7 External noxious substances as ADHD risk factors
- 7.1. Metals
- 7.2. Chemicals
- 7.2.1. Phthalates (+ 10 % to + 900 %)
- 7.2.2. Fluoridated drinking water (+ 510 % if 1 mg/L too high)
- 7.2.3. Polychlorinated biphenyls (PCBs) / polychlorinated biphenyl ethers (+ 26 % to + 92 %)
- 7.2.4. Perfluorooctane sulfonate (PFOS) (+ 77 %)
- 7.2.5. β-Hexachlorocyclohexane (β-HCH) (+ 75 %)
- 7.2.6. Chlorinated kerosenes (+ 57 % / quartile)
- 7.2.7. Benzene, toluene, ethylbenzene, xylene/xylene (BTEX) (+ 54 %)
- 7.2.8. Polyvinyl chloride (PVC)
- 7.2.9. Bisphenol A
- 7.2.10. Perfluoroalkyl compounds
- 7.2.11. Triclosan
- 7.2.12. Insecticides / pesticides
- 7.2.13. Spray disinfectant
- 7.3. Passive smoking - smokers in the environment in the first years of life (+ 42 % to + 170 %)
- 7.4. Air pollution in childhood
- 7.5. Pets (+ 58 % to + 66 %)
- 7.6. Printing ink on food paper
- 7.7. Synergy effects of neurotoxins
- 7.8. Factors without risk increase for ADHD
- 7.9. Factors with risk reduction for ADHD
7.1. Metals
7.1.1. Copper (up to + 1,546 % (?))
ADHD risk increased by 1,546% in the half of Spanish children (8 to 15 years) with higher urinary copper levels.1 Copper was also associated with increased inattention symptoms.
Note: If one were to assume an ADHD baseline prevalence of 6.47 % in children, this would result in a prevalence of 100 % in those 50 % children with the higher copper urine values.
7.1.2. Lead (+ 160 % to + 310 %)
According to several meta-analyses, lead exposure during development increased the risk of ADHD2 by
- 433 % in the half of Spanish children (8 to 15 years) with the higher urinary lead exposure.3
- 310 % increased) for the 20 % with the highest blood lead level compared to those with the lowest 20 %4
- 239 to 306 %5
- 160 to 260 %6
- 95 %7
- Every doubling of blood lead levels increased the risk of ADHD in children by 35%8 This was true even at very low lead levels of less than 10 mug/dl.
Mice that are chronically exposed to inorganic lead from birth show about three times more spontaneous motor activity than control mice. In addition, their behavioral responses to amphetamine, methylphenidate and phenobarbital are altered.9
Elevated lead urine levels correlated with increased inattention and hyperactivity symptoms and reduced IQ in children.10
Children with ADHD had significantly higher lead hair levels than children without ADHD.11
Elevated blood lead levels lead to an increased risk of ADHD.1213141516 With a blood lead level of ≥ 5 μg/dl, the risk of ADHD was found to be 2.33 times higher (OR 2.33).17
A review of k = 45 meta-analyses found a 1.96-fold ADHD risk.18
Leaded gasoline was found to be responsible for a 0.42 standard deviation increase in ADHD, particularly among those born between 1966 and 1986 (Generation X). The overall susceptibility to mental disorders in the population increased by 0.13 standard deviations and included an estimated increase of 151 million mental disorders due to lead exposure.19
Development paths:
Lead influenced the dopamine balance in many studies.
- Reduced dopamine signaling
- Increased dopamine signaling
The DRD2 gene variant rs1800497r is said to promote a link between ADHD and lead.24 A connection to certain MAO-A gene variants is also mentioned, which causes lower serotonin degradation.25 A study in rats suggests interactions between lead exposure and early stress on the dopaminergic system.26 A long-term study found no directly increased risk of ADHD in people with previous lead exposure, but increased externalizing behaviours and increased risk of addiction27
In one study, lead altered neostriatal serotonin and norepinephrine levels, increased anxiety and decreased open-field activity.28
Lead exposure during pregnancy may increase the risk of ADHD. See there.
Even a lead content in drinking water below the limit values is said to be problematic.29
Increased lead absorption can occur from old water pipes. In principle, lead water pipes are not very dangerous in areas with calcareous water, as lime forms a reliable protective layer in the pipes. However, if a water softening system is installed, this protective lime layer can be lost. If old lead pipes are still present, this can lead to increased lead absorption.
Lead is barely relevant as a toxin in Central Europe. In less developed countries, however, lead can be a serious problem.
In children who have been exposed to lead, succimer chelation can produce lasting cognitive benefits if chelation sufficiently reduces the lead concentration in the brain. At the same time, succimer treatment without lead exposure leads to permanent cognitive dysfunction.30
7.1.3. Cadmium (+ 269 % ?)
The risk of ADHD was 269% higher in the half of Spanish children (8 to 15 years) with higher urinary cadmium levels.1 Cadmium was also associated with increased inattention symptoms.
In a study of children with and without ADHD, children with ADHD-I had the highest urinary cadmium levels.10 Cadmium correlated negatively with IQ.
0 %: Cadmium exposure during development did not significantly increase the risk of ADHD, according to a meta-analysis7
7.1.4. Manganese (+ 163 % to + 257 %)
According to a meta-analysis, exposure to manganese during development increased the risk of ADHD by
- 163 % (meta-analysis, k = 47)31
- 79 %7
- Early exposure to manganese causes permanent attention problems via the mTOR pathway and an alteration of the catecholaminergic system32 as well as sensorimotor problems.33
Manganese intoxication shows a correlation with certain CYP2D6 gene variants34
MPH reduced the attention problems and sensorimotor problems caused by early manganese exposure in rats. 0.5 mg/kg/d completely improved the attention problems, but only after prolonged treatment, 3.0 mg/kg/d improved the sensorimotor deficits immediately. Selective antagonization of D1, D2 or α2A receptors did not affect the manganese-induced attention problems or their improvement by MPH. D2R antagonists reduced the sensorimotor deficits of manganese. D1 antagonists reduced the efficacy of MPH on sensorimotor deficits.33
In persons with ADHD, elevated manganese levels were only found in the hair, but not in the blood35, another study also found elevated manganese blood levels in schoolchildren with ADHD.36 Children with ADHD had manganese hair levels twice as high as children without ADHD. High manganese hair levels increased the risk of ADHD 3.57-fold (+257%).11
In children with ADHD, urinary manganese levels were slightly lower than in children without ADHD.10
A doubling of the manganese content in teeth from both the prenatal and postnatal period increased the risk of attention problems and ADHD symptoms in the school years by 5%. Manganese from childhood showed no influence.37
An animal model with developmental manganese exposure showed that manganese can cause permanent attention and sensorimotor deficits resembling ADHD-I. Oral methylphenidate was able to fully compensate for the deficits caused by early manganese exposure.30
An Israeli study addressed the fact that manganese levels in infant formula were often higher than in breast milk and recommended stricter limits.38
A study reports benefits of choline supplementation during pregnancy in rats to prevent manganese-induced developmental disorders in the offspring39
7.1.5. Antimony (up to + 204 %)
ADHD risk 204% higher in the half of Spanish children (8 to 15 years) with higher urinary antimony exposure.1
In a study of children with and without ADHD, children with ADHD-H/I showed the highest antimony urine levels.10 Antimony urine levels correlated with the severity of ADHD symptoms according to the teacher rating.
7.1.6. Mercury (up to + 168 %)
Exposure to methylmercury during development increased the risk of ADHD by 168% (meta-analysis, k = 47)40 One review found an increased risk of ADHD and ASD.41 Two reviews describe causality.426
Two meta-analyses found no significant increase in risk from mercury.72
Mercury urine levels were not significantly elevated in children with ADHD.10 Mercury blood levels were unchanged in children with ADHD.8
The correlation between mercury exposure and ADHD appears to be influenced by DNA methylation.43
There is weak evidence of relevance in ADHD.van de Bor (2019): Fetal toxicology. Handb Clin Neurol. 2019;162:31-55. doi: 10.1016/B978-0-444-64029-1.00002-3.}}44
A large study with n = 2073 participants was unable to establish a connection between amalgam and ADHD.45
Mercury is also suspected of being a possible contributory cause of Parkinson’s disease.46 This would be a clear indication of a damaging effect on the dopamine system.
7.1.7. Inorganic arsenic (+ 53 % to + 102 %)
Those children who were among the 20% with the highest urinary arsenic levels were found to have double the risk of ADHD (OR 2.02).17
According to a meta-analysis, exposure to arsenic during development increased the risk of ADHD by 53%7
Wistar rats given arsenic (50% of the lethal dose, 8 mg/kg) developed hyperactivity.47
7.1.8. Zinc
One study found elevated zinc blood levels in school children with ADHD.36
7.2. Chemicals
7.2.1. Phthalates (+ 10 % to + 900 %)
According to a meta-analysis, exposure to phthalates during development increased the risk of ADHD by 212% for girls and 254% for boys.48
Higher phthalate metabolites in children’s urine correlated with increased likelihood of ADHD by a factor of 3 to 9.49
Another study found that phthalate exposure in early childhood only increased ADHD symptoms by 10% in children with ASD, primarily in the direction of externalizing behaviors.50
A study of n = 67 drug-naïve children with ADHD aged 6 to 16 years found a significant correlation between urinary phthalate metabolites MEHHP (mono-[2-ethyl-5-hydroxyhexyl] phthalate) and MEOHP (mono-[2-ethyl-5-oxohexyl] phthalate) and commission error T-scores on the visual test for extended attention, which is a marker of impulsivity.51
Boys with ADHD showed significantly increased levels of mono-n-butylphthalate and ethylparaben, whereas girls with ADHD did not. Neither boys nor girls showed significant differences in testosterone, free testosterone, FSH, LH, estradiol, progesterone or SHBG compared to controls. In boys with ADHD, elevated urinary levels of monobenzyl phthalate and monoethylhexyl phthalate correlated with serum testosterone. In girls with ADHD, urinary levels of monoethyl phthalate correlated positively with serum levels of LH, testosterone and free testosterone.52
7.2.2. Fluoridated drinking water (+ 510 % if 1 mg/L too high)
A review of k = 45 meta-analyses found a 3.8-fold risk of cognitive deficits due to fluoride exposure in childhood.18
In Canada, a study found that a 1 mg/liter increase in fluoride levels in drinking water above acceptable limits increased the risk of ADHD by 6.1 times in 6- to 17-year-olds. In 14-year-olds living in areas where fluoride was added to drinking water, there was a 2.8-fold risk of an ADHD diagnosis compared to 14-year-olds in areas without fluoridated drinking water. Older children responded with a higher risk.53 Fluorine urine levels, on the other hand, did not correlate with ADHD (1,877 subjects).
A study in Mexico found a link between increased prenatal fluoride exposure and inattention and ADHD, but not hyperactivity54 and cognitive problems.55 Another study found similar results.56 A review summarizes the results.57
A longitudinal study over 22 years from Florida found a significant slight increase in the risk of ADHD and ASD as well as mental retardation and developmental delay for fluoridated water.58
A smaller long-term study in Sweden found an increased risk of ADHD due to fluorides.59
In Germany, 90% of drinking water has a fluoride content of 0.3 mg/liter. Drinking water is not fluoridated in Germany.60 However, fluoridated table salt is available.
A study found an inverse correlation between fluoride levels in the mother’s urine during pregnancy and cognitive problems in the offspring at the age of 11. The higher the pregnancy urine fluoride content, the lower the cognitive problems.61 This was not consistent with the results of other studies, which found an increased risk of ADHD with increased urine fluoride levels in the children themselves.
Sodium fluoride in drinking water (20 ppm to 100 ppm) led to a dose-dependent reduction of dopamine, noradrenaline and acetylcholine in the brain of rats, while the levels of adrenaline, histamine, serotonin and glutamate increased.62
7.2.3. Polychlorinated biphenyls (PCBs) / polychlorinated biphenyl ethers (+ 26 % to + 92 %)
Polychlorinated biphenyls and polychlorinated biphenyl ethers are suspected of causing ADHD.
PCBs are banned in many countries, in Germany since 1989. PCBs were used in particular as lubricants and coolants in electrical appliances and as building materials. Due to their chemical stability, many areas around the world are still contaminated with PCBs. Contaminated food, especially seafood from contaminated rivers and lakes, is the most common source of contamination today.6364
Even low levels of PCB exposure during development impair neurobiological, cognitive and behavioral functions.64
One study found a 26% to 92% increased risk of ADHD.65 Individual studies found contradictory or weak impairments,6667 however, the vast majority show evidence of relevance in ADHD.6869
Polychlorinated biphenyls affect the dopamine system.70 PCBs inhibit dopamine synthesis as well as the storage of dopamine in the vesicles and its release, thereby causing low dopamine levels7115 in the basal ganglia and PFC7273 71 74 75 76 , as well as reduced DAT in the striatum77, which overall corresponds quite closely to the picture of ADHD.
Exposure to polychlorinated biphenyls (PCBs) in childhood correlated with an increased risk of ADHD.6
Prenatal PCB exposure slightly reduced internalizing symptoms by 17% and slightly reduced ADHD risk by 16%.78
Rats that were exposed perinatally to A1221 (a PCB mixture) showed79
- a reduced sucrose preference (females only)
- a reduced reaction latency in the attention shifting task (males and females)
- TH+ cells in the VTA increased (males and females)
- DRD1 increased in the combined midbrain nuclei (males and females)
- Behavior correlated with number of dopaminergic cells in the VTA (in females)
- Behavior correlated with dopamine signaling genes (in males)
- Conditioned orientation and serum estradiol (E2) unchanged
Prenatal exposure to PCBs has an adverse effect:
- Hyperactivity (in rats even at subtoxic doses)7115
- IQ, memory, attention 80
- Memory, attention81
- Impulsivity (via corpus callosum)8283 in rats even at subtoxic doses7115
- Male and female offspring were trained as adults to perform asymptotically in a differential reinforcement of low rates (DRL) task. The PCB-exposed groups had a lower ratio of reinforced to non-reinforced responses than the control groups.70
- no effect on sustained attention66
Possible path of action: Gap junctions84
7.2.4. Perfluorooctane sulfonate (PFOS) (+ 77 %)
Perfluorooctane sulfonate (PFOS) in breast milk correlated with a 77% increased risk of ADHD per higher interquartile range.85
PFOS caused ADHD symptoms (hyperactivity, cognitive problems) in zebrafish, decreased dopamine levels and the number of dopaminergic neurons, and disrupted the transcriptional profiles of genes related to the dopaminergic system. MPH alleviated the ADHD symptoms induced by PFOS and restored DA levels, the number of dopaminergic neurons and the expression of genes associated with DA metabolism.86
7.2.5. β-Hexachlorocyclohexane (β-HCH) (+ 75 %)
β-Hexachlorocyclohexane (β-HCH) in breast milk correlated with a 75% increased risk of ADHD per higher interquartile range.85
7.2.6. Chlorinated kerosenes (+ 57 % / quartile)
Chlorinated kerosenes (CP) are widespread environmental and industrial pollutants.
