grandchildren of maternal smokers and underscore the clear potential thereof to benefit public health by informing the development and implementation of preventative measures, prophylactics, and treatments. To this end, the present review aims to encapsulate the burgeoning evidence linking maternal smoking to intergenerational epigenetic inheritance of neurodevelopmental abnormalities, to identify the strengths and weaknesses thereof, and to highlight areas of emphasis for future human and animal model research therein.
Keywords: Maternal smoking, pregnancy, nicotine, neurodevelopment, epigenetics, DNA methylation, children, grandchildren, intergenerational, heritable
Evidence from human and animal model research indicates that maternal smoking confers a constellation of neurodevelopmental anomalies which appear to undergo intergenerational transmission via a putative epigenetic mechanism.
Introduction
Approximately 10% of women in the United States smoke traditional cigarettes during pregnancy, and 14% use electronic cigarettes during pregnancy [1–3]. The incidences of maternal smoking (MS) are higher still in certain other countries [4]. Alarmingly, epidemiological studies report that, despite an absence of supporting evidence, the majority of those surveyed misperceive that electronic cigarettes are a safer and healthier surrogate for traditional cigarettes, and this misunderstanding is most pervasive among women of reproductive age [5–9]. Contrary to majority opinion, MS of traditional or electronic cigarettes or consumption of any tobacco- or nicotine-containing product during the gestational period or during the gestational and postpartum lactational periods results in the concurrent exposure of three generations to nicotine, namely the mother herself, her unborn or newborn child, and (via exposure of her unborn or newborn child’s germline) her grandchild(ren).
MS is associated with numerous fetal consequences including premature birth, low birth weight, birth defects, and sudden infant death syndrome [10–14]. Notably, an estimated 20 to 40% of all low-birthweight births are linked to MS, and low birthweight is the strongest predictor of mortality and morbidity in neonates [15]. Furthermore, 22% of sudden infant deaths in the United States are directly attributable to MS, and the risk of sudden infant death positively covaries with number of cigarettes consumed daily [16]. Circa 1983, approximately 9% ($272 million USD) of annual expenditures on neonatal intensive care in the United States were attributable to MS [15]. In 2020, the estimated aggregate costs of MS totaled $1.16 billion USD [17].
As will be detailed in the ensuing text, MS appears to be associated with heightened risk for neurodevelopmental disorders (NDDs) including attention-deficit/hyperactivity disorder (ADHD), autism, and schizophrenia in children and possibly grandchildren as well as a plethora of discrete transdiagnostic features of NDDs such as abnormal brain and neuronal development, behavioral anomalies such as hyperactivity and impulsivity, aberrant circadian rhythmicity, predisposition to smoking and other forms of substance abuse, alterations in catecholamine and cholinergic neurotransmitter systems, neurotrophic dysfunction, hypothalamic–pituitary–adrenal (HPA) axis deregulation, and epigenetic changes such as DNA methylome perturbations. Given the associations of MS and possibly grandmaternal smoking (GMS) with NDDs and the aforementioned transdiagnostic features thereof, it is also worthwhile to highlight the economic burdens attributable to NDDs, as a portion of these costs may be indirectly related to MS. Therein, the annual economic burdens of ADHD, autism, and schizophrenia were estimated at $12.76 billion USD in 2018, $250 billion USD circa 2014, and $155 billion USD in 2013, respectively, with the economic burden of autism projected to nearly double to $460 billion USD by 2025 [18–21].
While the economic burdens of MS as well as neurodevelopmental deficits and disorders are tremendous, the human and quality of life impacts of these phenomena are immeasurably greater. As such, it is of vital importance to better understand the specific brain and behavioral consequences as well as the broader public health implications of MS and GMS. Toward this objective, the present review endeavors to synthesize the states of the human and animal model literatures on the impacts of MS and GMS (or maternal nicotine exposure and grandmaternal nicotine exposure in the case of animal model research) on neurodevelopmental outcomes as well as the emerging evidence of a mechanistic role for DNA methylome perturbations therein.
State of the human literature on the neurodevelopmental impacts of MS
Association of MS with risk for NDDs
A substantial body of research associates MS with enhanced risk for NDDs including ADHD, autism, and schizophrenia in children, and there may be a causal pathway between MS and later neurodevelopmental problems [22–36]. However, some of the associations between MS and NDDs, namely autism and schizophrenia, no longer hold once controlling for gestational age and other confounding familial variables [27–30, 37–41]. Furthermore, while the literature supporting an association between MS and ADHD is relatively robust, there are also studies which suggest that MS may not be a direct risk factor independent of familial factors for ADHD [23, 26, 31–34, 42]. These mixed findings are in line with recent debate as to whether associations between MS and offspring NDD liability are indeed causal or instead due to unmeasured confounding variables [43]. For instance, genetic liability for both smoking and NDDs exists and may link MS and child outcomes without a direct teratogenic effect of smoking per se [22, 40]. In addition, there may be other familial variables, often conceptualized as more environmental in nature (e.g. parental education and parenting), that could also contribute to (and thus confound) the relationship between MS and offspring outcomes such as NDD diagnoses. In conjunction with the possible confounding of the associations between MS and NDD diagnoses by genetic and familial variables, heterogeneity in psychiatric diagnostic classification, which stems from the fact that clinical diagnoses for NDDs and other psychiatric conditions are not objective, discrete, or standardized measurements, undermines the utility and validity of clinical diagnoses as primary outcome measures for studies assessing the associations between MS and neurodevelopmental outcomes [44]. Accordingly, it is arguably more informative and rigorous to explore the relationships of MS with specific neurodevelopmental deficits and transdiagnostic markers of NDDs rather than inherently confounded NDD diagnoses.
Associations of MS with discrete neurobehavioral deficits
Epidemiological studies examining the relationships between MS and various transdiagnostic features of NDDs suggest a robust association of MS with a multitude of neurodevelopmental and behavioral alterations across the lifespan, with the majority of such studies focusing on the developmental periods of infancy, childhood, and adolescence [22, 45]. Therein, MS is associated with delayed infant psychomotor and mental developmental scores as measured by the Bayley Scales of Infant Development, atypical auditory brainstem responses, composite measures of neurodevelopmental impairment, and decreased motor scores between ages 18 and 21 months [37, 46–48]. Investigations of the relationship of MS with childhood and adolescent outcomes are considerably more common than those for infants and generally report the most consistent findings. For instance, MS is associated with intellectual impairment at age 4, inattention at age 6, hyperactivity at age 8, physical aggression during early childhood as well as middle childhood and adolescence, and increased externalizing disorders such as conduct disorder and ADHD [22–23, 26, 33–34, 49–56]. Many cognitive impairments are also associated with MS including deficits in sustained attention, response inhibition, overall cognitive function and memory, receptive language, verbal learning and design memory, problem solving, speech and language, school performance, and auditory processing as well as impulsivity [57–64]. Research also reveals dose–response relationships wherein MS-related relative risks for neurodevelopmental abnormalities increase with amount smoked, results which suggest the existence of vulnerable periods of fetal development [65–67]. Several studies have also reported that, similar to the associations of neurodevelopmental deficits and disorders with a proclivity to smoke, a tendency to start smoking earlier, and a penchant to smoke more, the children of maternal smokers are more likely to smoke, initiate smoking earlier, and smoke more [1, 12, 31, 68–77]. Importantly, many of the phenotypic aberrations associated with MS in the aforementioned studies are broadly characteristic of NDDs including ADHD, autism, and schizophrenia [78–93].
Associations of MS with abnormal brain and neuronal development
MS alters the structure of the developing human nervous system in part by perturbing neuronal differentiation, is associated with long-term neurological morbidity, results in lower proportions of neurons in the developing dorsolateral prefrontal cortex, is linked to reduced brain volume, reduced cortical gray matter volume, and cortical thinning in children, and is associated with hypoactivity of the ventral striata during reward anticipation in adolescents [94–100]. Taken together, these findings link MS with indicators of broad neuroteratogenicity and developmental encephalopathy in children and are reminiscent of the decreased total brain volume in ADHD and schizophrenia as well as the diminished cortical volume and thickness and impaired neuronal differentiation in ADHD, autism, and schizophrenia [101–107].
Associations of MS with cholinergic system anomalies
Nicotinic acetylcholine receptors (nAChRs) are expressed in a neurodevelopmental stage-specific manner under tight spatiotemporal regulation and function as key mediators of neuronal differentiation, maturation, and morphology as well as synaptogenesis in the developing brain [108–112]. Moreover, cholinergic signaling mediates NDD-related phenotypes including locomotor activity and impulsivity across the lifespan [113–117]. Notably, atypical nAChR expression patterns have been documented in the brains of the children as well as the placentas of maternal smokers [111, 118–120]. These findings concur with studies linking altered nAChR expression patterns and nAChR polymorphisms to ADHD, autism, and schizophrenia [121–130].
Associations of MS with catecholamine system perturbations
The catecholamines dopamine (DA) and norepinephrine (NE) are important mediators of many neurodevelopmental processes and exert modulatory influences on an array of neurobehavioral phenotypes relevant to NDDs as well as the neurodevelopmental consequences of MS [131–134]. Infants of maternal smokers have diminished norepinephrine content in cord blood at birth, and the children of maternal smokers exhibit highly significant associations between NE transporter (NET) polymorphisms and ADHD diagnosis, cognitive-behavioral measures relevant to ADHD, and response to methylphenidate treatment [135–136]. Furthermore, MS is associated with specific subtypes of ADHD in genetically susceptible children possessing variations in the DA transporter (DAT1) and/or DA receptor D4 (DRD4) loci [137]. Similarly, hyperactivity-impulsivity and oppositional behaviors are associated with a DAT polymorphism exclusively in children that were exposed to MS [138]. These limited findings suggest an emerging role of catecholamine system perturbations and polymorphisms in the effects of MS on neurodevelopmental outcomes in children and are congruent with the altered corticostriatal NE and DA neurotransmission, NE and DA receptor expression, and DAT and NET function as well as the DA and NE receptor and DAT and NET polymorphisms linked to NDDs including ADHD, autism, and schizophrenia [139–175].
Associations of MS with neurotrophic dysfunction
Neurotrophins such as brain-derived neurotrophic factor (BDNF) play vital roles in pregnancy wherein they mediate placental growth and development as well as offspring neurodevelopment [176–178]. Aberrant BDNF signaling is implicated in NDDs and pathological features including disrupted neuronal development and synaptogenesis, impaired dendritic arborization, altered synaptic plasticity, and aberrant synaptic pruning accompanied by learning and memory impairments, emotional dysregulation, and aberrant stress responsivity [179–192]. BDNF dysfunction may facilitate neurobehavioral deficits in part via excessive inhibition of inhibitory neurons resulting in disinhibition of and reduced capacity to downregulate excitatory circuits [193–195]. Notably, research suggests that the children of maternal smokers are subject to deregulation of BDNF signaling and display abnormal BDNF concentrations in umbilical cord sera [196–197]. Furthermore, BDNF moderates associations between MS and offspring behavioral disorders [195]. This nascent literature associates MS with alterations in BDNF signaling which are linked to neurodevelopmental deficits in offspring and that are reminiscent of the atypification of BDNF signaling implicated in NDDs including ADHD, autism, and schizophrenia [179–182].
Associations of MS with HPA axis dysregulation
HPA axis dysfunction disrupts neurodevelopment and neuronal maturation, perturbs synaptic plasticity, elicits hyperactivity, impulsivity, and aberrant stress responsivity, and promotes emotional deregulation [24, 198–204]. Multiple studies have identified markers of HPA axis dysfunction in neurodevelopmentally disordered children including hypocortisolemia and altered glucocorticoid (GC) receptor (GR) expression [205–209]. Similarly, the children of maternal smokers also display hypocortisolemia and aberrant GR expression [210–213]. Taken together, these findings indicate that MS confers HPA axis dysfunction characteristic of NDDs.
Emerging intergenerational consequences of MS
Alarmingly, contemporary research associates GMS with enhanced risk for ADHD diagnosis and increased autism symptoms [42, 214]. These findings imply that the heightened vulnerabilities to neurodevelopmental deficits and disorders associated with MS may be transmissible across multiple offspring generations. However, it should be noted that the preponderance of research examining the intergenerational influences of MS has focused on outcomes related to physical health such as asthma, while only a handful of studies have assessed neurodevelopmental deficits/disorders in relation to MS. Moreover, rigor and reproducibility in intergenerational human subjects research are extremely difficult to achieve given the amount of time and other resources needed to conduct such longitudinal family study designs coupled with common hurdles related to participant recruitment and retention. In the cases of MS and GMS studies, these inherent obstacles to intergenerational human subjects research often force investigators to make various inferences in light of incomplete or insufficient longitudinal data, such as assumptions about whether or not there was additional smoke exposure at other points in the pathway between the grandmother to the grandchild, including whether the mother smoked during her pregnancy. It therefore remains indeterminate whether the associations of GMS with neurodevelopmental deficits and disorders reported thus far will persist after controlling for confounders, or whether intergenerational influences of smoking during pregnancy affect other aspects of neurodevelopment.
In retrospect, there is extensive evidence from the human literature that associates MS with a variety of neurobehavioral outcomes in children, but animal model studies have yielded much of what is known about the neurobiological effects of MS on the developing brain. Therein, while the human research associating MS with cholinergic dysregulation, catecholamine dysfunction, BDNF dysregulation, and HPA axis aberrations is relatively sparse, the findings of studies characterizing maternal nicotine exposure (MNE) animal models of MS presented hereafter provide robust evidence of NDD-like anomalies in nAChR expression and function, DA and NE neurotransmission, BDNF signaling, and glucocorticoid hormone signaling as well as glucocorticoid receptor activity in the offspring of female rodents subjected to nicotine exposure paradigms designed to recapitulate MS patterns. Furthermore, a burgeoning body of animal model literature indicates that many MNE-induced NDD-like neurobiological perturbations undergo intergenerational co-transmission along with behavioral alterations that recapitulate phenotypes exhibited by the children of maternal smokers and neurodevelopmentally disordered children alike.
State of the animal model literature on the neurodevelopmental impacts of MS
Paradigms for animal modeling of the intergenerational impacts of MS
The information that animal models can provide to human research is significant [215]. For example, animal models enable intergenerational study designs that incorporate a specific controlled dose of a specific drug over a defined interval in a controlled setting. This experimental precision affords valuable insights into the exposure-, dosage-, and developmental stage-specific impacts of prenatal exposures on discrete behavioral phenotypes and accompanying biological changes at high resolution.
Translational validity is of the utmost importance in the design of animal models of the children and grandchildren of maternal smokers. To this end, a consensus paradigm for modeling the intergenerational impacts of MS utilizes a nicotine (or tobacco smoke) exposure regimen informed by the findings of epidemiological research that the majority of maternal smokers initiate regular smoking well in advance of conception and continue smoking throughout pregnancy and nursing [216, 217]. Therein, nicotine or tobacco smoke exposure of founder (F0) female dams (MNE) commences prior to mating with drug-naïve sires and continues until weaning of first-generation (F1) offspring. By this design, the F1 generation embryo/fetus and the maternal germ cells are subjected to direct nicotine or tobacco smoke exposure. Subsequent breeding of F1 generation progeny that were maternally exposed to nicotine or tobacco smoke during development (which model the children of maternal smokers and are referred to as MNE animals hereafter) with drug-naïve mates yields second-generation (F2) offspring that were exposed to nicotine or tobacco smoke exclusively via the maternal germline (which model the grandchildren of maternal smokers and are referred to as GMNE animals hereafter). Successive breeding of GMNE (F2) progeny with drug-naïve mates yields third-generation (F3) offspring that are entirely devoid of direct exposure to nicotine or tobacco smoke (which model the great grandchildren of maternal smokers). Reiteratively, MNE animals model the children of maternal smokers, GMNE animals model the grandchildren of maternal smokers, and F3 progeny model the great grandchildren of maternal smokers. As such, GMNE animal models provide insight into the intergenerational or multigenerational impacts of MS, while F3 generation progeny yield insight into the transgenerational effects of MS.
Importantly, despite its utility for modeling neurodevelopmental outcomes in the children and grandchildren of maternal smokers, the current MNE and GMNE animal model literature is limited in many facets. Perhaps most notably, whereas in the human literature the prevalences of traditional and electronic cigarette consumption among maternal smokers are comparable, an overwhelming majority of the MNE (F1) animal model studies referenced in the manuscript utilized pure nicotine exposure paradigms (which are more comparable to electronic than traditional cigarettes) rather than tobacco smoke exposure paradigms (which are more comparable to traditional than electronic cigarettes), and all GMNE (F2) animal model studies exclusively utilized nicotine exposure paradigms, a scenario that ultimately hinders the translatability of findings from animal model studies to the human condition. Owing further to this imbalance in the animal model literature, limited insights can be derived by evaluation of data from maternal tobacco smoke exposure animal model studies independent of data from MNE animal model studies. Nevertheless, an amalgamation of MNE (but not GMNE) animal model investigations which utilized different exposure paradigms and routes of administration including combustible tobacco inhalation chambers, aerosolized/vaporized nicotine apparatuses, oral nicotine delivery, nicotine injections (e.g. intraperitoneal or subcutaneous), and nicotine infusions (e.g. osmotic minipumps) have reported multiple NDD-related phenotypes that are common across study designs.
Intergenerational behavioral impacts of MNE
Consistent with behavioral anomalies documented in the children of maternal smokers, animal model research demonstrates that MNE mice exhibit hyperactivity, aberrant rhythmicity of activity, impulsivity-like and risk-taking behaviors, and inattention [50–52, 218–226]. These results recapitulate behavioral phenotypes shared among ADHD, autism, and schizophrenia patients [78–93]. Moreover, MS animal model studies also reveal reduced nicotine sensitivity, increased nicotine self-administration, and enhanced oral nicotine consumption and preference [219, 224, 227]. These findings are concordant with the proclivity to smoke, earlier initiation of smoking, and tendency to smoke more documented in the children of maternal smokers and linked to both clinical diagnoses and discrete symptoms (in the absence of a diagnosis) of ADHD, autism, and schizophrenia [1, 12, 31, 68–77]. Notably, contemporary research indicates that MNE-evoked hyperactivity is transgenerationally transmissible, while perturbed rhythmicity of activity, risk-taking behaviors, and enhanced oral nicotine consumption and preference appear to be intergenerationally transmissible [224]. These findings are consistent with human studies linking GMS to ADHD and autism in grandchildren [42, 214]. Altogether, the findings of animal model studies indicate that MNE elicits an array of behavioral anomalies that are consistent with NDDs including ADHD, autism, and schizophrenia and that appear to be transmissible to subsequent generations.
Interestingly, treatment with methylphenidate, a first-line medication for NDDs such as ADHD, rescues NDD-like phenotypes including hyperactivity, altered rhythmicity of activity, and risk-taking behaviors in both MNE and GMNE rodents [222–224, 228]. These findings support the validity of MNE and GMNE rodents as models for the intergenerationally transmissible impacts of MS. Similarly, voluntary oral nicotine consumption is elevated in MNE and GMNE rodents and exerts normalizing effects on hyperactivity and aberrant rhythmicity of activity that span both generations [224]. These results support the self-medication hypothesis for the comorbidity of smoking in NDDs, which postulates that the increased prevalence of smoking in NDDs such as ADHD and schizophrenia is attributable to therapeutic-like effects of nicotine [73, 224, 229–232].
Impacts of MNE on brain and neuronal development
MNE and GMNE rodents display several markers of impaired brain and neuronal development including premature neuronal differentiation, atypical neuronal morphology, neuronal apoptosis, disrupted neuronal maturation, aberrant neuronal migration, and reduced cortical volume and thickness [221–222, 233–239]. These results mirror the reduced total brain volume, decreased cortical volume, thickness, and neuronal content, and aberrant neurodifferentiation shared between neurodevelopmentally disordered children and the children of maternal smokers [94–107]. Notably, whether these developmental encephalopathies and indicators of neuroteratogenicity exhibited by MS animal models are intergenerationally transmissible remains indeterminate.
Intergenerational impacts of MNE on the cholinergic system
Animal model research indicates that the neuroteratogenic effects of MNE are attributable in part to MNE-evoked atemporal nAChR stimulation early in development [233–237]. Additional studies reveal alterations in both the expression and function of a variety of nAChR subtypes throughout the fetal, adolescent, and adult corticostriatothalamic circuitry of MNE rodents, suggesting that the impacts of MNE on corticostriatothalamic cholinergic signaling persist across the lifespan [238–247]. These findings mimic the perturbed nAChR expression patterns reported in the placenta and the children of maternal smokers, recapitulate the misconnectivity and cholinergic anomalies documented within the corticostriatothalamic neurocircuitry of neurodevelopmentally disordered individuals, and support the implication of corticostriatothalamic and cholinergic dysfunction in NDD-related phenotypes including locomotor hyperactivity, inattention, and impulsivity [111, 113, 115, 118–130, 248–258]. In furtherance of these findings, GMNE animal models exhibit an array of abnormalities in both the expression and function of multiple nAChR subpopulations within the frontal cortices, striata, and thalami, implying that MNE-induced alterations in the corticostriatothalamic cholinergic system are intergenerationally transmissible [224].