A large study (n = 122,965, including 7,139 with ADHD) investigated the ADHD risk of PM2.5, PM2.5-bound short-chain CPs (SCCPs), medium-chain CPs (MCCPs) and long-chain CPs (LCCPs). An increase in CP concentrations in the interquartile range (IQR) increased the risk of ADHD by 57% (OR = 1.57). The risk increased linearly in a dose-dependent manner and was particularly driven by SCCPs and MCCPs.87
7.2.7. Benzene, toluene, ethylbenzene, xylene/xylene (BTEX) (+ 54 %)
Higher exposure to these substances in the air correlated with a 1.54-fold increase in the risk of ADHD at kindergarten age.88
7.2.8. Polyvinyl chloride (PVC)
One review describes a suspected correlation between PVC exposure and ADHD.89
7.2.9. Bisphenol A
Bisphenol A is suspected of increasing the risk of ADHD.15 A connection with certain MAO-A gene variants that cause lower serotonin degradation25 and an influence on the thyroid balance is being discussed.90
A meta-analysis found a clear link between bisphenol exposure and ADD(H)S.91
A study of n = 67 drug-naïve children with ADHD aged 6 to 16 years found no significant correlation between bisphenol A, bisphenol F or bisphenol S in urine and ADHD symptoms.51
Possible path of action: Gap junctions84
7.2.10. Perfluoroalkyl compounds
Elevated levels of perfluoroalkyl compounds have been observed in ADHD.92
7.2.11. Triclosan
Triclosan is a polychlorinated phenoxyphenol.
Prolonged exposure to the environmental pollutant triclosan induced ADHD symptoms in rats. Triclosan appears to cause a reduction in dopamine levels in the PFC.93
A 60-day continuous exposure of rats to triclosan caused ADHD-like behavior in the offspring. It activated microglia in the PFC, which led to the release of inflammatory factors. In vitro, triclosan increased the levels of inflammatory cytokines, including IL-1β, IL-6 and TNF-α, in HMC3 cells. In addition, triclosan upregulated PKM2 via hnRNPA1, which affects the STAT3 signaling pathway and thus continuously activates microglia, promoting the release of inflammatory cytokines.94
7.2.12. Insecticides / pesticides
Elevated pesticide urine levels (here: Pentachlorophenol (PCP), 3,5,6-trichloro-2-pyridinol (TCPy) and carbofuranphenol (CFP)), particularly in early childhood, led to increased ADHD symptoms in later childhood.95
With regard to pesticides (especially organochlorine compounds, pyrethroids, organophosphates), there are indications of relevance in ADHD.6815
For pesticides during pregnancy and ADHD, see there.
7.2.12.1. Organochlorine compounds
With regard to organochlorine compounds, there are indications of relevance in ADHD.681596
A study of Greek schoolchildren with ADHD found no elevated blood serum levels of97
- Dichlorodiphenyltrichloroethane (DDT) Metabolites
- Hexachlorocyclohexane (HCH) isomers
- Cyclodienes
- Methoxychlorine
Other organochlorine compounds are:84
- Lindane
- Dieldrin
- Chlordane, endosulfan, heptachlor, aldrin
- Chlordecone
- Mirex
Possible path of action: Gap junctions84
7.2.12.2. Organophosphates
According to a large number of studies, organophosphate pesticides have a correlation between prenatal and postnatal exposure and ADHD 6498 99 100 96 101 or a theoretically possible increase in ADHD risk.102 One source suggests an increased risk of ADHD from organophosphates, particularly when coinciding with a particular MAO-A gene variant that causes lower serotonin degradation.25
In contrast, two larger studies found no influence 103104
With regard to hyperactivity, 2 studies found an association between organophosphates and hyperactivity, 4 studies found no association.105
Exposure to organophosphates in childhood correlated with an increased risk of ADHD.6
A study examined the urine of 186 Taiwanese children with and without ADHD for106
- organophosphate flame retardants (OPFRs)
- TDCPP (1,3-dichloro-2-propyl phosphate)
- TnBP (tri-n-butyl phosphate)
- TCEP (tris(2-chloroethyl) phosphate)
- TBEP (tris(2-butoxyethyl) phosphate)
- TPHP (triphenyl phosphate)
- and their metabolites
- BDCPP (bis(1,3-dichloro-2-propyl)phosphate)
- DNBP (di-n-butyl phosphate)
- BCEP (bis(2-chloroethyl)hydrogen phosphate)
- DBEP (di-(2-butoxyethyl) phosphate)
- DPHP (diphenyl phosphate)
In children with and without ADHD, BCEP and its metabolites were primarily found in the urine.
Children with ADHD showed significantly higher urinary levels of BDCPP, BCEP, DBEP, DPHP, TCEP, TBEP, TNBP, TPHP and Σ10OPFR. After controlling for age, gender, body mass index, PM2.5 exposure scenarios and phthalate metabolites, parabens, bisphenol-A and urinary creatinine, the levels of BDCPP, TDCPP and TBEP showed significant and dose-dependent effects on inattention. DNBP correlated positively with neuropsychological deficits (CPT recognizability, omission and commission). DPHP correlated negatively with CPT recognizability and commission.
Hyperactivity and impulsivity did not correlate with OPFRs or their metabolites.
Pathways of action (among others): Endocannabinoid pathway107
7.2.12.2.1. Chlorpyrifos
In children between 1 and 6 years of age, chlorpyrifos residues in the blood correlated with the risk of ADHD.108 Vitamin D reduced the risk.
Chlorpyrifos also significantly increases the risk of ADHD in the case of prenatal contamination of the mother during pregnancy.
One study found no correlation of chlorpyrifos with hyperactivity in rats109 while another found it in females.110
A study in rats was able to induce ADHD-like behaviors in Wistar and SHR rats by organophosphates and found strong circumstantial evidence that these are mediated by reductions in fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL) via the cannabinoid receptor.111
Blood values were measured in Egyptian adolescents, some of whom used pesticides, and the parents were asked about ADHD symptoms in the adolescents:112 There was no correlation with ADHD in relation to the organophosphate chlorpyrifos.
Higher vitamin D levels appear to reduce the negative effect of chlorpyrifos on the risk of ADHD.101
7.2.12.3. Pyrethroids (+ 142 %)
Various studies indicate a correlation between pyrethroid exposure in childhood and neurodevelopmental disorders such as ADHD with a 2.42-fold risk of ADHD 113 Other studies also found an association with ADHD11496 , ASD or developmental delay.64
Blood levels were measured in Egyptian adolescents, some of whom used pesticides, and the parents were asked about ADHD symptoms in the adolescents:112 A correlation to ADHD was found in relation to the pyrethroid λCH through the measured value Cis-DCCA (all persons with ADHD reported clinical ADHD symptoms).
The pyrethroid pesticide deltamethrin at low doses causes changes in ADHD- and NDD-related behaviors and in the striatal dopamine system during development in male mice.
Deltamethrin during development caused a multimodal biophenotype in the brain relevant to ADHD. Mouse mothers received deltamethrin (3 mg/kg or vehicle every 3 days) during gestation and lactation, which is well below the limits set by the EPA. Male offspring showed alterations in several canonical clock genes. Kinome analysis revealed alterations in the activity of several kinases involved in synaptic plasticity, including mitogen-activated protein kinase (MAP) ERK. Multiomics integration revealed a dysregulated protein-protein interaction network with primary clusters for MAP kinase cascades, regulation of apoptosis and synaptic function115
The annual number of ADHD cases caused by pyrethroids per million inhabitants was estimated at116
- 2189 for Israel
- 1710 for France
- 969 for Iceland
- 944 for Switzerland
- 209 for Germany
According to a meta-analysis, pyrethroid insecticides did not significantly increase the odds ratio for ADHD (0.99).48
7.2.12.4. Carbamate (-)
One review found no associations between carbamates and ADHD.96
7.2.12.5. Neonicotinoids (- ?)
One review found no links between neonicotinoids and ADHD, although there were few studies on this topic.96
7.2.13. Spray disinfectant
Spray disinfectants led to lung diseases in South Korea, as extreme use was widespread there.
One study found a link between spray sanitizer use, especially in early childhood or prolonged use, and ADHD.117
7.3. Passive smoking - smokers in the environment in the first years of life (+ 42 % to + 170 %)
Passive nicotine exposure increases the risk of ADHD.118 Nicotine exposure in children is associated with a 1.42-fold119 to 2.7-fold48 increase in ADHD.120 In one study, children with ADHD were twice as likely to have smokers in the family as children who were not affected.121
In the case of passive smoking, a connection is made to certain MAO-A gene variants that cause a lower level of serotonin degradation.25
In children, a linear association was found between salivary cotinine (a nicotine breakdown product) and hyperactivity and behavioral problems. This association remained significant after adjusting for family poverty, parental education, a history of ADHD, hostility, depression, caregiver IQ, and obstetric complications, and also after excluding from the calculation children of mothers who had smoked during pregnancy. This indicates that exposure to nicotine in the first years of life alone can increase hyperactivity and behavioral problems.122
One study found no link between smoke exposure after birth and ADHD.4
Tobacco smoke exposure can result from secondhand smoke, i.e. the involuntary inhalation of smoke from other people who are actively smoking, and also thirdhand smoke, i.e. involuntary exposure to smoke from objects that come into contact with tobacco smoke contaminants, such as skin, hair, furniture, clothing and dust.123 15.5% of children aged 0 to 17 years exposed to tobacco smoke showed ADHD, which corresponded to a 30% increased risk (OR 1.3).
The comorbidity of smoking with ADHD is 40%.124 In contrast, around 25% fewer of the total population smoke, namely 26.9% of women and 32.6% of men.125
7.4. Air pollution in childhood
Air pollution consists of a complex mixture of126
-
Particulate matter (PM2.5 and PM10)
-
Nitrogen oxide (NO)
-
Nitrogen dioxide (NO2)
-
Carbon monoxide (CO)
-
Sulphur dioxide (SO2)
-
Ozone (O3)
-
volatile organic compounds (VOC)
-
Nitrous oxide (laughing gas, N2O)127
-
Children in the top third of exposure to organic substances showed a 1.51-fold ADHD risk (+51%) compared to the bottom third128
-
Children in the top third of soot exposure showed a 1.29-fold ADHD risk (+29%) compared to the bottom third128
-
Children in the top third of sulfate exposure showed a 1.20-fold ADHD risk (+20%) compared to the bottom third128
A large Chinese study of 8,692 children aged 6 to 12 found a significant increase in the children’s risk of ADHD due to:129
- Home renovations
- Incense burning
- Cooking oil vapors
- Smokers in the household
Pathways of air pollution on ADHD
Pathways of air pollution on ADHD:126
- Inflammation
- Thyroid hormones
- Disorder of β-adrenergic, dopaminergic and glutamatergic (NMDAR) signaling pathways, resulting in disorder of G-protein/cAMP signaling, Ca2+ homeostasis and neurotransmitter pathways
- (reversible) inhibition of alpha-7-nicotinic acetylcholine receptors (α7 nAChRs) by N2O exposure
Particulate matter also acts via the gut microbiome. The intestinal microbiome in turn acts on the oxytocin system - in particular via L. reuteri. More on this under Microbiota against ASS in the article Gut-brain axis and ADHD
Since particles up to a maximum of 1000 nanometers = 1 micrometer can cross the blood-brain barrier (microplastics up to 200 nanometers133, extracellular vesicles up to 1000 nanometers - see Modulation of neurotransmitters by the microbiome In the article Gut-brain axis and ADHD), PM10 particulate matter (particulate matter of less than 10 micrometers, up to over 2.5 micrometers) should not be able to cross the blood-brain barrier directly. However, PM2.5 (particulate matter with 50% of 2.5 micrometers, a larger proportion below and a smaller proportion above) can also be smaller than 2.5 micrometers.134 If the studies do not differentiate between PM2.5 and PM1, it must therefore be assumed that PM2.5 also includes common blood-brain barrier particles.
On its surface, particulate matter carries foreign substances such as heavy metals, chemicals and volatile organic compounds into the body.135 Estimates that a person ingests 1 to 5 grams of microplastics (plastic up to a maximum of 5 mm) per week136 are criticized as being orders of magnitude too high137. A credit card weighs 5 grams. According to the same (criticized) WWF study, there will be 0.33 tons of plastic in the oceans for every ton of fish in 2025. However, the same amount of plastic, broken down into smaller particles the size of fine dust, has a much larger surface area. While a bank card has a surface area of 0.009 m² for 5 grams of plastic, 5 grams of particulate matter PM10 is likely to have a surface area of 2.5 to 7.5 m². 5 grams of PM1 are likely to have a surface area of around 500 square meters, i.e. more than 50,000 times as much. This shows that the input of pollutants via the surface of fine particulate matter is, depending on the type of particulate matter, much higher than that of microplastics.
Nevertheless, the input of polycyclic aromatic hydrocarbons via microplastics found in marine animals appears to be very low when ingested by humans 138137
9 % of microplastic dust up to 5 mm in Austria results from tire abrasion.139
Around 4% of the particles inhaled by humans are the result of microplastics, the other 96% are of natural origin.140
Mineral water from plastic bottles did not contain more microplastics than mineral water from glass bottles.137
The proportion of microplastics in cosmetics fell by 97% between 2012 and 2017.137 Microplastics for abrasion (microbeads) were banned by the EU in 2023, with long transition periods in some cases.
Plastic textiles lose considerable amounts of microplastics when worn and washed.
7.4.1. Particulate matter
The percentages indicate the second highest value of the collected studies.
The largest sources of particulate matter are the combustion of fuels in motor vehicles, combustion processes in industrial plants, forest fires and bush fires. 28 % of particulate matter results from diesel exhaust gases, which cause PM10 to PM30 in particular.126
A study of single nucleotide polymorphisms associated with air pollutants found141
- NO2, genetically predicted, causally caused an increased risk of
- Major depression (OR: 1.13, + 13 %)
- bipolar Disorder (OR: 1.26, + 26 %)
- Schizophrenia (OR: 1.57, + 57 %)
- ADHD (OR: 1.61, + 61 %)
- ASS (OR: 1.39, + 39 %)
- PM2.5, genetically predicted, causally caused an increased risk of
- severe depression (OR: 1.21, + 21 %)
- bipolar Disorder (OR: 1.32, + 32 %)
- ADHD (OR: 1.57, + 57 %)
- NOx, genetically predicted, causally caused an increased risk of
- ADHD (OR: 1.64, + 64 %)
- PM10, genetically predicted, causally caused an increased risk of
- ADHD (OR: 1.70, + 70 %)
7.4.1.1. Particulate matter PM10 (up to + 288 %)
One study found a 97% increase in the risk of ADHD in children with a 10 μg/m³ increase in PM10.142
- ADHD was found in143
- 11.0% of urban children living in cities who were exposed to air pollution, especially particulate matter (PM10)
- 2.7 % in the control group
- Risk thus + 307 %
- Children in the highest third of PM10 exposure had a 3.88-fold risk of ADHD (+288%) compared to children in the lowest tercile144
A Korean cohort study found a 44% increase in the risk of ADHD in children and adolescents for every 10 µg/m³ increase in PM10, with a tendency towards dose-dependent symptom severity.145 A Taiwanese register study came to comparable results.146
7.4.1.2. Particulate matter PM2.5 (up to + 82 %)
PM2.5 (airborne particles with a diameter of less than 2.5 μm) can easily penetrate biological systems. They are small enough to reach the alveoli and enter the brain via the bloodstream or the olfactory bulb.147148
- A review of k = 45 meta-analyses found a 1.82-fold ADHD risk (+82%) from PM2.5 exposure.18
- There is clear evidence of a link between PM2.5 and NO2 levels
- Children in the highest quintile of PM2.5 exposure had a 1.70-fold risk of ADHD (+70%) compared to children from the lowest quintile152
- Children in the top third of PM2.5 exposure showed a 1.37-fold ADHD risk (+37%) compared to the bottom third 128
Another cohort study found a 40% to 78% increased risk of ADHD from PM2.5 exposure in the first to third year of life. The risk was associated with PM2.5 >16 μg/m3 and increased sharply with PM2.5 > 50 μg/m3. There was no gender-dependent association.153
A meta-analysis of 12 studies found a correlation between particulate matter and ADHD in children in 9 of them.154
A longitudinal study of 2,750 children found an increased risk of ADHD and ASD from particulate matter PM2.5 and PM10, but not from ambient noise, ozone, sulfur dioxide, soot, nitrogen dioxide, or nitrogen oxide155
In one study, inhaled printer particles led to a 5-fold increase in dopamine levels in rats, although this was probably due to increased synthesis and not reduced degradation.156
A database genome study (n = 423,796) found that particulate matter (<2.5 pm) correlated with a 95% increased risk of ADHD. The risk for anxiety disorders was increased by 196%, for schizophrenia by 55% and for depression by 33%. Bipolar was negatively correlated, ASD, PTSD, obsessive-compulsive disorder and anorexia nervosa showed no significant correlation.157
7.4.1.3. Ultrafine dust 23nm
Ultrafine particulate matter 23nm has a size of about 1/100 of fine particulate matter PM2.5. Ultrafine particulate matter 23nm is a common lower limit of particle size measurement when analyzing vehicle emissions.
Plastic waste becomes microplastic through abrasion and decomposition and later also nanoplastic the size of ultrafine dust.
Chronic exposure to 23 nm polystyrene nanoplastics at a dose of 10 µg/day/kg (which corresponds to the reality in humans) caused158
- in case of exposure of the dams
- important developmental milestones of the offspring disrupted
- ADHD symptoms in offspring in adulthood
- Hyperactivity
- increased risk behavior
- impaired motor learning and executive functions
- Exposure of aging mice
- lower epilepsy threshold
- Development of epileptic seizures
- Behavioral changes were related to altered gene and synaptic protein expression associated with ADHD and epilepsy.
- lifelong exposure to nanoplastics
- lysosomal dysfunctions
- increased lipofuscin accumulation, which indicates accelerated aging of the brain
7.4.1.4. Pathways of particulate matter on ADHD
Pathways of particulate matter on ADHD
The toxicological profile of PM2.5 includes various toxic components that can trigger oxidative stress and neuroinflammation, such as126
- polycyclic aromatic hydrocarbons (PAHs)
- PAHs (e.g. benzo[a]pyrene, B[a]P) bind directly to noradrenergic β2A receptors, which are important for neurological developmental processes in the fetus
- PAHs can trigger desensitization of the β2AR via downstream signaling pathways159
- Disorder of β2AR signaling by PAHs (along with the aryl hydrocarbon receptor, AhR, to which PAHs also bind) may be a key mechanism for neurotoxicity.160 PAHs influence the gene regulation of NMDAR subunits.
- PAH disrupt Ca2+ homeostasis, which impairs BDNF signaling161
- Heavy metals
- organic and inorganic compounds
- reactive gases
PM2.5 exposure causes162
- reduced survival rate of nerve cells
- Morphology of the mitochondria disturbed
- ATP level reduced
- mRNA and protein expression reduced by
- Survival genes (CRB and Bcl-2)
- neuroprotective genes (PPARү and AMPK)
- increased oxidative stress
- Expression of inflammatory mediators increased in SH-SY5Y neurons (TNF-ɑ, IL-1β and NF-κB)
- Damage to the lungs147
- hypoxia-related damage
- Inflammatory reactions
PM2.5 exposure163
- reduced viability and increased apoptosis in hippocampal neurons
- disrupted synaptic ultrastructure and synapse-related protein expression
PKA/CREB/BDNF mediates the damaging effects of PM2.5 The neuronal and synaptic damage induced by PM2.5163
-
were aggravated by reduction of PKA/CREB/BDNF
-
were improved by increasing KA/CREB/BDNF
-
Neurotrophin-3 moderated just under 10% of the correlation between PM2.5 and ADHD and just under 6% of the correlation between PM2.5 and schizophrenia157
Exposure to particulate matter can126
- directly or indirectly cause oxidative stress and inflammation
- interfere with thyroid hormone signaling, with Consequences
- an underactive thyroid
- a reduced BDNF level
- can lead to dysregulation of GABAergic interneuron function
- impair synaptogenesis and cause dysfunction of the neuronal network
- Disrupt neurotransmitter signaling pathways
- β-adrenerg
- Dopamine
- Glutamate (NMDAR)
- Consequences: Disorder of G-protein/cAMP signaling, Ca2+ homeostasis and neurotransmitter pathways
7.4.2. Nitrogen oxides: nitrogen oxide (NO), nitrogen dioxide (NO2) (up to + 110 %)
Nitrogen dioxide (NO2) is a harmful, highly reactive gas that serves as a standard indicator for the group of nitrogen oxides (NOx). NO2 is mainly produced outdoors by the combustion of fuels in vehicles and power plants, and indoors by the combustion of fuels such as wood and gas.168
- Children in the highest third of NO exposure had a 2.1-fold risk of ADHD (+ 110%) compared to children in the lowest tercile144
- Children in the highest quintile of NO2 exposure had 1.63 times the risk of ADHD (+ 63%) compared to children from the lowest quintile152
A large cohort study found a statistically significant association between nitrogen oxides and particulate matter (<2.5 pm) in childhood and the development of ADHD.152 A smaller cohort study confirmed this for particulate matter, but not for nitrogen dioxide169
In another study, the risk of ADHD increased by 38% for every 10 μg/m3 increase in nitrogen oxide and by 51% for every 5 μg/m3 increase in particulate matter PM2.5. If both factors were considered together, the influence of nitrogen oxide predominated. A meta-analysis of k = 28 studies found similar results for the majority170
One study found no association between exposure to PM2.5 and NO2 at age 12 and ADHD at age 18, but with depression at age 18.171
One study found a 32% increase in the risk of ADHD in children due to a 10 μg/m3 increase in nitrogen oxide.142
A meta-analysis examined the effects of traffic-related air pollution on the neurological development of children in several countries using PM2.5 (particulate matter <2.5 µm), PM10, elemental carbon (EC), black carbon (BC), NO2 and NOx:172
Increased traffic-related air pollution correlated with an increase in ADHD, autism and impaired cognitive development.
PM2.5 reduced the expression of BDNF in the placenta.
Elevated PM2.5 concentrations impaired the cognitive development of adults (episodic memory) and increased major depressive disorders.
Elevated NO2 concentrations correlated with dementia, NOx with Parkinson’s disease.
Children in schools with higher levels of chronic traffic-related air pollution (elemental carbon, nitrogen dioxide and ultrafine particulate matter [10 to 700 nm]) in the schoolyard and classroom showed slower cognitive development. The improvement in working memory was only 7.4 % (compared to 11.5 %). Growth was reduced in all cognitive measures. Moving from the first to the fourth quartile of indoor elemental carbon levels reduced the gain in working memory by 13.0%.173 Another study found similar results.174
Exposure to nitrogen dioxide, ozone and sulfur dioxide has been linked to behavioral and developmental disorders, anxiety to particulate matter (PM10), ozone and sulfur dioxide, and overall pollution to ADHD and eating disorders175
Prenatal exposure to NO2 or PM2.5 did not additionally increase the risk of ADHD compared to exposure to the two pollutants in the first five years of life.152
Elevated levels of NO2 and particulate matter in schools appear to impair the development of working memory. For each interquartile range increase in exposure, the annual development of working memory decreased176
- by 20 % for NO2 outdoors
- by 19.9 % for ultrafine particulate matter indoors
Individual differences in susceptibility to air pollution appear to be related to the ε4 allele of the apolipoprotein E gene (APOE), which is the most important genetic risk factor for Alzheimer’s disease. PAH, EC and NO2 correlated significantly only in carriers of the APOEε4 allele177
- Behavioral problems,
- a smaller decrease in inattention over time
- a smaller caudate volume
A Polish study found a correlation between178
- long-term NO2 and PM10 exposure with poorer visual attention in children with ADHD
- short-term NO2 exposure with less efficient executive attention and higher impulsivity in TD children and more errors in children with ADHD
- short-term PM10 exposure with fewer omission errors in the CPT in TD children
One study found an increase in ADHD-related hospital admissions in adolescents after short-term exposure to nitrogen oxide (+68%), short-term exposure to sulphur dioxide (+29%) and short-term exposure to PM10 (+17%)179
Pathways of nitrogen oxides on ADHD
Even in non-toxic doses, nitrogen oxides have an influence on glutamatergic, opioidergic, cholinergic and dopaminergic neurotransmission in the brain.180
Emissions of nitrogen oxides in Germany fell by almost 2/3 between 1990 and 2020.181
Acute NO2 inhalation causes182
- oxidative stress with the formation of reactive oxygen species (ROS), which damage brain cells
- oxidative stress in conjunction with mitochondrial dysfunction impairs neuronal functions through
- impaired energy metabolism
- reduced ATP production
- impaired mitochondrial biogenesis
- these correlate with an increased risk of cognitive deficits, ischemic stroke and neurodevelopmental disorders
N2O exposure126
- reversibly inhibits human alpha-7 nicotinic acetylcholine receptors (α7-nAChRs). α7-nAChRs regulate synaptic neurotransmitter release in the CNS183 and the enhancement of glutamatergic activity that regulates working memory and attention-related pathways in the dlPFC184.
- Inhibition of α7-nAChR by N2O can trigger oxidative stress, which is characterized by increased NO in peripheral regions130
α7-nAChR on sympathetic nerves normally increase the release of noradrenaline, which leads to neuronal vasodilation. Inhibition of α7-nAChR mediated by N2O thus reduces sympathetic activity, which contributes to ADHD126
NO promotes parasympathetic activity.126
Excessive NO levels can induce a parasympathetic dominant state in ADHD [101]
N2O and NO consequently increase ADHD together.
NO2 in combination with other NOx reacts in the atmosphere (catalyzed by sunlight) with volatile organic compounds (VOC) and other chemicals to form PM and ozone.126
NO2 emissions in the USA have halved from 15 million tons in 2011 to just over 7.5 million tons in 2020126
7.4.3. Nitrous oxide (laughing gas)
Increasing exposure to N2O from air pollution could contribute significantly to the development of ADHD127
Environmentally relevant concentrations of N2O, even at trace levels, impair cognitive functions such as working memory in adult males126
7.4.4. Ozone
Children between the ages of 3 and 12 in China who were exposed to higher concentrations of ozone showed an increased risk of ADHD. This was further increased by sports.185
O3 exposure can constrict the airway muscles, trapping air in the alveoli and causing respiratory symptoms such as wheezing and shortness of breath. Ozone primarily attacks the lungs, but can also enter the bloodstream via the respiratory tract or nose and then into the brain, where it has a neurotoxic effect. Ozone can trigger inflammation in the brain (increased pro-inflammatory cytokines and activated microglia) 186126
High ozone exposure is a risk factor for neurological diseases.187 Long-term exposure to ozone leads to cognitive impairments such as memory problems or impaired executive functions.188
7.5. Pets (+ 58 % to + 66 %)
Preschool children aged 3 to 6 years had a 58% to 66% increased risk of ADHD if the children grew up with pets in a large Chinese population-based study based on parental reports.189
7.6. Printing ink on food paper
Using newspapers to wrap food 3 or more times a week increased the risk of ADHD by 105 times in an Egyptian study.11 It may depend on the approved printing inks.
With these figures, however, we ask ourselves whether contact with newspapers (when reading) should not already lead to a measurable increase in the risk of ADHD.
7.7. Synergy effects of neurotoxins
The synergistic effects of neurotoxins must be taken into account:68190
- Formaldehyde increases the toxicity of mercury.
- Amalgam increases the toxicity of PCBs and formaldehyde.