Intergenerational impacts of MNE on the catecholamine system
Concordant with the corticostriatal catecholamine system dysfunction and DA and NE receptor and transporter polymorphisms implicated in NDDs as well as the catecholamine system perturbations and polymorphisms implicated in the effects of MS on neurodevelopmental outcomes in children, MNE and GMNE animals exhibit a variety of anomalies in DA and NE signaling [135–175, 222, 224, 228, 233, 259–262]. Therein, adolescent and adult MNE rodents display decreased DA synthesis and release, altered DAT and NET as well as DA receptor expression, impaired DAT function, and impaired DA turnover within the corticostriatal circuitry [222, 224, 228, 233, 259–262]. In support and expansion of these findings, additional research demonstrates intergenerational transmission of dopaminergic and DAT dysfunction in animal models of the children and grandchildren of maternal smokers [139–165, 224]. These results are again consistent with the catecholamine system abnormalities documented in ADHD, autism, and schizophrenia and thus further support the notion that MNE-evoked NDD-like phenotypes are heritable.
Intergenerational impacts of MNE on neurotrophic signaling
Comparable to the perturbations of neurotrophic signaling documented in ADHD, autism, and schizophrenia patients and the implication of BDNF deregulation in neurodevelopmental deficits in the children of maternal smokers, animal model studies reveal aberrant BDNF signaling within the fetal, adolescent, and adult mesocorticolimbic circuitry as well as the juvenile brainstem of MNE rodents [179–183, 195–197, 259, 253–267]. Indicative of the intergenerational transmissibility thereof, recent research reveals that MNE and GMNE rodents exhibit deficiencies in frontal cortical and striatal BDNF content coupled with proportional accumulations of proBDNF, the proapoptotic peptide precursor of BDNF which functions in opposition to BDNF both during development and across the lifespan [185, 268–269]. Furthermore, the accumulations of proBDNF detected in MNE and GMNE rodents co-occur and are inversely correlated with downregulation of furin, a metalloprotease which catalyzes proteolytic conversion of proBDNF to BDNF and is downregulated in NDDs and thought to mediate proBDNF-BDNF imbalance therein [182, 184, 270–277]. These findings suggest that MNE and GMNE induce intergenerational transmission of NDD-like proBDNF-BDNF imbalance which appears to be driven by impairment of furin-mediated proBDNF proteolysis stemming from furin downregulation [268]. Collectively, these results provide strong evidence of neurotrophic dysfunction in MNE and GMNE animal models of the children and grandchildren of maternal smokers, respectively.
Intergenerational impacts of MNE on HPA axis function
Analogous to the HPA axis dysfunction such as hypocortisolemia and deficient GC signaling documented in neurodevelopmentally disordered children and the children of maternal smokers, MNE and GMNE rodents exhibit hypocorticosteronemia, aberrant GR expression in the frontal cortices and striata, and perturbed GR activity in the frontal cortices, striata, and hippocampi, suggesting that MNE-induced NDD-like HPA axis dysfunction is intergenerationally transmissible [24, 198, 208–213, 268, 278–281].
Overview of DNA methylation and the environmental epigenetics of MS
DNA methylation is a mechanism for the epigenetic regulation of gene expression wherein methyl moieties are covalently bound to nucleotide bases within DNA molecules. In turn, these methylated nucleotides can directly modulate the transcriptional accessibility of DNA sequences and/or indirectly influence chromatin architecture via interactions with histone modifiers.
Environmental epigenetics is a burgeoning field of study examining how environmental exposures such as MS affect epigenetic processes such as DNA methylation, particularly during critical and sensitive periods of development (i.e. the prenatal period) [282]. Therein, during the prenatal period methylation patterns of the germline and somatic cell lineages are beginning to be established [283–285]. These epigenetic marks are involved in modulating functional pathways and are key for the health and development of the embryo. Direct exposure to toxins, such as MS, is associated with changes in offspring methylation patterns in a variety of tissues [286–287]. Thus, prenatal smoke exposure at critical points of fetal development may exert important and persistent effects on DNA methylation which in turn influence gene expression and phenotypes later in life. As such, DNA methylation profiles may have utility as biomarkers of MS, as estimators of risk for behavioral deficits and disease, and as therapeutic targets [288–290]. In conjunction with its critical roles in brain and behavioral phenotypes during development and across the lifespan, the DNA methylome also mediates intergenerational phenotypic transmission via the germline.
As will be expounded in the subsequent passages, a plurality of the MS-associated as well as MNE- and GMNE-induced behavioral and neurobiological alterations reported in the aforementioned studies co-occurs with DNA methylome perturbations. Considering the implication of epigenetic changes in the induction by environmental exposures as well as the intergenerational transmission of a broad spectrum of brain and behavioral phenotypes, this co-transmission of DNA methylome and neurobehavioral anomalies in the children of maternal smokers as well as MNE and GMNE animal models suggests a putative mechanism wherein DNA methylome changes are substrates for the induction and intergenerational transmission of MS-associated as well as MNE- and GMNE-induced neurodevelopmental abnormalities. While the present transmittal focuses on DNA methylome alterations as epigenetic bases for these neurodevelopmental abnormalities, it is important to note that such phenotypes are likely also mediated by impacts on other epigenetic processes such as histone modifications.
State of the human literature on the DNA methylomic impacts of MS
DNA methylation mediates neurodevelopment, neuroplasticity, and synaptogenesis, neurodegenerative processes, learning, memory, and cognition, anxiety, stress responsivity, and emotional regulation, locomotor activity and impulsivity/risk-taking, the circadian rhythmicity of behavior, and cholinergic, dopaminergic, neurotrophic, and HPA axis signaling [179–180, 198, 208–209, 235, 272, 291–318]. As such, DNA methylome alterations may contribute to neurobehavioral deficits including the hyperactivity, impulsivity, inattention, and circadian anomalies, the disruption of brain and neuronal development, the malfunction of cholinergic and catecholamine systems, and the deregulation of BDNF and HPA axis signaling in neurodevelopmentally disordered individuals as well as the children of maternal smokers and/or animal models thereof. In support of this possibility, NDDs including ADHD, autism, and schizophrenia are associated with DNA methylome alterations [313, 319–332]. For instance, DNA methylation levels at birth are inversely related to symptom severity in pediatric ADHD patients, and DAT1 promoter methylation mediates hyperactivity and impulsivity in ADHD [236, 303, 319]. As will be detailed in the ensuing text, MS is also associated with DNA methylome perturbations, which may constitute a mechanistic basis for the increased incidences of neurodevelopmental deficits and disorders in the children and possibly the grandchildren of maternal smokers [98, 302, 333–338].
Associations of MS with DNA methylome perturbations in placental tissue
Both epigenome-wide association studies (EWAS) and gene-specific methylation studies have yielded significant associations between MS and placental methylation patterns. Summarily, DNA methylomic studies of placental tissue associate MS with aberrant global DNA methylation patterns as well as differential methylation of specific genes involved in brain and neuronal development, neuronal differentiation, synaptogenesis, and proliferation of various cell types within the central nervous system [335]. Candidate gene studies yielded associations between MS and methylation of genes involved in the metabolism of the potentially carcinogenic compounds found in cigarette smoke such as AHRR (CYP1A1) as well as the serotonin and glucocorticoid receptor subtypes HTR2A and NR3C1, respectively (Table 1) [339–341]. Notably, MS is associated with altered placental methylation of the NR3C1 promoter which mediates concomitant hypocortisolemia detected in corresponding salivary samples [341–342].
Table 1.
Glossary of gene symbols, gene names, and encoded protein functions for selected genes exhibiting differential methylation signatures in placental tissues, cord blood, and whole blood. Encoded protein functions were excerpted from Entrez Gene summaries
| Symbol | Name | Encoded Protein Function |
|---|---|---|
| AHRR | Aryl Hydrocarbon Receptor Repressor | Implicated in the regulation of cell growth and differentiation. Exhibits nuclear receptor and sequence-specific double-stranded DNA binding activities. Functions as a feedback modulator of AhR by repressing AhR-dependent gene expression. |
| NR3C1 | Nuclear Receptor Subfamily 3 Group C Member 1 | Functions as a transcription factor that binds to glucocorticoid response elements as well as a regulator of other transcription factors. Inactive form is found in the cytoplasm but translocates to the nucleus upon activation by glucocorticoid ligands. Involved in inflammation, proliferation, and differentiation. |
| DNMT3A | DNA Methyltransferase 3a | De novo DNA methyltransferase responsible for the establishment of DNA methylation patterns in embryos, the methylation of most imprinted loci in germ cells, and the establishment of DNA methylation patterns during development. Can actively repress transcription through the recruitment of HDAC activity. |
| MBD3 | Methyl-CpG Binding Domain Protein 3 | Acts as transcriptional repressor and plays a role in gene silencing. Does not bind to DNA by itself. Binds to DNA with a preference for sites containing methylated CpG dinucleotides (in vitro). Binds to a lesser degree DNA containing unmethylated CpG dinucleotides. Recruits histone deacetylases and DNMTs. |
| SLC6A3 | Solute Carrier Family 6 (Neurotransmitter Transporter, Dopamine), Member 3 | Member of the sodium- and chloride-dependent neurotransmitter transporter family. Terminates the action of dopamine by its high affinity sodium-dependent reuptake into presynaptic terminals. Variants associated with idiopathic epilepsy, attention-deficit hyperactivity disorder, dependence on alcohol and cocaine, susceptibility to Parkinson’s disease, and protection against nicotine dependence. |
| DRD1 | Dopamine Receptor D1 | Most abundant dopamine receptor in the central nervous system. Stimulates adenylyl cyclase and activates cyclic AMP-dependent protein kinases. Regulates neuronal growth and development and mediates some behavioral responses. |
| MAOB | Monoamine Oxidase B | Catalyzes the oxidative deamination of biogenic and xenobiotic amines and has important functions in the metabolism of neuroactive and vasoactive amines in the central nervous system and peripheral tissues. MAOB preferentially degrades benzylamine and phenylethylamine. Involved in dopamine metabolic processes. |
| CHRNA5 | Cholinergic Receptor Nicotinic Alpha 5 Subunit | Member of the superfamily of ligand-gated ion channels that mediate fast signal transmission at synapses. Contribute to acetylcholine receptor activity. Involved in regulation of synaptic vesicle exocytosis and response to nicotine. Localizes to acetylcholine-gated channel complexes and dopaminergic synapses. |
| CHRNB4 | Cholinergic Receptor Nicotinic Beta 4 Subunit | Exhibits acetylcholine receptor activity and acetylcholine-gated cation-selective channel activity. Involved in signal transduction and synaptic transmission. Localizes to acetylcholine-gated channel complex. |
| BDNF | Brain-Derived Neurotrophic Factor | During development, promotes the survival and differentiation of selected neuronal populations of the peripheral and central nervous systems. Participates in axonal growth, pathfinding and in the modulation of dendritic growth and morphology. Major regulator of synaptic transmission and plasticity at adult synapses in many regions of the CNS. |
| GDNF | Glial Cell Derived Neurotrophic Factor | Highly conserved neurotrophic factor involved in negative regulation of apoptotic processes; positive regulation of gene expression, and nervous system development, and survival and differentiation of dopaminergic neurons |
| FURIN | Furin (Paired Basic Amino Acid Cleaving Enzyme) | Calcium-dependent serine endoprotease that proteolytically activates different proprotein substrates traversing the secretory pathway. Involved in regulation of protein turnover and metabolism as well as regulation of transforming growth factor beta1 production and proteolytic conversion of proBDNF to mature BDNF. |
| DLGAP2 | DLG Associated Protein 2 | Molecular adaptor activity and protein domain specific binding activity. Involved in regulation of postsynaptic neurotransmitter receptor activity. Localizes to dendritic spines, glutamatergic synapses, and postsynaptic density. Knock-out confers hyperactivity, aggression, impaired reversal learning, decreased dendritic spine density and mESPC amplitude, synaptopathies, and enhanced paired-pulse ratio. |
| NRP2 | Neuropilin 2 | Exhibits semaphorin receptor activity. Localizes to axon; glutamatergic synapse; and integral component of postsynaptic membrane. Involved in nervous system development, neural crest cell migration, and axonal guidance. |
Associations of MS with DNA methylome perturbations in cord blood
Cord blood is a surrogate tissue that is widely utilized for EWAS of the effects of environmental exposures on the epigenome. Notably, the cord blood DNA methylome is shared between mother and child and in some instances may relate to phenotypes shared between maternal smokers and their children. However, it warrants consideration that the cord blood DNA methylome has not been shown to substantially relate to the DNA methylomes of specific cells and cell types within brain regions implicated in NDDs.
Studies examining cord blood from the children of maternal smokers intimate that MS confers global DNA hypomethylation [343]. The Pregnancy and Childhood Epigenetics Consortium (PACE) conducted a meta-analysis across 13 birth cohorts across the USA and Europe (N = 6685) examining the association between MS and newborn cord blood methylation at over 450 000 CpG sites [337]. Results suggested that 6073 CpGs were considered statistically significant when using FDR correction, while 568 CpGs remained significantly associated after Bonferroni correction for multiple testing [337]. Fewer statistically significant findings were found for any MS than for sustained MS, suggesting that comprehensive measurement of MS is an important consideration, with more sustained smoking over the entire pregnancy offering more power when compared to any smoking (yes/no) for studying epigenetic effects of MS [337].
Consistent with prior reports examining both MS-exposed offspring and adult smokers, the top finding of the PACE meta-analysis of MS and newborn cord blood methylation was for AHRR (aryl hydrocarbon receptor repressor), a gene that is involved in the detoxification of chemicals found in tobacco smoke (i.e. the xenobiotic response) and which acts as a feedback inhibitor of the aryl hydrocarbon receptor (AHR) [333, 337, 344–349]. Accordingly, AHRR demethylation and resultant upregulation may constitute an adaptive cellular response to the accumulation of chemicals found in cigarette smoke [345]. Furthermore, the methylation levels of several AHRR CpGs in cord blood of infants of maternal smokers are inversely related to cotinine levels, suggestive of a dose–response relationship between MS and AHRR demethylation [333]. Critically, GMS is also associated with AHRR demethylation in cord blood [349], findings which are rendered even more remarkable in light of the extensive involvement of AHR in phenotypes shared among neurodevelopmentally disordered individuals, the children of maternal smokers, and MNE and GMNE rodents. Therein, AHRs are expressed under tight spatiotemporal control within the developing brain, broadly regulate neuronal development, neuronal morphology, and synaptic maturation, neuronal migration and dendritic outgrowth, and neuronal differentiation, modify locomotor activity and the circadian rhythmicity thereof, mediate cholinergic influences on neurodevelopment by regulating acetylcholinesterase activity across various neurodevelopmental stages and processes, modulate DA synthesis by regulating tyrosine hydroxylase expression and thus indirectly regulates NE synthesis, influence BDNF expression in cortical neurons, and tune HPA axis function and the circadian rhythmicity thereof by regulating the activation of glucocorticoid receptors as well as the synthesis and release of cortisol [350–365]. In addition, AHR is subject to polymorphisms associated with increased severity of autism symptoms, and AHRR has expansive intragenic CpG islands which exhibit differential methylation that is dose-dependently associated with exacerbation of symptom severity in ADHD [366–368].
Interestingly, the aforementioned findings of decreased AHRR methylation in cord blood have not been replicated in placental tissue or saliva cells (345); however, this is not surprising given that AHRR is expressed at low levels in the placenta, while cord blood cells show high interindividual variation [345, 369]. Taken together, these results highlight the importance of examining the DNA methylomic impacts of MS across tissue types and considering differences in normative gene expression therein.
Additional evidence from studies of cord blood from maternal smokers reveals aberrant locus-specific DNA methylation signatures at CpGs located within genes (Table 1) including DNMT3A, DNMT1, and MBD3, SLC6A3 (DAT) and MAOB, CHRNA1 and CHRNA5, GDNF, NGFR, and FURIN, DLGAP2 (also known as SAPAP2), and NRP2 that encode proteins with functions (Table 1) relevant to phenotypes characteristic of the children of maternal smokers, neurodevelopmentally disordered children, and animal models thereof [337, 370–373]. For instance, DLGAP2 is known to mediate synaptic organization and neuronal signaling and is associated with both schizophrenia (370) and autism (371, 372), while NRP2 is expressed in neural crest cells, appears to be necessary for proper neuronal migration, and is associated with autism [337, 370–373]. Moreover, the differential methylation of SLC6A3 as well as FURIN are consistent with the DAT dysfunction as well as the furin downregulation and related proBDNF-BDNF imbalance, respectively, documented in MNE and GMNE animal models.
Complementing the locus-specific DNA methylome perturbations identified in the children and grandchildren of maternal smokers, functional annotation enrichment analyses have provided valuable insight into the overarching impacts of MS on the cord blood DNA methylome. As exemplified in Table 2, research reveals differential methylation signatures at CpG sites mapped to loci enriched for multiple functional annotations relevant to the neurodevelopmental impacts of MS as well as the neuropathologies of ADHD, autism, and schizophrenia [337]. Therein, MS is associated with differential methylation of gene sets enriched for GO biological process annotations such as behavior (GO:0007610), brain development (GO:0007420), regulation of nervous system development (GO:0051960), regulation of membrane potential (GO:0042391), neurotrophin signaling pathway (GO:0038179), response to steroid hormone (GO:0048545), and sequence-specific DNA binding (GO:0043565) [337]. These findings relate MS to DNA methylome alterations which disproportionately impact genes involved in a broad spectrum of biological processes with particular relevance to phenotypic alterations exhibited by the children of maternal smokers and animal models thereof as well as the pathosymptomatology of NDDs.
Table 2.
Exemplary enriched functional annotations of differentially methylated genes in cord blood from maternal smokers and their relation to neurodevelopmental disorder-like phenotypes in the children of maternal smokers and animal models thereof. GO, gene ontology; DMGs, differentially methylated genes. [337]
| GO Identifier: |
GO Biological Process Annotations of Differentially Methylated Genes in MS Cord Blood | n (DMGs) | Related Phenotypes Associated with MS in Humans and Animal Models |
|---|---|---|---|
| 0007610 | Behavior | 33 | Neurobehavioral Deficits |
| 0007611 | Learning or memory | 14 | Neurobehavioral Deficits |
| 0050890 | Cognition | 15 | Neurobehavioral Deficits |
| 0050795 | Regulation of behavior | 17 | Neurobehavioral Deficits |
| 0040017 | Positive regulation of locomotion | 36 | Neurobehavioral Deficits |
| 0048511 | Rhythmic process | 12 | Neurobehavioral Deficits |
| 0007420 | Brain development | 47 | Developmental Encephalopathy |
| 0030900 | Forebrain development | 29 | Developmental Encephalopathy |
| 0021987 | Cerebral cortex development | 9 | Developmental Encephalopathy |
| 0021761 | Limbic system development | 8 | Developmental Encephalopathy |
| 0021766 | Hippocampus development | 6 | Developmental Encephalopathy |
| 0030902 | Hindbrain development | 11 | Developmental Encephalopathy |
| 0051960 | Regulation of nervous system development | 47 | Neuroteratogenicity |
| 0050767 | Regulation of neurogenesis | 44 | Neuroteratogenicity |
| 0045664 | Regulation of neuron differentiation | 39 | Neuroteratogenicity |
| 0043005 | Neuron projection | 30 | Neuroteratogenicity |
| 0001764 | Neuron migration | 10 | Neuroteratogenicity |
| 0016358 | Dendrite development | 13 | Neuroteratogenicity |
| 0050770 | Regulation of axonogenesis | 13 | Neuroteratogenicity |
| 0070997 | Neuron death | 20 | Neuroteratogenicity |
| 0030522 | Intracellular receptor signaling pathway | 27 | Cholinergic and Catecholine Dysfunction |
| 0034765 | Regulation of ion transmembrane transport | 18 | Cholinergic and Catecholine Dysfunction |
| 0042391 | Regulation of membrane potential | 20 | Cholinergic and Catecholine Dysfunction |
| 0034765 | Regulation of ion transmembrane transport | 18 | Cholinergic and Catecholine Dysfunction |
| 0019199 | Transmembrane receptor protein kinase activity | 20 | Cholinergic and Catecholine Dysfunction |
| 0048015 | Phosphatidylinositol-mediated signaling | 28 | Cholinergic and Catecholine Dysfunction |
| 0038179 | Neurotrophin signaling pathway | 34 | Neurotrophic Dysfunction |
| 0048011 | Neurotrophin TRK receptor signaling pathway | 34 | Neurotrophic Dysfunction |
| 1,901,653 | Cellular response to peptide | 28 | Neurotrophic Dysfunction |
| 0019838 | Growth factor binding | 17 | Neurotrophic Dysfunction |
| 0090287 | Regulation of cellular response to growth factor stimulus | 18 | Neurotrophic Dysfunction |
| 0046879 | Hormone secretion | 21 | HPA Axis Dysregulation |
| 0009914 | Hormone transport | 21 | HPA Axis Dysregulation |
| 0048545 | Response to steroid hormone | 19 | HPA Axis Dysregulation |
| 0048732 | Gland development | 19 | HPA Axis Dysregulation |
| 0080135 | Regulation of cellular response to stress | 33 | HPA Axis Dysregulation |
| 0051169 | Nuclear transport | 37 | HPA Axis Dysregulation |
| 0043565 | Sequence-specific DNA binding | 61 | DNA Methylome Perturbations |
| 0000975 | Regulatory region DNA binding | 61 | DNA Methylome Perturbations |
| 0003714 | Transcription corepressor activity | 29 | DNA Methylome Perturbations |
| 0003713 | Transcription coactivator activity | 33 | DNA Methylome Perturbations |
| 0000785 | Chromatin | 29 | DNA Methylome Perturbations |
| 0001047 | Core promoter binding | 11 | DNA Methylome Perturbations |
| 0017053 | Transcriptional repressor complex | 11 | DNA Methylome Perturbations |
Associations of MS with DNA methylome perturbations in offspring whole blood
Comparable to the aforementioned findings in cord blood from maternal smokers, studies in whole blood from the children of maternal smokers show differential methylation across multiple CpG sites mapped to noteworthy specific genes (Table 1) such as AHRR, CHRNB4, SYTL1, DRD1, and BDNF [196, 374–378]. These studies also mapped differential methylation patterns in whole blood from the children of maternal smokers to gene sets enriched for functional annotations of numerous NDD-related processes including axonogenesis (GO:0050373), axon guidance (GO:0007411), neuron projection guidance (GO:0097485), and neuron projection development (GO:0031175) as well as locomotion (GO:0040011). Noteworthy pathway annotations of said data included dopamine receptor mediated signaling pathway (P05912), axon guidance mediated by semaphorins (P00007), and nicotine pharmacodynamics pathway (P06587) [196, 374–378]. Building upon this research, another in whole blood from the children of maternal smokers concluded that MS-associated DNA methylome alterations mediate the effect of MS on schizophrenia related outcomes [379].