- Mercury and PCBs potentiate each other’s effects.
7.8. Factors without risk increase for ADHD
- Dichlorodiphenyldichloroethylene did not affect the risk of ADHD78
- Hexachlorobenzene (HCB) showed a non-linear relationship with ADHD, with an increasing risk in the low exposure range, which turned into a decreasing risk at concentrations above 8 ng/g lipid.85 Another study found no effect on ADHD.78
- Organic pollutants (OP pesticides, PCBs, pyrethroid insecticides and trichlorophenol (TCP)) did not increase the odds ratio for ADHD (0.99)48
- Bismuth urine levels were slightly lower in children with ADHD than in children without ADHD.10
- Aluminum blood levels were unchanged in children with ADHD.8
7.9. Factors with risk reduction for ADHD
- p,p’-dichlorodiphenyltrichloroethane (p,p’-DDT) was associated with a 36% lower likelihood of ADHD85
Kaur S, Canals-Sans J, Morales-Hidalgo P, Alda JA, Arija V (2025): Investigating the Association Between Heavy Metals and Attention Deficit Hyperactivity Disorder in Children: An Exploratory Study. J Atten Disord. 2025 Apr;29(6):423-436. doi: 10.1177/10870547251315276. PMID: 39886771. n = 190 ↥ ↥ ↥
Farmani R, Mehrpour O, Kooshki A, Nakhaee S (2024): Exploring the link between toxic metal exposure and ADHD: a systematic review of pb and hg. J Neurodev Disord. 2024 Aug 1;16(1):44. doi: 10.1186/s11689-024-09555-8. PMID: 39090571; PMCID: PMC11292919. METASTUDY ↥ ↥
Kaur S, Canals-Sans J, Morales-Hidalgo P, Alda JA, Arija V (2025): Investigating the Association Between Heavy Metals and Attention Deficit Hyperactivity Disorder in Children: An Exploratory Study. J Atten Disord. 2025 Apr;29(6):423-436. doi: 10.1177/10870547251315276. PMID: 39886771. ↥
Braun JM, Kahn RS, Froehlich T, Auinger P, Lanphear BP (2006): Exposures to environmental toxicants and attention deficit hyperactivity disorder in U.S. children. Environ Health Perspect. 2006 Dec;114(12):1904-9. doi: 10.1289/ehp.9478. PMID: 17185283; PMCID: PMC1764142. n = 4.704) ↥ ↥
Nilsen FM, Tulve NS. A systematic review and meta-analysis examining the interrelationships between chemical and non-chemical stressors and inherent characteristics in children with ADHD. Environ Res. 2020 Jan;180:108884. doi: 10.1016/j.envres.2019.108884. PMID: 31706600; PMCID: PMC6937727. METASTUDY n = 47 Studien ↥
Dimitrov LV, Kaminski JW, Holbrook JR, Bitsko RH, Yeh M, Courtney JG, O’Masta B, Maher B, Cerles A, McGowan K, Rush M (2023): A Systematic Review and Meta-analysis of Chemical Exposures and Attention-Deficit/Hyperactivity Disorder in Children. Prev Sci. 2023 Dec 18. doi: 10.1007/s11121-023-01601-6. PMID: 38108946. METASTUDY ↥ ↥ ↥ ↥
Gu Q, Liu J, Zhang X, Huang A, Yu X, Wu K, Huang Y (2024): Association between heavy metals exposure and risk of attention deficit hyperactivity disorder (ADHD) in children: a systematic review and meta-analysis. Eur Child Adolesc Psychiatry. 2024 Aug 10. doi: 10.1007/s00787-024-02546-z. PMID: 39126497. METASTUDY ↥ ↥ ↥ ↥ ↥
Nicolescu R, Petcu C, Cordeanu A, Fabritius K, Schlumpf M, Krebs R, Krämer U, Winneke G (2010): Environmental exposure to lead, but not other neurotoxic metals, relates to core elements of ADHD in Romanian children: performance and questionnaire data. Environ Res. 2010 Jul;110(5):476-83. doi: 10.1016/j.envres.2010.04.002. PMID: 20434143. ↥ ↥ ↥
Silbergeld EK, Goldberg AM (1975): Pharmacological and neurochemical investigations of lead-induced hyperactivity. Neuropharmacology. 1975 May-Jun;14(5-6):431-44. doi: 10.1016/0028-3908(75)90026-x. PMID: 1171389. ↥
Lee MJ, Chou MC, Chou WJ, Huang CW, Kuo HC, Lee SY, Wang LJ (2018): Heavy Metals’ Effect on Susceptibility to Attention-Deficit/Hyperactivity Disorder: Implication of Lead, Cadmium, and Antimony. Int J Environ Res Public Health. 2018 Jun 10;15(6):1221. doi: 10.3390/ijerph15061221. PMID: 29890770; PMCID: PMC6025252. n = 122 ↥ ↥ ↥ ↥ ↥ ↥
Hussein RA, Refai RH, El-Zoka AH, Azouz HG, Hussein MF (2025): Association between some environmental risk factors and attention-deficit hyperactivity disorder among children in Egypt: a case-control study. Ital J Pediatr. 2025 Jan 29;51(1):19. doi: 10.1186/s13052-025-01843-w. PMID: 39875928; PMCID: PMC11776284. n = 252 ↥ ↥ ↥
Rosenauer V, Schwarz MI, Vlasak T, Barth A (2024): Childhood lead exposure increases the risk of attention-deficit-hyperactivity disorder: A meta-analysis. Sci Total Environ. 2024 Nov 15;951:175574. doi: 10.1016/j.scitotenv.2024.175574. PMID: 39153625. REVIEW ↥
Geier, Kern, Geier (2018): A cross-sectional study of the relationship between blood lead levels and reported attention deficit disorder: an assessment of the economic impact on the United States. Metab Brain Dis. 2018 Feb;33(1):201-208. doi: 10.1007/s11011-017-0146-6. n = 2109 ↥
Khalid, Abdollahi (2019): Epigenetic modifications associated with pathophysiological effects of lead exposure. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev. 2019 Aug 12:1-53. doi: 10.1080/10590501.2019.1640581. ↥
van de Bor (2019): Fetal toxicology. Handb Clin Neurol. 2019;162:31-55. doi: 10.1016/B978-0-444-64029-1.00002-3. ↥ ↥ ↥ ↥ ↥ ↥ ↥
Reuben A, Ward R, Rothbaum AO, Cornelison VL, Huffman S, McTeague LM, Schmidt MG, Specht AJ, Kilpatrick DG (2024): Who tests for lead and why? A 10-year analysis of blood lead screening, follow-up and CNS outcomes in a statewide US healthcare system. Occup Environ Med. 2024 Jan 25:oemed-2023-109210. doi: 10.1136/oemed-2023-109210. PMID: 38272665. ↥
Muñoz, Rubilar, Valdés, Muñoz-Quezad, Gómez, Saavedra, Iglesias (2020): Attention deficit hyperactivity disorder and its association with heavy metals in children from northern Chile. Int J Hyg Environ Health. 2020 May;226:113483. doi: 10.1016/j.ijheh.2020.113483. PMID: 32106053. ↥ ↥
Gao X, Zheng X, Wang X, Li Z, Yang L (2025): Environmental pollutant exposure and adverse neurodevelopmental outcomes: An umbrella review and evidence grading of meta-analyses. J Hazard Mater. 2025 Mar 5;491:137832. doi: 10.1016/j.jhazmat.2025.137832. PMID: 40068397. REVIEW ↥ ↥ ↥
McFarland MJ, Reuben A, Hauer M (2025): Contribution of childhood lead exposure to psychopathology in the US population over the past 75 years. J Child Psychol Psychiatry. 2025 May;66(5):659-666. doi: 10.1111/jcpp.14072. PMID: 39628272. ↥
Levin ED, Bowman RE, Wegert S, Vuchetich J (1987): Psychopharmacological investigations of a lead-induced long-term cognitive deficit in monkeys. Psychopharmacology (Berl). 1987;91(3):334-41. doi: 10.1007/BF00518187. PMID: 3104955. ↥
Cory-Slechta DA (1997): Relationships between Pb-induced changes in neurotransmitter system function and behavioral toxicity. Neurotoxicology. 1997;18(3):673-88. PMID: 9339816. REVIEW ↥
Zuch CL, O’Mara DJ, Cory-Slechta DA. Low-level lead exposure selectively enhances dopamine overflow in nucleus accumbens: an in vivo electrochemistry time course assessment. Toxicol Appl Pharmacol. 1998 May;150(1):174-85. doi: 10.1006/taap.1998.8396. PMID: 9630467. ↥
Jones, Miller (2008): The effects of environmental neurotoxicants on the dopaminergic system: A possible role in drug addiction. Biochem Pharmacol. 2008 Sep 1;76(5):569-81. doi: 10.1016/j.bcp.2008.05.010. PMID: 18555207. REVIEW ↥
Kim, Kim, Lee, Yun, Sohn, Shin, Kim, Chae, Roh, Kim (2018): Interaction between DRD2 and lead exposure on the cortical thickness of the frontal lobe in youth with attention-deficit/hyperactivity disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2018 Mar 2;82:169-176. doi: 10.1016/j.pnpbp.2017.11.018. ↥
Nilsen, Tulve (2019): A systematic review and meta-analysis examining the interrelationships between chemical and non-chemical stressors and inherent characteristics in children with ADHD. Environ Res. 2019 Nov 1:108884. doi: 10.1016/j.envres.2019.108884. REVIEW ↥ ↥ ↥ ↥
Amos-Kroohs, Graham, Grace, Braun, Schaefer, Skelton, Vorhees, Williams (2016): Developmental stress and lead (Pb): Effects of maternal separation and/or Pb on corticosterone, monoamines, and blood Pb in rats. Neurotoxicology. 2016 May;54:22-33. doi: 10.1016/j.neuro.2016.02.011. PMID: 26943976; PMCID: PMC4875812. ↥
Desrochers-Couture, Courtemanche, Forget-Dubois, Bélanger, Boucher, Ayotte, Cordier, Jacobson, Jacobson, Muckle (2019): Association between early lead exposure and externalizing behaviors in adolescence: A developmental cascade. Environ Res. 2019 Aug 19;178:108679. doi: 10.1016/j.envres.2019.108679. ↥
Sprowles JLN, Amos-Kroohs RM, Braun AA, Sugimoto C, Vorhees CV, Williams MT (2018): Developmental manganese, lead, and barren cage exposure have adverse long-term neurocognitive, behavioral and monoamine effects in Sprague-Dawley rats. Neurotoxicol Teratol. 2018 May-Jun;67:50-64. doi: 10.1016/j.ntt.2018.04.001. PMID: 29631003; PMCID: PMC5970996. ↥
http://www.adhs.org/genese/ mit weiteren Nachweisen ↥
Smith DR, Strupp BJ. Animal Models of Childhood Exposure to Lead or Manganese: Evidence for Impaired Attention, Impulse Control, and Affect Regulation and Assessment of Potential Therapies. Neurotherapeutics. 2023 Feb 28. doi: 10.1007/s13311-023-01345-9. PMID: 36853434. ↥ ↥
Nilsen FM, Tulve NS. A systematic review and meta-analysis examining the interrelationships between chemical and non-chemical stressors and inherent characteristics in children with ADHD. Environ Res. 2020 Jan;180:108884. doi: 10.1016/j.envres.2019.108884. PMID: 31706600; PMCID: PMC6937727. METASTUDY ↥
Santiago NA, He B, Howard SL, Beaudin S, Strupp BJ, Smith DR (2023):. Developmental Manganese Exposure Causes Lasting Attention Deficits Accompanied by Dysregulation of mTOR Signaling and Catecholaminergic Gene Expression in Brain Prefrontal Cortex. bioRxiv [Preprint]. 2023 Jul 18:2023.07.16.549215. doi: 10.1101/2023.07.16.549215. PMID: 37503220; PMCID: PMC10370122. ↥
Beaudin SA, Howard S, Santiago N, Strupp BJ, Smith DR (2023): Methylphenidate alleviates cognitive dysfunction from early Mn exposure: Role of catecholaminergic receptors. bioRxiv [Preprint]. 2023 Oct 4:2023.06.27.546786. doi: 10.1101/2023.06.27.546786. PMID: 37873333; PMCID: PMC10592804. ↥ ↥
Zheng YX, Chan P, Pan ZF, Shi NN, Wang ZX, Pan J, Liang HM, Niu Y, Zhou XR, He FS (2002): Polymorphism of metabolic genes and susceptibility to occupational chronic manganism. Biomarkers. 2002 Jul-Aug;7(4):337-46. doi: 10.1080/13547500210146740. PMID: 12171760. ↥
Shih, Zeng, Lin, Chen, Chen, Wu, Tseng, Wu (2018): Association between peripheral manganese levels and attention-deficit/hyperactivity disorder: a preliminary meta-analysis. Neuropsychiatr Dis Treat. 2018 Jul 18;14:1831-1842. doi: 10.2147/NDT.S165378. eCollection 2018. ↥
Yousef S, Adem A, Zoubeidi T, Kosanovic M, Mabrouk AA, Eapen V (2011): Attention deficit hyperactivity disorder and environmental toxic metal exposure in the United Arab Emirates. J Trop Pediatr. 2011 Dec;57(6):457-60. doi: 10.1093/tropej/fmq121. PMID: 21300623. ↥ ↥
Schildroth S, Bauer JA, Friedman A, Austin C, Coull BA, Placidi D, White RF, Smith D, Wright RO, Lucchini RG, Arora M, Horton M, Claus Henn B (2023): Early life manganese exposure and reported attention-related behaviors in Italian adolescents. Environ Epidemiol. 2023 Oct 19;7(6):e274. doi: 10.1097/EE9.0000000000000274. PMID: 38912396; PMCID: PMC11189689. ↥
Darr J, Hamama Z (2025): Manganese exposure assessment in formula-fed infants in Israel. Isr J Health Policy Res. 2025 Apr 15;14(1):24. doi: 10.1186/s13584-025-00688-2. PMID: 40234974; PMCID: PMC12001741. ↥
Howard SL, Beaudin SA, Strupp BJ, Smith DR (2023): Maternal choline supplementation: A potential therapy for developmental Manganese exposure? bioRxiv [Preprint]. 2023 Jun 26:2023.06.23.546356. doi: 10.1101/2023.06.23.546356. PMID: 37425833; PMCID: PMC10327095. ↥
Nilsen FM, Tulve NS. A systematic review and meta-analysis examining the interrelationships between chemical and non-chemical stressors and inherent characteristics in children with ADHD. Environ Res. 2020 Jan;180:108884. doi: 10.1016/j.envres.2019.108884. PMID: 31706600; PMCID: PMC6937727. METASTUDY, k = 47 ↥
O’Connor LE, Spill MK, Trivedi R, Saha S, Thoerig RC, Foster M, MacFarlane AJ (2025): Mercury exposure and childhood outcomes: An overview of systematic reviews. Environ Res. 2025 Feb 20;274:121196. doi: 10.1016/j.envres.2025.121196. PMID: 39986421. REVIEW ↥
Ke T, Tinkov AA, Skalny AV, Bowman AB, Rocha JBT, Santamaria A, Aschner M. Developmental exposure to methylmercury and ADHD, a literature review of epigenetic studies. Environ Epigenet. 2021 Nov 22;7(1):dvab014. doi: 10.1093/eep/dvab014. PMID: 34881051; PMCID: PMC8648069. REVIEW ↥
Ehlinger JV, Goodrich JM, Dolinoy DC, Watkins DJ, Cantoral A, Mercado-García A, Basu N, Téllez-Rojo MM, Peterson KE (2025): Interaction of mercury exposure and DNA methylation with sustained attention in children in a novel analysis of epigenetic susceptibility. Environ Epigenet. 2025 Apr 24;11(1):dvaf011. doi: 10.1093/eep/dvaf011. PMID: 40401167; PMCID: PMC12094074. ↥
Dreiem, Shan, Okoniewski, Sanchez-Morrissey, Seegal (2009): Methylmercury inhibits dopaminergic function in rat pup synaptosomes in an age-dependent manner. Neurotoxicol Teratol. 2009 Sep-Oct;31(5):312-7. doi: 10.1016/j.ntt.2009.05.001. PMID: 19464365. ↥
Lin, Wang, Chiang, Lai, Chang, Chi (2017): Risk of subsequent attention-deficit/hyperactivity disorder among children and adolescents with amalgam restorations: A nationwide longitudinal study. .Community Dent Oral Epidemiol. 2017 Aug 7. doi: 10.1111/cdoe.12327 ↥
Torrey EF, Simmons W (2023): Mercury and Parkinson’s Disease: Promising Leads, but Research Is Needed. Parkinsons Dis. 2023 Sep 16;2023:4709322. doi: 10.1155/2023/4709322. PMID: 37744289; PMCID: PMC10517869. REVIEW ↥
Mukhi S, Manjrekar PA, Srikantiah RM, Harish S, Kotian H, Rao YL, Sherly A (2024): Evaluation of the cognitive, physiological, and biomarker effects of heavy metal exposure in Wistar rats. Vet World. 2024 Aug;17(8):1855-1863. doi: 10.14202/vetworld.2024.1855-1863. Epub 2024 Aug 24. PMID: 39328457; PMCID: PMC11422626. ↥
Nilsen FM, Tulve NS. A systematic review and meta-analysis examining the interrelationships between chemical and non-chemical stressors and inherent characteristics in children with ADHD. Environ Res. 2020 Jan;180:108884. doi: 10.1016/j.envres.2019.108884. PMID: 31706600; PMCID: PMC6937727. METASTUDIE n = 47 Studien ↥ ↥ ↥ ↥
Ku, Tsai, Wang, Su, Sun, Wang, Wang (2019): Prenatal and childhood phthalate exposure and attention deficit hyperactivity disorder traits in child temperament: A 12-year follow-up birth cohort study. Sci Total Environ. 2019 Aug 29;699:134053. doi: 10.1016/j.scitotenv.2019.134053. ↥
Oh J, Kim K, Kannan K, Parsons PJ, Mlodnicka A, Schmidt RJ, Schweitzer JB, Hertz-Picciotto I, Bennett DH (2024): Early childhood exposure to environmental phenols and parabens, phthalates, organophosphate pesticides, and trace elements in association with attention deficit hyperactivity disorder (ADHD) symptoms in the CHARGE study. Environ Health. 2024 Mar 14;23(1):27. doi: 10.1186/s12940-024-01065-3. PMID: 38486233. ↥
Yeo KE, Lim S, Kim A, Lim YB, Lee CR, Kim JI, Kim BN (2025): Association Between Endocrine-Disrupting Chemicals Exposure and Attention-Deficit/Hyperactivity Disorder Symptoms in Children With Attention-Deficit/Hyperactivity Disorder. J Korean Acad Child Adolesc Psychiatry. 2025 Jan 1;36(1):18-25. doi: 10.5765/jkacap.240035. PMID: 39811025; PMCID: PMC11725660. n = 67 ↥ ↥
Tsai CS, Chou WJ, Lee SY, Lee MJ, Chou MC, Wang LJ (2020): Phthalates, Para-Hydroxybenzoic Acids, Bisphenol-A, and Gonadal Hormones’ Effects on Susceptibility to Attention-Deficit/Hyperactivity Disorder. Toxics. 2020 Aug 13;8(3):57. doi: 10.3390/toxics8030057. PMID: 32823738; PMCID: PMC7560246. ↥
Riddell, Malin, Flora, McCague, Till (2019): Association of water fluoride and urinary fluoride concentrations with attention deficit hyperactivity disorder in Canadian youth. Environ Int. 2019 Oct 22;133(Pt B):105190. doi: 10.1016/j.envint.2019.105190. n = 980 ↥
Bashash M, Marchand M, Hu H, Till C, Martinez-Mier EA, Sanchez BN, Basu N, Peterson KE, Green R, Schnaas L, Mercado-García A, Hernández-Avila M, Téllez-Rojo MM (2018): Prenatal fluoride exposure and attention deficit hyperactivity disorder (ADHD) symptoms in children at 6-12 years of age in Mexico City. Environ Int. 2018 Dec;121(Pt 1):658-666. doi: 10.1016/j.envint.2018.09.017. PMID: 30316181. ↥
Bashash M, Thomas D, Hu H, Martinez-Mier EA, Sanchez BN, Basu N, Peterson KE, Ettinger AS, Wright R, Zhang Z, Liu Y, Schnaas L, Mercado-García A, Téllez-Rojo MM, Hernández-Avila M (2017): Prenatal Fluoride Exposure and Cognitive Outcomes in Children at 4 and 6-12 Years of Age in Mexico. Environ Health Perspect. 2017 Sep 19;125(9):097017. doi: 10.1289/EHP655. PMID: 28937959; PMCID: PMC5915186. ↥
Wang A, Duan L, Huang H, Ma J, Zhang Y, Ma Q, Guo Y, Li Z, Cheng X, Zhu J, Zhou G, Ba Y (2022): Association between fluoride exposure and behavioural outcomes of school-age children: a pilot study in China. Int J Environ Health Res. 2022 Jan;32(1):232-241. doi: 10.1080/09603123.2020.1747601. PMID: 32281876. ↥
Fiore G, Veneri F, Di Lorenzo R, Generali L, Vinceti M, Filippini T (2023): Fluoride Exposure and ADHD: A Systematic Review of Epidemiological Studies. Medicina (Kaunas). 2023 Apr 19;59(4):797. doi: 10.3390/medicina59040797. PMID: 37109754; PMCID: PMC10143272. REVIEW ↥
Geier DA, Geier MR (2025): Tooth decay prevention and neurodevelopmental disorder risk following childhood fluoride exposure. BMC Pediatr. 2025 Apr 2;25(1):265. doi: 10.1186/s12887-025-05601-z. PMID: 40170137; PMCID: PMC11963426. n = 73.254 ↥
Kampouri M, Zander E, Gustin K, Sandin A, Barman M, Sandberg AS, Wold AE, Bölte S, Kippler M, Vahter M (2024): Associations of gestational and childhood exposure to lead, cadmium, and fluoride with cognitive abilities, behavior, and social communication at 4 years of age: NICE birth cohort study. Environ Res. 2024 Dec 15;263(Pt 2):120123. doi: 10.1016/j.envres.2024.120123. PMID: 39389199. ↥
Bundesinstitut für Risikobewertung: Information Nr. 037/2005 des BfR vom 12. Juli 2005: Durchschnittlicher Fluoridgehalt in Trinkwasser ist in Deutschland niedrig ↥
Ibarluzea J, Subiza-Pérez M, Arregi A, Molinuevo A, Arranz-Freijo E, Sánchez-de Miguel M, Jiménez A, Andiarena A, Santa-Marina L, Lertxundi A. Association of maternal prenatal urinary fluoride levels with ADHD symptoms in childhood. Environ Res. 2023 Jul 19;235:116705. doi: 10.1016/j.envres.2023.116705. PMID: 37479215. ↥
Reddy YP, Tiwari S, Tomar LK, Desai N, Sharma VK (2021): Fluoride-Induced Expression of Neuroinflammatory Markers and Neurophysiological Regulation in the Brain of Wistar Rat Model. Biol Trace Elem Res. 2021 Jul;199(7):2621-2626. doi: 10.1007/s12011-020-02362-x. PMID: 32865723. ↥
Mariussen E, Fonnum F (2006): Neurochemical targets and behavioral effects of organohalogen compounds: an update. Crit Rev Toxicol. 2006 Mar;36(3):253-89. doi: 10.1080/10408440500534164. PMID: 16686424. REVIEW ↥
Regan SL, Williams MT, Vorhees CV (2022): Review of rodent models of attention deficit hyperactivity disorder. Neurosci Biobehav Rev. 2022 Jan;132:621-637. doi: 10.1016/j.neubiorev.2021.11.041. PMID: 34848247; PMCID: PMC8816876.) REVIEW ↥ ↥ ↥ ↥
Sagiv SK, Thurston SW, Bellinger DC, Tolbert PE, Altshul LM, Korrick SA (2010): Prenatal organochlorine exposure and behaviors associated with attention deficit hyperactivity disorder in school-aged children. Am J Epidemiol. 2010 Mar 1;171(5):593-601. doi: 10.1093/aje/kwp427. PMID: 20106937; PMCID: PMC2842227. ↥
Bushnell PJ, Moser VC, MacPhail RC, Oshiro WM, Derr-Yellin EC, Phillips PM, Kodavanti PR (2002): Neurobehavioral assessments of rats perinatally exposed to a commercial mixture of polychlorinated biphenyls. Toxicol Sci. 2002 Jul;68(1):109-20. doi: 10.1093/toxsci/68.1.109. PMID: 12075116. ↥ ↥
Schantz SL (1996): Developmental neurotoxicity of PCBs in humans: what do we know and where do we go from here? Neurotoxicol Teratol. 1996 May-Jun;18(3):217-27; discussion 229-76. doi: 10.1016/s0892-0362(96)90001-x. PMID: 8725628. REVIEW ↥
Pessah, Lein, Seegal, Sagiv (2019): Neurotoxicity of polychlorinated biphenyls and related organohalogens. Acta Neuropathol. 2019 Apr 11. doi: 10.1007/s00401-019-01978-1. ↥
Sable HJ, Eubig PA, Powers BE, Wang VC, Schantz SL. Developmental exposure to PCBs and/or MeHg: effects on a differential reinforcement of low rates (DRL) operant task before and after amphetamine drug challenge. Neurotoxicol Teratol. 2009 May-Jun;31(3):149-58. doi: 10.1016/j.ntt.2008.12.006. Epub 2009 Jan 21. PMID: 19344642; PMCID: PMC2730353. ↥ ↥
Chishti, Fisher, Seegal (1996): Aroclors 1254 and 1260 reduce dopamine concentrations in rat striatal slices. Neurotoxicology. 1996 Fall-Winter;17(3-4):653-60. ↥ ↥ ↥ ↥
Faroon O, Jones D, de Rosa C (2000): Effects of polychlorinated biphenyls on the nervous system. Toxicol Ind Health. 2000 Sep;16(7-8):305-33. doi: 10.1177/074823370001600708. PMID: 11693948. REVIEW ↥
Seegal RF, Brosch KO, Okoniewski RJ (1997): Effects of in utero and lactational exposure of the laboratory rat to 2,4,2’,4’- and 3,4,3’,4’-tetrachlorobiphenyl on dopamine function. Toxicol Appl Pharmacol. 1997 Sep;146(1):95-103. doi: 10.1006/taap.1997.8226. PMID: 9299601. ↥
Seegal RF, Bush B, Brosch KO (1994): Decreases in dopamine concentrations in adult, non-human primate brain persist following removal from polychlorinated biphenyls. Toxicology. 1994 Jan 26;86(1-2):71-87. doi: 10.1016/0300-483x(94)90054-x. PMID: 8134924. ↥
Seegal RF, Bush B, Brosch KO (1991): Comparison of effects of Aroclors 1016 and 1260 on non-human primate catecholamine function. Toxicology. 1991 Feb;66(2):145-63. doi: 10.1016/0300-483x(91)90215-m. PMID: 2014516. ↥
Seegal RF, Bush B, Shain W (1990): Lightly chlorinated ortho-substituted PCB congeners decrease dopamine in nonhuman primate brain and in tissue culture. Toxicol Appl Pharmacol. 1990 Oct;106(1):136-44. doi: 10.1016/0041-008x(90)90113-9. PMID: 2123577. ↥
Caudle WM, Richardson JR, Delea KC, Guillot TS, Wang M, Pennell KD, Miller GW (2006): Polychlorinated biphenyl-induced reduction of dopamine transporter expression as a precursor to Parkinson’s disease-associated dopamine toxicity. Toxicol Sci. 2006 Aug;92(2):490-9. doi: 10.1093/toxsci/kfl018. Erratum in: Toxicol Sci. 2022 Feb 28;186(1):175. PMID: 16702228. ↥