Associations of MS with DNA methylome perturbations in other tissues
Minimal research has examined the effects of MS on DNA methylation patterns in tissues other than placental cells, umbilical cord blood, and whole blood. Nevertheless, research characterizing leukocyte samples from the children of maternal smokers demonstrates associations between MS and global DNA hypomethylation as well as demethylation of repetitive elements [380, 381].
A study examining postmortem fetal brain tissue from maternal smokers indicates that MS perturbs DNA methylation in the developing fetal brain and confers co-occurring deficits in prefrontal cortical neuronal content [98]. Taken together, these results suggest that exposures experienced throughout the course of life (from the prenatal period to adulthood) and accompanying epigenetic changes may be one important biological pathway by which MS affects later behavior and developmental outcomes.
Cumulatively, human research efforts to date demonstrate that MS confers a constellation of DNA methylome anomalies which are congruent with findings in NDDs. However, notwithstanding the aforementioned study associating GMS with differential AHRR methylation in grandchildren, there is a paucity of human research on the intergenerational effects of MS on DNA methylation in relation to both gene expression and behavior in children and especially grandchildren.
State of the animal model literature on the DNA methylomic impacts of MS
The available animal model research on the intergenerational as well as germline epigenomic impacts of nicotine is disproportionately more expansive for paternal versus MNE. For instance, several studies have traced intergenerational and transgenerational transmission of global and locus-specific DNA methylome anomalies from paternal sperm to offspring and grandoffspring brain that co-occur with a variety of NDD-like behavioral and neurobiological perturbations [382–388]. Moreover, the breadth and volume of human research characterizing the DNA methylomic impacts of MS far exceed that of MNE and GMNE animal model research. The direction of this disparity is quite unusual considering that in most instances the animal model research on a given topic tends to mature/progress more rapidly than corresponding human research, as is the case for the comparative states of the human and animal model literatures on the neurobiological and neurobehavioral impacts of MS. Despite the paucity of previous research investigating heritable DNA methylome perturbations in MNE and GMNE animal models, a modicum of pioneering studies in this realm have produced compelling and insightful results that warrant postliminary research.
MNE and GMNE rodents exhibit DNA methylome perturbations that co-occur with and in some cases have been shown to directly mediate or correlate with phenotypes encompassed by transdiagnostic features of NDDs including hyperactivity, risk-taking behaviors, anomalous circadian rhythmicity of activity, enhanced nicotine consumption and preference, aberrant DA system and nAChR function, BDNF deregulation and proBDNF-BDNF imbalance, and hypocorticosteronemia as well as dysregulation of GR activity [183, 259, 264, 278, 389–390]. The de novo DNA methylase DNMT3A and the DNA demethylase TET2 reciprocally regulate global and locus-specific DNA methylation patterns [391–392]. As such, downregulation of DNMT3A and/or upregulation of TET2 could underlie the abovementioned multigenerational global DNA methylome deficits and by extension numerous other phenotypic aberrations documented in MNE and GMNE rodents. In support of this possibility, research demonstrates the co-occurrence of global DNA hypomethylation in the frontal cortices and striata of MNE and GMNE mice, downregulation of DNMT3A in the frontal cortices, striata, and hippocampi of MNE mice and in the frontal cortices and striata of GMNE mice, and downregulation DNMT3B in the hippocampi of MNE mice [393–394]. Considering the absence of detectable changes in TET2 expression in MNE and GMNE mice, corticostriatal DNMT3A deficits may primarily mediate the intergenerational transmission of corticostriatal global DNA hypomethylation conferred by MNE [224]. Taken together with the results of human studies associating MS with differential methylation of DNMT1 and DNMT3A, these data imply that MS could elicit broad DNA methylome perturbations in part by interfering with key mediators of the establishment and maintenance of methylation marks across the epigenome.
In light of research implicating nicotine-evoked upregulation of ovarian nAChRs in the reduced successful pregnancy rates, decreased oocyte viability, and increased oocyte abnormalities documented in female smokers, it has recently been suggested that the intergenerational impacts of MNE may be facilitated in part by nicotine-evoked, embryonic ovarian nAChR-mediated DNA methylome perturbations in oocytes [395–399]. This hypothesis is supported by contemporary research demonstrating that oocytes from MNE mice exhibit global DNA hypomethylation, diminished expression of multiple DNMT subtypes, and altered chromatin configuration as well as compromised integrity and viability [400]. Taken together with the aforementioned findings of global DNA hypomethylation and DNMT3A downregulation in the frontal cortices and striata of MNE and GMNE mice, these data demonstrate DNA methylome perturbations in the female germline of MNE mice which mimic those reported in the frontal cortices and striata of GMNE mice [224]. By extension, these results suggest a plausible mechanism for the intergenerational impacts of MNE wherein MNE induces overexpression of ovarian nAChRs in MNE embryos, which in turn facilitates DNMT-mediated DNA methylome alterations in the oocytes (germline) thereof that are subsequently inherited by and manifest in the brains of GMNE progeny. Suggestive of a similar MNE-evoked intergenerational epigenetic cascade in the male lineage, recent research reveals impaired maturation of gonocytes to spermatogonia in male MNE mice [401]. These findings imply that the male germline is also susceptible to and may mediate the intergenerational transmission of the DNA methylomic impacts of MNE. Altogether, the aforementioned evidence suggests that intergenerational transmission of DNA methylome perturbations via the female and possibly the male germline may be a primary mechanism underlying the heritability of NDD-like brain and behavioral phenotypes documented in animal models of the children and grandchildren of maternal smokers.
Epigenetic factors beyond global and locus-specific DNA methylation patterns, deregulation of DNA methyltransferases, and the germline transmissibility thereof may also be involved in the heritability of phenotypic aberrations elicited by maternal exposure to nicotine and other drugs of abuse [402–403]. Two such factors are methyl-CpG-binding protein-2 (MeCP2) and histone deacetylase 2 (HDAC2), the former of which (MeCP2) binds to methylated DNA and recruits the latter (HDAC2) as part of a cascade that links the DNA methylome and chromatinome and ultimately leads to heterochromatization and epigenetic silencing of methylated loci [404–405]. As such, it is noteworthy that downregulation of both MeCP2 and HDAC2 co-occurs with global DNA hypomethylation and DNMT3A downregulation in both MNE and GMNE mice [393]. These findings are especially compelling in light of research linking alterations in MeCP2 and HDAC2 expression to disruption of neuronal development, synaptogenesis, and synaptoplasticity, perturbation of BDNF signaling, learning and memory deficits, aberrant stress responsivity, and HPA axis dysregulation [296, 405–421].
Cumulatively, the findings of animal model research to date imply that MNE pervasively alters the DNA methylome and consequently the broader epigenome in developing progeny and the germline thereof, thereby contributing to the induction and intergenerational transmission of myriad NDD-like phenotypic aberrations. While paternal nicotine exposure is rather robustly linked to transgenerational brain and behavioral consequences attributable in part to epigenomic alterations, at present there is insufficient evidence to assess the transgenerational transmissibility of phenotypic aberrations conferred by MNE.
Discussion
Interpretations and implications
As visualized in Figure 1, human and animal model research characterizing the children and grandchildren of maternal smokers has, to date, yielded varying strengths of evidence for associations of MS and GMS with transdiagnostic features of NDDs including developmental encephalopathies, markers of neuroteratogenicity, cognitive-behavioral deficits, catecholamine and cholinergic system anomalies, neurotrophic dysfunction, HPA axis dysregulation, and global as well as locus-specific DNA methylome perturbations. Summarily, evidence linking MS to an array of NDD-related neurobehavioral alterations is strong across both the human and animal model literatures, but the preponderance of evidence extending these findings to GMS is derived from GMNE animal models. Moreover, while a few human studies tentatively associate MS but not GMS with various NDD-related neurobiological anomalies, relatively thorough characterizations of neurobiological anomalies in MNE and GMNE animal models yield considerable evidence of multigenerational transmission of MNE-induced nAChR, dopaminergic, and DAT dysfunction, BDNF-proBDNF imbalance, and HPA axis downregulation including hypocorticosteronemia and GR hypoactivity. The human literature affords substantial evidence associating both global and locus-specific DNA methylome alterations with MS but almost entirely lacks evidence indicating whether GMS similarly impacts the DNA methylome. Conversely, the animal model literature provides significant evidence that MNE precipitates multigenerational global DNA methylome deficits but lacks sufficient epigenomic resolution to evaluate the intergenerational transmissibility of locus-specific DNA methylome anomalies identified in the human literature on MS.
Figure 1.

Survey of human and animal model research on the intergenerational impacts of maternal smoking. Dual-gradient heatmaps depicting arbitrary measures of the strength of evidence (based on quantity of studies) for various categories of neurodevelopmental anomalies from human studies of maternal smoking (MS) and grandmaternal smoking (GMS) as well as animal model studies of maternal nicotine exposure (MNE) and grandmaternal nicotine exposure (GMNE).
Despite the abovementioned weaknesses, the cumulative evidence from human and animal model studies links MS and to a lesser extent GMS to an ensemble of brain and behavioral phenotypes consistent with transdiagnostic features of NDDs and suggests a putative intergenerational epigenetic mechanism thereof wherein nicotine from MS aberrantly stimulates embryonic neuronal and embryonic ovarian nAChRs and thereby elicits self-propagating biological and epigenomic alterations in the developing brain and primordial germ cells, respectively, which in turn precipitate an array of neurodevelopmental deficits in offspring and enable germline epigenetic transmission of such phenotypes to grandoffspring.
Future directions
Toward the resolution of a pressing void in the human literature, future human studies should endeavor to delineate whether and to what extent maternal and GMS are associated with NDD-related neurobiological perturbations such as cholinergic and catecholamine system dysfunction, disruption of neurotrophic signaling, and HPA axis hypofunction. Furthermore, given the implication of DNA methylome anomalies in the NDD-related brain and behavioral phenotypes exhibited by the children of maternal smokers, multigenerational human studies should be conducted to assess the transmissibility of MS-related DNA methylomic alterations to the grandchildren of maternal smokers. In addition to evaluating the intergenerational impacts of MS on the DNA methylome, longitudinal human studies capable of measuring within-individual changes in DNA methylation in a variety of tissues over time should be conducted to garner complementary insights into the intragenerational plasticity of DNA methylation [422–423]. Importantly, these and other future human studies would benefit greatly from considering and directly evaluating the differential impacts of exposure windows, dosage, genotype, and other often-overlooked variables.
On the other hand, future animal model research is warranted to address the need for high-resolution, epigenome-wide profiling of locus-specific differential DNA methylation signatures across discrete brain regions and distinct cell types, the results of which will elucidate the gene-, cell type-, and brain regional-specificity of MNE-evoked multigenerational epigenetic alterations and the relevance to NDDs thereof. Subsequent animal model studies should also assess the transgenerational transmissibility of NDD-related phenotypic aberrations conferred by MNE and the possible roles of DNA methylome perturbations therein.
It is important to acknowledge that little is known about how animal research in the intergenerational arena translates to the human condition or about how human and animal studies can best account for complex human variables such as polysubstance use, variable dosing, differing modes of exposure (for instance traditional versus electronic cigarette consumption), and environmental as well as genetic risk factors. As such, in addition to advancing conventional epidemiological and animal model lines of inquiry, future research should implement translational epigenomic study designs that coalesce human subject and animal model experiments in an effort to better address key questions concerning the intergenerational transmissibility of MS-related epigenomic perturbations and consequent transcriptomic alterations as well as the relationships thereof with NDD-like brain and behavioral phenotypes. Notably, translational epigenomic research is also better equipped to inform the development of novel treatments and prophylactics intended to partially ameliorate or even prevent the intergenerational neurodevelopmental consequences of MS.
From a public health perspective, prevention and cessation of MS are of the utmost importance. Nevertheless, for maternal smokers who are unwilling or unable to quit, emerging evidence suggests that dietary supplementation may confer modest mitigation of or protection against the impacts of MS on the DNA methylome and, by extension, the brain and behavior. For instance, maternal vitamin C supplementation has shown partial efficacy toward the prevention of MS-related DNA methylome perturbations in a randomized clinical trial, and there is reasonable evidence that maternal and offspring folate supplementation may confer similar benefits [424–428]. However, these findings are preliminary and more research in this domain is necessary. As such, care should be taken not to misinform maternal smokers about the possible benefits of dietary interventions. Ultimately, abstinence is the only means of preventing the deleterious intergenerational impacts of MS.
Acknowledgments
N/A
Footnotes
†Grant Support: Financial support was provided by the National Institutes of Health (DA040228; DA017637; DA023134; DA17671).
Contributor Information
Jordan M Buck, Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA; Department of Integrative Physiology, University of Colorado, Boulder, Boulder, CO, USA.
Li Yu, Department of Human Development and Family Studies, Purdue University, West Lafayette, IN, USA.
Valerie S Knopik, Department of Human Development and Family Studies, Purdue University, West Lafayette, IN, USA.
Jerry A Stitzel, Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA; Department of Integrative Physiology, University of Colorado, Boulder, Boulder, CO, USA.
Author contributions
JB, LY, VK, and JS contributed to the preparation and editing of the manuscript.
Conflict of interest
JB, LY, VK, and JS declare no potential or actual conflicts of interest.
Data availability
No new data were generated or analyzed in support of this manuscript.
References
- 1.Centers for Disease Control and Prevention . Pregnancy Risk Assessment Monitoring System. Atlanta, GA: US department of health and human services, centers for disease control and prevention; 2011; 201:2568–9. [Google Scholar]
- 2.Curtin SC, Matthews TJ. Smoking prevalence and cessation before and during pregnancy: Data from the birth certificate, 2014. National vital statistics reports: from the Centers for Disease Control and Prevention, National Center for Health Statistics, National Vital Statistics System 2016; 65:1–14. [PubMed] [Google Scholar]
- 3.Oncken C, Ricci KA, Kuo CL, Dornelas E, Kranzler HR, Sankey HZ. Correlates of electronic cigarettes use before and during pregnancy. Nicotine Tob Res 2017; 19:585–590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Lange S, Probst C, Rehm J, Popova S. National, regional, and global prevalence of smoking during pregnancy in the general population: A systematic review and meta-analysis. Lancet Glob Health 2018; 6:e769–e776. [DOI] [PubMed] [Google Scholar]
- 5.Kahr MK, Padgett S, Shope CD, Griffin EN, Xie SS, Gonzalez PJ et al. A qualitative assessment of the perceived risks of electronic cigarette and hookah use in pregnancy. BMC Public Health 2015; 15:1273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Nguyen KH, Tong VT, Marynak KL, King BA. US Adults' perceptions of the harmful effects during pregnancy of using electronic vapor products versus smoking cigarettes, styles survey, 2015. Prev Chronic Dis 2016; 13:E175–E175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.McCubbin A, Fallin-Bennett A, Barnett J, Ashford K. Perceptions and use of electronic cigarettes in pregnancy. Health Educ Res 2017; 32:22–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Wagner NJ, Camerota M, Propper C. Prevalence and perceptions of electronic cigarette use during pregnancy. MCHJ 2017; 21:1655–1661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Whittington JR, Simmons PM, Phillips AM, Gammill SK, Cen R, Magann EF, Cardenas VM. The use of electronic cigarettes in pregnancy: A review of the literature. Obstet Gynecol Surv 2018; 73:544–549. [DOI] [PubMed] [Google Scholar]
- 10.Salihu HM, Wilson RE. Epidemiology of prenatal smoking and perinatal outcomes. Early Hum Dev 2007; 83:713–720. [DOI] [PubMed] [Google Scholar]
- 11.Knopik VS, Marceau K, Palmer RH, Smith TF, Heath AC. Maternal smoking during pregnancy and offspring birth weight: A genetically-informed approach comparing multiple raters. Behav Genet 2016; 46:353–364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Jaddoe VW, Troe EJW, Hofman A, Mackenbach JP, Moll HA, Steegers EA, Witteman JC. Active and passive maternal smoking during pregnancy and the risks of low birthweight and preterm birth: The generation R study. Paediatr Perinat Epidemiol 2008; 22:162–171. [DOI] [PubMed] [Google Scholar]
- 13.Vardavas CI, Chatzi L, Patelarou E, Plana E, Sarri K, Kafatos A et al. Smoking and smoking cessation during early pregnancy and its effect on adverse pregnancy outcomes and fetal growth. Eur J Pediatr 2010; 169:741–748. [DOI] [PubMed] [Google Scholar]
- 14.Hackshaw A, Rodeck C, Boniface S. Maternal smoking in pregnancy and birth defects: A systematic review based on 173 687 malformed cases and 11.7 million controls. Hum Reprod Update 2011; 17:589–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Oster G, Delea TE, Colditz GA. Maternal smoking during pregnancy and expenditures on neonatal health care. Am J Prev Med 1988; 4:216–219. [PubMed] [Google Scholar]
- 16.Anderson TM, Ferres JML, Ren SY, Moon RY, Goldstein RD, Ramirez JM, Mitchell EA. Maternal smoking before and during pregnancy and the risk of sudden unexpected infant death. Pediatrics 2019; 143:e20183325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Higgins ST, Slade EP, Shepard DS. Decreasing smoking during pregnancy: Potential economic benefit of reducing sudden unexpected infant death. Prev Med 2020; 140:106238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Sciberras E, Streatfeild J, Ceccato T, Pezzullo L, Scott JG, Middeldorp CM et al. Social and economic costs of attention-deficit/hyperactivity disorder across the lifespan. J Atten Disord 2020; 2020:1087054720961828. [DOI] [PubMed] [Google Scholar]
- 19.Buescher AV, Cidav Z, Knapp M, Mandell DS. Costs of autism spectrum disorders in the United Kingdom and the United States. JAMA Pediatr 2014; 168:721–728. [DOI] [PubMed] [Google Scholar]
- 20.Cloutier M, Aigbogun MS, Guerin A, Nitulescu R, Ramanakumar AV, Kamat SA et al. The economic burden of schizophrenia in the United States in 2013. J Clin Psychiatry 2016; 77:764–771. [DOI] [PubMed] [Google Scholar]
- 21.Leigh JP, Du J. Brief report: Forecasting the economic burden of autism in 2015 and 2025 in the United States. J Autism Dev Disord 2015; 45:4135–4139. [DOI] [PubMed] [Google Scholar]
- 22.Knopik VS. Maternal smoking during pregnancy and child outcomes: Real or spurious effect? Dev Neuropsychol 2009; 34:1–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Knopik VS, Marceau K, Bidwell LC, Palmer RH, Smith TF, Todorov A et al. Smoking during pregnancy and ADHD risk: A genetically informed, multiple-rater approach. Am J Med Genet B Neuropsychiatr Genet 2016; 171:971–981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.He X, Lu J, Dong W, Jiao Z, Zhang C, Yu Y et al. Prenatal nicotine exposure induces HPA axis-hypersensitivity in offspring rats via the intrauterine programming of up-regulation of hippocampal GAD67. Arch Toxicol 2017; 91:3927–3943. [DOI] [PubMed] [Google Scholar]
- 25.Huang L, Wang Y, Zhang L, Zheng Z, Zhu T, Qu Y, Mu D. Maternal smoking and attention-deficit/hyperactivity disorder in offspring: A meta-analysis. Pediatrics 2018; 141:e20172465. [DOI] [PubMed] [Google Scholar]
- 26.Marceau K, Bidwell LC, Karoly HC, Evans AS, Todorov AA, Palmer RH et al. Within-family effects of smoking during pregnancy on ADHD: The importance of phenotype. J Abnorm Child Psychol 2018; 46:685–699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Baguelin-Pinaud A, Robert S, Ménard J-F, Thibaut F. Prenatal exposure to tobacco and risk for schizophrenia: A retrospective epidemiological study. Compr Psychiatry 2010; 51:106–109. [DOI] [PubMed] [Google Scholar]
- 28.Kalkbrenner AE, Braun JM, Durkin MS, Maenner MJ, Cunniff C, Lee LC et al. Maternal smoking during pregnancy and the prevalence of autism spectrum disorders, using data from the autism and developmental disabilities monitoring network. Environ Health Perspect 2012; 120:1042–1048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Tran PL, Lehti V, Lampi KM, Helenius H, Suominen A, Gissler M, Brown AS, Sourander A. Smoking during pregnancy and risk of autism spectrum disorder in a Finnish National Birth Cohort. Paediatr Perinat Epidemiol 2013; 27:266–274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Meier SM, Mors O, Parner E. Familial confounding of the association between maternal smoking during pregnancy and schizophrenia. Am J Psychiatry 2017; 174:187–187. [DOI] [PubMed] [Google Scholar]
- 31.Kollins SH, McClernon FJ, Fuemmeler BF. Association between smoking and attention-deficit/hyperactivity disorder symptoms in a population-based sample of young adults. Arch Gen Psychiatry 2005; 62:1142–1147. [DOI] [PubMed] [Google Scholar]
- 32.Thakur GA, Sengupta SM, Grizenko N, Schmitz N, Pagé V, Joober R. Maternal smoking during pregnancy and ADHD: A comprehensive clinical and neurocognitive characterization. Nicotine Tob Res 2013; 15:149–157. [DOI] [PubMed] [Google Scholar]
- 33.Ekblad MO, Rolan E, Marceau K, Palmer R, Todorov A, Heath AC, Knopik VS. Disruptive behavior in siblings discordant for exposure to maternal smoking during pregnancy: A multi-rater approach. Nicotine Tob Res 2020; 22:1330–1338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Ekblad MO, Marceau K, Rolan E, Palmer RHC, Todorov A, Heath AC, Knopik VS. The effect of smoking during pregnancy on severity and directionality of externalizing and internalizing symptoms: A genetically informed approach. Int J Environ Res Public Health 2020; 17:7921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Stathopoulou A, Beratis IN, Beratis S. Prenatal tobacco smoke exposure, risk of schizophrenia, and severity of positive/negative symptoms. Schizophr Res 2013; 148:105–110. [DOI] [PubMed] [Google Scholar]
- 36.Lyall K, Schmidt RJ, Hertz-Picciotto I. Maternal lifestyle and environmental risk factors for autism spectrum disorders. Int J Epidemiol 2014; 43:443–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Robbins LS, Blanchard CT, Sinkey RG, Harris SL, Tita AT, Harper LM. Prenatal tobacco exposure and childhood neurodevelopment among infants born prematurely. Am J Perinatol 2020; 38:218–223. [DOI] [PubMed] [Google Scholar]
- 38.Trasti N, Vik T, Jacobsen G, Bakketeig LS. Smoking in pregnancy and children’s mental and motor development at age 1 and 5 years. Early Hum Dev 1999; 55:137–147. [DOI] [PubMed] [Google Scholar]
- 39.Caramaschi D, Taylor AE, Richmond RC, Havdahl KA, Golding J, Relton CL, Munafò MR, Davey Smith G, Rai D. Maternal smoking during pregnancy and autism: Using causal inference methods in a birth cohort study. Transl Psychiatry 2018; 8:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Kalkbrenner AE, Meier SM, Madley-Dowd P, Ladd-Acosta C, Fallin MD, Parner E, Schendel D. Familial confounding of the association between maternal smoking in pregnancy and autism spectrum disorder in offspring. Autism Res 2020; 13:134–144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Quinn PD, Rickert ME, Weibull CE, Johansson AL, Lichtenstein P, Almqvist C et al. Association between maternal smoking during pregnancy and severe mental illness in offspring. JAMA Psychiat 2017; 74:589–596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Gustavson K, Ystrom E, Stoltenberg C, Susser E, Surén P, Magnus P, Knudsen GP, Smith GD, Langley K, Rutter M, Aase H, Reichborn-Kjennerud T. Smoking in pregnancy and child ADHD. Pediatrics 2017; 139:e20162509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.D’Onofrio BM, Lahey BB, Turkheimer E, Lichtenstein P. Critical need for family-based, quasi-experimental designs in integrating genetic and social science research. Am J Public Health 2013; 103:S46–S55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Allsopp K, Read J, Corcoran R, Kinderman P. Heterogeneity in psychiatric diagnostic classification. Psychiatry Res 2019; 279:15–22. [DOI] [PubMed] [Google Scholar]
- 45.Polanska K, Jurewicz J, Hanke W. Smoking and alcohol drinking during pregnancy as the risk factors for poor child neurodevelopment – A review of epidemiological studies. Int J Occup Med Environ Health 2015; 28:419. [DOI] [PubMed] [Google Scholar]
- 46.Kiechl-Kohlendorfer U, Ralser E, Pupp Peglow U, Reiter G, Griesmaier E, Trawöger R. Smoking in pregnancy: A risk factor for adverse neurodevelopmental outcome in preterm infants? Acta Paediatr 2010; 99:1016–1019. [DOI] [PubMed] [Google Scholar]
- 47.Kable JA, Coles CD, Lynch ME, Carroll J. The impact of maternal smoking on fast auditory brainstem responses. Neurotoxicol Teratol 2009; 31:216–224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Ediger K, Hasan SU, Synnes A, Shah J, Creighton D, Isayama T, Shah PS, Lodha A. Maternal smoking and neurodevelopmental outcomes in infants <29 weeks gestation: A multicenter cohort study. J Perinatol 2019; 39:791–799. [DOI] [PubMed] [Google Scholar]
- 49.Olds D. Intellectual impairment in children of women who smoke cigarettes during pregnancy (vol 93, pg 221, 1994). Pediatrics 1994; 93:973–973. [PubMed] [Google Scholar]
- 50.Leech SL, Richardson GA, Goldschmidt L, Day NL. Prenatal substance exposure: Effects on attention and impulsivity of 6-year-olds. Neurotoxicol Teratol 1999; 21:109–118. [DOI] [PubMed] [Google Scholar]
- 51.Kotimaa AJ, Moilanen I, Taanila A, Ebeling H, Smalley SL, Mcgough JJ et al. Maternal smoking and hyperactivity in 8-year-old children. J Am Acad Child Adolesc Psychiatry 2003; 42:826–833. [DOI] [PubMed] [Google Scholar]
- 52.Huijbregts SC, Séguin JR, Zoccolillo M, Boivin M, Tremblay RE. Associations of maternal prenatal smoking with early childhood physical aggression, hyperactivity-impulsivity, and their co-occurrence. J Abnorm Child Psychol 2007; 35:203–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Huijbregts SCJ, Seguin JR, Zoccolillo M, Boivin M, Tremblay RE. Maternal prenatal smoking, parental antisocial behavior, and early childhood physical aggression. Dev Psychopathol 2008; 20:437–453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Malanchini M, Smith-Woolley E, Ayorech Z, Rimfeld K, Krapohl E, Vuoksimaa E, Korhonen T, Bartels M, van Beijsterveldt TCEM, Rose RJ, Lundström S, Anckarsäter H et al. Aggressive behaviour in childhood and adolescence: The role of smoking during pregnancy, evidence from four twin cohorts in the EU-ACTION consortium. Psychol Med 2019; 49:646–654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Abbott LC, Winzer-Serhan UH. Smoking during pregnancy: Lessons learned from epidemiological studies and experimental studies using animal models. Crit Rev Toxicol 2012; 42:279–303. [DOI] [PubMed] [Google Scholar]
- 56.Cornelius MD, Day NL. Developmental consequences of prenatal tobacco exposure. Curr Opin Neurol 2009; 22:121–125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Fried PA, O’Connell CM, Watkinson B. 60- and 72-month follow-up of children prenatally exposed to marijuana, cigarettes, and alcohol: Cognitive and language assessment. J Dev Behav Pediatr 1992; 13:383–391. [PubMed] [Google Scholar]
- 58.Fried PA, Watkinson B, Gray R. A follow-up study of attentional behavior in 6-year-old children exposed prenatally to marihuana, cigarettes, and alcohol. Neurotoxicol Teratol 1992; 14:299–311. [DOI] [PubMed] [Google Scholar]
- 59.Herrmann M, King K, Weitzman M. Prenatal tobacco smoke and postnatal secondhand smoke exposure and child neurodevelopment. Curr Opin Pediatr 2008; 20:184–190. [DOI] [PubMed] [Google Scholar]
- 60.Cornelius MD, Ryan CM, Day NL, Goldschmidt L, Willford JA. Prenatal tobacco effects on neuropsychological outcomes among preadolescents. J Dev Behav Pediatr 2001; 22:217–225. [DOI] [PubMed] [Google Scholar]
- 61.Oh K, Xu Y, Terrizzi BF, Lanphear B, Chen A, Kalkbrenner AE, Yolton K. Associations between early low-level tobacco smoke exposure and executive function at age 8 years. J Pediatr 2020; 221:174–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Makin J, Fried PA, Watkinson B. A comparison of active and passive smoking during pregnancy: Long-term effects. Neurotoxicol Teratol 1991; 13:5–12. [DOI] [PubMed] [Google Scholar]
- 63.Lambe M, Hultman C, Torrång A, Mac Cabe J, Cnattingius S. Maternal smoking during pregnancy and school performance at age 15. Epidemiology 2006; 17:524–530. [DOI] [PubMed] [Google Scholar]
- 64.Mccartney JS, Fried PA, Watkinson B. Central auditory processing in school-age children prenatally exposed to cigarette smoke. Neurotoxicol Teratol 1994; 16:269–276. [DOI] [PubMed] [Google Scholar]
- 65.Sexton M, Fox NL, Hebel JR. Prenatal exposure to tobacco: II effects on cognitive functioning at age three. Int J Epidemiol 1990; 19:72–77. [DOI] [PubMed] [Google Scholar]
- 66.Batstra L, Hadders-Algra M, Neeleman J. Effect of antenatal exposure to maternal smoking on behavioural problems and academic achievement in childhood: Prospective evidence from a Dutch birth cohort. Early Hum Dev 2003; 75:21–33. [DOI] [PubMed] [Google Scholar]
- 67.Ernst M, Moolchan ET, Robinson ML. Behavioral and neural consequences of prenatal exposure to nicotine. J Am Acad Child Adolesc Psychiatry 2001; 40:630–641. [DOI] [PubMed] [Google Scholar]
- 68.Kollins SH, McClernon FJ, Fuemmeler BF. Association between smoking and attention-deficit/hyperactivity disorder symptoms in a population-based sample of young adults. Arch Gen Psychiatry 2005; 62:1142–1147. [DOI] [PubMed] [Google Scholar]
- 69.Elkins IJ, Saunders GR, Malone SM, Keyes MA, Samek DR, McGue M, Iacono WG. Increased risk of smoking in female adolescents who had childhood ADHD. Am J Psychiatry 2018; 175:63–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Cornelius MD, Leech SL, Goldschmidt L, Day NL. Prenatal tobacco exposure: Is it a risk factor for early tobacco experimentation? Nicotine Tob Res 2000; 2:45–52. [DOI] [PubMed] [Google Scholar]
- 71.Rhodes JD, Pelham WE, Gnagy EM, Shiffman S, Derefinko KJ, Molina BS. Cigarette smoking and ADHD: An examination of prognostically relevant smoking behaviors among adolescents and young adults. Psychol Addict Behav 2016; 30:588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Hallerbäck MU, Lugnegård T, Gillberg C. ADHD and nicotine use in schizophrenia or asperger syndrome: A controlled study. J Atten Disord 2014; 18:425–433. [DOI] [PubMed] [Google Scholar]
- 73.Fang Y, Wang W, Zhu C, Lin GN, Cheng Y, Zou J, Cui D. Use of tobacco in schizophrenia: A double-edged sword. Brain Behav 2019; 9:e01433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Williams JM, Ziedonis DM, Abanyie F, Steinberg ML, Foulds J, Benowitz NL. Increased nicotine and cotinine levels in smokers with schizophrenia and schizoaffective disorder is not a metabolic effect. Schizophr Res 2005; 79:323–335. [DOI] [PubMed] [Google Scholar]
- 75.Cornelius MD, Goldschmidt L, Day NL. Prenatal cigarette smoking: Long-term effects on young adult behavior problems and smoking behavior. Neurotoxicol Teratol 2012; 34:554–559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Buka SL, Shenassa ED, Niaura R. Elevated risk of tobacco dependence among offspring of mothers who smoked during pregnancy: A 30-year prospective study. Am J Psychiatry 2003; 160:1978–1984. [DOI] [PubMed] [Google Scholar]
- 77.Agrawal A, Scherrer JF, Grant JD, Sartor CE, Pergadia ML, Duncan AE et al. The effects of maternal smoking during pregnancy on offspring outcomes. Prev Med 2010; 50:13–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Castillo RJ, Carlat DJ, Millon T, Millon CM, Meagher S, Grossman S, American Psychiatric Association . Diagnostic and statistical manual of mental disorders. Washington, DC: American Psychiatric Association Press; 2007. [Google Scholar]
- 79.Baird AL, Coogan AN, Siddiqui A, Donev RM, Thome J. Adult attention-deficit hyperactivity disorder is associated with alterations in circadian rhythms at the behavioural, endocrine and molecular levels. Mol Psychiatry 2012; 17:988–995. [DOI] [PubMed] [Google Scholar]
- 80.Van Veen MM, Kooij JS, Boonstra AM, Gordijn MC, Van Someren EJ. Delayed circadian rhythm in adults with attention-deficit/hyperactivity disorder and chronic sleep-onset insomnia. Biol Psychiatry 2010; 67:1091–1096. [DOI] [PubMed] [Google Scholar]
- 81.Imeraj L, Sonuga-Barke E, Antrop I, Roeyers H, Wiersema R, Bal S, Deboutte D. Altered circadian profiles in attention-deficit/hyperactivity disorder: An integrative review and theoretical framework for future studies. Neurosci Biobehav Rev 2012; 36:1897–1919. [DOI] [PubMed] [Google Scholar]
- 82.Bijlenga D, Van Someren EJ, Gruber R, Bron TI, Kruithof IF, Spanbroek EC, Kooij JS. Body temperature, activity and melatonin profiles in adults with attention-deficit/hyperactivity disorder and delayed sleep: A case–control study. J Sleep Res 2013; 22:607–616. [DOI] [PubMed] [Google Scholar]
- 83.Snitselaar MA, Smits MG, van der Heijden KB, Spijker J. Sleep and circadian rhythmicity in adult ADHD and the effect of stimulants: A review of the current literature. J Atten Disord 2017; 21:14–26. [DOI] [PubMed] [Google Scholar]
- 84.Tureck K, Matson JL, Cervantes P, Turygin N. Autism severity as a predictor of inattention and impulsivity in toddlers. Dev Neurorehabil 2015; 18:285–289. [DOI] [PubMed] [Google Scholar]
- 85.Lecavalier L. Behavioral and emotional problems in young people with pervasive developmental disorders: Relative prevalence, effects of subject characteristics, and empirical classification. J Autism Dev Disord 2006; 36:1101–1114. [DOI] [PubMed] [Google Scholar]
- 86.Gadow KD, DeVincent CJ, Pomeroy J. ADHD symptom subtypes in children with pervasive developmental disorder. J Autism Dev Disord 2006; 36:271–283. [DOI] [PubMed] [Google Scholar]
- 87.Hu VW, Sarachana T, Kim KS, Nguyen A, Kulkarni S, Steinberg ME et al. Gene expression profiling differentiates autism case–controls and phenotypic variants of autism spectrum disorders: Evidence for circadian rhythm dysfunction in severe autism. Autism Res 2009; 2:78–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Yang Z, Matsumoto A, Nakayama K, Jimbo EF, Kojima K, Nagata KI et al. Circadian-relevant genes are highly polymorphic in autism spectrum disorder patients. Brain Dev 2016; 38:91–99. [DOI] [PubMed] [Google Scholar]
- 89.Arican I, Bass N, Neelam K, Wolfe K, McQuillin A, Giaroli G. Prevalence of attention deficit hyperactivity disorder symptoms in patients with schizophrenia. Acta Psychiatr Scand 2019; 139:89–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Kaladjian A, Jeanningros R, Azorin JM, Anton JL, Mazzola-Pomietto P. Impulsivity and neural correlates of response inhibition in schizophrenia. Psychol Med 2011; 41:291–299. [DOI] [PubMed] [Google Scholar]
- 91.Nolan KA, 'Angelo DD, Hoptman MJ. Self-report and laboratory measures of impulsivity in patients with schizophrenia or schizoaffective disorder and healthy controls. Psychiatry Res 2011; 187:301–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Wulff K, Dijk DJ, Middleton B, Foster RG, Joyce EM. Sleep and circadian rhythm disruption in schizophrenia. Br J Psychiatry 2012; 200:308–316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Johansson AS, Owe-Larsson B, Hetta J, Lundkvist GB. Altered circadian clock gene expression in patients with schizophrenia. Schizophr Res 2016; 174:17–23. [DOI] [PubMed] [Google Scholar]