Mouatsou C, Margetaki K, Kampouri M, Roumeliotaki T, Rantakokko P, Kiviranta H, Karachaliou M, Stephanou EG, Chatzi L, Kogevinas M, Koutra K (2025): Prenatal exposure to persistent organic pollutants and emotional and behavioral outcomes from early childhood to adolescence: Rhea Cohort Study in Crete, Greece. Environ Epidemiol. 2025 Mar 24;9(2):e377. doi: 10.1097/EE9.0000000000000377. PMID: 40135050; PMCID: PMC11936570. n = 467 ↥ ↥ ↥
Hilz EN, Schnurer C, Bhamidipati S, Deka J, Thompson LM, Gore AC (2025): Cognitive effects of early life exposure to PCBs in rats: Sex-specific behavioral, hormonal and neuromolecular mechanisms involving the brain dopamine system. Horm Behav. 2025 Mar;169:105697. doi: 10.1016/j.yhbeh.2025.105697. PMID: 39923265; PMCID: PMC11908942. ↥
Jacobson JL, Jacobson SW. Intellectual impairment in children exposed to polychlorinated biphenyls in utero. N Engl J Med. 1996 Sep 12;335(11):783-9. doi: 10.1056/NEJM199609123351104. PMID: 8703183. ↥
Grandjean P, Weihe P, Burse VW, Needham LL, Storr-Hansen E, Heinzow B, Debes F, Murata K, Simonsen H, Ellefsen P, Budtz-Jørgensen E, Keiding N, White RF (2001): Neurobehavioral deficits associated with PCB in 7-year-old children prenatally exposed to seafood neurotoxicants. Neurotoxicol Teratol. 2001 Jul-Aug;23(4):305-17. doi: 10.1016/s0892-0362(01)00155-6. PMID: 11485834. ↥
Stewart P, Fitzgerald S, Reihman J, Gump B, Lonky E, Darvill T, Pagano J, Hauser P (2003): Prenatal PCB exposure, the corpus callosum, and response inhibition. Environ Health Perspect. 2003 Oct;111(13):1670-7. doi: 10.1289/ehp.6173. PMID: 14527849; PMCID: PMC1241692. ↥
Stewart P, Reihman J, Gump B, Lonky E, Darvill T, Pagano J (2005): Response inhibition at 8 and 9 1/2 years of age in children prenatally exposed to PCBs. Neurotoxicol Teratol. 2005 Nov-Dec;27(6):771-80. doi: 10.1016/j.ntt.2005.07.003. PMID: 16198536. ↥
Mesnil M, Defamie N, Naus C, Sarrouilhe D (2020): Brain Disorders and Chemical Pollutants: A Gap Junction Link? Biomolecules. 2020 Dec 31;11(1):51. doi: 10.3390/biom11010051. PMID: 33396565; PMCID: PMC7824109. REVIEW ↥ ↥ ↥ ↥
Lenters V, Iszatt N, Forns J, Čechová E, Kočan A, Legler J, Leonards P, Stigum H, Eggesbø M (2019): Early-life exposure to persistent organic pollutants (OCPs, PBDEs, PCBs, PFASs) and attention-deficit/hyperactivity disorder: A multi-pollutant analysis of a Norwegian birth cohort. Environ Int. 2019 Apr;125:33-42. doi: 10.1016/j.envint.2019.01.020. PMID: 30703609. n = 2.606 ↥ ↥ ↥ ↥
Zhang M, Gu X, Wu L, Wan N, Liu Y, Xin Z, Chen T, Liu S, Li M, Deng M, Wang Q (2023): A new mechanistic insight into the association between environmental perfluorooctane sulfonic acid (PFOS) exposure and attention deficit and hyperactivity disorder (ADHD)-like behavior. Neurotoxicology. 2023 Nov 10;99:254-263. doi: 10.1016/j.neuro.2023.11.004. PMID: 37952603. ↥
He WT, Huang JW, Zhang YT, Trevathan E, Qian Z, Boyd R, Elliott M, Lin LZ, Gui ZH, Liu RQ, Hu LW, Dong GH (2025): Chlorinated paraffins exposure in particulate matter increase the risk of attention-deficit/hyperactivity disorder symptoms in children and adolescents. Environ Pollut. 2025 Mar 27;373:126120. doi: 10.1016/j.envpol.2025.126120. PMID: 40157481. ↥
Dellefratte, Stingone, Claudio (2019): Combined association of BTEX and material hardship on ADHD-suggestive behaviours among a nationally representative sample of US children. Paediatr Perinat Epidemiol. 2019 Nov;33(6):482-489. doi: 10.1111/ppe.12594. n = 4.650 ↥
Xi, Wu (2021): A Review on the Mechanism Between Different Factors and the Occurrence of Autism and ADHD. Psychol Res Behav Manag. 2021 Apr 9;14:393-403. doi: 10.2147/PRBM.S304450. PMID: 33859505; PMCID: PMC8044340. REVIEW ↥
Salazar, Villaseca, Cisternas, Inestrosa (2021). Neurodevelopmental impact of the offspring by thyroid hormone system-disrupting environmental chemicals during pregnancy. Environ Res. 2021 Jun 1;200:111345. doi: 10.1016/j.envres.2021.111345. PMID: 34087190. ↥
Liu H, Wang J (2022): The association between bisphenol a exposure and attention deficit hyperactivity disorder in children: a meta-analysis of observational studies. Rev Environ Health. 2022 Dec 8. doi: 10.1515/reveh-2022-0184. PMID: 36480489. n = 5.710 ↥
Lee (2019): Potential health effects of emerging environmental contaminants perfluoroalkyl compounds. Yeungnam Univ J Med. 2018 Dec 31;35(2):156-164. doi: 10.12701/yujm.2018.35.2.156. ↥
Cui H, Shu C, Peng Y, Wei Z, Ni X, Zheng L, Shang J, Liu F, Liu J (2024): Long-life triclosan exposure induces ADHD-like behavior in rats via prefrontal cortex dopaminergic deficiency. Ecotoxicol Environ Saf. 2024 Jul 23;282:116766. doi: 10.1016/j.ecoenv.2024.116766. PMID: 39047361. ↥
Shu C, Cui H, Peng Y, Wei Z, Ni X, Zheng L, Shang J, Liu F, Liu J (2024): Understanding the molecular pathway of triclosan-induced ADHD-like behaviour: Involvement of the hnRNPA1-PKM2-STAT3 feedback loop. Environ Int. 2024 Sep;191:108966. doi: 10.1016/j.envint.2024.108966. PMID: 39167854. ↥
Ding J, Dai Y, Zhang L, Wang Z, Zhang B, Guo J, Qi X, Lu D, Chang X, Wu C, Zhang J, Zhou Z (2024): Identifying childhood pesticide exposure trajectories and critical window associated with behavioral problems at 10 years of age: Findings from SMBCS. Environ Int. 2024 Nov;193:109079. doi: 10.1016/j.envint.2024.109079. PMID: 39442318. ↥
Abreu-Villaça Y, Levin ED (2017): Developmental neurotoxicity of succeeding generations of insecticides. Environ Int. 2017 Feb;99:55-77. doi: 10.1016/j.envint.2016.11.019. Epub 2016 Nov 28. PMID: 27908457; PMCID: PMC5285268. REVIEW ↥ ↥ ↥ ↥ ↥
Makris, Chrousos, Anesiadou, Sabico, Abd-Alrahman, Al-Daghri, Chouliaras, Pervanidou (2019): Serum concentrations and detection rates of selected organochlorine pesticides in a sample of Greek school-aged children with neurodevelopmental disorders. Environ Sci Pollut Res Int. 2019 Aug;26(23):23739-23753. doi: 10.1007/s11356-019-05666-1. ↥
Sagiv SK, Kogut K, Harley K, Bradman A, Morga N, Eskenazi B (2021): Gestational Exposure to Organophosphate Pesticides and Longitudinally Assessed Behaviors Related to Attention-Deficit/Hyperactivity Disorder and Executive Function. Am J Epidemiol. 2021 Nov 2;190(11):2420-2431. doi: 10.1093/aje/kwab173. PMID: 34100072; PMCID: PMC8757311. ↥
(Marks, Harley, Bradman, Kogut, Barr, Johnson, Calderon, Eskenazi (2010): Organophosphate pesticide exposure and attention in young Mexican-American children: the CHAMACOS study. Environ Health Perspect. 2010 Dec;118(12):1768-74. doi: 10.1289/ehp.1002056. PMID: 21126939; PMCID: PMC3002198. ↥
Chang CH, Yu CJ, Du JC, Chiou HC, Chen HC, Yang W, Chung MY, Chen YS, Hwang B, Mao IF, Chen ML (2018): The interactions among organophosphate pesticide exposure, oxidative stress, and genetic polymorphisms of dopamine receptor D4 increase the risk of attention deficit/hyperactivity disorder in children. Environ Res. 2018 Jan;160:339-346. doi: 10.1016/j.envres.2017.10.011. PMID: 29054088. n = 207 ↥
Zhou W, Deng Y, Zhang C, Dai H, Guan L, Luo X, He W, Tian J, Zhao L (2022): Chlorpyrifos residue level and ADHD among children aged 1-6 years in rural China: A cross-sectional study. Front Pediatr. 2022 Oct 14;10:952559. doi: 10.3389/fped.2022.952559. PMID: 36313880; PMCID: PMC9616114. ↥ ↥
Mostafalou, Abdollahi (2018): The link of organophosphorus pesticides with neurodegenerative and neurodevelopmental diseases based on evidence and mechanisms. Toxicology. 2018 Nov 1;409:44-52. doi: 10.1016/j.tox.2018.07.014. ↥
van den Dries, Guxens, Pronk, Spaan, El Marroun, Jusko, Longnecker, Ferguson, Tiemeier (2019): Organophosphate pesticide metabolite concentrations in urine during pregnancy and offspring attention-deficit hyperactivity disorder and autistic traits. Environ Int. 2019 Jul 29;131:105002. doi: 10.1016/j.envint.2019.105002. n = 784 ↥
Quirós-Alcalá, Alkon, Boyce, Lippert, Davis, Bradman, Barr, Eskenazi (2011): Maternal prenatal and child organophosphate pesticide exposures and children’s autonomic function. Neurotoxicology. 2011 Oct;32(5):646-55. doi: 10.1016/j.neuro.2011.05.017. n > 500 ↥
Banhela N, Naidoo P, Naidoo S (2020): Association between pesticide exposure and paraoxonase-1 (PON1) polymorphisms, and neurobehavioural outcomes in children: a systematic review. Syst Rev. 2020 May 9;9(1):109. doi: 10.1186/s13643-020-01330-9. PMID: 32386510; PMCID: PMC7211330. ↥
Wang LJ, Chao HR, Chen CC, Chen CM, You HL, Tsai CC, Tsai CS, Chou WJ, Li CJ, Tsai KF, Cheng FJ, Kung CT, Li SH, Wang CC, Ou YC, Lee WC, Huang WT (2024): Effects of urinary organophosphate flame retardants in susceptibility to attention-deficit/hyperactivity disorder in school-age children. Ecotoxicol Environ Saf. 2024 Nov 15;287:117281. doi: 10.1016/j.ecoenv.2024.117281. PMID: 39509783. ↥
Terajima T, Inoue H, Shimomura K, Iwasaki F, Sasaki A, Ito Y, Kamijima M, Tomizawa M (2023): Organophosphate agent action at the fatty acid amide hydrolase enhancing anandamide-induced apoptosis in NG108-15 cells. J Toxicol Sci. 2023;48(7):421-428. doi: 10.2131/jts.48.421. PMID: 37394655. ↥
Zhou W, Deng Y, Zhang C, Dai H, Guan L, Luo X, He W, Tian J, Zhao L. Chlorpyrifos residue level and ADHD among children aged 1-6 years in rural China: A cross-sectional study. Front Pediatr. 2022 Oct 14;10:952559. doi: 10.3389/fped.2022.952559. PMID: 36313880; PMCID: PMC9616114. ↥
Berg EL, Ching TM, Bruun DA, Rivera JK, Careaga M, Ellegood J, Lerch JP, Wöhr M, Lein PJ, Silverman JL (2020): Translational outcomes relevant to neurodevelopmental disorders following early life exposure of rats to chlorpyrifos. J Neurodev Disord. 2020 Dec 16;12(1):40. doi: 10.1186/s11689-020-09342-1. PMID: 33327943; PMCID: PMC7745485. ↥
Gómez-Giménez B, Felipo V, Cabrera-Pastor A, Agustí A, Hernández-Rabaza V, Llansola M. Developmental Exposure to Pesticides Alters Motor Activity and Coordination in Rats: Sex Differences and Underlying Mechanisms. Neurotox Res. 2018 Feb;33(2):247-258. doi: 10.1007/s12640-017-9823-9. Epub 2017 Oct 3. PMID: 28975519. ↥
Ito, Tomizawa, Suzuki, Shirakawa, Ono, Adachi, Suzuki, Shimomura, Nabeshima, Kamijima (2020): Organophosphate agent induces ADHD-like behaviors via inhibition of brain endocannabinoid-hydrolyzing enzyme(s) in adolescent male rats. J Agric Food Chem. 2020 Jan 30;10.1021/acs.jafc.9b08195. doi: 10.1021/acs.jafc.9b08195. PMID: 31995978. ↥
Eadeh HM, Davis J, Ismail AA, Abdel Rasoul GM, Hendy OM, Olson JR, Bonner MR, Rohlman DS (2023): Evaluating how occupational exposure to organophosphates and pyrethroids impacts ADHD severity in Egyptian male adolescents. Neurotoxicology. 2023 Jan 5;95:75-82. doi: 10.1016/j.neuro.2023.01.001. PMID: 36621468. n = 226 ↥ ↥
Wagner-Schuman M, Richardson JR, Auinger P, Braun JM, Lanphear BP, Epstein JN, Yolton K, Froehlich TE. Association of pyrethroid pesticide exposure with attention-deficit/hyperactivity disorder in a nationally representative sample of U.S. children. Environ Health. 2015 May 28;14:44. doi: 10.1186/s12940-015-0030-y. PMID: 26017680; PMCID: PMC4458051. ↥
Richardson JR, Taylor MM, Shalat SL, Guillot TS 3rd, Caudle WM, Hossain MM, Mathews TA, Jones SR, Cory-Slechta DA, Miller GW (2015): Developmental pesticide exposure reproduces features of attention deficit hyperactivity disorder. FASEB J. 2015 May;29(5):1960-72. doi: 10.1096/fj.14-260901. PMID: 25630971; PMCID: PMC4415012. ↥