- 94.Roza SJ, Verburg BO, Jaddoe VWV, Hofman A, Mackenbach JP, Steegers EAP, Witteman JCM, Verhulst FC, Tiemeier H. Effects of maternal smoking in pregnancy on prenatal brain development. The generation R study. Eur J Neurosci 2007; 25:611–617. [DOI] [PubMed] [Google Scholar]
- 95.Ekblad M, Korkeila J, Lehtonen L. Smoking during pregnancy affects foetal brain development. Acta Paediatr 2015; 104:12–18. [DOI] [PubMed] [Google Scholar]
- 96.Shuffrey LC, Myers MM, Isler JR, Lucchini M, Sania A, Pini N, Nugent JD, Condon C, Ochoa T, Brink L, du Plessis C, Odendaal HJ et al. Association between prenatal exposure to alcohol and tobacco and neonatal brain activity: Results from the safe passage study. JAMA Netw Open 2020; 3:e204714–e204714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Gutvirtz G, Wainstock T, Landau D, Sheiner E. Maternal smoking during pregnancy and long-term neurological morbidity of the offspring. Addict Behav 2019; 88:86–91. [DOI] [PubMed] [Google Scholar]
- 98.Chatterton Z, Hartley BJ, Seok MH, Mendelev N, Chen S, Milekic M et al. In utero exposure to maternal smoking is associated with DNA methylation alterations and reduced neuronal content in the developing fetal brain. Epigenetics Chromatin 2017; 10:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.El Marroun H, Schmidt MN, Franken IH, Jaddoe VW, Hofman A, Van Der Lugt A et al. Prenatal tobacco exposure and brain morphology: A prospective study in young children. Neuropsychopharmacology 2014; 39:792–800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Müller KU, Mennigen E, Ripke S, Banaschewski T, Barker GJ, Büchel C et al. Altered reward processing in adolescents with prenatal exposure to maternal cigarette smoking. JAMA Psychiat 2013; 70:847–856. [DOI] [PubMed] [Google Scholar]
- 101.Mooney MA, Bhatt P, Hermosillo RJ, Ryabinin P, Nikolas M, Faraone SV, Nigg JT. Smaller total brain volume but not subcortical structure volume related to common genetic risk for ADHD. Psychol Med 2020; 59:1279–1288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Veijola J, Guo JY, Moilanen JS, Jääskeläinen E, Miettunen J, Kyllönen M et al. Longitudinal changes in total brain volume in schizophrenia: Relation to symptom severity, cognition and antipsychotic medication. PLoS One 2014; 9:e101689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Rimol LM, Nesvåg R, Hagler DJ Jr, Bergmann Ø, Fennema-Notestine C, Hartberg CB et al. Cortical volume, surface area, and thickness in schizophrenia and bipolar disorder. Biol Psychiatry 2012; 71:552–560. [DOI] [PubMed] [Google Scholar]
- 104.Libero LE, DeRamus TP, Deshpande HD, Kana RK. Surface-based morphometry of the cortical architecture of autism spectrum disorders: Volume, thickness, area, and gyrification. Neuropsychologia 2014; 62:1–10. [DOI] [PubMed] [Google Scholar]
- 105.Silk TJ, Beare R, Malpas C, Adamson C, Vilgis V, Vance A, Bellgrove MA. Cortical morphometry in attention deficit/hyperactivity disorder: Contribution of thickness and surface area to volume. Cortex 2016; 82:1–10. [DOI] [PubMed] [Google Scholar]
- 106.Martinez-Morga M, Quesada-Rico MP, Bueno C, Martinez S. Neurobiological bases of autistic spectrum disorder and attention deficit hyperactivity disorder: Neural differentiation and synaptogenesis. Rev Neurol 2018; 66:S97–S102. [PubMed] [Google Scholar]
- 107.Robicsek O, Karry R, Petit I, Salman-Kesner N, Müller FJ, Klein E et al. Abnormal neuronal differentiation and mitochondrial dysfunction in hair follicle-derived induced pluripotent stem cells of schizophrenia patients. Mol Psychiatry 2013; 18:1067–1076. [DOI] [PubMed] [Google Scholar]
- 108.Hellström-Lindahl E, Gorbounova O, Seiger Å, Mousavi M, Nordberg A. Regional distribution of nicotinic receptors during prenatal development of human brain and spinal cord. Developmental brain research 1998; 108:147–160. [DOI] [PubMed] [Google Scholar]
- 109.Cairns NJ, Wonnacott S. [3H](−) nicotine binding sites in fetal human brain. Brain Res 1988; 475:1–7. [DOI] [PubMed] [Google Scholar]
- 110.Agulhon C, Charnay Y, Vallet P, Bertrand D, Malafosse A. Distribution of mRNA for the α4 subunit of the nicotinic acetylcholine receptor in the human fetal brain. Mol Brain Res 1998; 58:123–131. [DOI] [PubMed] [Google Scholar]
- 111.Falk L, Nordberg A, Seiger Å, Kjaeldgaard A, Hellström-Lindahl E. Smoking during early pregnancy affects the expression pattern of both nicotinic and muscarinic acetylcholine receptors in human first trimester brainstem and cerebellum. Neuroscience 2005; 132:389–397. [DOI] [PubMed] [Google Scholar]
- 112.Abreu-Villaça Y, Filgueiras CC, Manhães AC. Developmental aspects of the cholinergic system. Behav Brain Res 2011; 221:367–378. [DOI] [PubMed] [Google Scholar]
- 113.Drenan RM, Grady SR, Steele AD, McKinney S, Patzlaff NE, McIntosh JM et al. Cholinergic modulation of locomotion and striatal dopamine release is mediated by α6α4* nicotinic acetylcholine receptors. J Neurosci 2010; 30:9877–9889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Cohen BN, Mackey ED, Grady SR, McKinney S, Patzlaff NE, Wageman CR et al. Nicotinic cholinergic mechanisms causing elevated dopamine release and abnormal locomotor behavior. Neuroscience 2012; 200:31–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Potter AS, Bucci DJ, Newhouse PA. Manipulation of nicotinic acetylcholine receptors differentially affects behavioral inhibition in human subjects with and without disordered baseline impulsivity. Psychopharmacology (Berl) 2012; 220:331–340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Ohmura Y, Tsutsui-Kimura I, Yoshioka M. Impulsive behavior and nicotinic acetylcholine receptors. J Pharmacol Sci 2012; 118:413–422. [DOI] [PubMed] [Google Scholar]
- 117.Tsutsui-Kimura I, Ohmura Y, Izumi T, Yamaguchi T, Yoshida T, Yoshioka M. Endogenous acetylcholine modulates impulsive action via α4β2 nicotinic acetylcholine receptors in rats. Eur J Pharmacol 2010; 641:148–153. [DOI] [PubMed] [Google Scholar]
- 118.Lavezzi AM. Toxic effect of cigarette smoke on brainstem nicotinic receptor expression: Primary cause of sudden unexplained perinatal death. Toxics 2018; 6:63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Machaalani R, Ghazavi E, Hinton T, Waters KA, Hennessy A. Cigarette smoking during pregnancy regulates the expression of specific nicotinic acetylcholine receptor (nAChR) subunits in the human placenta. Toxicol Appl Pharmacol 2014; 276:204–212. [DOI] [PubMed] [Google Scholar]
- 120.Alwazzan A, Mehboob R, Gilani SA, Hassan A, Perveen S, Tanvir I et al. Immunohistochemical expression of the alpha nicotinic acetylcholine receptor 7 in the human normal, diabetic, and Preeclamptic placenta and products of conception. Front Physiol 2020; 11:607239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Wallis D, Arcos-Burgos M, Jain M, Castellanos FX, Palacio JD, Pineda D et al. Polymorphisms in the neural nicotinic acetylcholine receptor α4 subunit (CHRNA4) are associated with ADHD in a genetic isolate. ADHD Attention Deficit and Hyperactivity Disorders 2009; 1:19–24. [DOI] [PubMed] [Google Scholar]
- 122.Lewis AS, Picciotto MR. High-affinity nicotinic acetylcholine receptor expression and trafficking abnormalities in psychiatric illness. Psychopharmacology (Berl) 2013; 229:477–485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Wigestrand MB, Mineur YS, Heath CJ, Fonnum F, Picciotto MR, Walaas SI. Decreased α4β2 nicotinic receptor number in the absence of mRNA changes suggests post-transcriptional regulation in the spontaneously hypertensive rat model of ADHD. J Neurochem 2011; 119:240–250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Bacchelli E, Battaglia A, Cameli C, Lomartire S, Tancredi R, Thomson S et al. Analysis of CHRNA7 rare variants in autism spectrum disorder susceptibility. Am J Med Genet A 2015; 167:715–723. [DOI] [PubMed] [Google Scholar]
- 125.Ray MA, Graham AJ, Lee M, Perry RH, Court JA, Perry EK. Neuronal nicotinic acetylcholine receptor subunits in autism: An immunohistochemical investigation in the thalamus. Neurobiol Dis 2005; 19:366–377. [DOI] [PubMed] [Google Scholar]
- 126.Martin-Ruiz CM, Lee M, Perry RH, Baumann M, Court JA, Perry EK. Molecular analysis of nicotinic receptor expression in autism. Mol Brain Res 2004; 123:81–90. [DOI] [PubMed] [Google Scholar]
- 127.D'Souza DC, Esterlis I, Carbuto M, Krasenics M, Seibyl J, Bois F et al. Lower β2*-nicotinic acetylcholine receptor availability in smokers with schizophrenia. Am J Psychiatry 2012; 169:326–334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Leonard S, Breese C, Adams C, Benhammou K, Gault J, Stevens K et al. Smoking and schizophrenia: Abnormal nicotinic receptor expression. Eur J Pharmacol 2000; 393:237–242. [DOI] [PubMed] [Google Scholar]
- 129.Guan ZZ, Zhang X, Blennow K, Nordberg A. Decreased protein level of nicotinic receptor α7 subunit in the frontal cortex from schizophrenic brain. Neuroreport 1999; 10:1779–1782. [DOI] [PubMed] [Google Scholar]
- 130.Stephens SH, Logel J, Barton A, Franks A, Schultz J, Short M et al. Association of the 5′-upstream regulatory region of the α7 nicotinic acetylcholine receptor subunit gene (CHRNA7) with schizophrenia. Schizophr Res 2009; 109:102–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Saboory E, Ghasemi M, Mehranfard N. Norepinephrine, neurodevelopment and behavior. Neurochem Int 2020; 135:104706. [DOI] [PubMed] [Google Scholar]
- 132.van Kesteren RE, Spencer GE. The role of neurotransmitters in neurite outgrowth and synapse formation. Rev Neurosci 2003; 14:217–231. [DOI] [PubMed] [Google Scholar]
- 133.Spencer GE, Klumperman J, Syed NI. Neurotransmitters and neurodevelopment. Role of dopamine in neurite outgrowth, target selection and specific synapse formation. Perspect Dev Neurobiol 1998; 5:451–467. [PubMed] [Google Scholar]
- 134.Antelman SM, Caggiula AR. Norepinephrine-dopamine interactions and behavior. Science 1977; 195:646–653. [DOI] [PubMed] [Google Scholar]
- 135.Oncken CA, Henry KM, Campbell WA, Kuhn CM, Slotkin TA, Kranzler HR. Effect of maternal smoking on fetal catecholamine concentrations at birth. Pediatr Res 2003; 53:119–124. [DOI] [PubMed] [Google Scholar]
- 136.Thakur GA, Sengupta SM, Grizenko N, Choudhry Z, Joober R. Comprehensive phenotype/genotype analyses of the norepinephrine transporter gene (SLC6A2) in ADHD: Relation to maternal smoking during pregnancy. PLoS One 2012; 7:e49616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Neuman RJ, Lobos E, Reich W, Henderson CA, Sun LW, Todd RD. Prenatal smoking exposure and dopaminergic genotypes interact to cause a severe ADHD subtype. Biol Psychiatry 2007; 61:1320–1328. [DOI] [PubMed] [Google Scholar]
- 138.Kahn RS, Khoury J, Nichols WC, Lanphear BP. Role of dopamine transporter genotype and maternal prenatal smoking in childhood hyperactive-impulsive, inattentive, and oppositional behaviors. J Pediatr 2003; 143:104–110. [DOI] [PubMed] [Google Scholar]
- 139.Volkow ND, Wang GJ, Newcorn JH, Kollins SH, Wigal TL, Telang F et al. Motivation deficit in ADHD is associated with dysfunction of the dopamine reward pathway. Mol Psychiatry 2011; 16:1147–1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Wu J, Xiao H, Sun H, Zou L, Zhu LQ. Role of dopamine receptors in ADHD: A systematic meta-analysis. Mol Neurobiol 2012; 45:605–620. [DOI] [PubMed] [Google Scholar]
- 141.Bédard AC, Schulz KP, Cook EH Jr, Fan J, Clerkin SM, Ivanov I et al. Dopamine transporter gene variation modulates activation of striatum in youth with ADHD. Neuroimage 2010; 53:935–942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Hesse S, Ballaschke O, Barthel H, von Cramon DY, Sabri O. The striatal dopamine transporter availability is reduced in adults with attention-deficit/hyperactivity disorder. J Nucl Med 2006; 47:142P–142P. [Google Scholar]
- 143.Spencer TJ, Biederman J, Madras BK, Dougherty DD, Bonab AA, Livni E et al. Further evidence of dopamine transporter dysregulation in ADHD: A controlled PET imaging study using altropane. Biol Psychiatry 2007; 62:1059–1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Heijtz RD, Kolb B, Forssberg H. Motor inhibitory role of dopamine D1 receptors: Implications for ADHD. Physiol Behav 2007; 92:155–160. [DOI] [PubMed] [Google Scholar]
- 145.Jucaite A, Fernell E, Halldin C, Forssberg H, Farde L. Reduced midbrain dopamine transporter binding in male adolescents with attention-deficit/hyperactivity disorder: Association between striatal dopamine markers and motor hyperactivity. Biol Psychiatry 2005; 57:229–238. [DOI] [PubMed] [Google Scholar]
- 146.Bobb AJ, Addington AM, Sidransky E, Gornick MC, Lerch JP, Greenstein DK et al. Support for association between ADHD and two candidate genes: NET1 and DRD1. Am J Med Genet B Neuropsychiatr Genet 2005; 134:67–72. [DOI] [PubMed] [Google Scholar]
- 147.Lou HC, Rosa P, Pryds O, Karrebæk H, Lunding J, Cumming P, Gjedde A. ADHD: Increased dopamine receptor availability linked to attention deficit and low neonatal cerebral blood flow. Dev Med Child Neurol 2004; 46:179–183. [DOI] [PubMed] [Google Scholar]
- 148.Lee Y, Kim H, Kim JE, Park JY, Choi J, Lee JE et al. Excessive D1 dopamine receptor activation in the dorsal striatum promotes autistic-like behaviors. Mol Neurobiol 2018; 55:5658–5671. [DOI] [PubMed] [Google Scholar]
- 149.Angelakos CC, Watson AJ, O'Brien WT, Krainock KS, Nickl-Jockschat T, Abel T. Hyperactivity and male-specific sleep deficits in the 16p11. 2 deletion mouse model of autism. Autism Res 2017; 10:572–584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Bowton E, Saunders C, Reddy IA, Campbell NG, Hamilton PJ, Henry LK et al. SLC6A3 coding variant Ala559Val found in two autism probands alters dopamine transporter function and trafficking. Transl Psychiatry 2014; 4:e464–e464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Rogers TD, Dickson PE, McKimm E, Heck DH, Goldowitz D, Blaha CD, Mittleman G. Reorganization of circuits underlying cerebellar modulation of prefrontal cortical dopamine in mouse models of autism spectrum disorder. The Cerebellum 2013; 12:547–556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Staal WG, de Krom M, de Jonge MV. Brief report: The dopamine-3-receptor gene (DRD3) is associated with specific repetitive behavior in autism spectrum disorder (ASD). J Autism Dev Disord 2012; 42:885–888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Nakasato A, Nakatani Y, Seki Y, Tsujino N, Umino M, Arita H. Swim stress exaggerates the hyperactive mesocortical dopamine system in a rodent model of autism. Brain Res 2008; 1193:128–135. [DOI] [PubMed] [Google Scholar]
- 154.Mittleman G, Goldowitz D, Heck DH, Blaha CD. Cerebellar modulation of frontal cortex dopamine efflux in mice: Relevance to autism and schizophrenia. Synapse 2008; 62:544–550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Ernst M, Zametkin AJ, Matochik JA, Pascualvaca D, Cohen RM. Low medial prefrontal dopaminergic activity in autistic children. The Lancet 1997; 350:638. [DOI] [PubMed] [Google Scholar]
- 156.Maia TV, Frank MJ. An integrative perspective on the role of dopamine in schizophrenia. Biol Psychiatry 2017; 81:52–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Hamilton PJ, Campbell NG, Sharma S, Erreger K, Hansen FH, Saunders C et al. De novo mutation in the dopamine transporter gene associates dopamine dysfunction with autism spectrum disorder. Mol Psychiatry 2013; 18:1315–1323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Winterer G. Prefrontal dopamine signaling in schizophrenia-the corticocentric model. Pharmacopsychiatry 2007; 40:S45–S53. [Google Scholar]
- 159.Schmitt GJE, Frodl T, Dresel S, La Fougère C, Bottlender R, Koutsouleris N et al. Striatal dopamine transporter availability is associated with the productive psychotic state in first episode, drug–naive schizophrenic patients. Eur Arch Psychiatry Clin Neurosci 2006; 256:115–121. [DOI] [PubMed] [Google Scholar]
- 160.Baracskay KL, Haroutunian V, Meador-Woodruff JH. Dopamine receptor signaling molecules are altered in elderly schizophrenic cortex. Synapse 2006; 60:271–279. [DOI] [PubMed] [Google Scholar]
- 161.Glenthoj BY, Mackeprang T, Svarer C, Rasmussen H, Pinborg LH, Friberg L et al. Frontal dopamine D2/3 receptor binding in drug-naive first-episode schizophrenic patients correlates with positive psychotic symptoms and gender. Biol Psychiatry 2006; 60:621–629. [DOI] [PubMed] [Google Scholar]
- 162.Meyer-Lindenberg A, Miletich RS, Kohn PD, Esposito G, Carson RE, Quarantelli M et al. Reduced prefrontal activity predicts exaggerated striatal dopaminergic function in schizophrenia. Nat Neurosci 2002; 5:267–271. [DOI] [PubMed] [Google Scholar]
- 163.Laakso A, Bergman J, Haaparanta M, Vilkman H, Solin O, Syvälahti E, Hietala J. Decreased striatal dopamine transporter binding in vivo in chronic schizophrenia. Schizophr Res 2001; 52:115–120. [DOI] [PubMed] [Google Scholar]
- 164.Abi-Dargham A, Rodenhiser J, Printz D, Zea-Ponce Y, Gil R, Kegeles LS et al. Increased baseline occupancy of D2 receptors by dopamine in schizophrenia. Proc Natl Acad Sci 2000; 97:8104–8109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Stefanis NC, Bresnick JN, Kerwin RW, Schofield WN, McAllister G. Elevation of D4 dopamine receptor mRNA in postmortem schizophrenic brain. Mol Brain Res 1998; 53:112–119. [DOI] [PubMed] [Google Scholar]
- 166.Bast N, Poustka L, Freitag CM. The locus coeruleus–norepinephrine system as pacemaker of attention–a developmental mechanism of derailed attentional function in autism spectrum disorder. Eur J Neurosci 2018; 47:115–125. [DOI] [PubMed] [Google Scholar]
- 167.Lake CR, Ziegler MG, Murphy DL. Increased norepinephrine levels and decreased dopamine-β-hydroxylase activity in primary autism. Arch Gen Psychiatry 1977; 34:553–556. [DOI] [PubMed] [Google Scholar]
- 168.Barr CL, Kroft J, Feng Y, Wigg K, Roberts W, Malone M et al. The norepinephrine transporter gene and attention-deficit hyperactivity disorder. Am J Med Genet 2002; 114:255–259. [DOI] [PubMed] [Google Scholar]
- 169.Sigurdardottir HL, Kranz GS, Rami-Mark C, James GM, Vanicek T, Gryglewski G, Lanzenberger R. Association of norepinephrine transporter methylation with in vivo NET expression and hyperactivity–impulsivity symptoms in ADHD measured with PET. Mol Psychiatry 2019; 26:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Sigurdardottir HL, Kranz GS, Rami-Mark C, James GM, Vanicek T, Gryglewski G et al. Effects of norepinephrine transporter gene variants on NET binding in ADHD and healthy controls investigated by PET. Hum Brain Mapp 2016; 37:884–895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Sengupta SM, Grizenko N, Thakur GA, Bellingham J, DeGuzman R, Robinson S et al. Differential association between the norepinephrine transporter gene and ADHD: Role of sex and subtype. Journal of psychiatry & neuroscience: JPN 2012; 37:129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Breier A, Wolkowitz OM, Roy A, Potter WZ, Pickar D. Plasma norepinephrine in chronic schizophrenia. Am J Psychiatry 1990; 147:1467–1470. [DOI] [PubMed] [Google Scholar]
- 173.Mäki-Marttunen V, Andreassen OA, Espeseth T. The role of norepinephrine in the pathophysiology of schizophrenia. Neurosci Biobehav Rev 2020; 118:298–314. [DOI] [PubMed] [Google Scholar]
- 174.Farley IJ, Price KS, McCullough E, Deck JH, Hordynski W, Hornykiewicz O. Norepinephrine in chronic paranoid schizophrenia: Above-normal levels in limbic forebrain. Science 1978; 200:456–458. [DOI] [PubMed] [Google Scholar]
- 175.Launay JM, Bursztejn C, Ferrari P, Dreux C, Braconnier A, Zarifian E et al. Catecholamines metabolism in infantile autism: A controlled study of 22 autistic children. J Autism Dev Disord 1987; 17:333–347. [DOI] [PubMed] [Google Scholar]
- 176.D’Angelo A, Ceccanti M, Petrella C, Greco A, Tirassa P, Rosso P et al. Role of neurotrophins in pregnancy, delivery and postpartum. Eur J Obstet Gynecol Reprod Biol 2020; 247:32–41. [DOI] [PubMed] [Google Scholar]
- 177.Sahay AS, Sundrani DP, Joshi SR. Neurotrophins: Role in placental growth and development. Vitam Horm 2017; 104:243–261. [DOI] [PubMed] [Google Scholar]
- 178.Huang EJ, Reichardt LF. Neurotrophins: Roles in neuronal development and function. Annu Rev Neurosci 2001; 24:677–736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Corominas-Roso M, Ramos-Quiroga JA, Ribases M, Sanchez-Mora C, Palomar G, Valero S et al. Decreased serum levels of brain-derived neurotrophic factor in adults with attention-deficit hyperactivity disorder. Int J Neuropsychopharmacol 2013; 16:1267–1275. [DOI] [PubMed] [Google Scholar]
- 180.Autry AE, Monteggia LM. Brain-derived neurotrophic factor and neuropsychiatric disorders. Pharmacol Rev 2012; 64:238–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.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; 52:1467–1476. [DOI] [PubMed] [Google Scholar]
- 182.Garcia KL, Yu G, Nicolini C, Michalski B, Garzon DJ, Chiu VS et al. Altered balance of proteolytic isoforms of pro-brain-derived neurotrophic factor in autism. J Neuropathol Exp Neurol 2012; 71:289–297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Pei Y, Jiao Z, Dong W, Pei L, He X, Wang H, Xu D. Excitotoxicity and compensatory upregulation of GAD67 in fetal rat hippocampus caused by prenatal nicotine exposure are associated with inhibition of the BDNF pathway. Food Chem Toxicol 2019; 123:314–325. [DOI] [PubMed] [Google Scholar]