Nguyen JH, Curtis MA, Imami AS, Ryan WG, Alganem K, Neifer KL, Saferin N, Nawor CN, Kistler BP, Miller GW, Shukla R, McCullumsmith RE, Burkett JP (2023): Developmental pyrethroid exposure disrupts molecular pathways for circadian rhythms and MAP kinase in mouse brain. bioRxiv [Preprint]. 2023 Nov 28:2023.08.28.555113. doi: 10.1101/2023.08.28.555113. PMID: 37745438; PMCID: PMC10515776. ↥
Purece A, Thomsen ST, Plass D, Spyropoulou A, Machera K, Palmont P, Crépet A, Benchrih R, Devleesschauwer B, Wieland N, Scheepers P, Deepika D, Kumar V, Sanchez G, Bessems J, Piselli D, Buekers J (2024): A preliminary estimate of the environmental burden of disease associated with exposure to pyrethroid insecticides and ADHD in Europe based on human biomonitoring. Environ Health. 2024 Oct 23;23(1):91. doi: 10.1186/s12940-024-01131-w. PMID: 39443952; PMCID: PMC11515492. ↥
Choi H, Lee H, Ahn YS (2025): Prenatal and Childhood Exposure to Humidifier Disinfectants and Attention-Deficit/Hyperactivity Disorder (ADHD): Insights from a Retrospective Cohort Design. Toxics. 2025 Jan 23;13(2):78. doi: 10.3390/toxics13020078. PMID: 39997894; PMCID: PMC11860939. ↥
Ou XX, Wang X, Zhan XL, Shen SL, Karatela S, Jing J, Cai L, Liu RQ, Lin LZ, Dong GH (2024): The associations of secondhand smoke exposure with neurodevelopmental disorders and critical time window identification: A systematic review and meta-analysis. Sci Total Environ. 2024 Feb 25;913:169649. doi: 10.1016/j.scitotenv.2023.169649. PMID: 38159763. METASTUDY ↥
Lee S, Lee W (2023): The association between attention deficit hyperactivity disorder (ADHD) and smoking experience or exposure to environmental tobacco smoke among children and adolescents. Tob Induc Dis. 2023 Jan 30;21:15. doi: 10.18332/tid/157209. PMID: 36762265; PMCID: PMC9885444. n = 16.434 ↥
Deng, Yang, Wang, Zhou, Wang, Zhang, Niu (2022): Identification and Characterization of Influential Factors in Susceptibility to Attention Deficit Hyperactivity Disorder Among Preschool-Aged Children. Front Neurosci. 2022 Jan 31;15:709374. doi: 10.3389/fnins.2021.709374. PMID: 35173570; PMCID: PMC8841729. n = 7.938 ↥
Abdel Hamed, Hammad, Salama, Yassa, Awaga (2019): Secondhand smoke as a risk factor for attention deficit hyperactivity disorder in children. Inhal Toxicol. 2019 Sep – Oct;31(11-12):420-427. doi: 10.1080/08958378.2019.1705440. ↥
Gatzke-Kopp, Willoughby, Warkentien, Petrie, Mills-Koonce, Blair (2019): Association between environmental tobacco smoke exposure across the first four years of life and manifestation of externalizing behavior problems in school-aged children. J Child Psychol Psychiatry. 2019 Dec 3. doi: 10.1111/jcpp.13157. n = 1.096 ↥
Salehi M, Saeidi M, Kasulis N, Barias T, Kainth T, Gunturu S (2024): Tobacco Smoke Exposure in Children and Adolescents: Prevalence, Risk Factors and Co-Morbid Neuropsychiatric Conditions in a US Nationwide Study. Healthcare (Basel). 2024 Oct 22;12(21):2102. doi: 10.3390/healthcare12212102. PMID: 39517315; PMCID: PMC11545687. ↥
Müller, Candrian, Kropotov (2011): ADHS – Neurodiagnostik in der Praxis, Springer, Seite 88 ↥
http://de.statista.com/statistik/daten/studie/261015/umfrage/praevalenz-des-rauchens-in-deutschland-nach-geschlecht/ ↥
Lane M, Oyster E, Luo Y, Wang H (2025): The Effects of Air Pollution on Neurological Diseases: A Narrative Review on Causes and Mechanisms. Toxics. 2025 Mar 13;13(3):207. doi: 10.3390/toxics13030207. PMID: 40137534; PMCID: PMC11946816. REVIEW ↥ ↥ ↥ ↥ ↥ ↥ ↥ ↥ ↥ ↥ ↥ ↥
Fluegge K (2020): Do Toxic Synergies of Underlying Etiologies Predispose the Positive Association Between Traumatic Brain Injury and ADHD? J Atten Disord. 2020 Sep;24(11):1616-1619. doi: 10.1177/1087054716633858. PMID: 26957549. ↥ ↥
Zeng HX, Meng WJ, Zeng QG, Wei J, Liu LS, Wu QZ, Zhao B, Oudin A, Yang M, Jalava P, Dong GH, Zeng XW (2025): Long-term effects of PM2.5 constituents on childhood attention deficit hyperactivity disorder: evidence from a large population-based study in the Pearl River Delta Region, China. Environ Res. 2025 Jul 15;277:121641. doi: 10.1016/j.envres.2025.121641. PMID: 40250580. n = 110.818 ↥ ↥ ↥ ↥
Chen YC, Gui ZH, Bao WW, Liang JH, Zhang SX, Zhao Y, Jiang N, Chen YJ (2022): Chronic Exposure to Indoor Air Pollutants in Association with Attention-Deficit/Hyperactivity Disorder Symptoms in Chinese Schoolchildren: A Cross-Sectional Study. Neurotoxicology. 2022 Dec 9:S0161-813X(22)00196-6. doi: 10.1016/j.neuro.2022.12.003. PMID: 36509211. n = 8.692 ↥
Joseph N, Zhang-James Y, Perl A, Faraone SV (2015): Oxidative Stress and ADHD: A Meta-Analysis. J Atten Disord. 2015 Nov;19(11):915-24. doi: 10.1177/1087054713510354. PMID: 24232168; PMCID: PMC5293138. METASTUDY ↥ ↥
Si ML, Lee TJ (2002): Alpha7-nicotinic acetylcholine receptors on cerebral perivascular sympathetic nerves mediate choline-induced nitrergic neurogenic vasodilation. Circ Res. 2002 Jul 12;91(1):62-9. doi: 10.1161/01.res.0000024417.79275.23. PMID: 12114323. ↥
Chowdhary S, Vaile JC, Fletcher J, Ross HF, Coote JH, Townend JN (2000): Nitric oxide and cardiac autonomic control in humans. Hypertension. 2000 Aug;36(2):264-9. doi: 10.1161/01.hyp.36.2.264. PMID: 10948088. ↥
Kopatz V, Wen K, Kovács T, Keimowitz AS, Pichler V, Widder J, Vethaak AD, Hollóczki O, Kenner L (2023): Micro- and Nanoplastics Breach the Blood-Brain Barrier (BBB): Biomolecular Corona’s Role Revealed. Nanomaterials (Basel). 2023 Apr 19;13(8):1404. doi: 10.3390/nano13081404. PMID: 37110989; PMCID: PMC10141840. ↥
Deutscher Wetterdienst; Größenverteilung luftgetragener Partikel und ihre Entstehungsprozessenach Whitby und Cantrell, 1976 german ↥
Emecheta EE, Borda DB, Pfohl PM, Wohlleben W,Hutzler C, Haase A, Roloff A (2022): A comparative investigation of the sorption of polycyclic aromatic hydrocarbons to various polydisperse micro- and nanoplastics using a novel third-phase partition method. Micropl.&Nanopl. 2, 29 (2022). https://doi.org/10.1186/s43591-022-00049-9 ↥
Deutsches Bundesinstitut für Risikobewertung: Mikroplastik: Fakten, Forschung und offene Fragen german ↥ ↥ ↥ ↥
Emecheta EE, Pfohl PM, Wohlleben W, Haase A, Roloff A (2024): Desorption of Polycyclic Aromatic Hydrocarbons from Microplastics in Human Gastrointestinal Fluid Simulants-Implications for Exposure Assessment. ACS Omega. 2024 May 24;9(23):24281-24290. doi: 10.1021/acsomega.3c09380. PMID: 38882100; PMCID: PMC11170755. ↥
Prenner S, Allesch A, Staudner M, Rexeis M, Schwingshackl M, Huber-Humer M, Part F (2021): Static modelling of the material flows of micro- and nanoplastic particles caused by the use of vehicle tyres. Environ Pollut. 2021 Dec 1;290:118102. doi: 10.1016/j.envpol.2021.118102. PMID: 34523518. ↥
Vianello A, Jensen RL, Liu L, Vollertsen J (2019): Simulating human exposure to indoor airborne microplastics using a Breathing Thermal Manikin. Sci Rep. 2019 Jun 17;9(1):8670. doi: 10.1038/s41598-019-45054-w. PMID: 31209244; PMCID: PMC6573036. ↥
Ma YY, Li QY, Shi AY, Li JL, Wang YJ, Li X (2024): Association of air pollutants with psychiatric disorders: a two-sample Mendelian randomization. Ecotoxicol Environ Saf. 2024 Oct 15;285:117105. doi: 10.1016/j.ecoenv.2024.117105. PMID: 39332193. ↥
Markevych I, Tesch F, Datzmann T, Romanos M, Schmitt J, Heinrich J (2018): Outdoor air pollution, greenspace, and incidence of ADHD: A semi-individual study. Sci Total Environ. 2018 Nov 15;642:1362-1368. doi: 10.1016/j.scitotenv.2018.06.167. PMID: 30045516. n = 2.044 ↥ ↥
Siddique S, Banerjee M, Ray MR, Lahiri T (2011): Attention-deficit hyperactivity disorder in children chronically exposed to high level of vehicular pollution. Eur J Pediatr. 2011 Jul;170(7):923-9. doi: 10.1007/s00431-010-1379-0. PMID: 21191614. ↥
Min JY, Min KB (2017): Exposure to ambient PM10 and NO2 and the incidence of attention-deficit hyperactivity disorder in childhood. Environ Int. 2017 Feb;99:221-227. doi: 10.1016/j.envint.2016.11.022. PMID: 27939018. ↥ ↥
Shim JI, Byun G, Lee JT (2022): Exposure to Particulate Matter as a Potential Risk Factor for Attention-Deficit/Hyperactivity Disorder in Korean Children and Adolescents (KNHANES 2008-2018). Int J Environ Res Public Health. 2022 Oct 27;19(21):13966. doi: 10.3390/ijerph192113966. PMID: 36360844. n = 1.120 n = 1.120 ↥
Fan HC, Chen CM, Tsai JD, Chiang KL, Tsai SC, Huang CY, Lin CL, Hsu CY, Chang KH (2022): Association between Exposure to Particulate Matter Air Pollution during Early Childhood and Risk of Attention-Deficit/Hyperactivity Disorder in Taiwan. Int J Environ Res Public Health. 2022 Dec 2;19(23):16138. doi: 10.3390/ijerph192316138. PMID: 36498210. ↥
Oberdörster G, Sharp Z, Atudorei V, Elder A, Gelein R, Kreyling W, Cox C (2004): Translocation of inhaled ultrafine particles to the brain. Inhal Toxicol. 2004 Jun;16(6-7):437-45. doi: 10.1080/08958370490439597. PMID: 15204759. ↥ ↥
Li T, Yu Y, Sun Z, Duan J (2022): A comprehensive understanding of ambient particulate matter and its components on the adverse health effects based from epidemiological and laboratory evidence. Part Fibre Toxicol. 2022 Nov 29;19(1):67. doi: 10.1186/s12989-022-00507-5. PMID: 36447278; PMCID: PMC9707232. REVIEW ↥
Mazahir FA, Shukla A, Albastaki NA (2025): The association of particulate matter PM2.5 and nitrogen oxides from ambient air pollution and mental health of children and young adults- a systematic review. Rev Environ Health. 2025 Mar 14. doi: 10.1515/reveh-2024-0120. PMID: 40074563. REVIEW ↥ ↥
Ahmad S, K G N, Mani Babu A, Ranjan R, Kumar P (2024): Association Between Ambient Air Pollution and Attention-Deficit/Hyperactivity Disorder (ADHD) in Children: A Systematic Review and Meta-Analysis. Cureus. 2024 Oct 15;16(10):e71527. doi: 10.7759/cureus.71527. PMID: 39544605; PMCID: PMC11562299. METASTUDY ↥ ↥
Zhou J, Lu Z, Xu K, Zhao G, Zhu Y, Yuan R, Sun Y, Zhang Y, Yue W (2025): Air pollution is the risk factor for psychiatric disorders: a two-step Mendelian randomization study. J Affect Disord. 2025 Nov 1;388:119475. doi: 10.1016/j.jad.2025.119475. PMID: 40436208. n = 423.796 ↥
Thygesen, Holst, Hansen, Geels, Kalkbrenner, Schendel, Brandt, Pedersen, Dalsgaard (2019): Exposure to air pollution in early childhood and the association with Attention-Deficit Hyperactivity Disorder. Environ Res. 2019 Nov 22:108930. doi: 10.1016/j.envres.2019.108930. n = 809.654 ↥ ↥ ↥ ↥
Chang YC, Chen WT, Su SH, Jung CR, Hwang BF (2022): PM2.5 exposure and incident attention-deficit/hyperactivity disorder during the prenatal and postnatal periods: A birth cohort study. Environ Res. 2022 Jun 28:113769. doi: 10.1016/j.envres.2022.113769. PMID: 35777438. n = 425.736 ↥
Donzelli, Llopis-Gonzalez, Llopis-Morales, Cioni, Morales-Suárez-Varela (2019): Particulate Matter Exposure and Attention-Deficit/Hyperactivity Disorder in Children: A Systematic Review of Epidemiological Studies. Int J Environ Res Public Health. 2019 Dec 20;17(1). pii: E67. doi: 10.3390/ijerph17010067. n = 181.144 REVIEW ↥