- 184.Ethell IM, Ethell DW. Matrix metalloproteinases in brain development and remodeling: Synaptic functions and targets. J Neurosci Res 2007; 85:2813–2823. [DOI] [PubMed] [Google Scholar]
- 185.Orefice LL, Shih CC, Xu H, Waterhouse EG, Xu B. Control of spine maturation and pruning through proBDNF synthesized and released in dendrites. Mol Cell Neurosci 2016; 71:66–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Menshanov PN, Lanshakov DA, Dygalo NN. proBDNF is a major product of bdnf gene expressed in the perinatal rat cortex. Physiol Res 2015; 64:925–934. [DOI] [PubMed] [Google Scholar]
- 187.Li JY, Liu J, Manaph NPA, Bobrovskaya L, Zhou XF. ProBDNF inhibits proliferation, migration and differentiation of mouse neural stem cells. Brain Res 2017; 1668:46–55. [DOI] [PubMed] [Google Scholar]
- 188.Yang J, Harte-Hargrove LC, Siao CJ, Marinic T, Clarke R, Ma Q et al. proBDNF negatively regulates neuronal remodeling, synaptic transmission, and synaptic plasticity in hippocampus. Cell Rep 2014; 7:796–806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Guo J, Ji Y, Ding Y, Jiang W, Sun Y, Lu B, Nagappan G. BDNF pro-peptide regulates dendritic spines via caspase-3. Cell Death Dis 2016; 7:e2264–e2264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Zhu Y, Jiang X, Ji W. The mechanism of cortico-striato-thalamo-cortical neurocircuitry in response inhibition and emotional responding in attention deficit hyperactivity disorder with comorbid disruptive behavior disorder. Neurosci Bull 2018; 34:566–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Wetsel WC, Rodriguiz RM, Guillemot J, Rousselet E, Essalmani R, Kim IH et al. Disruption of the expression of the proprotein convertase PC7 reduces BDNF production and affects learning and memory in mice. Proc Natl Acad Sci 2013; 110:17362–17367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Bai YY, Ruan CS, Yang CR, Li JY, Kang ZL, Zhou L et al. ProBDNF signaling regulates depression-like behaviors in rodents under chronic stress. Neuropsychopharmacology 2016; 41:2882–2892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Hong EJ, McCord AE, Greenberg ME. A biological function for the neuronal activity-dependent component of Bdnf transcription in the development of cortical inhibition. Neuron 2008; 60:610–624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Sakata K, Woo NH, Martinowich K, Greene JS, Schloesser RJ, Shen L, Lu B. Critical role of promoter IV-driven BDNF transcription in GABAergic transmission and synaptic plasticity in the prefrontal cortex. Proc Natl Acad Sci 2009; 106:5942–5947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Talati A, Odgerel Z, Wickramaratne PJ, Weissman MM. Brain derived neurotrophic factor moderates associations between maternal smoking during pregnancy and offspring behavioral disorders. Psychiatry Res 2016; 245:387–391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Toledo-Rodriguez M, Lotfipour S, Leonard G, Perron M, Richer L, Veillette S et al. Maternal smoking during pregnancy is associated with epigenetic modifications of the brain-derived neurotrophic factor-6 exon in adolescent offspring. Am J Med Genet B Neuropsychiatr Genet 2010; 153:1350–1354. [DOI] [PubMed] [Google Scholar]
- 197.Spulber S, Rantamäki T, Nikkilä O, Castrén E, Weihe P, Grandjean P, Ceccatelli S. Effects of maternal smoking and exposure to methylmercury on brain-derived neurotrophic factor concentrations in umbilical cord serum. Toxicol Sci 2010; 117:263–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Paquette AG, Lester BM, Lesseur C, Armstrong DA, Guerin DJ, Appleton AA, Marsit CJ. Placental epigenetic patterning of glucocorticoid response genes is associated with infant neurodevelopment. Epigenomics 2015; 7:767–779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.2 Wang C, Shen M, Guillaume B, Chong YS, Chen H, Fortier MV et al. FKBP5 moderates the association between antenatal maternal depressive symptoms and neonatal brain morphology. Neuropsychopharmacology 2018; 43:564–570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Zheng D, Sabbagh JJ, Blair LJ, Darling AL, Wen X, Dickey CA. MicroRNA-511 binds to FKBP5 mRNA, which encodes a chaperone protein, and regulates neuronal differentiation. J Biol Chem 2016; 291:17897–17906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Cruceanu C, Dony L, Krontira AC, Fischer DS, Roeh S, Di Giaimo R, Binder EB. Cell-type specific impact of glucocorticoid receptor activation on the developing brain. bioRxiv 2020, January 09, 2020. 10.1101/2020.01.09.897868 preprint: not peer reviewed. [DOI] [PubMed] [Google Scholar]
- 202.Qiu B, Xu Y, Wang J, Liu M, Dou L, Deng R et al. Loss of FKBP5 affects neuron synaptic plasticity: An electrophysiology insight. Neuroscience 2019; 402:23–36. [DOI] [PubMed] [Google Scholar]
- 203.Challet E, Le Maho Y, Robin JP, Malan A. Involvement of corticosterone in the fasting-induced rise in protein utilization and locomotor activity. Pharmacol Biochem Behav 1995; 50:405–412. [DOI] [PubMed] [Google Scholar]
- 204.Watanabe K, Hiroshige T. Phase relation between episodic fluctuations of spontaneous locomotor acitivty and plasma corticosterone in rats with Suprachiasmatic nuclei lesions. Neuroendocrinology 1981; 33:52–59. [DOI] [PubMed] [Google Scholar]
- 205.Binder EB. The role of FKBP5, a co-chaperone of the glucocorticoid receptor in the pathogenesis and therapy of affective and anxiety disorders. Psychoneuroendocrinology 2009; 34:S186–S195. [DOI] [PubMed] [Google Scholar]
- 206.Zannas AS, Wiechmann T, Gassen NC, Binder EB. Gene–stress–epigenetic regulation of FKBP5: Clinical and translational implications. Neuropsychopharmacology 2016; 41:261–274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.O’Leary III, John C, Zhang B, Koren III, Blair L, Dickey A, C. The role of FKBP5 in mood disorders: Action of FKBP5 on steroid hormone receptors leads to questions about its evolutionary importance. CNS & Neurological Disorders-Drug Targets (Formerly Current Drug Targets-CNS & Neurological Disorders) 2013; 12:1157–1162. [PMC free article] [PubMed] [Google Scholar]
- 208.Patel N, Crider A, Pandya CD, Ahmed AO, Pillai A. Altered mRNA levels of glucocorticoid receptor, mineralocorticoid receptor, and co-chaperones (FKBP5 and PTGES3) in the middle frontal gyrus of autism spectrum disorder subjects. Mol Neurobiol 2016; 53:2090–2099. [DOI] [PubMed] [Google Scholar]
- 209.Ma L, Chen YH, Chen H, Liu YY, Wang YX. The function of hypothalamus–pituitary–adrenal axis in children with ADHD. Brain Res 2011; 1368:159–162. [DOI] [PubMed] [Google Scholar]
- 210.McDonald SD, Walker M, Perkins SL, Beyene J, Murphy K, Gibb W, Ohlsson A. The effect of tobacco exposure on the fetal hypothalamic–pituitary–adrenal axis. BJOG 2006; 113:1289–1295. [DOI] [PubMed] [Google Scholar]
- 211.Dušková M, Hruškovičová H, Šimůnková K, Starka L, Pařízek A. The effects of smoking on steroid metabolism and fetal programming. J Steroid Biochem Mol Biol 2014; 139:138–143. [DOI] [PubMed] [Google Scholar]
- 212.Haslinger C, Bamert H, Rauh M, Burkhardt T, Schäffer L. Effect of maternal smoking on stress physiology in healthy neonates. J Perinatol 2018; 38:132–136. [DOI] [PubMed] [Google Scholar]
- 213.Stroud LR, Papandonatos GD, Shenassa E, Rodriguez D, Niaura R, LeWinn KZ et al. Prenatal glucocorticoids and maternal smoking during pregnancy independently program adult nicotine dependence in daughters: A 40-year prospective study. Biol Psychiatry 2014; 75:47–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Golding J, Ellis G, Gregory S, Birmingham K, Iles-Caven Y, Rai D, Pembrey M. Grand-maternal smoking in pregnancy and grandchild’s autistic traits and diagnosed autism. Sci Rep 2017; 7:46179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.England LJ, Aagaard K, Bloch M, Conway K, Cosgrove K, Grana R, Gould TJ, Hatsukami D, Jensen F, Kandel D, Lanphear B, Leslie F et al. Developmental toxicity of nicotine: A transdisciplinary synthesis and implications for emerging tobacco products. Neurosci Biobehav Rev 2017; 72:176–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Cooper S, Orton S, Leonardi-Bee J, Brotherton E, Vanderbloemen L, Bowker K et al. Smoking and quit attempts during pregnancy and postpartum: A longitudinal UK cohort. BMJ Open 2017; 7:e018746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Linnet KM, Dalsgaard S, Obel C, Wisborg K, Henriksen TB, Rodriguez A et al. Maternal lifestyle factors in pregnancy risk of attention deficit hyperactivity disorder and associated behaviors: Review of the current evidence. Am J Psychiatry 2003; 160:1028–1040. [DOI] [PubMed] [Google Scholar]
- 218.Ajarem JS, Ahmad M. Prenatal nicotine exposure modifies behavior of mice through early development. Pharmacol Biochem Behav 1998; 59:313–318. [DOI] [PubMed] [Google Scholar]
- 219.Pauly JR, Sparks JA, Hauser KF, Pauly TH. In utero nicotine exposure causes persistent, gender-dependant changes in locomotor activity and sensitivity to nicotine in C57Bl/6 mice. Int J Dev Neurosci 2004; 22:329–337. [DOI] [PubMed] [Google Scholar]
- 220.Paz R, Barsness B, Martenson T, Tanner D, Allan A. Behavioral teratogenicity induced by nonforced maternal nicotine consumption. Neuropsychopharmacology 2007; 32:693–699. [DOI] [PubMed] [Google Scholar]
- 221.Heath CJ, Horst NK, Picciotto MR. Oral nicotine consumption does not affect maternal care or early development in mice but results in modest hyperactivity in adolescence. Physiol Behav 2010; 101:764–769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Zhu J, Zhang X, Xu Y, Spencer TJ, Biederman J, Bhide PG. Prenatal nicotine exposure mouse model showing hyperactivity, reduced cingulate cortex volume, reduced dopamine turnover, and responsiveness to oral methylphenidate treatment. J Neurosci 2012; 32:9410–9418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Zhu J, Fan F, McCarthy DM, Zhang L, Cannon EN, Spencer TJ et al. A prenatal nicotine exposure mouse model of methylphenidate responsive ADHD-associated cognitive phenotypes. Int J Dev Neurosci 2017; 58:26–34. [DOI] [PubMed] [Google Scholar]
- 224.Buck JM, Sanders KN, Wageman CR, Knopik VS, Stitzel JA, O’Neill HC. Developmental nicotine exposure precipitates multigenerational maternal transmission of nicotine preference and ADHD-like behavioral, rhythmometric, neuropharmacological, and epigenetic anomalies in adolescent mice. Neuropharmacology 2019; 149:66–82. 10.1016/j.neuropharm.2019.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Zhu J, Zhang X, Xu Y, Spencer TJ, Biederman J, Bhide PG. Prenatal nicotine exposure mouse model showing hyperactivity, reduced cingulate cortex volume, reduced dopamine turnover, and responsiveness to oral methylphenidate treatment. J Neurosci 2012; 32:9410–9418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Lee H, Chung S, Noh J. Maternal nicotine exposure during late gestation and lactation increases anxiety-like and impulsive decision-making behavior in adolescent offspring of rat. Toxicol Res 2016; 32:275–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Chistyakov V, Patkina N, Tammimäki A, Talka R, Salminen O, Belozertseva I et al. Nicotine exposure throughout early development promotes nicotine self-administration in adolescent mice and induces long-lasting behavioural changes. Eur J Pharmacol 2010; 640:87–93. [DOI] [PubMed] [Google Scholar]
- 228.Alkam T, Mamiya T, Kimura N, Yoshida A, Kihara D, Tsunoda Y et al. Prenatal nicotine exposure decreases the release of dopamine in the medial frontal cortex and induces atomoxetine-responsive neurobehavioral deficits in mice. Psychopharmacology (Berl) 2017; 234:1853–1869. [DOI] [PubMed] [Google Scholar]
- 229.Mowla SJ, Farhadi HF, Pareek S, Atwal JK, Morris SJ, Seidah NG, Murphy RA. Biosynthesis and post-translational processing of the precursor to brain-derived neurotrophic factor. J Biol Chem 2001; 276:12660–12666. [DOI] [PubMed] [Google Scholar]
- 230.van Amsterdam J, van der Velde B, Schulte M, van den Brink W. Causal factors of increased smoking in ADHD: A systematic review. Subst Use Misuse 2018; 53:432–445. [DOI] [PubMed] [Google Scholar]
- 231.Lucatch AM, Lowe DJ, Clark RC, Kozak K, George TP. Neurobiological determinants of tobacco smoking in schizophrenia. Front Psych 2018; 9:672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Kumari V, Postma P. Nicotine use in schizophrenia: The self medication hypotheses. Neurosci Biobehav Rev 2005; 29:1021–1034. [DOI] [PubMed] [Google Scholar]
- 233.Smith AM, Dwoskin LP, Pauly JR. Early exposure to nicotine during critical periods of brain development: Mechanisms and consequences. Journal of pediatric biochemistry 2010; 1:125–141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Blood-Siegfried J, Rende EK. The long-term effects of prenatal nicotine exposure on neurologic development. J Midwifery Womens Health 2010; 55:143–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Slotkin TA, Cho H, Whitmore WL. Effects of prenatal nicotine exposure on neuronal development: Selective actions on central and peripheral catecholaminergic pathways. Brain Res Bull 1987; 18:601–611. [DOI] [PubMed] [Google Scholar]
- 236.Slotkin TA, Pinkerton KE, Auman JT, Qiao D, Seidler FJ. Perinatal exposure to environmental tobacco smoke upregulates nicotinic cholinergic receptors in monkey brain. Developmental brain research 2002; 133:175–179. [DOI] [PubMed] [Google Scholar]
- 237.Slotkin TA. Nicotine and the adolescent brain: Insights from an animal model. Neurotoxicol Teratol 2002; 24:369–384. [DOI] [PubMed] [Google Scholar]
- 238.Bailey CD, Tian MK, Kang L, O'Reilly R, Lambe EK. Chrna5 genotype determines the long-lasting effects of developmental in vivo nicotine exposure on prefrontal attention circuitry. Neuropharmacology 2014; 77:145–155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Yanai J. Neurobehavioral teratology. Amsterdam: Elsevier Publishing Company; 1984. [Google Scholar]
- 240.Gold AB, Keller AB, Perry DC. Prenatal exposure of rats to nicotine causes persistent alterations of nicotinic cholinergic receptors. Brain Res 2009; 1250:88–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Lv J, Mao C, Zhu L, Zhang H, Pengpeng H, Xu F et al. The effect of prenatal nicotine on expression of nicotine receptor subunits in the fetal brain. Neurotoxicology 2008; 29:722–726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Britton AF, Vann RE, Robinson SE. Perinatal nicotine exposure eliminates peak in nicotinic acetylcholine receptor response in adolescent rats. Journal of Pharmacology and Experimental Therapeutics 2007; 320:871–876. [DOI] [PubMed] [Google Scholar]
- 243.Huang LZ, Winzer-Serhan UH. Chronic neonatal nicotine upregulates heteromeric nicotinic acetylcholine receptor binding without change in subunit mRNA expression. Brain Res 2006; 1113:94–109. [DOI] [PubMed] [Google Scholar]
- 244.Miao H, Liu C, Bishop K, Gong ZH, Nordberg A, Zhang X. Nicotine exposure during a critical period of development leads to persistent changes in nicotinic acetylcholine receptors of adult rat brain. J Neurochem 1998; 70:752–762. [DOI] [PubMed] [Google Scholar]
- 245.Tizabi Y, Perry DC. Prenatal nicotine exposure is associated with an increase in [125I] epibatidine binding in discrete cortical regions in rats. Pharmacol Biochem Behav 2000; 67:319–323. [DOI] [PubMed] [Google Scholar]
- 246.Tizabi Y, Popke EJ, Rahman MA, Nespor SM, Grunberg NE. Hyperactivity induced by prenatal nicotine exposure is associated with an increase in cortical nicotinic receptors. Pharmacol Biochem Behav 1997; 58:141–146. [DOI] [PubMed] [Google Scholar]
- 247.Van de Kamp JL, Collins AC. Prenatal nicotine alters nicotinic receptor development in the mouse brain. Pharmacol Biochem Behav 1994; 47:889–900. [DOI] [PubMed] [Google Scholar]
- 248.del Campo N, Chamberlain SR, Sahakian BJ, Robbins TW. The roles of dopamine and noradrenaline in the pathophysiology and treatment of attention-deficit/hyperactivity disorder. Biol Psychiatry 2011; 69:e145–e157. [DOI] [PubMed] [Google Scholar]
- 249.Zhu Y, Jiang X, Ji W. The mechanism of cortico-striato-thalamo-cortical neurocircuitry in response inhibition and emotional responding in attention deficit hyperactivity disorder with comorbid disruptive behavior disorder. Neurosci Bull 2018; 34:566–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Clerkin SM, Schulz KP, Berwid OG, Fan J, Newcorn JH, Tang CY, Halperin JM. Thalamo-cortical activation and connectivity during response preparation in adults with persistent and remitted ADHD. Am J Psychiatry 2013; 170:1011–1019. [DOI] [PubMed] [Google Scholar]
- 251.Mills KL, Bathula D, Costa Dias TG, Iyer SP, Fenesy MC, Musser ED et al. Altered cortico-striatal–thalamic connectivity in relation to spatial working memory capacity in children with ADHD. Front Psych 2012; 3:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Li W, Pozzo-Miller L. Dysfunction of the corticostriatal pathway in autism spectrum disorders. J Neurosci Res 2019; 11:2130–2147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Abbott AE, Linke AC, Nair A, Jahedi A, Alba LA, Keown CL et al. Repetitive behaviors in autism are linked to imbalance of corticostriatal connectivity: A functional connectivity MRI study. Soc Cogn Affect Neurosci 2018; 13:32–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Di Martino A, Kelly C, Grzadzinski R, Zuo XN, Mennes M, Mairena MA et al. Aberrant striatal functional connectivity in children with autism. Biol Psychiatry 2011; 69:847–856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Avram M, Brandl F, Bäuml J, Sorg C. Cortico-thalamic hypo-and hyperconnectivity extend consistently to basal ganglia in schizophrenia. Neuropsychopharmacology 2018; 43:2239–2248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Levitt JJ, Nestor PG, Levin L, Pelavin P, Lin P, Kubicki M et al. Reduced structural connectivity in frontostriatal white matter tracts in the associative loop in schizophrenia. Am J Psychiatry 2017; 174:1102–1111. [DOI] [PubMed] [Google Scholar]
- 257.Jones EG. Cortical development and thalamic pathology in schizophrenia. Schizophr Bull 1997; 23:483–501. [DOI] [PubMed] [Google Scholar]
- 258.Ding JB, Guzman JN, Peterson JD, Goldberg JA, Surmeier DJ. Thalamic gating of corticostriatal signaling by cholinergic interneurons. Neuron 2010; 67:294–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Yochum C, Doherty-Lyon S, Hoffman C, Hossain MM, Zelikoff JT, Richardson JR. Prenatal cigarette smoke exposure causes hyperactivity and aggressive behavior: Role of altered catecholamines and BDNF. Exp Neurol 2014; 254:145–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Muneoka K, Nakatsu T, Fuji JI, Ogawa T, Takigawa M. Prenatal administration of nicotine results in dopaminergic alterations in the neocortex. Neurotoxicol Teratol 1999; 21:603–609. [DOI] [PubMed] [Google Scholar]
- 261.Dwyer JB, Cardenas A, Franke RM, Chen Y, Bai Y, Belluzzi JD et al. Prenatal nicotine sex-dependently alters adolescent dopamine system development. Transl Psychiatry 2019; 9:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Fung YK, Lau YS. Effects of prenatal nicotine exposure on rat striatal dopaminergic and nicotinic systems. Pharmacol Biochem Behav 1989; 33:1–6. [DOI] [PubMed] [Google Scholar]
- 263.Tang S, Machaalani R, Waters KA. Brain-derived neurotrophic factor (BDNF) and TrkB in the piglet brainstem after post-natal nicotine and intermittent hypercapnic hypoxia. Brain Res 2008; 1232:195–205. [DOI] [PubMed] [Google Scholar]
- 264.Harrod SB, Lacy RT, Zhu J, Hughes BA, Perna MK, Brown RW. Gestational IV nicotine produces elevated brain-derived neurotrophic factor in the mesocorticolimbic dopamine system of adolescent rat offspring. Synapse 2011; 65:1382–1392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Lacy RT, Brown RW, Morgan AJ, Mactutus CF, Harrod SB. Intravenous prenatal nicotine exposure alters METH-induced hyperactivity, conditioned hyperactivity, and BDNF in adult rat offspring. Dev Neurosci 2016; 38:171–185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Xiao L, Kish VL, Benders KM, Wu ZX. Prenatal and early postnatal exposure to cigarette smoke decreases BDNF/TrkB signaling and increases abnormal behaviors later in life. Int J Neuropsychopharmacol 2016; 19:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Machaalani R, Thawley M, Huang J, Chen H. Effects of prenatal cigarette smoke exposure on BDNF, PACAP, microglia and gliosis expression in the young male mouse brainstem. Neurotoxicology 2019; 74:40–46. [DOI] [PubMed] [Google Scholar]
- 268.Buck JM, O'Neill HC, Stitzel JA. Developmental nicotine exposure elicits multigenerational disequilibria in proBDNF proteolysis and glucocorticoid signaling in the frontal cortices, striata, and hippocampi of adolescent mice. Biochem Pharmacol 2019; 168:438–451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.Fayard B, Loeffler S, Weis J, Vögelin E, Krüttgen A. The secreted brain-derived neurotrophic factor precursor pro-BDNF binds to TrkB and p75NTR but not to TrkA or TrkC. J Neurosci Res 2005; 80:18–28. [DOI] [PubMed] [Google Scholar]
- 270.Hou Y, Liang W, Zhang J, Li Q, Ou H, Wang Z et al. Schizophrenia-associated rs4702 G allele-specific downregulation of FURIN expression by miR-338-3p reduces BDNF production. Schizophr Res 2018; 199:176–180. [DOI] [PubMed] [Google Scholar]
- 271.Costa RO, Perestrelo T, Almeida RD. PROneurotrophins and CONSequences. Mol Neurobiol 2018; 55:2934–2951. [DOI] [PubMed] [Google Scholar]
- 272.Seidah NG, Benjannet S, Pareek S, Chrétien M, Murphy RA. Cellular processing of the neurotrophin precursors of NT3 and BDNF by the mammalian proprotein convertases. FEBS Lett 1996; 379:247–250. [DOI] [PubMed] [Google Scholar]
- 273.Mowla SJ, Farhadi HF, Pareek S, Atwal JK, Morris SJ, Seidah NG, Murphy RA. Biosynthesis and post-translational processing of the precursor to brain-derived neurotrophic factor. J Biol Chem 2001; 276:12660–12666. [DOI] [PubMed] [Google Scholar]
- 274.Chen Y, Zhang J, Deng M. Furin mediates brain-derived neurotrophic factor upregulation in cultured rat astrocytes exposed to oxygen–glucose deprivation. J Neurosci Res 2015; 93:189–194. [DOI] [PubMed] [Google Scholar]
- 275.Abdallah MW, Michel TM. Matrix metalloproteinases in autism spectrum disorders. Journal of molecular psychiatry 2013; 1:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276.Tsai SJ. Role of neurotrophic factors in attention deficit hyperactivity disorder. Cytokine Growth Factor Rev 2017; 34:35–41. [DOI] [PubMed] [Google Scholar]
- 277.Fromer M, Roussos P, Sieberts SK, Johnson JS, Kavanagh DH, Perumal TM et al. Gene expression elucidates functional impact of polygenic risk for schizophrenia. Nat Neurosci 2016; 19:1442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Suter MA, Abramovici AR, Griffin E, Branch DW, Lane RH, Mastrobattista J et al. In utero nicotine exposure epigenetically alters fetal chromatin structure and differentially regulates transcription of the glucocorticoid receptor in a rat model. Birth Defects Res A Clin Mol Teratol 2015; 103:583–588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279.Xu D, Liang G, Yan YE, He WW, Liu YS, Chen LB et al. Nicotine-induced over-exposure to maternal glucocorticoid and activated glucocorticoid metabolism causes hypothalamic–pituitary–adrenal axis-associated neuroendocrine metabolic alterations in fetal rats. Toxicol Lett 2012; 209:282–290. [DOI] [PubMed] [Google Scholar]
- 280.Yan YE, Liu L, Wang JF, Liu F, Li XH, Qin HQ, Wang H. Prenatal nicotinic exposure suppresses fetal adrenal steroidogenesis via steroidogenic factor 1 (SF-1) deacetylation. Toxicol Appl Pharmacol 2014; 277:231–241. [DOI] [PubMed] [Google Scholar]
- 281.Xu D, Xia LP, Shen L, Lei YY, Liu L, Zhang L et al. Prenatal nicotine exposure enhances the susceptibility to metabolic syndrome in adult offspring rats fed high-fat diet via alteration of HPA axis-associated neuroendocrine metabolic programming. Acta Pharmacol Sin 2013; 34:1526–1534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282.Reamon-Buettner SM, Mutschler V, Borlak J. The next innovation cycle in toxicogenomics: Environmental epigenetics. Mutation Research/Reviews in Mutation Research 2008; 659:158–165. [DOI] [PubMed] [Google Scholar]
- 283.Jaenisch R. DNA methylation and imprinting: Why bother? Trends Genet 1997; 13:323–329. [DOI] [PubMed] [Google Scholar]
- 284.Maccani MA, Marsit CJ. REVIEW ARTICLE: Epigenetics in the placenta. Am J Reprod Immunol 2009; 62:78–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285.Reik W. Stability and flexibility of epigenetic gene regulation in mammalian development. Nature 2007; 447:425–432. [DOI] [PubMed] [Google Scholar]
- 286.Yang IV, Schwartz DA. Epigenetic mechanisms and the development of asthma. J Allergy Clin Immunol 2012; 130:1243–1255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287.Zhou Z, Enoch M-A, Goldman D. Chapter Ten—Gene Expression in the Addicted Brain. In: Hitzemann R, Mcweeney S (eds.), International Review of Neurobiology, vol. 116. Academic Press; 2014; 116:251–273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288.Ladd-Acosta C. Epigenetic signatures as biomarkers of exposure. Current Environmental Health Reports 2015; 2:117–125. [DOI] [PubMed] [Google Scholar]
- 289.Maccani JZ, Koestler DC, Houseman EA, Marsit CJ, Kelsey KT. Placental DNA methylation alterations associated with maternal tobacco smoking at the RUNX3 gene are also associated with gestational age. Epigenomics 2013; 5:619–630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290.Pajer K, Andrus BM, Gardner W, Lourie A, Strange B, Campo J, Bridge J, Blizinsky K, Dennis K, Vedell P, Churchill GA, Redei EE. Discovery of blood transcriptomic markers for depression in animal models and pilot validation in subjects with early-onset major depression. Transl Psychiatry 2012; 2:e101–e101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 291.LaPlant Q, Vialou V, Covington HE III, Dumitriu D, Feng J, Warren BL et al. Dnmt3a regulates emotional behavior and spine plasticity in the nucleus accumbens. Nat Neurosci 2010; 13:1137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292.Morris MJ, Adachi M, Na ES, Monteggia LM. Selective role for DNMT3a in learning and memory. Neurobiol Learn Mem 2014; 115:30–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293.Gontier G, Iyer M, Shea JM, Bieri G, Wheatley EG, Ramalho-Santos M, Villeda SA. Tet2 rescues age-related regenerative decline and enhances cognitive function in the adult mouse brain. Cell Rep 2018; 22:1974–1981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294.Dawlaty MM, Breiling A, Le T, Barrasa MI, Raddatz G, Gao Q et al. Loss of Tet enzymes compromises proper differentiation of embryonic stem cells. Dev Cell 2014; 29:102–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 295.Al-Mahdawi S, Virmouni SA, Pook MA. The emerging role of 5-hydroxymethylcytosine in neurodegenerative diseases. Front Neurosci 2014; 8:397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 296.Wang J, Zhang KX, Lu GZ, Zhao XH. Research progress on 5hmC and TET dioxygenases in neurodevelopment and neurological diseases. Yi chuan= Hereditas 2017; 39:1138–1149. [DOI] [PubMed] [Google Scholar]
- 297.Antunes C, Sousa N, Pinto L, Marques CJ. TET enzymes in neurophysiology and brain function. Neurosci Biobehav Rev 2019; 102:337–344. [DOI] [PubMed] [Google Scholar]
- 298.Nguyen T, Li GE, Chen H, Cranfield CG, McGrath KC, Gorrie CA. Maternal E-cigarette exposure results in cognitive and epigenetic alterations in offspring in a mouse model. Chem Res Toxicol 2018; 31:601–611. [DOI] [PubMed] [Google Scholar]
- 299.Alaghband Y, Bredy TW, Wood MA. The role of active DNA demethylation and Tet enzyme function in memory formation and cocaine action. Neurosci Lett 2016; 625:40–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300.Cheng Y, Sun M, Chen L, Li Y, Lin L, Yao B et al. Ten-eleven translocation proteins modulate the response to environmental stress in mice. Cell Rep 2018; 25:3194–3203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301.Mill J, Tang T, Kaminsky Z, Khare T, Yazdanpanah S, Bouchard L et al. Epigenomic profiling reveals DNA-methylation changes associated with major psychosis. The American Journal of Human Genetics 2008; 82:696–711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 302.Jung Y, Hsieh LS, Lee AM, Zhou Z, Coman D, Heath CJ et al. An epigenetic mechanism mediates developmental nicotine effects on neuronal structure and behavior. Nat Neurosci 2016; 19:905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303.Ding K, Yang J, Reynolds GP, Chen B, Shao J, Liu R et al. DAT1 methylation is associated with methylphenidate response on oppositional and hyperactive-impulsive symptoms in children and adolescents with ADHD. World J Biol Psychiatry 2017; 18:291–299. [DOI] [PubMed] [Google Scholar]
- 304.Saunderson EA, Spiers H, Mifsud KR, Gutierrez-Mecinas M, Trollope AF, Shaikh A et al. Stress-induced gene expression and behavior are controlled by DNA methylation and methyl donor availability in the dentate gyrus. Proc Natl Acad Sci 2016; 113:4830–4835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 305.Azzi A, Dallmann R, Casserly A, Rehrauer H, Patrignani A, Maier B et al. Circadian behavior is light-reprogrammed by plastic DNA methylation. Nat Neurosci 2014; 17:377–382. [DOI] [PubMed] [Google Scholar]
- 306.Aizawa S, Sensui N, Yamamuro Y. Induction of cholinergic differentiation by 5-azacytidine in NG108-15 neuronal cells. Neuroreport 2009; 20:157–160. [DOI] [PubMed] [Google Scholar]
- 307.Watanabe H, Zoli M, Changeux JP. Promoter analysis of the neuronal nicotinic acetylcholine receptor α4 gene: Methylation and expression of the transgene. Eur J Neurosci 1998; 10:2244–2253. [DOI] [PubMed] [Google Scholar]
- 308.Chan A, Tchantchou F, Graves V, Rozen R, Shea TB. Dietary and genetic compromise in folate availability reduces acetylcholine, cognitive performance and increases aggression: Critical role of S-adenosyl methionine. The Journal of Nutrition Health and Aging 2008; 12:252–261. [DOI] [PubMed] [Google Scholar]
- 309.Kuncl RW, Drachman DB, Adams R. Inhibition of methyltransferase reduces the turnover of acetylcholine receptors. Proc Natl Acad Sci 1988; 85:4032–4036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 310.Ricceri L, De Filippis B, Fuso A, Laviola G. Cholinergic hypofunction in MeCP2-308 mice: Beneficial neurobehavioural effects of neonatal choline supplementation. Behav Brain Res 2011; 221:623–629. [DOI] [PubMed] [Google Scholar]
- 311.Cermak JM, Holler T, Jackson DA, Blusztajn JK. Prenatal availability of choline modifies development of the hippocampal cholinergic system. FASEB J 1998; 12:349–357. [DOI] [PubMed] [Google Scholar]
- 312.Busche A, Bagorda A, Lehmann K, Neddens J, Teuchert-Noodt G. The maturation of the acetylcholine system in the dentate gyrus of gerbils (Meriones unguiculatus) is affected by epigenetic factors. J Neural Transm 2006; 113:113–124. [DOI] [PubMed] [Google Scholar]
- 313.Shumay E, Fowler JS, Volkow ND. Genomic features of the human dopamine transporter gene and its potential epigenetic states: Implications for phenotypic diversity. PLoS One 2010; 5:e11067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 314.Wiers CE, Lohoff FW, Lee J, Muench C, Freeman C, Zehra A et al. Methylation of the dopamine transporter gene in blood is associated with striatal dopamine transporter availability in ADHD: A preliminary study. Eur J Neurosci 2018; 48:1884–1895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 315.Kordi-Tamandani DM, Sahranavard R, Torkamanzehi A. DNA methylation and expression profiles of the brain-derived neurotrophic factor (BDNF) and dopamine transporter (DAT1) genes in patients with schizophrenia. Mol Biol Rep 2012; 39:10889–10893. [DOI] [PubMed] [Google Scholar]
- 316.Wang S, Zhang R, Claret FX, Yang H. Involvement of microRNA-24 and DNA methylation in resistance of nasopharyngeal carcinoma to ionizing radiation. Mol Cancer Ther 2014; 13:3163–3174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 317.Cubero-Millán I, Ruiz-Ramos MJ, Molina-Carballo A, Martínez-Serrano S, Fernández-López L, Machado-Casas I et al. BDNF concentrations and daily fluctuations differ among ADHD children and respond differently to methylphenidate with no relationship with depressive symptomatology. Psychopharmacology (Berl) 2017; 234:267–279. [DOI] [PubMed] [Google Scholar]
- 318.Ikegame T, Bundo M, Sunaga F, Asai T, Nishimura F, Yoshikawa A et al. DNA methylation analysis of BDNF gene promoters in peripheral blood cells of schizophrenia patients. Neurosci Res 2013; 77:208–214. [DOI] [PubMed] [Google Scholar]
- 319.Hing B, Ramos E, Braun P, McKane M, Jancic D, Tamashiro KL et al. Adaptation of the targeted capture methyl-Seq platform for the mouse genome identifies novel tissue-specific DNA methylation patterns of genes involved in neurodevelopment. Epigenetics 2015; 10:581–596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 320.van Mil NH, Steegers-Theunissen RP, Bouwland-Both MI, Verbiest MM, Rijlaarsdam J, Hofman A et al. DNA methylation profiles at birth and child ADHD symptoms. J Psychiatr Res 2014; 49:51–59. [DOI] [PubMed] [Google Scholar]
- 321.Walton E, Pingault JB, Cecil CA, Gaunt TR, Relton CL, Mill J, Barker ED. Epigenetic profiling of ADHD symptoms trajectories: A prospective, methylome-wide study. Mol Psychiatry 2017; 22:250–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 322.Ding K, Yang J, Reynolds GP, Chen B, Shao J, Liu R et al. DAT1 methylation is associated with methylphenidate response on oppositional and hyperactive-impulsive symptoms in children and adolescents with ADHD. World J Biol Psychiatry 2017; 18:291–299. [DOI] [PubMed] [Google Scholar]
- 323.Nardone, S., Sams, D. S., Zito, A., Reuveni, E., & Elliott, E. Dysregulation of cortical neuron DNA methylation profile in autism spectrum disorder. Cereb Cortex 2017; 27:5739–5754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 324.James SJ, Melnyk S, Jernigan S, Pavliv O, Trusty T, Lehman S et al. A functional polymorphism in the reduced folate carrier gene and DNA hypomethylation in mothers of children with autism. Am J Med Genet B Neuropsychiatr Genet 2010; 153:1209–1220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 325.Ladd-Acosta C, Hansen KD, Briem E, Fallin MD, Kaufmann WE, Feinberg AP. Common DNA methylation alterations in multiple brain regions in autism. Mol Psychiatry 2014; 19:862–871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 326.Ryan J, Saffery R. Crucial timing in schizophrenia: Role of DNA methylation in early neurodevelopment. Genome Biol 2014; 15:495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 327.Jaffe AE, Gao Y, Deep-Soboslay A, Tao R, Hyde TM, Weinberger DR, Kleinman JE. Mapping DNA methylation across development, genotype and schizophrenia in the human frontal cortex. Nat Neurosci 2016; 19:40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 328.Chen C, Zhang C, Cheng L, Reilly JL, Bishop JR, Sweeney JA et al. Correlation between DNA methylation and gene expression in the brains of patients with bipolar disorder and schizophrenia. Bipolar Disord 2014; 16:790–799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 329.Numata S, Ye T, Herman M, Lipska BK. DNA methylation changes in the postmortem dorsolateral prefrontal cortex of patients with schizophrenia. Front Genet 2014; 5:280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 330.Hannon E, Dempster E, Viana J, Burrage J, Smith AR, Macdonald R et al. An integrated genetic-epigenetic analysis of schizophrenia: Evidence for co-localization of genetic associations and differential DNA methylation. Genome Biol 2016; 17:176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 331.Wockner LF, Noble EP, Lawford BR, Young RM, Morris CP, Whitehall VL, Voisey J. Genome-wide DNA methylation analysis of human brain tissue from schizophrenia patients. Transl Psychiatry 2014; 4:e339–e339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 332.Huang HS, Akbarian S. GAD1 mRNA expression and DNA methylation in prefrontal cortex of subjects with schizophrenia. PLoS One 2007; 2:e809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 333.Joubert BR, Håberg SE, Nilsen RM, Wang X, Vollset SE, Murphy SK et al. 450K epigenome-wide scan identifies differential DNA methylation in newborns related to maternal smoking during pregnancy. Environ Health Perspect 2012; 120:1425–1431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 334.Guerrero-Preston R, Goldman LR, Brebi-Mieville P, Ili-Gangas C, LeBron C, Witter FR et al. Global DNA hypomethylation is associated with in utero exposure to cotinine and perfluorinated alkyl compounds. Epigenetics 2010; 5:539–546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 335.Maccani JZ, Maccani MA. Altered placental DNA methylation patterns associated with maternal smoking: Current perspectives. Advances in genomics and genetics 2015; 2015:205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 336.Richmond RC, Simpkin AJ, Woodward G, Gaunt TR, Lyttleton O, McArdle WL et al. Prenatal exposure to maternal smoking and offspring DNA methylation across the lifecourse: Findings from the Avon longitudinal study of parents and children (ALSPAC). Hum Mol Genet 2015; 24:2201–2217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 337.Chatterton Z, Hartley BJ, Seok MH, Mendelev N, Chen S, Milekic M et al. In utero exposure to maternal smoking is associated with DNA methylation alterations and reduced neuronal content in the developing fetal brain. Epigenetics Chromatin 2017; 10:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 338.Knopik VS, Marceau K, Bidwell LC, Rolan E. Prenatal substance exposure and offspring development: Does DNA methylation play a role? Neurotoxicol Teratol 2019; 71:50–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 339.Suter M, Abramovici A, Showalter L, Hu M, Shope CD, Varner M, Aagaard-Tillery K. In utero tobacco exposure epigenetically modifies placental CYP1A1 expression. Metabolism - Clinical and Experimental 2010; 59:1481–1490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 340.Paquette AG, Lesseur C, Armstrong DA, Koestler DC, Appleton AA, Lester BM, Marsit CJ. Placental HTR2A methylation is associated with infant neurobehavioral outcomes. Epigenetics 2013; 8:796–801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 341.Stroud LR, Papandonatos GD, Salisbury AL, Phipps MG, Huestis MA, Niaura R, Padbury JF, Marsit CJ, Lester BM. Epigenetic regulation of placental NR3C1: Mechanism underlying prenatal programming of infant Neurobehavior by maternal smoking? Child Dev 2016; 87:49–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 342.Stroud LR, Papandonatos GD, Rodriguez D, McCallum M, Salisbury AL, Phipps MG et al. Maternal smoking during pregnancy and infant stress response: Test of a prenatal programming hypothesis. Psychoneuroendocrinology 2014; 48:29–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 343.Ivorra C, Fraga MF, Bayón GF, Fernández AF, Garcia-Vicent C, Chaves FJ, Redon J, Lurbe E. DNA methylation patterns in newborns exposed to tobacco in utero. J Transl Med 2015; 13:25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 344.758 Küpers LK, Xu X, Jankipersadsing SA, Vaez A, la Bastide-van Gemert S, Scholtens S, Nolte IM, Richmond RC, Relton CL, Felix JF, Duijts L, van Meurs JB et al. DNA methylation mediates the effect of maternal smoking during pregnancy on birthweight of the offspring. Int J Epidemiol 2015; 44:1224–1237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 345.Novakovic B, Ryan J, Pereira N, Boughton B, Craig JM, Saffery R. Postnatal stability, tissue, and time specific effects of AHRR methylation change in response to maternal smoking in pregnancy. Epigenetics 2014; 9:377–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 346.Markunas Christina A, Zongli X, Sophia H, Wade Paul A, Lie Rolv T, Taylor Jack A, Wilcox Allen J. Identification of DNA methylation changes in newborns related to maternal smoking during pregnancy. Environ Health Perspect 2014; 122:1147–1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 347.Philibert RA, Beach SRH, Lei M-K, Brody GH. Changes in DNA methylation at the aryl hydrocarbon receptor repressor may be a new biomarker for smoking. Clin Epigenetics 2013; 5:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 348.Shenker NS, Polidoro S, van Veldhoven K, Sacerdote C, Ricceri F, Birrell MA, Belvisi MG, Brown R, Vineis P, Flanagan JM. Epigenome-wide association study in the European prospective investigation into cancer and nutrition (EPIC-Turin) identifies novel genetic loci associated with smoking. Hum Mol Genet 2013; 22:843–851. [DOI] [PubMed] [Google Scholar]
- 349.Patil VK, Mukherjee N, Lockett G, Zhang H, Karmaus W, Holloway JW, Arshad SH. Transgenerational epigenetics: Maternal and grandmaternal gestational smoking influences DNA methylation of the AHRR gene. European Respiratory Journal 2016; 48:PA1200. [Google Scholar]
- 350.Kimura E, Tohyama C. Embryonic and postnatal expression of aryl hydrocarbon receptor mRNA in mouse brain. Frontiers in neuroanatomy 2017; 11:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 351.Qin H, Powell-Coffman JA. The Caenorhabditis elegans aryl hydrocarbon receptor, AHR-1, regulates neuronal development. Dev Biol 2004; 270:64–75. [DOI] [PubMed] [Google Scholar]
- 352.de la Parra J, Cuartero MI, Pérez-Ruiz A, García-Culebras A, Martín R, Sánchez-Prieto J et al. AhR deletion promotes aberrant morphogenesis and synaptic activity of adult-generated granule neurons and impairs hippocampus-dependent memory. Eneuro 2018; 5:1076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 353.Kimura E, Ding Y, Tohyama C. AhR signaling activation disrupts migration and dendritic growth of olfactory interneurons in the developing mouse. Sci Rep 2016; 6:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 354.Kimura E, Kubo KI, Endo T, Ling W, Nakajima K, Kakeyama M, Tohyama C. Impaired dendritic growth and positioning of cortical pyramidal neurons by activation of aryl hydrocarbon receptor signaling in the developing mouse. PLoS One 2017; 12:e0183497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 355.Kimura E, Kubo KI, Matsuyoshi C, Benner S, Hosokawa M, Endo T et al. Developmental origin of abnormal dendritic growth in the mouse brain induced by in utero disruption of aryl hydrocarbon receptor signaling. Neurotoxicol Teratol 2015; 52:42–50. [DOI] [PubMed] [Google Scholar]
- 356.Wu PY, Chuang PY, Chang GD, Chan YY, Tsai TC, Wang BJ et al. Novel endogenous ligands of aryl hydrocarbon receptor mediate neural development and differentiation of neuroblastoma. ACS Chem Nerosci 2019; 10:4031–4042. [DOI] [PubMed] [Google Scholar]
- 357.Li W. Aryl Hydocarbon Receptor Signaling Modulates Circadian Locomotor Activity and Expression of Clock-Associated Proteins. 2005.