Li Y, Xie T, Cardoso Melo RD, de Vries M, Lakerveld J, Zijlema W, Hartman CA (2023): Longitudinal effects of environmental noise and air pollution exposure on autism spectrum disorder and attention-deficit/hyperactivity disorder during adolescence and early adulthood: The TRAILS study. Environ Res. 2023 Mar 20;227:115704. doi: 10.1016/j.envres.2023.115704. PMID: 36940817. n = 2.750 ↥
Carll, Salatini, Pirela, Wang, Xie, Lorkiewicz, Naeem, Qian, Castranova, Godleski, Demokritou (2020): Inhalation of printer-emitted particles impairs cardiac conduction, hemodynamics, and autonomic regulation and induces arrhythmia and electrical remodeling in rats. Part Fibre Toxicol. 2020 Jan 29;17(1):7. doi: 10.1186/s12989-019-0335-z. PMID: 31996220; PMCID: PMC6990551. ↥
Zhang Y, Wang W, Zhang X, Jing R, Wen X, Xiao P, Liu X, Zhao Z, Chang T, Li Y, Liu W, Sun C, Yang X, Yang L, Lu M (2025): Neurotrophin-3 as a mediator in the link between PM2.5 exposure and psychiatric disorders: A Mendelian randomization study. Ecotoxicol Environ Saf. 2025 Jan 1;289:117658. doi: 10.1016/j.ecoenv.2024.117658. PMID: 39765118. ↥ ↥
Vignon AN, Dudon G, Oliva G, Thirard S, Alenda UG, Brugoux A, Cazevieille C, Imbert J, Bellières C, Lehmann S, Crozet C, Torrent J, Bertaso F, Le Merrer J, Becker JAJ, Perrier V (2025): Lifelong exposure to polystyrene-nanoplastics induces an attention-deficit hyperactivity disorder-like phenotype and impairs brain aging in mice. J Hazard Mater. 2025 Aug 15;494:138640. doi: 10.1016/j.jhazmat.2025.138640. PMID: 40403375. ↥
Mayati A, Podechard N, Rineau M, Sparfel L, Lagadic-Gossmann D, Fardel O, Le Ferrec E (2017): Benzo(a)pyrene triggers desensitization of β2-adrenergic pathway. Sci Rep. 2017 Jun 12;7(1):3262. doi: 10.1038/s41598-017-03646-4. PMID: 28607424; PMCID: PMC5468268. ↥
Chepelev NL, Moffat ID, Bowers WJ, Yauk CL (2015): Neurotoxicity may be an overlooked consequence of benzo[a]pyrene exposure that is relevant to human health risk assessment. Mutat Res Rev Mutat Res. 2015 Apr-Jun;764:64-89. doi: 10.1016/j.mrrev.2015.03.001. PMID: 26041267. REVIEW ↥
Mayati A, Levoin N, Paris H, N’Diaye M, Courtois A, Uriac P, Lagadic-Gossmann D, Fardel O, Le Ferrec E (2012): Induction of intracellular calcium concentration by environmental benzo(a)pyrene involves a β2-adrenergic receptor/adenylyl cyclase/Epac-1/inositol 1,4,5-trisphosphate pathway in endothelial cells. J Biol Chem. 2012 Feb 3;287(6):4041-52. doi: 10.1074/jbc.M111.319970. PMID: 22167199; PMCID: PMC3281724. ↥
Lin CH, Nicol CJB, Wan C, Chen SJ, Huang RN, Chiang MC (2022): Exposure to PM2.5 induces neurotoxicity, mitochondrial dysfunction, oxidative stress and inflammation in human SH-SY5Y neuronal cells. Neurotoxicology. 2022 Jan;88:25-35. doi: 10.1016/j.neuro.2021.10.009. PMID: 34718062. ↥
Liu J, Liu B, Yuan P, Cheng L, Sun H, Gui J, Pan Y, Huang D, Chen H, Jiang L (2021): Role of PKA/CREB/BDNF signaling in PM2.5-induced neurodevelopmental damage to the hippocampal neurons of rats. Ecotoxicol Environ Saf. 2021 May;214:112005. doi: 10.1016/j.ecoenv.2021.112005. PMID: 33640725. ↥ ↥
Yokota S, Mizuo K, Moriya N, Oshio S, Sugawara I, Takeda K (2009): Effect of prenatal exposure to diesel exhaust on dopaminergic system in mice. Neurosci Lett. 2009 Jan 2;449(1):38-41. doi: 10.1016/j.neulet.2008.09.085. PMID: 18938223. ↥
Yokota S, Moriya N, Iwata M, Umezawa M, Oshio S, Takeda K (2013): Exposure to diesel exhaust during fetal period affects behavior and neurotransmitters in male offspring mice. J Toxicol Sci. 2013 Feb;38(1):13-23. doi: 10.2131/jts.38.13. PMID: 23358136. ↥
Suzuki T, Oshio S, Iwata M, Saburi H, Odagiri T, Udagawa T, Sugawara I, Umezawa M, Takeda K (2010): In utero exposure to a low concentration of diesel exhaust affects spontaneous locomotor activity and monoaminergic system in male mice. Part Fibre Toxicol. 2010 Mar 23;7:7. doi: 10.1186/1743-8977-7-7. PMID: 20331848; PMCID: PMC2853486. ↥ ↥ ↥
Bernal-Meléndez E, Callebert J, Bouillaud P, Persuy MA, Olivier B, Badonnel K, Chavatte-Palmer P, Baly C, Schroeder H (2021): Dopaminergic and serotonergic changes in rabbit fetal brain upon repeated gestational exposure to diesel engine exhaust. Arch Toxicol. 2021 Sep;95(9):3085-3099. doi: 10.1007/s00204-021-03110-3. PMID: 34189592. ↥ ↥ ↥
Zhu N, Li H, Han M, Guo L, Chen L, Yun Y, Guo Z, Li G, Sang N (2012): Environmental nitrogen dioxide (NO2) exposure influences development and progression of ischemic stroke. Toxicol Lett. 2012 Oct 17;214(2):120-30. doi: 10.1016/j.toxlet.2012.08.021. PMID: 22960396. ↥
Yuchi, Brauer, Czekajlo, Davies, Davis, Guhn, Jarvis, Jerrett, Nesbitt, Oberlander, Sbihi, Su, van den Bosch (2022): Neighborhood environmental exposures and incidence of attention deficit/hyperactivity disorder: A population-based cohort study. Environ Int. 2022 Feb 7;161:107120. doi: 10.1016/j.envint.2022.107120. PMID: 35144157. ↥
Aghaei, Janjani, Yousefian, Jamal, Yunesian (2019): Association between ambient gaseous and particulate air pollutants and attention deficit hyperactivity disorder (ADHD) in children; a systematic review. Environ Res. 2019 Jun;173:135-156. doi: 10.1016/j.envres.2019.03.030. REVIEW ↥
Roberts S, Arseneault L, Barratt B, Beevers S, Danese A, Odgers CL, Moffitt TE, Reuben A, Kelly FJ, Fisher HL (2019): Exploration of NO2 and PM2.5 air pollution and mental health problems using high-resolution data in London-based children from a UK longitudinal cohort study. Psychiatry Res. 2019 Feb;272:8-17. doi: 10.1016/j.psychres.2018.12.050. PMID: 30576995; PMCID: PMC6401205. n = 284 ↥
Sram RJ, Veleminsky M Jr, Veleminsky M Sr, Stejskalová J (2017): The impact of air pollution to central nervous system in children and adults. Neuro Endocrinol Lett. 2017 Dec;38(6):389-396. PMID: 29298278. REVIEW ↥
Sunyer J, Esnaola M, Alvarez-Pedrerol M, Forns J, Rivas I, López-Vicente M, Suades-González E, Foraster M, Garcia-Esteban R, Basagaña X, Viana M, Cirach M, Moreno T, Alastuey A, Sebastian-Galles N, Nieuwenhuijsen M, Querol X (2015): Association between traffic-related air pollution in schools and cognitive development in primary school children: a prospective cohort study. PLoS Med. 2015 Mar 3;12(3):e1001792. doi: 10.1371/journal.pmed.1001792. PMID: 25734425; PMCID: PMC4348510. n = 2.715 ↥
Forns J, Dadvand P, Foraster M, Alvarez-Pedrerol M, Rivas I, López-Vicente M, Suades-Gonzalez E, Garcia-Esteban R, Esnaola M, Cirach M, Grellier J, Basagaña X, Querol X, Guxens M, Nieuwenhuijsen MJ, Sunyer J (2016): Traffic-Related Air Pollution, Noise at School, and Behavioral Problems in Barcelona Schoolchildren: A Cross-Sectional Study. Environ Health Perspect. 2016 Apr;124(4):529-35. doi: 10.1289/ehp.1409449. PMID: 26241036; PMCID: PMC4829987. ↥
Mota-Bertran A, Coenders G, Plaja P, Saez M, Barceló MA (2024): Air pollution and children’s mental health in rural areas: compositional spatio-temporal model. Sci Rep. 2024 Aug 21;14(1):19363. doi: 10.1038/s41598-024-70024-2. PMID: 39169039; PMCID: PMC11339296. ↥
Forns J, Dadvand P, Esnaola M, Alvarez-Pedrerol M, López-Vicente M, Garcia-Esteban R, Cirach M, Basagaña X, Guxens M, Sunyer J (2017): Longitudinal association between air pollution exposure at school and cognitive development in school children over a period of 3.5 years. Environ Res. 2017 Nov;159:416-421. doi: 10.1016/j.envres.2017.08.031. PMID: 28858754. ↥
Alemany S, Vilor-Tejedor N, García-Esteban R, Bustamante M, Dadvand P, Esnaola M, Mortamais M, Forns J, van Drooge BL, Álvarez-Pedrerol M, Grimalt JO, Rivas I, Querol X, Pujol J, Sunyer J. Traffic-Related Air Pollution, APOEε4 Status, and Neurodevelopmental Outcomes among School Children Enrolled in the BREATHE Project (Catalonia, Spain). Environ Health Perspect. 2018 Aug 2;126(8):087001. doi: 10.1289/EHP2246. PMID: 30073950; PMCID: PMC6108838. ↥
Compa M, Baumbach C, Kaczmarek-Majer K, Buczyłowska D, Gradys GO, Skotak K, Degórska A, Bratkowski J, Wierzba-Łukaszyk M, Mysak Y, Sitnik-Warchulska K, Lipowska M, Izydorczyk B, Grellier J, Asanowicz D, Markevych I, Szwed M (2023): Air pollution and attention in Polish schoolchildren with and without ADHD. Sci Total Environ. 2023 Jun 9;892:164759. doi: 10.1016/j.scitotenv.2023.164759. PMID: 37302611. ↥
Park J, Sohn JH, Cho SJ, Seo HY, Hwang IU, Hong YC, Kim KN (2020): Association between short-term air pollution exposure and attention-deficit/hyperactivity disorder-related hospital admissions among adolescents: A nationwide time-series study. Environ Pollut. 2020 Nov;266(Pt 1):115369. doi: 10.1016/j.envpol.2020.115369. PMID: 32810816. ↥
Fluegge (2016): Does environmental exposure to the greenhouse gas, N2O, contribute to etiological factors in neurodevelopmental disorders? A mini-review of the evidence. Environ Toxicol Pharmacol. 2016 Oct;47:6-18. doi: 10.1016/j.etap.2016.08.013. PMID: 27566494. REVIEW ↥
Umweltbundesamt, abgerufen 30.11.23 ↥
Yan W, Ji X, Shi J, Li G, Sang N (2015): Acute nitrogen dioxide inhalation induces mitochondrial dysfunction in rat brain. Environ Res. 2015 Apr;138:416-24. doi: 10.1016/j.envres.2015.02.022. PMID: 25791864. ↥
Suzuki T, Ueta K, Sugimoto M, Uchida I, Mashimo T (2003): Nitrous oxide and xenon inhibit the human (alpha 7)5 nicotinic acetylcholine receptor expressed in Xenopus oocyte. Anesth Analg. 2003 Feb;96(2):443-8, table of contents. doi: 10.1097/00000539-200302000-00028. PMID: 12538194. ↥
Yang Y, Paspalas CD, Jin LE, Picciotto MR, Arnsten AF, Wang M (2013): Nicotinic α7 receptors enhance NMDA cognitive circuits in dorsolateral prefrontal cortex. Proc Natl Acad Sci U S A. 2013 Jul 16;110(29):12078-83. doi: 10.1073/pnas.1307849110. PMID: 23818597; PMCID: PMC3718126. ↥
Zhou P, Zhang W, Xu YJ, Liu RQ, Qian Z, McMillin SE, Bingheim E, Lin LZ, Zeng XW, Yang BY, Hu LW, Chen W, Chen G, Yu Y, Dong GH (2022): Association between long-term ambient ozone exposure and attention-deficit/hyperactivity disorder symptoms among Chinese children. Environ Res. 2022 Oct 18;216(Pt 2):114602. doi: 10.1016/j.envres.2022.114602. PMID: 36265606. n = 35.103 ↥
Fu P, Yung KKL (2020): Air Pollution and Alzheimer’s Disease: A Systematic Review and Meta-Analysis. J Alzheimers Dis. 2020;77(2):701-714. doi: 10.3233/JAD-200483. PMID: 32741830. METASTUDY ↥
Liu RM, Chong Z, Chen JC (2020): Ozone and Particulate Matter Exposure and Alzheimer’s Disease: A Review of Human and Animal Studies. J Alzheimers Dis. 2020;76(3):807-824. doi: 10.3233/JAD-200435. PMID: 32568209. REVIEW ↥
Gao Q, Zang E, Bi J, Dubrow R, Lowe SR, Chen H, Zeng Y, Shi L, Chen K (2022): Long-term ozone exposure and cognitive impairment among Chinese older adults: A cohort study. Environ Int. 2022 Feb;160:107072. doi: 10.1016/j.envint.2021.107072. PMID: 34979350; PMCID: PMC8821373. ↥
Zhang Y, Qiu S, Guo VY, Chen W, Han X, Yang W (2025): Association of only-child status and household pet ownership with attention-deficit/hyperactivity disorder among Chinese preschool children: a population-based study. Front Public Health. 2025 Feb 11;12:1450216. doi: 10.3389/fpubh.2024.1450216. PMID: 40012582; PMCID: PMC11862913. n = 63.282 ↥
Dórea (2019): Environmental exposure to low-level lead (Pb) co-occurring with other neurotoxicants in early life and neurodevelopment of children. Environ Res. 2019 Oct;177:108641. doi: 10.1016/j.envres.2019.108641. ↥