- 358.Tischkau SA. Mechanisms of circadian clock interactions with aryl hydrocarbon receptor signalling. Eur J Neurosci 2020; 51:379–395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 359.Williams EG, Mouchiroud L, Frochaux M, Pandey A, Andreux PA, Deplancke B, Auwerx J. An evolutionarily conserved role for the aryl hydrocarbon receptor in the regulation of movement. PLoS Genet 2014; 10:e1004673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 360.Ma Y, Fu H, Xu T, Luo Y, Liu Y, Chen Y et al. New perspective on the regulation of acetylcholinesterase via the aryl hydrocarbon receptor. J Neurochem 2020; 00:1–9. [DOI] [PubMed] [Google Scholar]
- 361.Xie HQ, Xu HM, Fu HL, Hu Q, Tian WJ, Pei XH, Zhao B. AhR-mediated effects of dioxin on neuronal acetylcholinesterase expression in vitro. Environ Health Perspect 2013; 121:613–618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 362.Akahoshi E, Yoshimura S, Uruno S, Ishihara-Sugano M. Effect of dioxins on regulation of tyrosine hydroxylase gene expression by aryl hydrocarbon receptor: A neurotoxicology study. Environ Health 2009; 8:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 363.Lin CH, Chen CC, Chou CM, Wang CY, Hung CC, Chen JY et al. Knockdown of the aryl hydrocarbon receptor attenuates excitotoxicity and enhances NMDA-induced BDNF expression in cortical neurons. J Neurochem 2009; 111:777–789. [DOI] [PubMed] [Google Scholar]
- 364.Wang SH, Liang CT, Liu YW, Huang MC, Huang SC, Hong WF, Su JGJ. Crosstalk between activated forms of the aryl hydrocarbon receptor and glucocorticoid receptor. Toxicology 2009; 262:87–97. [DOI] [PubMed] [Google Scholar]
- 365.Aluru N, Vijayan MM. Aryl hydrocarbon receptor activation impairs cortisol response to stress in rainbow trout by disrupting the rate-limiting steps in steroidogenesis. Endocrinology 2006; 147:1895–1903. [DOI] [PubMed] [Google Scholar]
- 366.Fujisawa TX, Nishitani S, Iwanaga R, Matsuzaki J, Kawasaki C, Tochigi M et al. Association of aryl hydrocarbon receptor-related gene variants with the severity of autism spectrum disorders. Front Psych 2016; 7:184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 367.Sengupta SM, Smith AK, Grizenko N, Joober R. Locus-specific DNA methylation changes and phenotypic variability in children with attention-deficit hyperactivity disorder. Psychiatry Res 2017; 256:298–304. [DOI] [PubMed] [Google Scholar]
- 368.Monick MM, Beach SRH, Plume J, Sears R, Gerrard M, Brody GH, Philibert RA. Coordinated changes in AHRR methylation in lymphoblasts and pulmonary macrophages from smokers. Am J Med Genet B Neuropsychiatr Genet 2012; 159B:141–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 369.Yamamoto J, Ihara K, Nakayama H, Hikino S, Satoh K, Kubo N, Iida T, Fujii Y, Hara T. Characteristic expression of aryl hydrocarbon receptor repressor gene in human tissues: Organ-specific distribution and variable induction patterns in mononuclear cells. Life Sci 2004; 74:1039–1049. [DOI] [PubMed] [Google Scholar]
- 370.Li J-M, Lu C-L, Cheng M-C, Luu S-U, Hsu S-H, Hu T-M, Tsai H-Y, Chen C-H. Role of the DLGAP2 gene encoding the SAP90/PSD-95-associated protein 2 in schizophrenia. PLoS One 2014; 9:e85373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 371.Chien W-H, Gau S-F, Liao H-M, Chiu Y-N, Wu Y-Y, Huang Y-S, Tsai W-C, Tsai H-M, Chen C-H. Deep exon resequencing of DLGAP2 as a candidate gene of autism spectrum disorders. Mol Autism 2013; 4:26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 372.Pinto D, Pagnamenta AT, Klei L, Anney R, Merico D, Regan R, Conroy J, Magalhaes TR, Correia C, Abrahams BS, Almeida J, Bacchelli E et al. Functional impact of global rare copy number variation in autism spectrum disorders. Nature 2010; 466:368–372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 373.Wu S, Yue W, Jia M, Ruan Y, Lu T, Gong X, Shuang M, Liu J, Yang X, Zhang D. Association of the neuropilin-2 (NRP2) gene polymorphisms with autism in Chinese Han population. Am J Med Genet B Neuropsychiatr Genet 2007; 144B:492–495. [DOI] [PubMed] [Google Scholar]
- 374.Richmond RC, Suderman M, Langdon R, Relton CL, Davey Smith G. DNA methylation as a marker for prenatal smoke exposure in adults. Int J Epidemiol 2018; 47:1120–1130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 375.Rzehak P, Saffery R, Reischl E, Covic M, Wahl S, Grote V, Xhonneux A, Langhendries J-P, Ferre N, Closa-Monasterolo R, Verduci E, Riva E et al. Maternal smoking during pregnancy and DNA-methylation in children at age 5.5 years: Epigenome-wide-analysis in the European Childhood Obesity Project (CHOP)-study. PLoS One 2016; 11:e0155554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 376.Lee Ken WK, Rebecca R, Hu P, Leon F, Jean S, Celine B, Eva R, Melanie W, Sonja Z, Tom G, Wendy MA, Susan R et al. Prenatal exposure to maternal cigarette smoking and DNA methylation: Epigenome-wide association in a discovery sample of adolescents and replication in an independent cohort at birth through 17 years of age. Environ Health Perspect 2015; 123:193–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 377.Rauschert S, Melton PE, Burdge G, Craig JM, Godfrey KM, Holbrook JD, Lillycrop K, Mori TA, Beilin LJ, Oddy WH, Pennell C, Huang R-C. Maternal smoking during pregnancy induces persistent epigenetic changes into adolescence, independent of postnatal smoke exposure and is associated with cardiometabolic risk. Front Genet 2019; 10:770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 378.Kheirkhah Rahimabad P, Anthony TM, Jones AD, Eslamimehr S, Mukherjee N, Ewart S et al. Nicotine and its downstream metabolites in maternal and cord sera: Biomarkers of prenatal smoking exposure associated with offspring DNA methylation. Int J Environ Res Public Health 2020; 17:9552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 379.Wiklund P, Karhunen V, Richmond RC, Parmar P, Rodriguez A, De Silva M et al. DNA methylation links prenatal smoking exposure to later life health outcomes in offspring. Clin Epigenetics 2019; 11:1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 380.Terry MB, Ferris JS, Pilsner R, Flom JD, Tehranifar P, Santella RM, Gamble MV, Susser E. Genomic DNA methylation among women in a multiethnic New York City birth cohort. Cancer Epidemiology and Prevention Biomarkers 2008; 17:2306–2310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 381.Flom JD, Ferris JS, Liao Y, Tehranifar P, Richards CB, Cho YH, Gonzalez K, Santella RM, Terry MB. Prenatal smoke exposure and genomic DNA methylation in a multiethnic birth cohort. Cancer Epidemiology and Prevention Biomarkers 2011; 20:2518–2523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 382.Schrott R, Rajavel M, Acharya K, Huang Z, Acharya C, Hawkey A et al. Sperm DNA methylation altered by THC and nicotine: Vulnerability of neurodevelopmental genes with bivalent chromatin. Sci Rep 2020; 10:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 383.Dai J, Wang Z, Xu W, Zhang M, Zhu Z, Zhao X et al. Paternal nicotine exposure defines different behavior in subsequent generation via hyper-methylation of mmu-miR-15b. Sci Rep 2017; 7:1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 384.McCarthy DM, Morgan TJ Jr, Lowe SE, Williamson MJ, Spencer TJ, Biederman J, Bhide PG. Nicotine exposure of male mice produces behavioral impairment in multiple generations of descendants. PLoS Biol 2018; 16:e2006497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 385.Murphy PJ, Guo J, Jenkins TG, James ER, Hoidal JR, Huecksteadt T et al. Paternal cigarette smoke alters DNA methylation in sperm and gene expression in offspring brain. bioRxiv 2019; 750638. [Google Scholar]
- 386.Goldberg LR, Zeid D, Kutlu MG, Cole RD, Lallai V, Sebastian A et al. Paternal nicotine enhances fear memory, reduces nicotine administration, and alters hippocampal genetic and neural function in offspring. Addict Biol 2021; 26:e12859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 387.Kutlu MG, Cole R, Tumolo JM, Parikh V, Gould TJ. Paternal nicotine exposure trans-generationally alters fear response and cholinergic function: potential epigenetic mechanisms.
- 388.Zhu J, Lee KP, Spencer TJ, Biederman J, Bhide PG. Transgenerational transmission of hyperactivity in a mouse model of ADHD. J Neurosci 2014; 34:2768–2773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 389.Lkhagvadorj K, Zeng Z, Meyer KF, Verweij LP, Kooistra W, Reinders-Luinge M et al. Postnatal smoke exposure further increases the hepatic nicotine metabolism in prenatally smoke exposed male offspring and is linked with aberrant Cyp2a5 methylation. Int J Mol Sci 2021; 22:164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 390.Lkhagvadorj K, Meyer KF, Verweij LP, Kooistra W, Reinders-Luinge M, Dijkhuizen HW et al. Prenatal smoke exposure induces persistent Cyp2a5 methylation and increases nicotine metabolism in the liver of neonatal and adult male offspring. Epigenetics 2020; 15:1370–1385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 391.Hermann A, Gowher H, Jeltsch A. Biochemistry and biology of mammalian DNA methyltransferases. Cellular and Molecular Life Sciences CMLS 2004; 61:2571–2587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 392.Zhenwei J, Shuxin G, Yongchun Z, Xianhua Z. Mechanisms of TET protein-mediated DNA demethylation and its role in the regulation of mouse development. Yi chuan= Hereditas 2015; 37:34–40. [DOI] [PubMed] [Google Scholar]
- 393.Buck JM, O’Neill HC, Stitzel JA. Developmental nicotine exposure engenders intergenerational downregulation and aberrant posttranslational modification of cardinal epigenetic factors in the frontal cortices, striata, and hippocampi of adolescent mice. Epigenetics Chromatin 2020; 13:1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 394.Nguyen T, Li GE, Chen H, Cranfield CG, McGrath KC, Gorrie CA. Maternal e-cigarette exposure results in cognitive and epigenetic alterations in offspring in a mouse model. Chem Res Toxicol 2018; 31:601–611. [DOI] [PubMed] [Google Scholar]
- 395.Harrison KL, Breen TM, Hennessey JF. The effect of patient smoking habit on the outcome of IVF and GIFT treatment. Australian and New Zealand Journal of Obstetrics and Gynaecology 1990; 30:340–342. [DOI] [PubMed] [Google Scholar]
- 396.Rosevear SK, Ford WCL, Wardle PG, Hull MGR, Holt DW, Lee TD. Smoking and decreased fertilisation rates in vitro. The Lancet 1992; 340:1195–1196. [DOI] [PubMed] [Google Scholar]
- 397.Oyesanya OA, Zenzes MT, Wang P, Casper RF. Cigarette smoking may affect meiotic maturation of human oocytes. Hum Reprod 1995; 10:3213–3217. [DOI] [PubMed] [Google Scholar]
- 398.Van Voorhis BJ, Dawson JD, Stovall DW, Sparks AE, Syrop CH. The effects of smoking on ovarian function and fertility during assisted reproduction cycles. Obstetrics & Gyne 1996; 88:785–791. [DOI] [PubMed] [Google Scholar]
- 399.Zeid D, Gould TJ. Impact of nicotine, alcohol, and cocaine exposure on germline integrity and epigenome. Neuropharmacology 2020; 173:108127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 400.Gao J, Yang J, Gong L, Lin A, Zhang Y, Bu W. Effects of cigarette smoke on chromatin configuration, DNA methylation and expression of related genes in mice oocytes. Wei sheng yan jiu= Journal of hygiene research 2017; 46:213–218. [PubMed] [Google Scholar]
- 401.Vigueras-Villaseñor RM, Fuentes-Cano MA, Saldaña MC, Espinosa LR, Reynoso-Robles R, Rojas P et al. Fetal and postnatal nicotine exposure modifies maturation of gonocytes to spermatogonia in mice. Anal Cell Pathol 2020; 2020:8892217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 402.Yohn NL, Bartolomei MS, Blendy JA. Multigenerational and transgenerational inheritance of drug exposure: The effects of alcohol, opiates, cocaine, marijuana, and nicotine. Prog Biophys Mol Biol 2015; 118:21–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 403.Moulaei N, Mondanizadeh M, Salmani ME, Palizvan MR, Khansarinejad B, Sadegh M. Transgenerational consequences of prepregnancy chronic morphine use on spatial learning and hippocampal Mecp2 and Hdac2 expression. Neuroreport 2018; 29:739–744. [DOI] [PubMed] [Google Scholar]
- 404.Krämer OH. HDAC2: A critical factor in health and disease. Trends Pharmacol Sci 2009; 30:647–655. [DOI] [PubMed] [Google Scholar]
- 405.Kavalali ET, Nelson ED, Monteggia LM. Role of MeCP2, DNA methylation, and HDACs in regulating synapse function. J Neurodev Disord 2011; 3:250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 406.Irwin RE, Pentieva K, Cassidy T, Lees-Murdock DJ, McLaughlin M, Prasad G et al. The interplay between DNA methylation, folate and neurocognitive development. Epigenomics 2016; 8:863–879. [DOI] [PubMed] [Google Scholar]
- 407.Jaworska J, Ziemka-Nalecz MALGORZATA, Zalewska T. Histone deacetylases 1 and 2 are required for brain development. Int J Dev Biol 2015; 59:171–177. [DOI] [PubMed] [Google Scholar]
- 408.Lv J, Xin Y, Zhou W, Qiu Z. The epigenetic switches for neural development and psychiatric disorders. J Genet Genomics 2013; 40:339–346. [DOI] [PubMed] [Google Scholar]
- 409.Fasolino M, Zhou Z. The crucial role of DNA methylation and MeCP2 in neuronal function. Gen 2017; 8:141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 410.Peng S, Yuan Z. HDAC2: A potential target for neurological diseases. Therapeutic Targets for Neurological Diseases 2015; 2:e723. [Google Scholar]
- 411.Sartor GC, Malvezzi AM, Kumar A, Andrade NS, Wiedner HJ, Vilca SJ et al. Enhancement of BDNF expression and memory by HDAC inhibition requires BET bromodomain reader proteins. J Neurosci 2019; 39:612–626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 412.Wu J, Liu C, Zhang L, Qu CH, Sui XL, Zhu H et al. Histone deacetylase-2 is involved in stress-induced cognitive impairment via histone deacetylation and PI3K/AKT signaling pathway modification. Mol Med Rep 2017; 16:1846–1854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 413.Penney J, Tsai LH. Histone deacetylases in memory and cognition. Sci Signal 2014; 7:re12–re12. [DOI] [PubMed] [Google Scholar]
- 414.Panayotis N, Sheinin A, Dagan SY, Tsoory MM, Rother F, Vadhvani M et al. Importin α5 regulates anxiety through MeCP2 and sphingosine kinase 1. Cell Rep 2018; 25:3169–3179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 415.Fyffe SL, Neul JL, Samaco RC, Chao HT, Ben-Shachar S, Moretti P et al. Deletion of Mecp2 in Sim1-expressing neurons reveals a critical role for MeCP2 in feeding behavior, aggression, and the response to stress. Neuron 2008; 59:947–958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 416.Moonat S, Sakharkar AJ, Zhang H, Tang L, Pandey SC. Aberrant histone deacetylase2–mediated histone modifications and synaptic plasticity in the amygdala predisposes to anxiety and alcoholism. Biol Psychiatry 2013; 73:763–773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 417.Su M, Hong J, Zhao Y, Liu S, Xue X. MeCP2 controls hippocampal brain-derived neurotrophic factor expression via homeostatic interactions with microRNA-132 in rats with depression. Mol Med Rep 2015; 12:5399–5406. [DOI] [PubMed] [Google Scholar]
- 418.Zimmermann CA, Hoffmann A, Raabe F, Spengler D. Role of mecp2 in experience-dependent epigenetic programming. Gen 2015; 6:60–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 419.Bhave SA, Uht RM. CpG methylation and the methyl CpG binding protein 2 (MeCP2) are required for restraining corticotropin releasing hormone (CRH) gene expression. Mol Cell Endocrinol 2017; 454:158–164. [DOI] [PubMed] [Google Scholar]
- 420.Ito K, Yamamura S, Essilfie-Quaye S, Cosio B, Ito M, Barnes PJ, Adcock IM. Histone deacetylase 2–mediated deacetylation of the glucocorticoid receptor enables NF-κB suppression. J Exp Med 2006; 203:7–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 421.La Noce M, Mele L, Laino L, Iolascon G, Pieretti G, Papaccio G et al. Cytoplasmic interactions between the glucocorticoid receptor and hdac2 regulate osteocalcin expression in vpa-treated MSCs. Cell 2019; 8:217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 422.Knopik VS, Maccani MA, Francazio S, McGeary JE. The epigenetics of maternal cigarette smoking during pregnancy and effects on child development. Dev Psychopathol 2012; 24:1377–1390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 423.Knopik VS, Marceau K, Bidwell LC, Rolan E. Prenatal substance exposure and offspring development: Does DNA methylation play a role? Neurotoxicol Teratol 2019; 71:50–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 424.Shorey-Kendrick LE, McEvoy CT, Ferguson B, Burchard J, Park BS, Gao L et al. Vitamin C prevents offspring DNA methylation changes associated with maternal smoking in pregnancy. Am J Respir Crit Care Med 2017; 196:745–755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 425.Slomski A. More folic acid for pregnant smokers. JAMA 2019; 321:1965–1965. [DOI] [PubMed] [Google Scholar]
- 426.Li W, Li Z, Li S, Wang X, Wilson JX, Huang G. Periconceptional folic acid supplementation benefit to development of early sensory-motor function through increase DNA methylation in rat offspring. Nutrients 2018; 10:292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 427.Wang X, Li Z, Zhu Y, Yan J, Liu H, Huang G, Li W. Maternal folic acid impacts DNA methylation profile in male rat offspring implicated in neurodevelopment and learning/memory abilities. Genes Nutr 2021; 16:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 428.Van Wersch JWJ, Janssens Y, Zandvoort JA. Folic acid, vitamin B12, and homocysteine in smoking and non-smoking pregnant women. Eur J Obstet Gynecol Reprod Biol 2002; 103:18–21. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No new data were generated or analyzed in support of this manuscript.
